1
|
Charlick JN, Bozadzhieva D, Butler AS, Wilkinson KA, Marrion NV. A single coiled-coil domain mutation in hIKCa channel subunits disrupts preferential formation of heteromeric hSK1:hIKCa channels. Eur J Neurosci 2024; 59:3-16. [PMID: 38018635 PMCID: PMC10952195 DOI: 10.1111/ejn.16189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 09/22/2023] [Accepted: 10/25/2023] [Indexed: 11/30/2023]
Abstract
The expression of IKCa (SK4) channel subunits overlaps with that of SK channel subunits, and it has been proposed that the two related subunits prefer to co-assemble to form heteromeric hSK1:hIKCa channels. This implicates hSK1:hIKCa heteromers in physiological roles that might have been attributed to activation of SK channels. We have used a mutation approach to confirm formation of heterometric hSK1:hIKCa channels. Introduction of residues within hSK1 that were predicted to impart sensitivity to the hIKCa current blocker TRAM-34 changed the pharmacology of functional heteromers. Heteromeric channels formed between wildtype hIKCa and mutant hSK1 subunits displayed a significantly higher sensitivity and maximum block to addition of TRAM-34 than heteromers formed between wildtype subunits. Heteromer formation was disrupted by a single point mutation within one COOH-terminal coiled-coil domain of the hIKCa channel subunit. This mutation only disrupted the formation of hSK1:hIKCa heteromeric channels, without affecting the formation of homomeric hIKCa channels. Finally, the Ca2+ gating sensitivity of heteromeric hSK1:hIKCa channels was found to be significantly lower than the Ca2+ gating sensitivity of homomeric hIKCa channels. These data confirmed the preferred formation of heteromeric channels that results from COOH-terminal interactions between subunits. The distinct sensitivity of the heteromer to activation by Ca2+ suggests that heteromeric channels fulfil a distinct function within those neurons that express both subunits.
Collapse
Affiliation(s)
- James N. Charlick
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Daniella Bozadzhieva
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Andrew S. Butler
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Kevin A. Wilkinson
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Neil V. Marrion
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| |
Collapse
|
2
|
Moore SJ, Cazares VA, Temme SJ, Murphy GG. Age-related deficits in neuronal physiology and cognitive function are recapitulated in young mice overexpressing the L-type calcium channel, Ca V 1.3. Aging Cell 2023; 22:e13781. [PMID: 36703244 PMCID: PMC10014069 DOI: 10.1111/acel.13781] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/21/2022] [Accepted: 12/30/2022] [Indexed: 01/28/2023] Open
Abstract
The calcium dysregulation hypothesis of brain aging posits that an age-related increase in neuronal calcium concentration is responsible for alterations in a variety of cellular processes that ultimately result in learning and memory deficits in aged individuals. We previously generated a novel transgenic mouse line, in which expression of the L-type voltage-gated calcium, CaV 1.3, is increased by ~50% over wild-type littermates. Here, we show that, in young mice, this increase is sufficient to drive changes in neuronal physiology and cognitive function similar to those observed in aged animals. Specifically, there is an increase in the magnitude of the postburst afterhyperpolarization, a deficit in spatial learning and memory (assessed by the Morris water maze), a deficit in recognition memory (assessed in novel object recognition), and an overgeneralization of fear to novel contexts (assessed by contextual fear conditioning). While overexpression of CaV 1.3 recapitulated these key aspects of brain aging, it did not produce alterations in action potential firing rates, basal synaptic communication, or spine number/density. Taken together, these results suggest that increased expression of CaV 1.3 in the aged brain is a crucial factor that acts in concert with age-related changes in other processes to produce the full complement of structural, functional, and behavioral outcomes that are characteristic of aged animals.
Collapse
Affiliation(s)
- Shannon J. Moore
- Michigan Neuroscience InstituteAnn ArborMichiganUSA
- Molecular & Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Victor A. Cazares
- Molecular & Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
- Department of PsychologyWilliams CollegeWilliamstownMassachusettsUSA
| | | | - Geoffrey G. Murphy
- Michigan Neuroscience InstituteAnn ArborMichiganUSA
- Molecular & Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
3
|
Maziar A, Critch TNRHY, Ghosh S, Rajani V, Flynn CM, Qin T, Reinhardt C, Man KNM, Lee A, Hell JW, Yuan Q. Aging differentially affects LTCC function in hippocampal CA1 and piriform cortex pyramidal neurons. Cereb Cortex 2023; 33:1489-1503. [PMID: 35437602 PMCID: PMC9930631 DOI: 10.1093/cercor/bhac152] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/25/2022] [Accepted: 03/27/2022] [Indexed: 11/14/2022] Open
Abstract
Aging is associated with cognitive decline and memory loss in humans. In rats, aging-associated neuronal excitability changes and impairments in learning have been extensively studied in the hippocampus. Here, we investigated the roles of L-type calcium channels (LTCCs) in the rat piriform cortex (PC), in comparison with those of the hippocampus. We employed spatial and olfactory tasks that involve the hippocampus and PC. LTCC blocker nimodipine administration impaired spontaneous location recognition in adult rats (6-9 months). However, the same blocker rescued the spatial learning deficiency in aged rats (19-23 months). In an odor-associative learning task, infusions of nimodipine into either the PC or dorsal CA1 impaired the ability of adult rats to learn a positive odor association. Again, in contrast, nimodipine rescued odor associative learning in aged rats. Aged CA1 neurons had higher somatic expression of LTCC Cav1.2 subunits, exhibited larger afterhyperpolarization (AHP) and lower excitability compared with adult neurons. In contrast, PC neurons from aged rats showed higher excitability and no difference in AHP. Cav1.2 expression was similar in adult and aged PC somata, but relatively higher in PSD95- puncta in aged dendrites. Our data suggest unique features of aging-associated changes in LTCCs in the PC and hippocampus.
Collapse
Affiliation(s)
- Aida Maziar
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| | - Tristian N R H Y Critch
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| | - Sourav Ghosh
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| | - Vishaal Rajani
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| | - Cassandra M Flynn
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| | - Tian Qin
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| | - Camila Reinhardt
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| | - Kwun Nok Mimi Man
- Department of Pharmacology, School of Medicine, University of California-Davis, Sacramento, CA 95817, United States
| | - Amy Lee
- Department of Neuroscience, University of Texas-Austin, Austin, TX 78712, United States
| | - Johannes W Hell
- Department of Pharmacology, School of Medicine, University of California-Davis, Sacramento, CA 95817, United States
| | - Qi Yuan
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL A1B 3V6, Canada
| |
Collapse
|
4
|
Scott DN, Frank MJ. Adaptive control of synaptic plasticity integrates micro- and macroscopic network function. Neuropsychopharmacology 2023; 48:121-144. [PMID: 36038780 PMCID: PMC9700774 DOI: 10.1038/s41386-022-01374-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 11/09/2022]
Abstract
Synaptic plasticity configures interactions between neurons and is therefore likely to be a primary driver of behavioral learning and development. How this microscopic-macroscopic interaction occurs is poorly understood, as researchers frequently examine models within particular ranges of abstraction and scale. Computational neuroscience and machine learning models offer theoretically powerful analyses of plasticity in neural networks, but results are often siloed and only coarsely linked to biology. In this review, we examine connections between these areas, asking how network computations change as a function of diverse features of plasticity and vice versa. We review how plasticity can be controlled at synapses by calcium dynamics and neuromodulatory signals, the manifestation of these changes in networks, and their impacts in specialized circuits. We conclude that metaplasticity-defined broadly as the adaptive control of plasticity-forges connections across scales by governing what groups of synapses can and can't learn about, when, and to what ends. The metaplasticity we discuss acts by co-opting Hebbian mechanisms, shifting network properties, and routing activity within and across brain systems. Asking how these operations can go awry should also be useful for understanding pathology, which we address in the context of autism, schizophrenia and Parkinson's disease.
Collapse
Affiliation(s)
- Daniel N Scott
- Cognitive Linguistic, and Psychological Sciences, Brown University, Providence, RI, USA.
- Carney Institute for Brain Science, Brown University, Providence, RI, USA.
| | - Michael J Frank
- Cognitive Linguistic, and Psychological Sciences, Brown University, Providence, RI, USA.
- Carney Institute for Brain Science, Brown University, Providence, RI, USA.
| |
Collapse
|
5
|
Modulation of L-type calcium channels in Alzheimer's disease: A potential therapeutic target. Comput Struct Biotechnol J 2022; 21:11-20. [PMID: 36514335 PMCID: PMC9719069 DOI: 10.1016/j.csbj.2022.11.049] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 11/28/2022] Open
Abstract
Calcium plays a fundamental role in various signaling pathways and cellular processes in the human organism. In the nervous system, voltage-gated calcium channels such as L-type calcium channels (LTCCs) are critical elements in mediating neurotransmitter release, synaptic integration and plasticity. Dysfunction of LTCCs has been implicated in both aging and Alzheimer's Disease (AD), constituting a key component of calcium hypothesis of AD. As such, LTCCs are a promising drug target in AD. However, due to their structural and functional complexity, the mechanisms by which LTCCs contribute to AD are still unclear. In this review, we briefly summarize the structure, function, and modulation of LTCCs that are the backbone for understanding pathological processes involving LTCCs. We suggest targeting molecular pathways up-regulating LTCCs in AD may be a more promising approach, given the diverse physiological functions of LTCCs and the ineffectiveness of LTCC blockers in clinical studies.
Collapse
Key Words
- AC, adenylyl cyclase
- AD, Alzheimer’s Disease
- AHP, afterhyperpolarization
- AR, adrenoceptor
- Aging
- Alzheimer’s disease
- Aβ, β-amyloid
- BIN1, bridging integrator 1
- BTZs, benzothiazepines
- CDF, calcium-dependent facilitation
- CDI, calcium-dependent inactivation
- CaMKII, calmodulin-dependent protein kinase II
- DHP, dihydropyridine
- L-type calcium channel
- LTCC, L-type calcium channels
- LTD, long-term depression
- LTP, long-term potentiation
- NFT, neurofibrillary tangles
- NMDAR, N-methyl-D-aspartate receptor
- PAA, phenylalkylamines
- PKA, protein kinase A
- PKC, protein kinase C
- PKG, protein kinase G
- SFK, Src family kinase
- Tau
- VSD, voltage sensing domain
- β-Amyloid
Collapse
|
6
|
Tau isoform-specific enhancement of L-type calcium current and augmentation of afterhyperpolarization in rat hippocampal neurons. Sci Rep 2022; 12:15231. [PMID: 36075936 PMCID: PMC9458744 DOI: 10.1038/s41598-022-18648-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/17/2022] [Indexed: 11/08/2022] Open
Abstract
Accumulation of tau is observed in dementia, with human tau displaying 6 isoforms grouped by whether they display either 3 or 4 C-terminal repeat domains (3R or 4R) and exhibit no (0N), one (1N) or two (2N) N terminal repeats. Overexpression of 4R0N-tau in rat hippocampal slices enhanced the L-type calcium (Ca2+) current-dependent components of the medium and slow afterhyperpolarizations (AHPs). Overexpression of both 4R0N-tau and 4R2N-tau augmented CaV1.2-mediated L-type currents when expressed in tsA-201 cells, an effect not observed with the third 4R isoform, 4R1N-tau. Current enhancement was only observed when the pore-forming subunit was co-expressed with CaVβ3 and not CaVβ2a subunits. Non-stationary noise analysis indicated that enhanced Ca2+ channel current arose from a larger number of functional channels. 4R0N-tau and CaVβ3 were found to be physically associated by co-immunoprecipitation. In contrast, the 4R1N-tau isoform that did not augment expressed macroscopic L-type Ca2+ current exhibited greatly reduced binding to CaVβ3. These data suggest that physical association between tau and the CaVβ3 subunit stabilises functional L-type channels in the membrane, increasing channel number and Ca2+ influx. Enhancing the Ca2+-dependent component of AHPs would produce cognitive impairment that underlie those seen in the early phases of tauopathies.
Collapse
|
7
|
Sahu G, Turner RW. The Molecular Basis for the Calcium-Dependent Slow Afterhyperpolarization in CA1 Hippocampal Pyramidal Neurons. Front Physiol 2022; 12:759707. [PMID: 35002757 PMCID: PMC8730529 DOI: 10.3389/fphys.2021.759707] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/01/2021] [Indexed: 12/02/2022] Open
Abstract
Neuronal signal transmission depends on the frequency, pattern, and timing of spike output, each of which are shaped by spike afterhyperpolarizations (AHPs). There are classically three post-spike AHPs of increasing duration categorized as fast, medium and slow AHPs that hyperpolarize a cell over a range of 10 ms to 30 s. Intensive early work on CA1 hippocampal pyramidal cells revealed that all three AHPs incorporate activation of calcium-gated potassium channels. The ionic basis for a fAHP was rapidly attributed to the actions of big conductance (BK) and the mAHP to small conductance (SK) or Kv7 potassium channels. In stark contrast, the ionic basis for a prominent slow AHP of up to 30 s duration remained an enigma for over 30 years. Recent advances in pharmacological, molecular, and imaging tools have uncovered the expression of a calcium-gated intermediate conductance potassium channel (IK, KCa3.1) in central neurons that proves to contribute to the slow AHP in CA1 hippocampal pyramidal cells. Together the data show that the sAHP arises in part from a core tripartite complex between Cav1.3 (L-type) calcium channels, ryanodine receptors, and IK channels at endoplasmic reticulum-plasma membrane junctions. Work on the sAHP in CA1 pyramidal neurons has again quickened pace, with identified contributions by both IK channels and the Na-K pump providing answers to several mysteries in the pharmacological properties of the sAHP.
Collapse
Affiliation(s)
- Giriraj Sahu
- National Institute of Pharmaceutical Education and Research Ahmedabad, Ahmedabad, India
| | - Ray W Turner
- Department Cell Biology & Anatomy, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
8
|
Calcium-induced calcium release and type 3 ryanodine receptors modulate the slow afterhyperpolarising current, sIAHP, and its potentiation in hippocampal pyramidal neurons. PLoS One 2020; 15:e0230465. [PMID: 32559219 PMCID: PMC7304577 DOI: 10.1371/journal.pone.0230465] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/03/2020] [Indexed: 12/21/2022] Open
Abstract
The slow afterhyperpolarising current, sIAHP, is a Ca2+-dependent current that plays an important role in the late phase of spike frequency adaptation. sIAHP is activated by voltage-gated Ca2+ channels, while the contribution of calcium from ryanodine-sensitive intracellular stores, released by calcium-induced calcium release (CICR), is controversial in hippocampal pyramidal neurons. Three types of ryanodine receptors (RyR1-3) are expressed in the hippocampus, with RyR3 showing a predominant expression in CA1 neurons. We investigated the specific role of CICR, and particularly of its RyR3-mediated component, in the regulation of the sIAHP amplitude and time course, and the activity-dependent potentiation of the sIAHP in rat and mouse CA1 pyramidal neurons. Here we report that enhancement of CICR by caffeine led to an increase in sIAHP amplitude, while inhibition of CICR by ryanodine caused a small, but significant reduction of sIAHP. Inhibition of ryanodine-sensitive Ca2+ stores by ryanodine or depletion by the SERCA pump inhibitor cyclopiazonic acid caused a substantial attenuation in the sIAHP activity-dependent potentiation in both rat and mouse CA1 pyramidal neurons. Neurons from mice lacking RyR3 receptors exhibited a sIAHP with features undistinguishable from wild-type neurons, which was similarly reduced by ryanodine. However, the lack of RyR3 receptors led to a faster and reduced activity-dependent potentiation of sIAHP. We conclude that ryanodine receptor-mediated CICR contributes both to the amplitude of the sIAHP at steady state and its activity-dependent potentiation in rat and mouse hippocampal pyramidal neurons. In particular, we show that RyR3 receptors play an essential and specific role in shaping the activity-dependent potentiation of the sIAHP. The modulation of activity-dependent potentiation of sIAHP by RyR3-mediated CICR contributes to plasticity of intrinsic neuronal excitability and is likely to play a critical role in higher cognitive functions, such as learning and memory.
Collapse
|
9
|
Moore SJ, Murphy GG. The role of L-type calcium channels in neuronal excitability and aging. Neurobiol Learn Mem 2020; 173:107230. [PMID: 32407963 DOI: 10.1016/j.nlm.2020.107230] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 03/09/2020] [Accepted: 04/12/2020] [Indexed: 12/11/2022]
Abstract
Over the last two decades there has been significant progress towards understanding the neural substrates that underlie age-related cognitive decline. Although many of the exact molecular and cellular mechanisms have yet to be fully understood, there is consensus that alterations in neuronal calcium homeostasis contribute to age-related deficits in learning and memory. Furthermore, it is thought that the age-related changes in calcium homeostasis are driven, at least in part, by changes in calcium channel expression. In this review, we focus on the role of a specific class of calcium channels: L-type voltage-gated calcium channels (LVGCCs). We provide the reader with a general introduction to voltage-gated calcium channels, followed by a more detailed description of LVGCCs and how they serve to regulate neuronal excitability via the post burst afterhyperpolarization (AHP). We conclude by reviewing studies that link the slow component of the AHP to learning and memory, and discuss how age-related increases in LVGCC expression may underlie cognitive decline by mediating a decrease in neuronal excitability.
Collapse
Affiliation(s)
- Shannon J Moore
- Michigan Neuroscience Institute, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109, United States; Department of Molecular and Integrative Physiology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109, United States
| | - Geoffrey G Murphy
- Michigan Neuroscience Institute, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109, United States; Department of Molecular and Integrative Physiology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109, United States.
| |
Collapse
|
10
|
Calcium Signaling in Neurons and Glial Cells: Role of Cav1 channels. Neuroscience 2019; 421:95-111. [DOI: 10.1016/j.neuroscience.2019.09.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 11/18/2022]
|
11
|
Plumbly W, Brandon N, Deeb TZ, Hall J, Harwood AJ. L-type voltage-gated calcium channel regulation of in vitro human cortical neuronal networks. Sci Rep 2019; 9:13810. [PMID: 31554851 PMCID: PMC6761148 DOI: 10.1038/s41598-019-50226-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 09/02/2019] [Indexed: 11/09/2022] Open
Abstract
The combination of in vitro multi-electrode arrays (MEAs) and the neuronal differentiation of stem cells offers the capability to study human neuronal networks from patient or engineered human cell lines. Here, we use MEA-based assays to probe synaptic function and network interactions of hiPSC-derived neurons. Neuronal network behaviour first emerges at approximately 30 days of culture and is driven by glutamate neurotransmission. Over a further 30 days, inhibitory GABAergic signalling shapes network behaviour into a synchronous regular pattern of burst firing activity and low activity periods. Gene mutations in L-type voltage gated calcium channel subunit genes are strongly implicated as genetic risk factors for the development of schizophrenia and bipolar disorder. We find that, although basal neuronal firing rate is unaffected, there is a dose-dependent effect of L-type voltage gated calcium channel inhibitors on synchronous firing patterns of our hiPSC-derived neural networks. This demonstrates that MEA assays have sufficient sensitivity to detect changes in patterns of neuronal interaction that may arise from hypo-function of psychiatric risk genes. Our study highlights the utility of in vitro MEA based platforms for the study of hiPSC neural network activity and their potential use in novel compound screening.
Collapse
Affiliation(s)
- William Plumbly
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Nick Brandon
- Neuroscience, IMED Biotech Unit, AstraZeneca, 35 Gatehouse Dr, Waltham, MA, 02451, USA
| | - Tarek Z Deeb
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Adrian J Harwood
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK.
| |
Collapse
|
12
|
Junctophilin Proteins Tether a Cav1-RyR2-KCa3.1 Tripartite Complex to Regulate Neuronal Excitability. Cell Rep 2019; 28:2427-2442.e6. [DOI: 10.1016/j.celrep.2019.07.075] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/20/2019] [Accepted: 07/19/2019] [Indexed: 12/12/2022] Open
|
13
|
Kshatri AS, Gonzalez-Hernandez A, Giraldez T. Physiological Roles and Therapeutic Potential of Ca 2+ Activated Potassium Channels in the Nervous System. Front Mol Neurosci 2018; 11:258. [PMID: 30104956 PMCID: PMC6077210 DOI: 10.3389/fnmol.2018.00258] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/06/2018] [Indexed: 12/21/2022] Open
Abstract
Within the potassium ion channel family, calcium activated potassium (KCa) channels are unique in their ability to couple intracellular Ca2+ signals to membrane potential variations. KCa channels are diversely distributed throughout the central nervous system and play fundamental roles ranging from regulating neuronal excitability to controlling neurotransmitter release. The physiological versatility of KCa channels is enhanced by alternative splicing and co-assembly with auxiliary subunits, leading to fundamental differences in distribution, subunit composition and pharmacological profiles. Thus, understanding specific KCa channels’ mechanisms in neuronal function is challenging. Based on their single channel conductance, KCa channels are divided into three subtypes: small (SK, 4–14 pS), intermediate (IK, 32–39 pS) and big potassium (BK, 200–300 pS) channels. This review describes the biophysical characteristics of these KCa channels, as well as their physiological roles and pathological implications. In addition, we also discuss the current pharmacological strategies and challenges to target KCa channels for the treatment of various neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Aravind S Kshatri
- Department of Basic Medical Sciences, Medical School, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologias Biomedicas, Universidad de La Laguna, Tenerife, Spain
| | - Alberto Gonzalez-Hernandez
- Department of Basic Medical Sciences, Medical School, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologias Biomedicas, Universidad de La Laguna, Tenerife, Spain
| | - Teresa Giraldez
- Department of Basic Medical Sciences, Medical School, Universidad de La Laguna, Tenerife, Spain.,Instituto de Tecnologias Biomedicas, Universidad de La Laguna, Tenerife, Spain
| |
Collapse
|
14
|
Grossberg S. Acetylcholine Neuromodulation in Normal and Abnormal Learning and Memory: Vigilance Control in Waking, Sleep, Autism, Amnesia and Alzheimer's Disease. Front Neural Circuits 2017; 11:82. [PMID: 29163063 PMCID: PMC5673653 DOI: 10.3389/fncir.2017.00082] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 10/12/2017] [Indexed: 01/30/2023] Open
Abstract
Adaptive Resonance Theory, or ART, is a neural model that explains how normal and abnormal brains may learn to categorize and recognize objects and events in a changing world, and how these learned categories may be remembered for a long time. This article uses ART to propose and unify the explanation of diverse data about normal and abnormal modulation of learning and memory by acetylcholine (ACh). In ART, vigilance control determines whether learned categories will be general and abstract, or specific and concrete. ART models how vigilance may be regulated by ACh release in layer 5 neocortical cells by influencing after-hyperpolarization (AHP) currents. This phasic ACh release is mediated by cells in the nucleus basalis (NB) of Meynert that are activated by unexpected events. The article additionally discusses data about ACh-mediated tonic control of vigilance. ART proposes that there are often dynamic breakdowns of tonic control in mental disorders such as autism, where vigilance remains high, and medial temporal amnesia, where vigilance remains low. Tonic control also occurs during sleep-wake cycles. Properties of Up and Down states during slow wave sleep arise in ACh-modulated laminar cortical ART circuits that carry out processes in awake individuals of contrast normalization, attentional modulation, decision-making, activity-dependent habituation, and mismatch-mediated reset. These slow wave sleep circuits interact with circuits that control circadian rhythms and memory consolidation. Tonic control properties also clarify how Alzheimer's disease symptoms follow from a massive structural degeneration that includes undermining vigilance control by ACh in cortical layers 3 and 5. Sleep disruptions before and during Alzheimer's disease, and how they contribute to a vicious cycle of plaque formation in layers 3 and 5, are also clarified from this perspective.
Collapse
Affiliation(s)
- Stephen Grossberg
- Center for Adaptive Systems, Graduate Program in Cognitive and Neural Systems, Departments of Mathematics & Statistics, Psychological & Brain Sciences and Biomedical Engineering, Boston University, Boston, MA, United States
| |
Collapse
|
15
|
Activity-Dependent Facilitation of Ca V1.3 Calcium Channels Promotes KCa3.1 Activation in Hippocampal Neurons. J Neurosci 2017; 37:11255-11270. [PMID: 29038242 DOI: 10.1523/jneurosci.0967-17.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 10/02/2017] [Accepted: 10/07/2017] [Indexed: 11/21/2022] Open
Abstract
CaV1 L-type calcium channels are key to regulating neuronal excitability, with the range of functional roles enhanced by interactions with calmodulin, accessory proteins, or CaMKII that modulate channel activity. In hippocampal pyramidal cells, a prominent elevation of CaV1 activity is apparent in late channel openings that can last for seconds following a depolarizing stimulus train. The current study tested the hypothesis that a reported interaction among CaV1.3 channels, the scaffolding protein densin, and CaMKII could generate a facilitation of channel activity that outlasts a depolarizing stimulus. We found that CaV1.3 but not CaV1.2 channels exhibit a long-duration calcium-dependent facilitation (L-CDF) that lasts up to 8 s following a brief 50 Hz stimulus train, but only when coexpressed with densin and CaMKII. To test the physiological role for CaV1.3 L-CDF, we coexpressed the intermediate-conductance KCa3.1 potassium channel, revealing a strong functional coupling to CaV1.3 channel activity that was accentuated by densin and CaMKII. Moreover, the CaV1.3-densin-CaMKII interaction gave rise to an outward tail current of up to 8 s duration following a depolarizing stimulus in both tsA-201 cells and male rat CA1 pyramidal cells. A slow afterhyperpolarization in pyramidal cells was reduced by a selective block of CaV1 channels by isradipine, a CaMKII blocker, and siRNA knockdown of densin, and spike frequency increased upon selective block of CaV1 channel conductance. The results are important in revealing a CaV1.3-densin-CaMKII interaction that extends the contribution of CaV1.3 calcium influx to a time frame well beyond a brief input train.SIGNIFICANCE STATEMENT CaV1 L-type calcium channels play a key role in regulating the output of central neurons by providing calcium influx during repetitive inputs. This study identifies a long-duration calcium-dependent facilitation (L-CDF) of CaV1.3 channels that depends on the scaffolding protein densin and CaMKII and that outlasts a depolarizing stimulus by seconds. We further show a tight functional coupling between CaV1.3 calcium influx and the intermediate-conductance KCa3.1 potassium channel that promotes an outward tail current of up to 8 s following a depolarizing stimulus. Tests in CA1 hippocampal pyramidal cells reveal that a slow AHP is reduced by blocking different components of the CaV1.3-densin-CaMKII interaction, identifying an important role for CaV1.3 L-CDF in regulating neuronal excitability.
Collapse
|
16
|
Gholami Pourbadie H, Naderi N, Janahmadi M, Mehranfard N, Motamedi F. Calcium channel blockade attenuates abnormal synaptic transmission in the dentate gyrus elicited by entorhinal amyloidopathy. Synapse 2016; 70:408-17. [DOI: 10.1002/syn.21915] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 05/24/2016] [Accepted: 05/25/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Hamid Gholami Pourbadie
- Department of Physiology and Pharmacology; Pasteur Institute of Iran; Tehran Iran
- Neuroscience Research Center; Shahid Beheshti University of Medical Sciences; Tehran Iran
- Department of Physiology; Faculty of Medicine, Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Nima Naderi
- Neuroscience Research Center; Shahid Beheshti University of Medical Sciences; Tehran Iran
- Department of Pharmacology and Toxicology; School of Pharmacy, Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Mahyar Janahmadi
- Neurophysiology Research Center; Faculty of Medicine, Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Nasrin Mehranfard
- Neuroscience Research Center; Shahid Beheshti University of Medical Sciences; Tehran Iran
- Department of Physiology; Faculty of Medicine, Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Fereshteh Motamedi
- Neuroscience Research Center; Shahid Beheshti University of Medical Sciences; Tehran Iran
- Department of Physiology; Faculty of Medicine, Shahid Beheshti University of Medical Sciences; Tehran Iran
| |
Collapse
|
17
|
Krueger JN, Moore SJ, Parent R, McKinney BC, Lee A, Murphy GG. A novel mouse model of the aged brain: Over-expression of the L-type voltage-gated calcium channel Ca V1.3. Behav Brain Res 2016; 322:241-249. [PMID: 27368417 DOI: 10.1016/j.bbr.2016.06.054] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 06/09/2016] [Accepted: 06/27/2016] [Indexed: 11/16/2022]
Abstract
The aged population is growing rapidly, which has sparked tremendous interest in elucidating mechanisms of aging in both the body and the brain. Animal models have become an indispensable tool in biomedical science, but because of the cost and extended timeframe associated with aging animals to appropriate time points, studies that rely on using aged animals are often not feasible. Somewhat surprisingly, there are relatively few animal models that have been specifically engineered to mimic physiological changes known to occur during "normal" aging. Developing transgenic animal models that faithfully mimic key aspects of aging would likely be of great utility in studying both age-related deficits in the absence of overt pathology as well as an adjunct for transgenic models of diseases where aging is a primary risk factor. In particular, there are several alterations in the aged brain that are amenable to being modeled genetically. We have focused on one key aspect that has been repeatedly demonstrated in aged animals - an increase in the L-type voltage-gated calcium channel CaV1.3. Here we present a novel transgenic mouse line in which expression of CaV1.3 is increased by approximately 50% in the forebrain of young mice. These mice do not display any overt physical or non-cognitive deficits, exhibiting normal exploratory behavior, motor function, and affective-like responses, suggesting that these mice can be successfully deployed to assess the impact of an "aged brain" in a variety of conditions.
Collapse
Affiliation(s)
- Jamie N Krueger
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Shannon J Moore
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States.
| | - Rachel Parent
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Brandon C McKinney
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Amy Lee
- Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, United States
| | - Geoffrey G Murphy
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States; Department of Physiology, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
18
|
King B, Rizwan AP, Asmara H, Heath NC, Engbers JDT, Dykstra S, Bartoletti TM, Hameed S, Zamponi GW, Turner RW. IKCa channels are a critical determinant of the slow AHP in CA1 pyramidal neurons. Cell Rep 2016; 11:175-82. [PMID: 25865881 DOI: 10.1016/j.celrep.2015.03.026] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 01/30/2015] [Accepted: 03/10/2015] [Indexed: 12/23/2022] Open
Abstract
Control over the frequency and pattern of neuronal spike discharge depends on Ca2+-gated K+ channels that reduce cell excitability by hyperpolarizing the membrane potential. The Ca2+-dependent slow afterhyperpolarization (sAHP) is one of the most prominent inhibitory responses in the brain, with sAHP amplitude linked to a host of circuit and behavioral functions, yet the channel that underlies the sAHP has defied identification for decades. Here, we show that intermediate-conductance Ca2+-dependent K+ (IKCa) channels underlie the sAHP generated by trains of synaptic input or postsynaptic stimuli in CA1 hippocampal pyramidal cells. These findings are significant in providing a molecular identity for the sAHP of central neurons that will identify pharmacological tools capable of potentially modifying the several behavioral or disease states associated with the sAHP.
Collapse
|
19
|
Reversal of Aging-Related Neuronal Ca2+ Dysregulation and Cognitive Impairment by Delivery of a Transgene Encoding FK506-Binding Protein 12.6/1b to the Hippocampus. J Neurosci 2015. [PMID: 26224869 DOI: 10.1523/jneurosci.1248-15.2015] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
UNLABELLED Brain Ca2+ regulatory processes are altered during aging, disrupting neuronal, and cognitive functions. In hippocampal pyramidal neurons, the Ca2+ -dependent slow afterhyperpolarization (sAHP) exhibits an increase with aging, which correlates with memory impairment. The increased sAHP results from elevated L-type Ca2+ channel activity and ryanodine receptor (RyR)-mediated Ca2+ release, but underlying molecular mechanisms are poorly understood. Previously, we found that expression of the gene encoding FK506-binding protein 12.6/1b (FKBP1b), a small immunophilin that stabilizes RyR-mediated Ca2+ release in cardiomyocytes, declines in hippocampus of aged rats and Alzheimer's disease subjects. Additionally, knockdown/disruption of hippocampal FKBP1b in young rats augments neuronal Ca2+ responses. Here, we test the hypothesis that declining FKBP1b underlies aging-related hippocampal Ca2+ dysregulation. Using microinjection of adeno-associated viral vector bearing a transgene encoding FKBP1b into the hippocampus of aged male rats, we assessed the critical prediction that overexpressing FKBP1b should reverse Ca2+ -mediated manifestations of brain aging. Immunohistochemistry and qRT-PCR confirmed hippocampal FKBP1b overexpression 4-6 weeks after injection. Compared to aged vector controls, aged rats overexpressing FKBP1b showed dramatic enhancement of spatial memory, which correlated with marked reduction of sAHP magnitude. Furthermore, simultaneous electrophysiological recording and Ca2+ imaging in hippocampal neurons revealed that the sAHP reduction was associated with a decrease in parallel RyR-mediated Ca2+ transients. Thus, hippocampal FKBP1b overexpression reversed key aspects of Ca2+ dysregulation and cognitive impairment in aging rats, supporting the novel hypothesis that declining FKBP1b is a molecular mechanism underlying aging-related Ca2+ dysregulation and unhealthy brain aging and pointing to FKBP1b as a potential therapeutic target. SIGNIFICANCE STATEMENT This paper reports critical tests of a novel hypothesis that proposes a molecular mechanism of unhealthy brain aging and possibly, Alzheimer's disease. For more than 30 years, evidence has been accumulating that brain aging is associated with dysregulation of calcium in neurons. Recently, we found that FK506-binding protein 12.6/1b (FKBP1b), a small protein that regulates calcium, declines with aging in the hippocampus, a brain region important for memory. Here we used gene therapy approaches and found that raising FKBP1b reversed calcium dysregulation and memory impairment in aging rats, allowing them to perform a memory task as well as young rats. These studies identify a potential molecular mechanism of brain aging and may also have implications for treatment of Alzheimer's disease.
Collapse
|
20
|
Balaraman Y, Lahiri DK, Nurnberger JI. Variants in Ion Channel Genes Link Phenotypic Features of Bipolar Illness to Specific Neurobiological Process Domains. MOLECULAR NEUROPSYCHIATRY 2015; 1:23-35. [PMID: 27602355 DOI: 10.1159/000371886] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/05/2015] [Indexed: 11/19/2022]
Abstract
Recent advances in genome-wide association studies are pointing towards a major role for voltage-gated ion channels in neuropsychiatric disorders and, in particular, bipolar disorder (BD). The phenotype of BD is complex, with symptoms during mood episodes and deficits persisting between episodes. We have tried to elucidate the common neurobiological mechanisms associated with ion channel signaling in order to provide a new perspective on the clinical symptoms and possible endophenotypes seen in BD patients. We propose a model in which the multiple variants in genes coding for ion channel proteins would perturb motivational circuits, synaptic plasticity, myelination, hypothalamic-pituitary-adrenal axis function, circadian neuronal rhythms, and energy regulation. These changes in neurobiological mechanisms would manifest in endophenotypes of aberrant reward processing, white matter hyperintensities, deficits in executive function, altered frontolimbic connectivity, increased amygdala activity, increased melatonin suppression, decreased REM latency, and aberrant myo-inositol/ATP shuttling. The endophenotypes result in behaviors of poor impulse control, motivational changes, cognitive deficits, abnormal stress response, sleep disturbances, and energy changes involving different neurobiological process domains. The hypothesis is that these disturbances start with altered neural circuitry during development, following which multiple environmental triggers may disrupt the neuronal excitability balance through an activity-dependent molecular process, resulting in clinical mood episodes.
Collapse
Affiliation(s)
- Yokesh Balaraman
- Institute of Psychiatric Research, Department of Psychiatry, Neuroscience Research Center, Indiana University School of Medicine, Indianapolis, Ind., USA
| | - Debomoy K Lahiri
- Institute of Psychiatric Research, Department of Psychiatry, Neuroscience Research Center, Indiana University School of Medicine, Indianapolis, Ind., USA
| | - John I Nurnberger
- Institute of Psychiatric Research, Department of Psychiatry, Neuroscience Research Center, Indiana University School of Medicine, Indianapolis, Ind., USA
| |
Collapse
|
21
|
Simão D, Pinto C, Piersanti S, Weston A, Peddie CJ, Bastos AE, Licursi V, Schwarz SC, Collinson LM, Salinas S, Serra M, Teixeira AP, Saggio I, Lima PA, Kremer EJ, Schiavo G, Brito C, Alves PM. Modeling Human Neural Functionality In Vitro: Three-Dimensional Culture for Dopaminergic Differentiation. Tissue Eng Part A 2015; 21:654-68. [DOI: 10.1089/ten.tea.2014.0079] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- Daniel Simão
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Catarina Pinto
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Stefania Piersanti
- Dipartimento di Biologia e Biotecnologie “Charles Darwin,” Università di Roma La Sapienza, Rome, Italy
| | - Anne Weston
- Lincoln's Inn Fields Laboratories, Cancer Research UK London Research Institute, London, United Kingdom
| | - Christopher J. Peddie
- Lincoln's Inn Fields Laboratories, Cancer Research UK London Research Institute, London, United Kingdom
| | - André E.P. Bastos
- NOVA Medical School, Faculdade de Ciências Médicas da Universidade Nova de Lisboa, Lisboa, Portugal
- Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade de Lisboa, Lisboa, Portugal
| | - Valerio Licursi
- Dipartimento di Biologia e Biotecnologie “Charles Darwin,” Università di Roma La Sapienza, Rome, Italy
| | | | - Lucy M. Collinson
- Lincoln's Inn Fields Laboratories, Cancer Research UK London Research Institute, London, United Kingdom
| | - Sara Salinas
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier, France
- Université Montpellier I and II, Montpellier, France
| | - Margarida Serra
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Ana P. Teixeira
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Isabella Saggio
- Dipartimento di Biologia e Biotecnologie “Charles Darwin,” Università di Roma La Sapienza, Rome, Italy
- Istituto Pasteur Fondazione Cenci Bolognetti, Università di Roma La Sapienza, Rome, Italy
- Istituto di Biologia e Patologia Molecolari del CNR, Università di Roma La Sapienza, Rome, Italy
| | - Pedro A. Lima
- NOVA Medical School, Faculdade de Ciências Médicas da Universidade Nova de Lisboa, Lisboa, Portugal
| | - Eric J. Kremer
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier, France
- Université Montpellier I and II, Montpellier, France
| | - Giampietro Schiavo
- Lincoln's Inn Fields Laboratories, Cancer Research UK London Research Institute, London, United Kingdom
- Sobell Department of Motor Neuroscience and Movement Disorders, Institute of Neurology, University College London, London, United Kingdom
| | - Catarina Brito
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Paula M. Alves
- iBET—Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
22
|
Church TW, Weatherall KL, Corrêa SAL, Prole DL, Brown JT, Marrion NV. Preferential assembly of heteromeric small conductance calcium-activated potassium channels. Eur J Neurosci 2014; 41:305-15. [PMID: 25421315 DOI: 10.1111/ejn.12789] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/16/2014] [Accepted: 10/20/2014] [Indexed: 11/27/2022]
Abstract
The activation of small conductance calcium-dependent (SK) channels regulates membrane excitability by causing membrane hyperpolarization. Three subtypes (SK1-3) have been cloned, with each subtype expressed within the nervous system. The locations of channel subunits overlap, with SK1 and SK2 subunits often expressed in the same brain region. We showed that expressed homomeric rat SK1 subunits did not form functional channels, because subunits accumulated in the Golgi. This raised the question of whether heteromeric channels could form with SK1 subunits. The co-expression of SK1 and SK2 subunits in HEK293 cells preferentially co-assembled to produce heteromeric channels with a fixed stoichiometry of alternating subunits. The expression in hippocampal CA1 neurons of mutant rat SK1 subunits [rat SK1(LV213/4YA)] that produced an apamin-sensitive current changed the amplitude and pharmacology of the medium afterhyperpolarization. The overexpression of rat SK1(LV213/4YA) subunits reduced the sensitivity of the medium afterhyperpolarization to apamin, substantiating the preferential co-assembly of SK1 and SK2 subunits to form heteromeric channels. Species-specific channel assembly occurred as the co-expression of human SK1 with rat SK2 did not form functional heteromeric channels. The replacement of two amino acids within the C-terminus of rat SK2 with those from human SK2 permitted the assembly of heteromeric channels when co-expressed with human SK1. These data showed that species-specific co-assembly was mediated by interaction between the C-termini of SK channel subunits. The finding that SK channels preferentially co-assembled to form heteromeric channels suggested that native heteromeric channels will predominate in cells expressing multiple SK channel subunits.
Collapse
Affiliation(s)
- Timothy W Church
- School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| | | | | | | | | | | |
Collapse
|
23
|
The role of L-type voltage-gated calcium channels Cav1.2 and Cav1.3 in normal and pathological brain function. Cell Tissue Res 2014; 357:463-76. [DOI: 10.1007/s00441-014-1936-3] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 05/27/2014] [Indexed: 10/25/2022]
|
24
|
Abstract
The sodium-potassium ATPase (i.e., the "sodium pump") plays a central role in maintaining ionic homeostasis in all cells. Although the sodium pump is intrinsically electrogenic and responsive to dynamic changes in intracellular sodium concentration, its role in regulating neuronal excitability remains unclear. Here we describe a physiological role for the sodium pump in regulating the excitability of mouse neocortical layer 5 and hippocampal CA1 pyramidal neurons. Trains of action potentials produced long-lasting (∼20 s) afterhyperpolarizations (AHPs) that were insensitive to blockade of voltage-gated calcium channels or chelation of intracellular calcium, but were blocked by tetrodotoxin, ouabain, or the removal of extracellular potassium. Correspondingly, the AHP time course was similar to the decay of activity-induced increases in intracellular sodium, whereas intracellular calcium decayed at much faster rates. To determine whether physiological patterns of activity engage the sodium pump, we replayed in vitro a place-specific burst of 15 action potentials recorded originally in vivo in a CA1 "place cell" as the animal traversed the associated place field. In both layer 5 and CA1 pyramidal neurons, this "place cell train" generated small, long-lasting AHPs capable of reducing neuronal excitability for many seconds. Place-cell-train-induced AHPs were blocked by ouabain or removal of extracellular potassium, but not by intracellular calcium chelation. Finally, we found calcium contributions to the AHP to be temperature dependent: prominent at room temperature, but largely absent at 35°C. Our results demonstrate a previously unappreciated role for the sodium-potassium ATPase in regulating the excitability of neocortical and hippocampal pyramidal neurons.
Collapse
|
25
|
The slow afterhyperpolarization: a target of β1-adrenergic signaling in hippocampus-dependent memory retrieval. J Neurosci 2013; 33:5006-16. [PMID: 23486971 DOI: 10.1523/jneurosci.3834-12.2013] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
In rodents, adrenergic signaling by norepinephrine (NE) in the hippocampus is required for the retrieval of intermediate-term memory. NE promotes retrieval via the stimulation of β1-adrenergic receptors, the production of cAMP, and the activation of both protein kinase A (PKA) and the exchange protein activated by cAMP. However, a final effector for this signaling pathway has not been identified. Among the many targets of adrenergic signaling in the hippocampus, the slow afterhyperpolarization (sAHP) is an appealing candidate because its reduction by β1 signaling enhances excitatory neurotransmission. Here we report that reducing the sAHP is critical for the facilitation of retrieval by NE. Direct blockers of the sAHP, as well as blockers of the L-type voltage-dependent calcium influx that activates the sAHP, rescue retrieval in mutant mice lacking either NE or the β1 receptor. Complementary to this, a facilitator of L-type calcium influx impairs retrieval in wild-type mice. In addition, we examined the role of NE in the learning-related reduction of the sAHP observed ex vivo in hippocampal slices. We find that this reduction in the sAHP depends on the induction of persistent PKA activity specifically in conditioned slices. Interestingly, this persistent PKA activity is induced by NE/β1 signaling during slice preparation rather than during learning. These observations suggest that the reduction in the sAHP may not be present autonomously in vivo, but is likely induced by neuromodulatory input, which is consistent with the idea that NE is required in vivo for reduction of the sAHP during memory retrieval.
Collapse
|
26
|
Palma J, Grossberg S, Versace M. Persistence and storage of activity patterns in spiking recurrent cortical networks: modulation of sigmoid signals by after-hyperpolarization currents and acetylcholine. Front Comput Neurosci 2012; 6:42. [PMID: 22754524 PMCID: PMC3386521 DOI: 10.3389/fncom.2012.00042] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 06/11/2012] [Indexed: 11/13/2022] Open
Abstract
Many cortical networks contain recurrent architectures that transform input patterns before storing them in short-term memory (STM). Theorems in the 1970's showed how feedback signal functions in rate-based recurrent on-center off-surround networks control this process. A sigmoid signal function induces a quenching threshold below which inputs are suppressed as noise and above which they are contrast-enhanced before pattern storage. This article describes how changes in feedback signaling, neuromodulation, and recurrent connectivity may alter pattern processing in recurrent on-center off-surround networks of spiking neurons. In spiking neurons, fast, medium, and slow after-hyperpolarization (AHP) currents control sigmoid signal threshold and slope. Modulation of AHP currents by acetylcholine (ACh) can change sigmoid shape and, with it, network dynamics. For example, decreasing signal function threshold and increasing slope can lengthen the persistence of a partially contrast-enhanced pattern, increase the number of active cells stored in STM, or, if connectivity is distance-dependent, cause cell activities to cluster. These results clarify how cholinergic modulation by the basal forebrain may alter the vigilance of category learning circuits, and thus their sensitivity to predictive mismatches, thereby controlling whether learned categories code concrete or abstract features, as predicted by Adaptive Resonance Theory. The analysis includes global, distance-dependent, and interneuron-mediated circuits. With an appropriate degree of recurrent excitation and inhibition, spiking networks maintain a partially contrast-enhanced pattern for 800 ms or longer after stimuli offset, then resolve to no stored pattern, or to winner-take-all (WTA) stored patterns with one or multiple winners. Strengthening inhibition prolongs a partially contrast-enhanced pattern by slowing the transition to stability, while strengthening excitation causes more winners when the network stabilizes.
Collapse
Affiliation(s)
| | - Stephen Grossberg
- Graduate Program in Cognitive and Neural Systems, Center for Adaptive Systems, Center of Excellence for Learning in Education, Science, and Technology, Center for Computational Neuroscience and Neural Technology, Boston University, BostonMA, USA
| | | |
Collapse
|
27
|
The effect of neural noise on spike time precision in a detailed CA3 neuron model. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2012; 2012:595398. [PMID: 22778784 PMCID: PMC3388596 DOI: 10.1155/2012/595398] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 12/21/2011] [Accepted: 01/23/2012] [Indexed: 11/26/2022]
Abstract
Experimental and computational studies emphasize the role of the millisecond precision of neuronal spike times as an important coding mechanism for transmitting and representing information in the central nervous system. We investigate the spike time precision of a multicompartmental pyramidal neuron model of the CA3 region of the hippocampus under the influence of various sources of neuronal noise. We describe differences in the contribution to noise originating from voltage-gated ion channels, synaptic vesicle release, and vesicle quantal size. We analyze the effect of interspike intervals and the voltage course preceding the firing of spikes on the spike-timing jitter. The main finding of this study is the ranking of different noise sources according to their contribution to spike time precision. The most influential is synaptic vesicle release noise, causing the spike jitter to vary from 1 ms to 7 ms of a mean value 2.5 ms. Of second importance was the noise incurred by vesicle quantal size variation causing the spike time jitter to vary from 0.03 ms to 0.6 ms. Least influential was the voltage-gated channel noise generating spike jitter from 0.02 ms to 0.15 ms.
Collapse
|
28
|
The role of metaplasticity mechanisms in regulating memory destabilization and reconsolidation. Neurosci Biobehav Rev 2012; 36:1667-707. [PMID: 22484475 DOI: 10.1016/j.neubiorev.2012.03.008] [Citation(s) in RCA: 144] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 03/09/2012] [Accepted: 03/21/2012] [Indexed: 12/13/2022]
Abstract
Memory allows organisms to predict future events based on prior experiences. This requires encoded information to persist once important predictors are extracted, while also being modifiable in response to changes within the environment. Memory reconsolidation may allow stored information to be modified in response to related experience. However, there are many boundary conditions beyond which reconsolidation may not occur. One interpretation of these findings is that the event triggering memory retrieval must contain new information about a familiar stimulus in order to induce reconsolidation. Presently, the mechanisms that affect the likelihood of reconsolidation occurring under these conditions are not well understood. Here we speculate on a number of systems that may play a role in protecting memory from being destabilized during retrieval. We conclude that few memories may enter a state in which they cannot be modified. Rather, metaplasticity mechanisms may serve to alter the specific reactivation cues necessary to destabilize a memory. This might imply that destabilization mechanisms can differ depending on learning conditions.
Collapse
|
29
|
Zaitsev AV, Anwyl R. Inhibition of the slow afterhyperpolarization restores the classical spike timing-dependent plasticity rule obeyed in layer 2/3 pyramidal cells of the prefrontal cortex. J Neurophysiol 2011; 107:205-15. [PMID: 21975445 DOI: 10.1152/jn.00452.2011] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The induction of long-term potentiation (LTP) and long-term depression (LTD) of excitatory postsynaptic currents was investigated in proximal synapses of layer 2/3 pyramidal cells of the rat medial prefrontal cortex. The spike timing-dependent plasticity (STDP) induction protocol of negative timing, with postsynaptic leading presynaptic stimulation of action potentials (APs), induced LTD as expected from the classical STDP rule. However, the positive STDP protocol of presynaptic leading postsynaptic stimulation of APs predominantly induced a presynaptically expressed LTD rather than the expected postsynaptically expressed LTP. Thus the induction of plasticity in layer 2/3 pyramidal cells does not obey the classical STDP rule for positive timing. This unusual STDP switched to a classical timing rule if the slow Ca(2+)-dependent, K(+)-mediated afterhyperpolarization (sAHP) was inhibited by the selective blocker N-trityl-3-pyridinemethanamine (UCL2077), by the β-adrenergic receptor agonist isoproterenol, or by the cholinergic agonist carbachol. Thus we demonstrate that neuromodulators can affect synaptic plasticity by inhibition of the sAHP. These findings shed light on a fundamental question in the field of memory research regarding how environmental and behavioral stimuli influence LTP, thereby contributing to the modulation of memory.
Collapse
Affiliation(s)
- Aleksey V Zaitsev
- Dept. of Physiology, Trinity College Dublin, College Green, Dublin 2, Ireland
| | | |
Collapse
|
30
|
Palma J, Versace M, Grossberg S. After-hyperpolarization currents and acetylcholine control sigmoid transfer functions in a spiking cortical model. J Comput Neurosci 2011; 32:253-80. [PMID: 21779754 DOI: 10.1007/s10827-011-0354-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Revised: 06/09/2011] [Accepted: 07/06/2011] [Indexed: 10/18/2022]
Abstract
Recurrent networks are ubiquitous in the brain, where they enable a diverse set of transformations during perception, cognition, emotion, and action. It has been known since the 1970's how, in rate-based recurrent on-center off-surround networks, the choice of feedback signal function can control the transformation of input patterns into activity patterns that are stored in short term memory. A sigmoid signal function may, in particular, control a quenching threshold below which inputs are suppressed as noise and above which they may be contrast enhanced before the resulting activity pattern is stored. The threshold and slope of the sigmoid signal function determine the degree of noise suppression and of contrast enhancement. This article analyses how sigmoid signal functions and their shape may be determined in biophysically realistic spiking neurons. Combinations of fast, medium, and slow after-hyperpolarization (AHP) currents, and their modulation by acetylcholine (ACh), can control sigmoid signal threshold and slope. Instead of a simple gain in excitability that was previously attributed to ACh, cholinergic modulation may cause translation of the sigmoid threshold. This property clarifies how activation of ACh by basal forebrain circuits, notably the nucleus basalis of Meynert, may alter the vigilance of category learning circuits, and thus their sensitivity to predictive mismatches, thereby controlling whether learned categories code concrete or abstract information, as predicted by Adaptive Resonance Theory.
Collapse
Affiliation(s)
- Jesse Palma
- Center for Adaptive Systems, Department of Cognitive and Neural Systems, and Center of Excellence for Learning in Education, Science, and Technology, Boston University, Boston, MA 02215, USA
| | | | | |
Collapse
|
31
|
Moore SJ, Throesch BT, Murphy GG. Of mice and intrinsic excitability: genetic background affects the size of the postburst afterhyperpolarization in CA1 pyramidal neurons. J Neurophysiol 2011; 106:1570-80. [PMID: 21697442 DOI: 10.1152/jn.00257.2011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
As the use of genetically engineered mice has become increasingly prevalent in neurobiological research, evidence has steadily accumulated that substantial differences exist between strains. Although a number of studies have reported effects of genetic background on behavior, few have focused on differences in neurophysiology. The postburst afterhyperpolarization (AHP) is an important determinant of intrinsic neuronal excitability and has been suggested to play a critical role in the cellular mechanisms underlying learning and memory. Using whole cell current-clamp recordings of CA1 pyramidal neurons, we examined the magnitude of different phases of the AHP (peak, medium, and slow) in two commonly used genetic backgrounds, C57BL/6 (B6) and 129SvEv (129), as well as in an F2 hybrid B6:129 background (F2). We found that neurons from B6 and F2 animals exhibited a significantly larger AHP compared with 129 animals and that this difference was consistent across all phases. Furthermore, our recordings revealed a marked dichotomy in the shape of the AHP waveform, which was independent of genetic background. Approximately 60% of cells exhibited an AHP with a sharp transition between the peak AHP and medium AHP, whereas the remaining 40% exhibited a more gradual transition. Our data add to the growing body of work suggesting that genetic background can affect neuronal function as well as behavior. In addition, these results highlight the innate heterogeneity of CA1 pyramidal neurons, even within a single genetic background. These differences should be taken into consideration during the analysis and comparison of experimental results.
Collapse
Affiliation(s)
- Shannon J Moore
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | | | | |
Collapse
|
32
|
Hirtz JJ, Boesen M, Braun N, Deitmer JW, Kramer F, Lohr C, Müller B, Nothwang HG, Striessnig J, Löhrke S, Friauf E. Cav1.3 calcium channels are required for normal development of the auditory brainstem. J Neurosci 2011; 31:8280-94. [PMID: 21632949 PMCID: PMC6622878 DOI: 10.1523/jneurosci.5098-10.2011] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Revised: 03/14/2011] [Accepted: 04/10/2011] [Indexed: 12/18/2022] Open
Abstract
Within the Ca(v)1 family of voltage-gated calcium channels, Ca(v)1.2 and Ca(v)1.3 channels are the predominant subtypes in the brain. Whereas specific functions for each subtype were described in the adult brain, their role in brain development is poorly understood. Here we assess the role of Ca(v)1.3 subunits in the activity-dependent development of the auditory brainstem. We used Ca(v)1.3-deficient (Ca(v)1.3(-/-)) mice because these mice lack cochlea-driven activity that deprives the auditory centers from peripheral input. We found a drastically reduced volume in all auditory brainstem centers (range 25-59%, total 35%), which was manifest before hearing onset. A reduction was not obvious outside the auditory system. The lateral superior olive (LSO) was strikingly malformed in Ca(v)1.3(-/-) mice and had fewer neurons (1/3 less). The remaining LSO neurons displayed normal dendritic trees and received functional glutamatergic input, yet they fired action potentials predominantly with a multiple pattern upon depolarization, in contrast to the single firing pattern prevalent in controls. The latter finding appears to be due to a reduction of dendrototoxin-sensitive potassium conductances, presumably mediated through the K(v)1.2 subtype. Fura2 imaging provided evidence for functional Ca(v)1.3 channels in the LSO of wild-type mice. Our results imply that Ca(v)1.3 channels are indispensable for the development of the central auditory system. We propose that the unique LSO phenotype in Ca(v)1.3(-/-) mice, which hitherto was not described in other hereditary deafness models, is caused by the synergistic contribution of two factors: on-site loss of Ca(v)1.3 channels in the neurons plus lack of peripheral input.
Collapse
Affiliation(s)
| | | | | | - Joachim W. Deitmer
- General Zoology, Department of Biology, University of Kaiserslautern, D-67653 Kaiserslautern, Germany
| | | | - Christian Lohr
- General Zoology, Department of Biology, University of Kaiserslautern, D-67653 Kaiserslautern, Germany
| | | | - Hans Gerd Nothwang
- Animal Physiology Group and
- Department of Neurogenetics, Institute for Biology and Environmental Sciences, University of Oldenburg, D-26129 Oldenburg, Germany, and
| | - Jörg Striessnig
- Institute of Pharmacy, Pharmacology and Toxicology, Center of Molecular Biosciences, University of Innsbruck, A-6020 Innsbruck, Austria
| | | | | |
Collapse
|
33
|
Gamelli AE, McKinney BC, White JA, Murphy GG. Deletion of the L-type calcium channel Ca(V) 1.3 but not Ca(V) 1.2 results in a diminished sAHP in mouse CA1 pyramidal neurons. Hippocampus 2011; 21:133-41. [PMID: 20014384 DOI: 10.1002/hipo.20728] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Trains of action potentials in CA1 pyramidal neurons are followed by a prolonged calcium-dependent postburst afterhyperpolarization (AHP) that serves to limit further firing to a sustained depolarizing input. A reduction in the AHP accompanies acquisition of several types of learning and increases in the AHP are correlated with age-related cognitive impairment. The AHP develops primarily as the result of activation of outward calcium-activated potassium currents; however, the precise source of calcium for activation of the AHP remains unclear. There is substantial experimental evidence suggesting that calcium influx via voltage-gated L-type calcium channels (L-VGCCs) contributes to the generation of the AHP. Two L-VGCC subtypes are predominately expressed in the hippocampus, Ca(V) 1.2 and Ca(V) 1.3; however, it is not known which L-VGCC subtype is involved in generation of the AHP. This ambiguity is due in large part to the fact that at present there are no subunit-specific agonists or antagonists. Therefore, using mice in which the gene encoding Ca(V) 1.2 or Ca(V) 1.3 was deleted, we sought to determine the impact of alterations in levels of these two L-VCGG subtypes on neuronal excitability. No differences in any AHP measure were seen between neurons from Ca(V) 1.2 knockout mice and controls. However, the total area of the AHP was significantly smaller in neurons from Ca(V) 1.3 knockout mice as compared with neurons from wild-type controls. A significant reduction in the amplitude of the AHP was also seen at the 1 s time point in neurons from Ca(V) 1.3 knockout mice as compared with those from controls. Reductions in both the area and 1 s amplitude suggest the involvement of calcium influx via Ca(V) 1.3 in the slow AHP (sAHP). Thus, the results of our study demonstrate that deletion of Ca(V) 1.3, but not Ca(V) 1.2, significantly impacts the generation of the sAHP.
Collapse
Affiliation(s)
- Amy E Gamelli
- Molecular & Behavioral Neuroscience Institute, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | | | |
Collapse
|
34
|
Lu CB, Hamilton JB, Powell AD, Toescu EC, Vreugdenhil M. Effect of ageing on CA3 interneuron sAHP and gamma oscillations is activity-dependent. Neurobiol Aging 2011; 32:956-65. [PMID: 19523715 DOI: 10.1016/j.neurobiolaging.2009.05.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Revised: 05/05/2009] [Accepted: 05/07/2009] [Indexed: 01/21/2023]
Abstract
Normal ageing-associated spatial memory impairment has been linked to subtle changes in the hippocampal network. Here we test whether the age-dependent reduction in gamma oscillations can be explained by the changes in intrinsic properties of hippocampal interneurons. Kainate-induced gamma oscillations, but not spontaneous gamma oscillations, were reduced in slices from aged mice. CA3 interneurons were recorded in slices from young and aged mice using Fura-2-filled pipettes. Passive membrane properties, firing properties, medium- and slow-afterhyperpolarisation amplitudes, basal [Ca(2+)](i) and firing-induced [Ca(2+)](i) transients were not different with ageing. Kainate caused a larger depolarisation and increase in [Ca(2+)](i) signal in aged interneurons than in young ones. In contrast to young interneurons, kainate increased the medium- and slow-afterhyperpolarisation and underlying [Ca(2+)](i) transient in aged interneurons. Modulating the slow-afterhyperpolarisation by modulating L-type calcium channels with BAY K 8644 and nimodipine suppressed and potentiated, respectively, kainate-induced gamma oscillations in young slices. The age-dependent and stimulation-dependent increase in basal [Ca(2+)](i), firing-induced [Ca(2+)](i) transient and associated afterhyperpolarisation may reduce interneuron excitability and contribute to an age-dependent impairment of hippocampal gamma oscillations.
Collapse
Affiliation(s)
- Cheng B Lu
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | | | | | | | | |
Collapse
|
35
|
Location and function of the slow afterhyperpolarization channels in the basolateral amygdala. J Neurosci 2011; 31:526-37. [PMID: 21228162 DOI: 10.1523/jneurosci.1045-10.2011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The basolateral amygdala (BLA) assigns emotional significance to sensory stimuli. This association results in a change in the output (action potentials) of BLA projection neurons in response to the stimulus. Neuronal output is controlled by the intrinsic excitability of the neuron. A major determinant of intrinsic excitability in these neurons is the slow afterhyperpolarization (sAHP) that follows action potential (AP) trains and produces spike-frequency adaptation. The sAHP is mediated by a slow calcium-activated potassium current (sI(AHP)), but little is known about the channels that underlie this current. Here, using whole-cell patch-clamp recordings and high-speed calcium imaging from rat BLA projection neurons, we examined the location and function of these channels. We determined the location of the sI(AHP) by applying a hyperpolarizing voltage step during the sI(AHP) and measuring the time needed for the current to adapt to the new command potential, a function of its electrotonic distance from the somatic recording electrode. Channel location was also probed by focally uncaging calcium using a UV laser. Both methodologies indicated that, in BLA neurons, the sI(AHP) is primarily located in the dendritic tree. EPSPs recorded at the soma were smaller, decayed faster, and showed less summation during the sAHP. Adrenergic stimulation and buffering calcium reduced the sAHP and the attenuation of the EPSP during the sAHP. The sAHP also modulated the AP in the dendrite, reducing the calcium response evoked by a single AP. Thus, in addition to mediating spike-frequency adaptation, the sI(AHP) modulates communication between the soma and the dendrite.
Collapse
|
36
|
Opposite effects of low and high doses of Abeta42 on electrical network and neuronal excitability in the rat prefrontal cortex. PLoS One 2009; 4:e8366. [PMID: 20027222 PMCID: PMC2791225 DOI: 10.1371/journal.pone.0008366] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2009] [Accepted: 11/24/2009] [Indexed: 12/20/2022] Open
Abstract
Changes in neuronal synchronization have been found in patients and animal models of Alzheimer's disease (AD). Synchronized behaviors within neuronal networks are important to such complex cognitive processes as working memory. The mechanisms behind these changes are not understood but may involve the action of soluble beta-amyloid (Abeta) on electrical networks. In order to determine if Abeta can induce changes in neuronal synchronization, the activities of pyramidal neurons were recorded in rat prefrontal cortical (PFC) slices under calcium-free conditions using multi-neuron patch clamp technique. Electrical network activities and synchronization among neurons were significantly inhibited by low dose Abeta42 (1 nM) and initially by high dose Abeta42 (500 nM). However, prolonged application of high dose Abeta42 resulted in network activation and tonic firing. Underlying these observations, we discovered that prolonged application of low and high doses of Abeta42 induced opposite changes in action potential (AP)-threshold and after-hyperpolarization (AHP) of neurons. Accordingly, low dose Abeta42 significantly increased the AP-threshold and deepened the AHP, making neurons less excitable. In contrast, high dose Abeta42 significantly reduced the AP-threshold and shallowed the AHP, making neurons more excitable. These results support a model that low dose Abeta42 released into the interstitium has a physiologic feedback role to dampen electrical network activity by reducing neuronal excitability. Higher concentrations of Abeta42 over time promote supra-synchronization between individual neurons by increasing their excitability. The latter may disrupt frontal-based cognitive processing and in some cases lead to epileptiform discharges.
Collapse
|
37
|
McKinney BC, Sze W, Lee B, Murphy GG. Impaired long-term potentiation and enhanced neuronal excitability in the amygdala of Ca(V)1.3 knockout mice. Neurobiol Learn Mem 2009; 92:519-28. [PMID: 19595780 PMCID: PMC2747027 DOI: 10.1016/j.nlm.2009.06.012] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Revised: 06/24/2009] [Accepted: 06/26/2009] [Indexed: 11/21/2022]
Abstract
Previously, we demonstrated that mice in which the gene for the L-type voltage-gated calcium channel Ca(V)1.3 is deleted (Ca(V)1.3 knockout mice) exhibit an impaired ability to consolidate contextually-conditioned fear. Given that this form of Pavlovian fear conditioning is critically dependent on the basolateral complex of the amygdala (BLA), we were interested in the mechanisms by which Ca(V)1.3 contributes to BLA neurophysiology. In the present study, we used in vitro amygdala slices prepared from Ca(V)1.3 knockout mice and wild-type littermates to explore the role of Ca(V)1.3 in long-term potentiation (LTP) and intrinsic neuronal excitability in the BLA. We found that LTP in the lateral nucleus (LA) of the BLA, induced by high-frequency stimulation of the external capsule, was significantly reduced in Ca(V)1.3 knockout mice. Additionally, we found that BLA principal neurons from Ca(V)1.3 knockout mice were hyperexcitable, exhibiting significant increases in firing rates and decreased interspike intervals in response to prolonged somatic depolarization. This aberrant increase in neuronal excitability appears to be at least in part due to a concomitant reduction in the slow component of the post-burst afterhyperpolarization. Together, these results demonstrate altered neuronal function in the BLA of Ca(V)1.3 knockout mice which may account for the impaired ability of these mice to consolidate contextually-conditioned fear.
Collapse
Affiliation(s)
- Brandon C. McKinney
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI 48109-0069
- Neuroscience Program, University of Michigan, Ann Arbor, MI 48109-0069
| | - Wilson Sze
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109-0069
| | - Benjamin Lee
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109-0069
| | - Geoffrey G. Murphy
- Neuroscience Program, University of Michigan, Ann Arbor, MI 48109-0069
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109-0069
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109-0069
| |
Collapse
|
38
|
Leitch B, Szostek A, Lin R, Shevtsova O. Subcellular distribution of L-type calcium channel subtypes in rat hippocampal neurons. Neuroscience 2009; 164:641-57. [PMID: 19665524 DOI: 10.1016/j.neuroscience.2009.08.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2009] [Revised: 08/03/2009] [Accepted: 08/03/2009] [Indexed: 01/01/2023]
Abstract
L-type calcium channels play an essential role in synaptic activity-dependent gene expression and are implicated in long-term alterations in synaptic efficacy underlying learning and memory in the hippocampus. The two principal pore-forming subunits of L-type Ca2+ channels expressed in neurons are the Ca(v)1.2 (alpha(1C)) or Ca(v)1.3 (alpha(1D)) subtypes. Experimental evidence suggests that calcium entry through Ca(v)1.2 and Ca(v)1.3 Ca2+ channels occurs in close proximity to key signalling molecules responsible for triggering signalling pathways leading to transcriptional responses. Determining the subcellular distribution of Ca(v)1.2 and Ca(v)1.3 L-type channels in neurons is clearly important for unravelling the molecular mechanisms underlying long-term alterations in neuronal function. In this study, we used immunogold-labelling techniques and electron-microscopy (EM) to analyse the subcellular distribution and density of both Ca(v)1.2 and Ca(v)1.3 Ca2+ channels in rat hippocampal CA1 pyramidal cells in vivo. We confirm that both Ca(v)1.2 and Ca(v)1.3 channel subtypes are predominantly but not exclusively located in postsynaptic dendritic processes and somata. Both Ca(v)1.2 and Ca(v)1.3 are distributed throughout the dendritic tree. However, the smallest (distal) dendritic processes and spines have proportionally more calcium channels inserted into their plasma membrane than located within cytoplasmic compartments indicating the potential targeting of calcium channels to microdomains within neurons. Ca(v)1.2 and Ca(v)1.3 Ca2+ channels are located at the postsynaptic density and also at extra-synaptic sites. The location of L-type Ca(v)1.2 and Ca(v)1.3 channels in distal dendrites and spines would thus place them at appropriate sites where they could initiate synapse to nucleus signalling.
Collapse
Affiliation(s)
- B Leitch
- Department of Anatomy and Structural Biology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand.
| | | | | | | |
Collapse
|
39
|
Vatanparast J, Janahmadi M. Contribution of apamin-sensitive SK channels to the firing precision but not to the slow afterhyperpolarization and spike frequency adaptation in snail neurons. Brain Res 2008; 1255:57-66. [PMID: 19100724 DOI: 10.1016/j.brainres.2008.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2008] [Revised: 12/01/2008] [Accepted: 12/02/2008] [Indexed: 10/21/2022]
Abstract
Apamin-sensitive small conductance Ca(2+)-dependent K(+)(SK) channels are generally accepted as responsible for the medium afterhyperpolarization (mAHP) after single or train of action potentials. Here, we examined the functional involvement of these channels in the firing precision, post train AHP and spike frequency adaptation (SFA) in neurons of snail Caucasotachea atrolabiata. Apamin, a selective SK channel antagonist, reduced the duration of single-spike AHP and disrupted the spontaneous rhythmic activity. High frequency trains of evoked action potentials showed a time-dependent decrease in the action potential discharge rate (spike frequency adaptation) and followed by a prominent post stimulus inhibitory period (PSIP) as a marker of slow AHP (sAHP). Neither sAHP nor SFA was attenuated by apamin, suggesting that apamin-sensitive SK channels can strongly affect the rhythmicity, but are probably not involved in the SFA and sAHP. Nifedipine, antagonist of L-type Ca(2+) channels, decreased the firing frequency and neuronal rhythmicity. When PSIP was normalized to the background interspike interval, a suppressing effect of nifedipine on PSIP was also observed. Intracellular iontophoretic injection of BAPTA, a potent Ca(2+) chelator, dramatically suppressed PSIP that confirms the intracellular Ca(2+) dependence of the sAHP, but had no discernable effect on the SFA. During train-evoked activity a reduction in the action potential overshoot and maximum depolarization rate was also observed, along with a decrease in the firing frequency, while the action potential threshold increased, which indicated that Na(+) channels, rather than Ca(2+)-dependent K(+) channels, are involved in the SFA.
Collapse
Affiliation(s)
- Jafar Vatanparast
- Department of Biology, College of Sciences, Shiraz University, Adabiat Intersection, Shiraz 71454, Iran.
| | | |
Collapse
|
40
|
Liebmann L, Karst H, Sidiropoulou K, van Gemert N, Meijer OC, Poirazi P, Joëls M. Differential Effects of Corticosterone on the Slow Afterhyperpolarization in the Basolateral Amygdala and CA1 Region: Possible Role of Calcium Channel Subunits. J Neurophysiol 2008; 99:958-68. [DOI: 10.1152/jn.01137.2007] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The stress hormone corticosterone increases the amplitude of the slow afterhyperpolarization (sAHP) in CA1 pyramidal neurons, without affecting resting membrane potential, input resistance, or action potential characteristics. We here examined how corticosterone affects these properties in the basolateral amygdala (BLA). In the amygdala, corticosterone does not change the AHP amplitude, nor any of the passive and active membrane properties studied. The lack of effect on the AHP is surprising since in both areas corticosterone increases high-voltage–activated sustained calcium currents, which supposedly regulate the sAHP. We wondered whether corticosterone targets different calcium channel subunits in the two areas because currents through only one of the subunits (Cav1.3) are thought to alter the AHP amplitude. In situ hybridization studies revealed that CA1 cells express Cav1.2 and Cav1.3 subunits; corticosterone does not transcriptionally regulate Cav1.2 but increases Cav1.3 expression compared with vehicle treatment. In the BLA, Cav1.3 expression was not detectable, both after control and corticosterone treatment. Cav1.2 is moderately expressed. In a modeling study, we examined putative consequences of changes in specific calcium channel subunit expression and calcium extrusion by corticosterone for the AHP in CA1 and amygdala neurons. A differential distribution and transcriptional regulation of Cav1.2 and Cav1.3 in the CA1 area versus BLA partly explain the observed differences in AHP amplitude. The functional implication of these findings could be that stress-induced arousal of activity in the BLA is more prolonged than that in the CA1 hippocampal area, so that information with an emotional component is more effectively encoded.
Collapse
|