1
|
Powell SK, O'Shea C, Townsley K, Prytkova I, Dobrindt K, Elahi R, Iskhakova M, Lambert T, Valada A, Liao W, Ho SM, Slesinger PA, Huckins LM, Akbarian S, Brennand KJ. Induction of dopaminergic neurons for neuronal subtype-specific modeling of psychiatric disease risk. Mol Psychiatry 2023; 28:1970-1982. [PMID: 34493831 PMCID: PMC8898985 DOI: 10.1038/s41380-021-01273-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/22/2021] [Accepted: 08/19/2021] [Indexed: 11/08/2022]
Abstract
Dopaminergic neurons are critical to movement, mood, addiction, and stress. Current techniques for generating dopaminergic neurons from human induced pluripotent stem cells (hiPSCs) yield heterogenous cell populations with variable purity and inconsistent reproducibility between donors, hiPSC clones, and experiments. Here, we report the rapid (5 weeks) and efficient (~90%) induction of induced dopaminergic neurons (iDANs) through transient overexpression of lineage-promoting transcription factors combined with stringent selection across five donors. We observe maturation-dependent increase in dopamine synthesis and electrophysiological properties consistent with midbrain dopaminergic neuron identity, such as slow-rising after- hyperpolarization potentials, an action potential duration of ~3 ms, tonic sub-threshold oscillatory activity, and spontaneous burst firing at a frequency of ~1.0-1.75 Hz. Transcriptome analysis reveals robust expression of genes involved in fetal midbrain dopaminergic neuron identity. Specifically expressed genes in iDANs, as well as those from isogenic induced GABAergic and glutamatergic neurons, were enriched in loci conferring heritability for cannabis use disorder, schizophrenia, and bipolar disorder; however, each neuronal subtype demonstrated subtype-specific heritability enrichments in biologically relevant pathways, and iDANs alone were uniquely enriched in autism spectrum disorder risk loci. Therefore, iDANs provide a critical tool for modeling midbrain dopaminergic neuron development and dysfunction in psychiatric disease.
Collapse
Affiliation(s)
- Samuel K Powell
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Molecular Psychiatry, Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Callan O'Shea
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Molecular Psychiatry, Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Kayla Townsley
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Iya Prytkova
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristina Dobrindt
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Molecular Psychiatry, Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Rahat Elahi
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marina Iskhakova
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tova Lambert
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aditi Valada
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Will Liao
- New York Genome Center, New York, NY, USA
| | - Seok-Man Ho
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paul A Slesinger
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura M Huckins
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Schahram Akbarian
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Kristen J Brennand
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomics, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Division of Molecular Psychiatry, Department of Psychiatry, Yale University, New Haven, CT, USA.
| |
Collapse
|
2
|
Skiteva O, Yao N, Mantas I, Zhang X, Perlmann T, Svenningsson P, Chergui K. Aberrant somatic calcium channel function in cNurr1 and LRRK2-G2019S mice. NPJ Parkinsons Dis 2023; 9:56. [PMID: 37029193 PMCID: PMC10082048 DOI: 10.1038/s41531-023-00500-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 03/23/2023] [Indexed: 04/09/2023] Open
Abstract
In Parkinson's disease (PD), axons of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNc) degenerate before their cell bodies. Calcium influx during pacemaker firing might contribute to neuronal loss, but it is not known if dysfunctions of voltage-gated calcium channels (VGCCs) occur in DA neurons somata and axon terminals. We investigated T-type and L-type VGCCs in SNc-DA neurons of two mouse models of PD: mice with a deletion of the Nurr1 gene in DA neurons from an adult age (cNurr1 mice), and mice bearing the G2019S mutation in the gene coding for LRRK2 (G2019S mice). Adult cNurr1 mice displayed motor and DA deficits, while middle-aged G2019S mice did not. The number and morphology of SNc-DA neurons, most of their intrinsic membrane properties and pacemaker firing were unaltered in cNurr1 and G2019S mice compared to their control and wild-type littermates. L-type VGCCs contributed to the pacemaker firing of SNc-DA neurons in G2019S mice, but not in control, wild-type, and cNurr1 mice. In cNurr1 mice, but not G2019S mice, the contribution of T-type VGCCs to the pacemaker firing of SNc-DA neurons was reduced, and somatic dopamine-D2 autoreceptors desensitized more. Altered contribution of L-type and T-type VGCCs to the pacemaker firing was not observed in the presence of a LRRK2 kinase inhibitor in G2019S mice, and in the presence of a flavonoid with antioxidant activity in G2019S and cNurr1 mice. The role of L-type and T-type VGCCs in controlling dopamine release from axon terminals in the striatum was unaltered in cNurr1 and G2019S mice. Our findings uncover opposite changes, linked to oxidative stress, in the function of two VGCCs in DA neurons somata, but not axon terminals, in two different experimental PD models.
Collapse
Affiliation(s)
- Olga Skiteva
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ning Yao
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ioannis Mantas
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Xiaoqun Zhang
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Perlmann
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Karima Chergui
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
3
|
Multi-timescale analysis of midbrain dopamine neuronal firing activities. J Theor Biol 2023; 556:111310. [PMID: 36279959 DOI: 10.1016/j.jtbi.2022.111310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/21/2022] [Accepted: 10/10/2022] [Indexed: 11/05/2022]
Abstract
Midbrain dopamine (DA) neurons exhibit spiking and bursting patterns under physiological conditions. Based on the data on electrophysiological recordings, Yu et al. developed a 13-dimensional mathematical model to capture the detailed characteristics of the DA neuronal firing activities. We use the fitting method to simplify the original model into a 4-dimensional model. Then, the spiking-to-bursting transition is detected from a simple and robust mathematical condition. Physiologically, this condition is a balance of the restorative and the regenerative ion channels at resting potential. Geometrically, this condition imposes a transcritical bifurcation. Moreover, we combine singularity theory and singular perturbation methods to capture the geometry of three-timescale firing attractors in a universal unfolding of a cusp singularity. In particular, the planar description of the corresponding firing patterns can generate the corresponding firing attractors. This analysis provides a new idea for understanding the firing activities of the DA neuron and the specific mechanisms for the switching and dynamic regulation among different patterns.
Collapse
|
4
|
Birtele M, Storm P, Sharma Y, Kajtez J, Wahlestedt JN, Sozzi E, Nilsson F, Stott S, He XL, Mattsson B, Ottosson DR, Barker RA, Fiorenzano A, Parmar M. Single-cell transcriptional and functional analysis of dopaminergic neurons in organoid-like cultures derived from human fetal midbrain. Development 2022; 149:285890. [PMID: 36305490 PMCID: PMC10114107 DOI: 10.1242/dev.200504] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 09/12/2022] [Indexed: 11/06/2022]
Abstract
Significant efforts are ongoing to develop refined differentiation protocols to generate midbrain dopamine (DA) neurons from pluripotent stem cells for application in disease modeling, diagnostics, drug screening and cell-based therapies for Parkinson's disease. An increased understanding of the timing and molecular mechanisms that promote the generation of distinct subtypes of human midbrain DA during development will be essential for guiding future efforts to generate molecularly defined and subtype-specific DA neurons from pluripotent stem cells. Here, we use droplet-based single-cell RNA sequencing to transcriptionally profile the developing human ventral midbrain (VM) when the DA neurons are generated (6-11 weeks post-conception) and their subsequent differentiation into functional mature DA neurons in primary fetal 3D organoid-like cultures. This approach reveals that 3D cultures are superior to monolayer conditions for their ability to generate and maintain mature DA neurons; hence, they have the potential to be used for studying human VM development. These results provide a unique transcriptional profile of the developing human fetal VM and functionally mature human DA neurons that can be used to guide stem cell-based therapies and disease modeling approaches in Parkinson's disease.
Collapse
Affiliation(s)
- Marcella Birtele
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, and Lund Stem Cell Centre, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| | - Petter Storm
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, and Lund Stem Cell Centre, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| | - Yogita Sharma
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, and Lund Stem Cell Centre, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| | - Janko Kajtez
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, and Lund Stem Cell Centre, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| | - Jenny Nelander Wahlestedt
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, and Lund Stem Cell Centre, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| | - Edoardo Sozzi
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, and Lund Stem Cell Centre, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| | - Fredrik Nilsson
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, and Lund Stem Cell Centre, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| | - Simon Stott
- Department of Clinical Neuroscience and Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0PY, UK
| | - Xiaoling L He
- Department of Clinical Neuroscience and Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0PY, UK
| | - Bengt Mattsson
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, and Lund Stem Cell Centre, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| | - Daniella Rylander Ottosson
- Regenerative Neurophysiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| | - Roger A Barker
- Department of Clinical Neuroscience and Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0PY, UK
| | - Alessandro Fiorenzano
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, and Lund Stem Cell Centre, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| | - Malin Parmar
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, and Lund Stem Cell Centre, Department of Experimental Medical Science, Lund University, Lund 223 62, Sweden
| |
Collapse
|
5
|
Sanchez-Conde FG, Jimenez-Vazquez EN, Auerbach DS, Jones DK. The ERG1 K+ Channel and Its Role in Neuronal Health and Disease. Front Mol Neurosci 2022; 15:890368. [PMID: 35600076 PMCID: PMC9113952 DOI: 10.3389/fnmol.2022.890368] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022] Open
Abstract
The ERG1 potassium channel, encoded by KCNH2, has long been associated with cardiac electrical excitability. Yet, a growing body of work suggests that ERG1 mediates physiology throughout the human body, including the brain. ERG1 is a regulator of neuronal excitability, ERG1 variants are associated with neuronal diseases (e.g., epilepsy and schizophrenia), and ERG1 serves as a potential therapeutic target for neuronal pathophysiology. This review summarizes the current state-of-the-field regarding the ERG1 channel structure and function, ERG1’s relationship to the mammalian brain and highlights key questions that have yet to be answered.
Collapse
Affiliation(s)
| | - Eric N. Jimenez-Vazquez
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - David S. Auerbach
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY, United States
- *Correspondence: David S. Auerbach,
| | - David K. Jones
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
- David K. Jones,
| |
Collapse
|
6
|
Jefri M, Bell S, Peng H, Hettige N, Maussion G, Soubannier V, Wu H, Silveira H, Theroux JF, Moquin L, Zhang X, Aouabed Z, Krishnan J, O'Leary LA, Antonyan L, Zhang Y, McCarty V, Mechawar N, Gratton A, Schuppert A, Durcan TM, Fon EA, Ernst C. Stimulation of L-type calcium channels increases tyrosine hydroxylase and dopamine in ventral midbrain cells induced from somatic cells. Stem Cells Transl Med 2020; 9:697-712. [PMID: 32154672 PMCID: PMC7214648 DOI: 10.1002/sctm.18-0180] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 01/03/2020] [Indexed: 01/29/2023] Open
Abstract
Making high‐quality dopamine (DA)‐producing cells for basic biological or small molecule screening studies is critical for the development of novel therapeutics for disorders of the ventral midbrain. Currently, many ventral midbrain assays have low signal‐to‐noise ratio due to low levels of cellular DA and the rate‐limiting enzyme of DA synthesis, tyrosine hydroxylase (TH), hampering discovery efforts. Using intensively characterized ventral midbrain cells derived from human skin, which demonstrate calcium pacemaking activity and classical electrophysiological properties, we show that an L‐type calcium agonist can significantly increase TH protein levels and DA content and release. Live calcium imaging suggests that it is the immediate influx of calcium occurring simultaneously in all cells that drives this effect. Genome‐wide expression profiling suggests that L‐type calcium channel stimulation has a significant effect on specific genes related to DA synthesis and affects expression of L‐type calcium receptor subunits from the CACNA1 and CACNA2D families. Together, our findings provide an advance in the ability to increase DA and TH levels to improve the accuracy of disease modeling and small molecule screening for disorders of the ventral midbrain, including Parkinson's disease.
Collapse
Affiliation(s)
- Malvin Jefri
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Scott Bell
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Huashan Peng
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Nuwan Hettige
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Gilles Maussion
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Vincent Soubannier
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Hanrong Wu
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Heika Silveira
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Jean-Francois Theroux
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Luc Moquin
- Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Xin Zhang
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Zahia Aouabed
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Jeyashree Krishnan
- Institute for Computational Biomedicine, Aachen University, Aachen, Germany
| | - Liam A O'Leary
- Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | | | - Ying Zhang
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Vincent McCarty
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Naguib Mechawar
- Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Alain Gratton
- Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Andreas Schuppert
- Institute for Computational Biomedicine, Aachen University, Aachen, Germany
| | - Thomas M Durcan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Edward A Fon
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Carl Ernst
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Lee HJ, Choi JS, Choi BH, Hahn SJ. Effects of norquetiapine, the active metabolite of quetiapine, on cloned hERG potassium channels. Neurosci Lett 2017; 664:66-73. [PMID: 29133173 DOI: 10.1016/j.neulet.2017.11.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/06/2017] [Accepted: 11/09/2017] [Indexed: 12/25/2022]
Abstract
Quetiapine is an atypical antipsychotic drug that is widely used for the treatment of schizophrenia. It is mainly metabolized by a cytochrome P450 system in the liver. Norquetiapine is a major active metabolite in humans with a pharmacological profile that differs distinctly from that of quetiapine. We used the whole-cell patch-clamp technique to investigate the effects of norquetiapine on hERG channels that are stably expressed in HEK cells. Quetiapine and norquetiapine inhibited the hERG tail currents at -50mV in a concentration-dependent manner with IC50 values of 8.3 and 10.8μM, respectively, which suggested equal potency. The block of hERG currents by norquetiapine was voltage-dependent with a steep increase over a range of voltages for channel activation. However, at more depolarized potentials where the channels were fully activated, the block by norquetiapine was voltage-independent. The steady-state inactivation curve of the hERG currents was shifted to the hyperpolarizing direction in the presence of norquetiapine. Norquetiapine did not produce a use-dependent block. A fast application of norquetiapine inhibited the hERG current elicited by a 5s depolarizing pulse to +60mV, which fully inactivated the hERG currents, suggesting an inactivated-state block. During a repolarizing pulse wherein the hERG current was slowly deactivated, albeit remaining in an open state, a fast application of norquetiapine rapidly and reversibly inhibited the open state of the hERG current. Our results indicated that quetiapine and norquetiapine had equal potency in inhibiting hERG tail currents. Norquetiapine inhibited the hERG current by preferentially interacting with the open and/or inactivated states of the channels.
Collapse
Affiliation(s)
- Hong Joon Lee
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jin-Sung Choi
- College of Pharmacy, Integrated Research Institute of Pharmaceutical, The Catholic University of Korea, Gyeonggi-do, Republic of Korea
| | - Bok Hee Choi
- Department of Pharmacology, Institute for Medical Science, Chonbuk National University Medical School, Jeonju, Jeonbuk 54097, Republic of Korea
| | - Sang June Hahn
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea.
| |
Collapse
|
8
|
Huang CS, Wang GH, Tai CH, Hu CC, Yang YC. Antiarrhythmics cure brain arrhythmia: The imperativeness of subthalamic ERG K + channels in parkinsonian discharges. SCIENCE ADVANCES 2017; 3:e1602272. [PMID: 28508055 PMCID: PMC5425237 DOI: 10.1126/sciadv.1602272] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 03/15/2017] [Indexed: 06/07/2023]
Abstract
ERG K+ channels have long been known to play a crucial role in shaping cardiac action potentials and, thus, appropriate heart rhythms. The functional role of ERG channels in the central nervous system, however, remains elusive. We demonstrated that ERG channels exist in subthalamic neurons and have similar gating characteristics to those in the heart. ERG channels contribute crucially not only to the setting of membrane potential and, consequently, the firing modes, but also to the configuration of burst discharges and, consequently, the firing frequency and automaticity of the subthalamic neurons. Moreover, modulation of subthalamic discharges via ERG channels effectively modulates locomotor behaviors. ERG channel inhibitors ameliorate parkinsonian symptoms, whereas enhancers render normal animals hypokinetic. Thus, ERG K+ channels could be vital to the regulation of both cardiac and neuronal rhythms and may constitute an important pathophysiological basis and pharmacotherapeutic target for the growing list of neurological disorders related to "brain arrhythmias."
Collapse
Affiliation(s)
- Chen-Syuan Huang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Guan-Hsun Wang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Chun-Hwei Tai
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Chang Hu
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Ya-Chin Yang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Tao-Yuan, Taiwan
| |
Collapse
|
9
|
Cocaine increases dopaminergic neuron and motor activity via midbrain α1 adrenergic signaling. Neuropsychopharmacology 2015; 40:1151-62. [PMID: 25374094 PMCID: PMC4367457 DOI: 10.1038/npp.2014.296] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/07/2014] [Accepted: 10/25/2014] [Indexed: 11/09/2022]
Abstract
Cocaine reinforcement is mediated by increased extracellular dopamine levels in the forebrain. This neurochemical effect was thought to require inhibition of dopamine reuptake, but cocaine is still reinforcing even in the absence of the dopamine transporter. Here, we demonstrate that the rapid elevation in dopamine levels and motor activity elicited by cocaine involves α1 receptor activation within the ventral midbrain. Activation of α1 receptors increases dopaminergic neuron burst firing by decreasing the calcium-activated potassium channel current (SK), as well as elevates dopaminergic neuron pacemaker firing through modulation of both SK and the hyperpolarization-activated cation currents (Ih). Furthermore, we found that cocaine increases both the pacemaker and burst-firing frequency of rat ventral-midbrain dopaminergic neurons through an α1 adrenergic receptor-dependent mechanism within the ventral tegmental area and substantia nigra pars compacta. These results demonstrate the mechanism underlying the critical role of α1 adrenergic receptors in the regulation of dopamine neurotransmission and behavior by cocaine.
Collapse
|
10
|
Yu N, Canavier CC. A Mathematical Model of a Midbrain Dopamine Neuron Identifies Two Slow Variables Likely Responsible for Bursts Evoked by SK Channel Antagonists and Terminated by Depolarization Block. JOURNAL OF MATHEMATICAL NEUROSCIENCE 2015; 5:5. [PMID: 25852980 PMCID: PMC4385104 DOI: 10.1186/s13408-015-0017-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 02/03/2015] [Indexed: 06/04/2023]
Abstract
Midbrain dopamine neurons exhibit a novel type of bursting that we call "inverted square wave bursting" when exposed to Ca(2+)-activated small conductance (SK) K(+) channel blockers in vitro. This type of bursting has three phases: hyperpolarized silence, spiking, and depolarization block. We find that two slow variables are required for this type of bursting, and we show that the three-dimensional bifurcation diagram for inverted square wave bursting is a folded surface with upper (depolarized) and lower (hyperpolarized) branches. The activation of the L-type Ca(2+) channel largely supports the separation between these branches. Spiking is initiated at a saddle node on an invariant circle bifurcation at the folded edge of the lower branch and the trajectory spirals around the unstable fixed points on the upper branch. Spiking is terminated at a supercritical Hopf bifurcation, but the trajectory remains on the upper branch until it hits a saddle node on the upper folded edge and drops to the lower branch. The two slow variables contribute as follows. A second, slow component of sodium channel inactivation is largely responsible for the initiation and termination of spiking. The slow activation of the ether-a-go-go-related (ERG) K(+) current is largely responsible for termination of the depolarized plateau. The mechanisms and slow processes identified herein may contribute to bursting as well as entry into and recovery from the depolarization block to different degrees in different subpopulations of dopamine neurons in vivo.
Collapse
Affiliation(s)
- Na Yu
- />Department of Cell Biology and Anatomy, Louisiana State University School of Medicine, New Orleans, LA 70112 USA
- />Department of Mathematics and Computer Science, Lawrence Technological University, 21000 West 10 Mile Road, Southfield, MI 48075 USA
| | - Carmen C. Canavier
- />Department of Cell Biology and Anatomy, Louisiana State University School of Medicine, New Orleans, LA 70112 USA
| |
Collapse
|
11
|
Mitcheson J, Arcangeli A. The Therapeutic Potential of hERG1 K+ Channels for Treating Cancer and Cardiac Arrhythmias. ION CHANNEL DRUG DISCOVERY 2014. [DOI: 10.1039/9781849735087-00258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
hERG potassium channels present pharmacologists and medicinal chemists with a dilemma. On the one hand hERG is a major reason for drugs being withdrawn from the market because of drug induced long QT syndrome and the associated risk of inducing sudden cardiac death, and yet hERG blockers are still widely used in the clinic to treat cardiac arrhythmias. Moreover, in the last decade overwhelming evidence has been provided that hERG channels are aberrantly expressed in cancer cells and that they contribute to tumour cell proliferation, resistance to apoptosis, and neoangiogenesis. Here we provide an overview of the properties of hERG channels and their role in excitable cells of the heart and nervous system as well as in cancer. We consider the therapeutic potential of hERG, not only with regard to the negative impact due to drug induced long QT syndrome, but also its future potential as a treatment in the fight against cancer.
Collapse
Affiliation(s)
- John Mitcheson
- University of Leicester, Department of Cell Physiology and Pharmacology, Medical Sciences Building University Road Leicester LE1 9HN UK
| | - Annarosa Arcangeli
- Department of Experimental Pathology and Oncology, University of Florence Viale GB Morgagni, 50 50134 Firenze Italy
| |
Collapse
|
12
|
Implications of cellular models of dopamine neurons for schizophrenia. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 123:53-82. [PMID: 24560140 DOI: 10.1016/b978-0-12-397897-4.00011-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Midbrain dopamine neurons are pacemakers in vitro, but in vivo they fire less regularly and occasionally in bursts that can lead to a temporary cessation in firing produced by depolarization block. The therapeutic efficacy of antipsychotic drugs used to treat the positive symptoms of schizophrenia has been attributed to their ability to induce depolarization block within a subpopulation of dopamine neurons. We summarize the results of experiments characterizing the physiological mechanisms underlying the ability of these neurons to enter depolarization block in vitro, and our computational simulations of those experiments. We suggest that the inactivation of voltage-dependent Na(+) channels, and, in particular, the slower component of this inactivation, is critical in controlling entry into depolarization block. In addition, an ether-a-go-related gene (ERG) K(+) current also appears to be involved by delaying entry into and speeding recovery from depolarization block. Since many antipsychotic drugs share the ability to block this current, ERG channels may contribute to the therapeutic effects of these drugs.
Collapse
|
13
|
D'Adamo MC, Catacuzzeno L, Di Giovanni G, Franciolini F, Pessia M. K(+) channelepsy: progress in the neurobiology of potassium channels and epilepsy. Front Cell Neurosci 2013; 7:134. [PMID: 24062639 PMCID: PMC3772396 DOI: 10.3389/fncel.2013.00134] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 08/06/2013] [Indexed: 12/19/2022] Open
Abstract
K(+) channels are important determinants of seizure susceptibility. These membrane proteins, encoded by more than 70 genes, make the largest group of ion channels that fine-tune the electrical activity of neuronal and non-neuronal cells in the brain. Their ubiquity and extremely high genetic and functional diversity, unmatched by any other ion channel type, place K(+) channels as primary targets of genetic variations or perturbations in K(+)-dependent homeostasis, even in the absence of a primary channel defect. It is therefore not surprising that numerous inherited or acquired K(+) channels dysfunctions have been associated with several neurologic syndromes, including epilepsy, which often generate confusion in the classification of the associated diseases. Therefore, we propose to name the K(+) channels defects underlying distinct epilepsies as "K(+) channelepsies," and introduce a new nomenclature (e.g., Kx.y-channelepsy), following the widely used K(+) channel classification, which could be also adopted to easily identify other channelopathies involving Na(+) (e.g., Nav x.y-phenotype), Ca(2+) (e.g., Cav x.y-phenotype), and Cl(-) channels. Furthermore, we discuss novel genetic defects in K(+) channels and associated proteins that underlie distinct epileptic phenotypes in humans, and analyze critically the recent progress in the neurobiology of this disease that has also been provided by investigations on valuable animal models of epilepsy. The abundant and varied lines of evidence discussed here strongly foster assessments for variations in genes encoding for K(+) channels and associated proteins in patients with idiopathic epilepsy, provide new avenues for future investigations, and highlight these proteins as critical pharmacological targets.
Collapse
Key Words
- Potassium channels: [Kv1, Kv2, Kv3, Kv4, Kv8, Kv11(HERG), KCa1.1, Kvβ1, Kvβ2, KChIP LGI1, Kir1-Kir7 (GIRK, KATP)]
- autism–epilepsy
- channelopathies
- temporal lobe epilepsy
Collapse
Affiliation(s)
- Maria Cristina D'Adamo
- Faculty of Medicine, Section of Human Physiology, Department of Internal Medicine, University of Perugia Perugia, Italy ; Istituto Euro Mediterraneo di Scienza e Tecnologia, IEMEST Palermo, Italy
| | | | | | | | | |
Collapse
|
14
|
Ha J, Kuznetsov A. Interaction of NMDA receptor and pacemaking mechanisms in the midbrain dopaminergic neuron. PLoS One 2013; 8:e69984. [PMID: 23894569 PMCID: PMC3716766 DOI: 10.1371/journal.pone.0069984] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 06/14/2013] [Indexed: 11/18/2022] Open
Abstract
Dopamine neurotransmission has been found to play a role in addictive behavior and is altered in psychiatric disorders. Dopaminergic (DA) neurons display two functionally distinct modes of electrophysiological activity: low- and high-frequency firing. A puzzling feature of the DA neuron is the following combination of its responses: N-methyl-D-aspartate receptor (NMDAR) activation evokes high-frequency firing, whereas other tonic excitatory stimuli (α-amino-3-hydroxyl-5-methyl-4-isoxazolepropionate receptor (AMPAR) activation or applied depolarization) block firing instead. We suggest a new computational model that reproduces this combination of responses and explains recent experimental data. Namely, somatic NMDAR stimulation evokes high-frequency firing and is more effective than distal dendritic stimulation. We further reduce the model to a single compartment and analyze the mechanism of the distinct high-frequency response to NMDAR activation vs. other stimuli. Standard nullcline analysis shows that the mechanism is based on a decrease in the amplitude of calcium oscillations. The analysis confirms that the nonlinear voltage dependence provided by the magnesium block of the NMDAR determine its capacity to elevate the firing frequency. We further predict that the moderate slope of the voltage dependence plays the central role in the frequency elevation. Additionally, we suggest a repolarizing current that sustains calcium-independent firing or firing in the absence of calcium-dependent repolarizing currents. We predict that the ether-a-go-go current (ERG), which has been observed in the DA neuron, is the best fit for this critical role. We show that a calcium-dependent and a calcium-independent oscillatory mechanisms form a structure of interlocked negative feedback loops in the DA neuron. The structure connects research of DA neuron firing with circadian biology and determines common minimal models for investigation of robustness of oscillations, which is critical for normal function of both systems.
Collapse
Affiliation(s)
- Joon Ha
- Laboratory of Biological Modeling, The National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institute of Health, Bethesda, Maryland, United States of America
| | - Alexey Kuznetsov
- Department of Mathematical Sciences and Center for Mathematical Biosciences, Indiana University, Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| |
Collapse
|
15
|
Pissadaki EK, Bolam JP. The energy cost of action potential propagation in dopamine neurons: clues to susceptibility in Parkinson's disease. Front Comput Neurosci 2013; 7:13. [PMID: 23515615 PMCID: PMC3600574 DOI: 10.3389/fncom.2013.00013] [Citation(s) in RCA: 231] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 02/20/2013] [Indexed: 11/15/2022] Open
Abstract
Dopamine neurons of the substantia nigra pars compacta (SNc) are uniquely sensitive to degeneration in Parkinson's disease (PD) and its models. Although a variety of molecular characteristics have been proposed to underlie this sensitivity, one possible contributory factor is their massive, unmyelinated axonal arbor that is orders of magnitude larger than other neuronal types. We suggest that this puts them under such a high energy demand that any stressor that perturbs energy production leads to energy demand exceeding supply and subsequent cell death. One prediction of this hypothesis is that those dopamine neurons that are selectively vulnerable in PD will have a higher energy cost than those that are less vulnerable. We show here, through the use of a biology-based computational model of the axons of individual dopamine neurons, that the energy cost of axon potential propagation and recovery of the membrane potential increases with the size and complexity of the axonal arbor according to a power law. Thus SNc dopamine neurons, particularly in humans, whose axons we estimate to give rise to more than 1 million synapses and have a total length exceeding 4 m, are at a distinct disadvantage with respect to energy balance which may be a factor in their selective vulnerability in PD.
Collapse
Affiliation(s)
- Eleftheria K Pissadaki
- Medical Research Council Anatomical Neuropharmacology Unit, Department of Pharmacology and Oxford Parkinson's Disease Centre, University of Oxford Oxford, UK
| | | |
Collapse
|
16
|
Ji H, Tucker KR, Putzier I, Huertas MA, Horn JP, Canavier CC, Levitan ES, Shepard PD. Functional characterization of ether-à-go-go-related gene potassium channels in midbrain dopamine neurons - implications for a role in depolarization block. Eur J Neurosci 2012; 36:2906-16. [PMID: 22780096 PMCID: PMC4042402 DOI: 10.1111/j.1460-9568.2012.08190.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Bursting activity by midbrain dopamine neurons reflects the complex interplay between their intrinsic pacemaker activity and synaptic inputs. Although the precise mechanism responsible for the generation and modulation of bursting in vivo has yet to be established, several ion channels have been implicated in the process. Previous studies with nonselective blockers suggested that ether-à-go-go-related gene (ERG) K(+) channels are functionally significant. Here, electrophysiology with selective chemical and peptide ERG channel blockers (E-4031 and rBeKm-1) and computational methods were used to define the contribution made by ERG channels to the firing properties of midbrain dopamine neurons in vivo and in vitro. Selective ERG channel blockade increased the frequency of spontaneous activity as well as the response to depolarizing current pulses without altering spike frequency adaptation. ERG channel block also accelerated entry into depolarization inactivation during bursts elicited by virtual NMDA receptors generated with the dynamic clamp, and significantly prolonged the duration of the sustained depolarization inactivation that followed pharmacologically evoked bursts. In vivo, somatic ERG blockade was associated with an increase in bursting activity attributed to a reduction in doublet firing. Taken together, these results show that dopamine neuron ERG K(+) channels play a prominent role in limiting excitability and in minimizing depolarization inactivation. As the therapeutic actions of antipsychotic drugs are associated with depolarization inactivation of dopamine neurons and blockade of cardiac ERG channels is a prominent side effect of these drugs, ERG channels in the central nervous system may represent a novel target for antipsychotic drug development.
Collapse
Affiliation(s)
- Huifang Ji
- Department of Psychiatry and the Maryland Psychiatry Research Center, University of Maryland School of Medicine, Baltimore, Maryland 21228
| | - Kristal R. Tucker
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Ilva Putzier
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Marco A. Huertas
- Department of Cell Biology and Anatomy and the Neuroscience Center of Excellence, LSU Health Sciences Center, 2020 Gravier Street, Suite D, New Orleans, LA, 70112, USA
| | - John P. Horn
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Carmen C. Canavier
- Department of Cell Biology and Anatomy and the Neuroscience Center of Excellence, LSU Health Sciences Center, 2020 Gravier Street, Suite D, New Orleans, LA, 70112, USA
| | - Edwin S. Levitan
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Paul D. Shepard
- Department of Psychiatry and the Maryland Psychiatry Research Center, University of Maryland School of Medicine, Baltimore, Maryland 21228
| |
Collapse
|
17
|
Mrejeru A, Wei A, Ramirez JM. Calcium-activated non-selective cation currents are involved in generation of tonic and bursting activity in dopamine neurons of the substantia nigra pars compacta. J Physiol 2011; 589:2497-514. [PMID: 21486760 DOI: 10.1113/jphysiol.2011.206631] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Nigral dopamine neurons are transiently activated by high frequency glutamatergic inputs relaying reward-predicting sensory information. The tonic firing pattern of dopamine cells responds to these inputs with a transient burst of spikes that requires NMDA receptors. Here, we show that NMDA receptor activation further excites the cell by recruiting a calcium-activated non-selective cation current (ICAN) capable of generating a plateau potential. Burst firing in vitro is eliminated after blockade of ICAN with flufenamic acid, 9-phenanthrol, or intracellular BAPTA. ICAN is likely to be mediated by a transient receptor potential (TRP) channel, and RT-PCR was used to confirm expression of TRPM2 and TRPM4mRNA in substantia nigra pars compacta.We propose that ICAN is selectively activated during burst firing to boost NMDA currents and allow plateau potentials. This boost mechanism may render DA cells vulnerable to excitotoxicity.
Collapse
Affiliation(s)
- Ana Mrejeru
- Committee on Neurobiology, University of Chicago, Chicago, IL 60637, USA
| | | | | |
Collapse
|
18
|
Atalar F, Acuner TT, Cine N, Oncu F, Yesilbursa D, Ozbek U, Turkcan S. Two four-marker haplotypes on 7q36.1 region indicate that the potassium channel gene HERG1 (KCNH2, Kv11.1) is related to schizophrenia: a case control study. Behav Brain Funct 2010; 6:27. [PMID: 20507645 PMCID: PMC2890623 DOI: 10.1186/1744-9081-6-27] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2009] [Accepted: 05/28/2010] [Indexed: 11/10/2022] Open
Abstract
Background The pathobiology of schizophrenia is still unclear. Its current treatment mainly depends on antipsychotic drugs. A leading adverse effect of these medications is the acquired long QT syndrome, which results from the blockade of cardiac HERG1 channels (human ether-a-go-go-related gene potassium channels 1) by antipsychotic agents. The HERG1 channel is encoded by HERG1 (KCNH2, Kv11.1) gene and is most highly expressed in heart and brain. Genetic variations in HERG1 predispose to acquired long QT syndrome. We hypothesized that the blockade of HERG1 channels by antipsychotics might also be significant for their therapeutic mode of action, indicating a novel mechanism in the pathogenesis of schizophrenia. Methods We genotyped four single nucleotide polymorphisms (SNPs) in 7q36.1 region (two SNPs, rs1805123 and rs3800779, located on HERG1, and two SNPs, rs885684 and rs956642, at the 3'-downstream intergenic region) and then performed single SNP and haplotype association analyses in 84 patients with schizophrenia and 74 healthy controls after the exclusion of individuals having prolonged or shortened QT interval on electrocardiogram. Results Our analyses revealed that both genotype and allele frequencies of rs3800779 (c.307+585G>T) were significantly different between populations (P = 0.023 and P = 0.018, respectively). We also identified that two previously undescribed four-marker haplotypes which are nearly allelic opposite of each other and located in chr7:150225599-150302147bp position encompassing HERG1 were either overrepresented (A-A-A-T, the at-risk haplotype, P = 0.0007) or underrepresented (C-A-C-G, the protective haplotype, P = 0.005) in patients compared to controls. Conclusions Our results indicate that the potassium channel gene HERG1 is related to schizophrenia. Our findings may also implicate the whole family of HERG channels (HERG1, HERG2 and HERG3) in the pathogenesis of psychosis and its treatment.
Collapse
Affiliation(s)
- Fatmahan Atalar
- Endocrinology Laboratory, Department of Growth, Development and Pediatric Endocrinology, Child Health Institute, Istanbul University, Istanbul, Turkey.
| | | | | | | | | | | | | |
Collapse
|
19
|
Hardman RM, Forsythe ID. Ether-à-go-go-related gene K+ channels contribute to threshold excitability of mouse auditory brainstem neurons. J Physiol 2009; 587:2487-97. [PMID: 19359372 PMCID: PMC2714015 DOI: 10.1113/jphysiol.2009.170548] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2009] [Accepted: 03/31/2009] [Indexed: 12/21/2022] Open
Abstract
The ionic basis of excitability requires identification and characterisation of expressed channels and their specific roles in native neurons. We have exploited principal neurons of the medial nucleus of the trapezoid body (MNTB) as a model system for examining voltage-gated K(+) channels, because of their known function and simple morphology. Here we show that channels of the ether-à-go-go-related gene family (ERG, Kv11; encoded by kcnh) complement Kv1 channels in regulating neuronal excitability around threshold voltages. Using whole-cell patch clamp from brainstem slices, the selective ERG antagonist E-4031 reduced action potential (AP) threshold and increased firing on depolarisation. In P12 mice, under voltage-clamp with elevated [K(+)](o) (20 mm), a slowly deactivating current was blocked by E-4031 or terfenadine (V(0.5,act) = -58.4 +/- 0.9 mV, V(0.5,inact) = -76.1 +/- 3.6 mV). Deactivation followed a double exponential time course (tau(slow) = 113.8 +/- 6.9 ms, tau(fast) = 33.2 +/- 3.8 ms at -110 mV, tau(fast) 46% peak amplitude). In P25 mice, deactivation was best fitted by a single exponential (tau(fast) = 46.8 +/- 5.8 ms at -110 mV). Quantitative RT-PCR showed that ERG1 and ERG3 were the predominant mRNAs and immunohistochemistry showed expression as somatic plasma membrane puncta on principal neurons. We conclude that ERG currents complement Kv1 currents in limiting AP firing at around threshold; ERG may have a particular role during periods of high activity when [K(+)](o) is elevated. These ERG currents suggest a potential link between auditory hyperexcitability and acoustic startle triggering of cardiac events in familial LQT2.
Collapse
|
20
|
Ji H, Hougaard C, Herrik KF, Strøbaek D, Christophersen P, Shepard PD. Tuning the excitability of midbrain dopamine neurons by modulating the Ca2+ sensitivity of SK channels. Eur J Neurosci 2009; 29:1883-95. [PMID: 19473240 DOI: 10.1111/j.1460-9568.2009.06735.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Small conductance Ca(2+) -activated K(+) (SK) channels play a prominent role in modulating the spontaneous activity of dopamine (DA) neurons as well as their response to synaptically-released glutamate. SK channel gating is dependent on Ca(2+) binding to constitutively bound calmodulin, which itself is subject to endogenous and exogenous modulation. In the present study, patch-clamp recording techniques were used to examine the relationship between the apparent Ca(2+) affinity of cloned SK3 channels expressed in cultured human embryonic kidney 293 cells and the excitability of DA neurons in slices from rat substantia nigra using the positive and negative SK channel modulators, 6,7-dichloro-1H-indole-2,3-dione-3-oxime and R-N-(benzimidazol-2-yl)-1,2,3,4-tetrohydro-1-naphtylamine. Increasing the apparent Ca(2+) affinity of SK channels decreased the responsiveness of DA neurons to depolarizing current pulses, enhanced spike frequency adaptation and slowed spontaneous firing, effects attributable to an increase in the amplitude and duration of an apamin-sensitive afterhyperpolarization. In contrast, decreasing the apparent Ca(2+) affinity of SK channels enhanced DA neuronal excitability and changed the firing pattern from a pacemaker to an irregular or bursting discharge. Both the reduction in apparent Ca(2+) affinity and the bursting associated with negative SK channel modulation were gradually surmounted by co-application of the positive SK channel modulator. These results underscore the importance of SK channels in 'tuning' the excitability of DA neurons and demonstrate that gating modulation, in a manner analogous to physiological regulation of SK channels in vivo, represents a means of altering the response of DA neurons to membrane depolarization.
Collapse
Affiliation(s)
- Huifang Ji
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | | | | | | | | | | |
Collapse
|
21
|
Functional properties of human neuronal Kv11 channels. Pflugers Arch 2009; 458:689-700. [DOI: 10.1007/s00424-009-0651-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Accepted: 02/12/2009] [Indexed: 11/25/2022]
|
22
|
Electrophysiological characteristics of dopamine neurons: a 35-year update. JOURNAL OF NEURAL TRANSMISSION. SUPPLEMENTUM 2009:103-19. [PMID: 20411771 DOI: 10.1007/978-3-211-92660-4_8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
This chapter consists of four sections. The first section provides a general description of the electrophysiological characteristics of dopamine (DA) neurons in both the substantia nigra and ventral tegmental area. Emphasis is placed on the differences between DA and neighboring non-DA neurons. The second section discusses the ionic mechanisms underlying the generation of action potential in DA cells. Evidence is provided to suggest that these mechanisms differ not only between DA and non-DA neurons but also between DA cells located in different areas, with different projection sites and at different developmental stages. Some of the differences may play a critical role in the vulnerability of a DA neuron to cell death. The third section describes the firing patterns of DA cells. Data are presented to show that the current "80/160 ms" criteria for burst identification need to be revised and that the burst firing, originally described by Bunney et al., can be described as slow oscillations in firing rate. In the ventral tegmental area, the slow oscillations are, at least partially, derived from the prefrontal cortex and part of prefrontal information is transferred to DA cells indirectly through inhibitory neurons. The final section focuses on the feedback regulation of DA cells. New evidence suggests that DA autoreceptors are coupled to multiple effectors, and both D1 and D2-like receptors are involved in long-loop feedback control of DA neurons. Because of the presence of multiple feedback and nonfeedback pathways, the effect of a drug on a DA neuron can be far more complex than an inhibition or excitation. A better understanding of the intrinsic properties of DA neurons and their regulation by afferent input will, in time, help to point to the way to more effective and safer treatments for disorders including schizophrenia, drug addiction, and Parkinson's disease.
Collapse
|
23
|
Pessia M, Servettini I, Panichi R, Guasti L, Grassi S, Arcangeli A, Wanke E, Pettorossi VE. ERG voltage-gated K+ channels regulate excitability and discharge dynamics of the medial vestibular nucleus neurones. J Physiol 2008; 586:4877-90. [PMID: 18718985 PMCID: PMC2614050 DOI: 10.1113/jphysiol.2008.155762] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2008] [Accepted: 08/14/2008] [Indexed: 11/08/2022] Open
Abstract
The discharge properties of the medial vestibular nucleus neurones (MVNn) critically depend on the activity of several ion channel types. In this study we show, immunohistochemically, that the voltage-gated K(+) channels ERG1A, ERG1B, ERG2 and ERG3 are highly expressed within the vestibular nuclei of P10 and P60 mice. The role played by these channels in the spike-generating mechanisms of the MVNn and in temporal information processing was investigated electrophysiologically from mouse brain slices, in vitro, by analysing the spontaneous discharge and the response to square-, ramp- and sinusoid-like intracellular DC current injections in extracellular and whole-cell patch-clamp studies. We show that more than half of the recorded MVNn were responsive to ERG channel block (WAY-123,398, E4031), displaying an increase in spontaneous activity and discharge irregularity. The response to step and ramp current injection was also modified by ERG block showing a reduction of first spike latency, enhancement of discharge rate and reduction of the slow spike-frequency adaptation process. ERG channels influence the interspike slope without affecting the spike shape. Moreover, in response to sinusoid-like current, ERG channel block caused frequency-dependent gain enhancement and phase-lead shift. Taken together, the data demonstrate that ERG channels control the excitability of MVNn, their discharge regularity and probably their resonance properties.
Collapse
Affiliation(s)
- Mauro Pessia
- Department of Internal Medicine, Section of Human Physiology, University of Perugia, Perugia, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Antipsychotic drugs are thought to exert their therapeutic action by antagonizing dopamine receptors but are also known to produce side effects in the heart by inhibiting cardiac ether-a-go-go-related gene (ERG) K(+) channels. Recently, it has been discovered that the same channels are present in the brain, including midbrain dopamine neurons. ERG channels are most active after the cessation of intense electrical activity, and blockade of these channels prolongs plateau potentials in bursting dopamine neurons. This change in excitability would be expected to alter dopamine release. Therefore, the therapeutic action of antipsychotic drugs may depend on inhibition of both postsynaptic dopamine receptors and presynaptic ERG K(+) channels.
Collapse
Affiliation(s)
- Paul D Shepard
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228, USA.
| | | | | |
Collapse
|
25
|
Segev D, Korngreen A. Kinetics of two voltage-gated K+ conductances in substantia nigra dopaminergic neurons. Brain Res 2007; 1173:27-35. [PMID: 17826751 DOI: 10.1016/j.brainres.2007.08.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2007] [Revised: 08/01/2007] [Accepted: 08/02/2007] [Indexed: 10/23/2022]
Abstract
The substantia nigra (SN) is part of the basal ganglia which is a section in the movement circuit in the brain. Dopaminergic neurons (DA) constitute the bulk of substantia nigra neurons and are related to diseases such as Parkinson's disease. Aiming at describing the mechanism of action potential firing in these cells, the current research examined the biophysical characteristics of voltage-gated K+ conductances in the dopaminergic neurons of the SN. The outside-out configuration of the patch-clamp technique was used to measure from dopaminergic neurons in acute brain slices. Two types of K+ voltage-gated conductances, a fast-inactivating A-type-like K+ conductance (K(fast)) and a slow-inactivating delayed rectifier-like K+ conductance (K(slow)), were isolated in these neurons using kinetic separation protocols. The data suggested that a fast-inactivating conductance was due to 69% of the total voltage-gated K+ conductances, while the remainder of the voltage-gated K+ conductance was due to the activation of a slow-inactivating K+ conductance. The two voltage-gated K+ conductances were analyzed assuming a Hodgkin-Huxley model with two activation and one inactivation gate. The kinetic models obtained from this analysis were used in a numerical simulation of the action potential. This simulation suggested that K(fast) may be involved in the modulation of the height and width of action potentials initiated at different resting membrane potentials while K(slow) may participate in action potential repolarization. This mechanism may indicate that SN dopaminergic neurons may perform analog coding by modulation of action potential shape.
Collapse
Affiliation(s)
- Dekel Segev
- The Mina & Everard Goodman Faculty of Life Sciences and the Susan & Leslie Gonda Multidiciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | | |
Collapse
|
26
|
Canavier CC, Oprisan SA, Callaway JC, Ji H, Shepard PD. Computational model predicts a role for ERG current in repolarizing plateau potentials in dopamine neurons: implications for modulation of neuronal activity. J Neurophysiol 2007; 98:3006-22. [PMID: 17699694 DOI: 10.1152/jn.00422.2007] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Blocking the small-conductance (SK) calcium-activated potassium channel promotes burst firing in dopamine neurons both in vivo and in vitro. In vitro, the bursting is unusual in that spiking persists during the hyperpolarized trough and frequently terminates by depolarization block during the plateau. We focus on the underlying plateau potential oscillation generated in the presence of both apamin and TTX, so that action potentials are not considered. We find that although the plateau potentials are mediated by a voltage-gated Ca(2+) current, they do not depend on the accumulation of cytosolic Ca(2+), then use a computational model to test the hypothesis that the slowly voltage-activated ether-a-go-go-related gene (ERG) potassium current repolarizes the plateaus. The model, which includes a material balance on calcium, is able to reproduce the time course of both membrane potential and somatic calcium concentration, and can also mimic the induction of plateau potentials by the calcium chelator BAPTA. The principle of separation of timescales was used to gain insight into the mechanisms of oscillation and its modulation using nullclines in the phase space. The model predicts that the plateau will be elongated and ultimately result in a persistent depolarization as the ERG current is reduced. This study suggests that the ERG current may play a role in burst termination and the relief of depolarization block in vivo.
Collapse
Affiliation(s)
- Carmen C Canavier
- Neuroscience Center, Louisiana State University Health Sciences Center, New Orleans 70112, USA.
| | | | | | | | | |
Collapse
|
27
|
Rodriguez M, Gonzalez S, Morales I, Sabate M, Gonzalez-Hernandez T, Gonzalez-Mora JL. Nigrostriatal cell firing action on the dopamine transporter. Eur J Neurosci 2007; 25:2755-65. [PMID: 17561841 DOI: 10.1111/j.1460-9568.2007.05510.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The influence of nigrostriatal cell firing on the dopamine transporter (DAT) activity of the rat striatum was studied in vivo with amperometric methods. Data were obtained after preventing dopamine (DA) release with alpha-methyl-L-tyrosine and replenishing extracellular DA with local injections. The DA cell stimulation, which under basal conditions increased extracellular DA, decreased DA after this pre-treatment, suggesting that firing activity facilitates the DA cell uptake of DA under these circumstances (drain response). Cocaine and GBR13069 markedly decreased the drain response, suggesting that it is dependent on DAT activation. Data obtained after haloperidol and apomorphine administration showed that the drain response was facilitated by pre-synaptic DA receptor stimulation but that receptors are not a necessary requirement. Two components in the drain response were observed, one with a short latency and duration that needed high-frequency stimuli, and the other with a long latency and duration that was even induced by low-frequency stimuli. This is the first evidence showing that DAT can be activated by the firing activity in nigrostriatal cells in a direct way and without the participation of pre-synaptic DA receptors.
Collapse
Affiliation(s)
- Manuel Rodriguez
- Laboratory of Neurobiology and Experimental Neurology, Department of Physiology, Faculty of Medicine, University of La Laguna, La Laguna, 38320 Tenerife, Spain.
| | | | | | | | | | | |
Collapse
|
28
|
Abstract
The action potential of the squid giant axon is formed by just two voltage-dependent conductances in the cell membrane, yet mammalian central neurons typically express more than a dozen different types of voltage-dependent ion channels. This rich repertoire of channels allows neurons to encode information by generating action potentials with a wide range of shapes, frequencies and patterns. Recent work offers an increasingly detailed understanding of how the expression of particular channel types underlies the remarkably diverse firing behaviour of various types of neurons.
Collapse
Affiliation(s)
- Bruce P Bean
- Harvard Medical School, Department of Neurobiology, 220 Longwood Avenue, Boston, Massachusetts 02115, USA.
| |
Collapse
|
29
|
Atherton JF, Bevan MD. Ionic mechanisms underlying autonomous action potential generation in the somata and dendrites of GABAergic substantia nigra pars reticulata neurons in vitro. J Neurosci 2006; 25:8272-81. [PMID: 16148235 PMCID: PMC6725542 DOI: 10.1523/jneurosci.1475-05.2005] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Through their repetitive discharge, GABAergic neurons of the substantia nigra pars reticulata (SNr) tonically inhibit the target nuclei of the basal ganglia and the dopamine neurons of the midbrain. As the repetitive firing of SNr neurons persists in vitro, perforated, whole-cell and cell-attached patch-clamp recordings were made from rat brain slices to determine the mechanisms underlying this activity. The spontaneous activity of SNr neurons was not perturbed by the blockade of fast synaptic transmission, demonstrating that it was autonomous in nature. A subthreshold, slowly inactivating, voltage-dependent, tetrodotoxin (TTX)-sensitive Na+ current and a TTX-insensitive inward current that was mediated in part by Na+ were responsible for depolarization to action potential (AP) threshold. An apamin-sensitive spike afterhyperpolarization mediated by small-conductance Ca2+-dependent K+ (SK) channels was critical for the precision of autonomous activity. SK channels were activated, in part, by Ca(2+) flowing throughomega-conotoxin GVIA-sensitive, class 2.2 voltage-dependent Ca2+ channels. Although Cs+/ZD7288 (4-ethylphenylamino-1,2-dimethyl-6-methylaminopyrimidinium chloride)-sensitive hyperpolarization-activated currents were also observed in SNr neurons, they were activated at voltages that were in general more hyperpolarized than those associated with autonomous activity. Simultaneous somatic and dendritic recordings revealed that autonomously generated APs were observed first at the soma before propagating into dendrites up to 120 microm from the somatic recording site. Backpropagation of autonomously generated APs was reliable with no observable incidence of failure. Together, these data suggest that the resting inhibitory output of the basal ganglia relies, in large part, on the intrinsic firing properties of the neurons that convey this signal.
Collapse
Affiliation(s)
- Jeremy F Atherton
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | |
Collapse
|
30
|
Kuznetsov AS, Kopell NJ, Wilson CJ. Transient high-frequency firing in a coupled-oscillator model of the mesencephalic dopaminergic neuron. J Neurophysiol 2005; 95:932-47. [PMID: 16207783 DOI: 10.1152/jn.00691.2004] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Dopaminergic neurons of the midbrain fire spontaneously at rates <10/s and ordinarily will not exceed this range even when driven with somatic current injection. When driven at higher rates, these cells undergo spike failure through depolarization block. During spontaneous bursting of dopaminergic neurons in vivo, bursts related to reward expectation in behaving animals, and bursts generated by dendritic application of N-methyl-d-aspartate (NMDA) agonists, transient firing attains rates well above this range. We suggest a way such high-frequency firing may occur in response to dendritic NMDA receptor activation. We have extended the coupled oscillator model of the dopaminergic neuron, which represents the soma and dendrites as electrically coupled compartments with different natural spiking frequencies, by addition of dendritic AMPA (voltage-independent) or NMDA (voltage-dependent) synaptic conductance. Both soma and dendrites contain a simplified version of the calcium-potassium mechanism known to be the mechanism for slow spontaneous oscillation and background firing in dopaminergic cells. The compartments differ only in diameter, and this difference is responsible for the difference in natural frequencies. We show that because of its voltage dependence, NMDA receptor activation acts to amplify the effect on the soma of the high-frequency oscillation of the dendrites, which is normally too weak to exert a large influence on the overall oscillation frequency of the neuron. During the high-frequency oscillations that result, sodium inactivation in the soma is removed rapidly after each action potential by the hyperpolarizing influence of the dendritic calcium-dependent potassium current, preventing depolarization block of the spike mechanism, and allowing high-frequency spiking.
Collapse
Affiliation(s)
- Alexey S Kuznetsov
- Center for BioDynamics and Mathematics Department, Boston University, Boston, Massachusetts, USA.
| | | | | |
Collapse
|
31
|
Sturm P, Wimmers S, Schwarz JR, Bauer CK. Extracellular potassium effects are conserved within the rat erg K+ channel family. J Physiol 2005; 564:329-45. [PMID: 15705650 PMCID: PMC1464433 DOI: 10.1113/jphysiol.2004.078840] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The biophysical properties of native cardiac erg1 and recombinant HERG1 channels have been shown to be influenced by the extracellular K(+) concentration ([K(+)](o)). The erg1 conductance, for example, increases dramatically with a rise in [K(+)](o). In the brain, where local [K(+)](o) can change considerably with the extent of physiological and pathophysiological neuronal activity, all three erg channel subunits are expressed. We have now investigated and compared the effects of an increase in [K(+)](o) from 2 to 10 mm on the three rat erg channels heterologously expressed in CHO cells. Upon increasing [K(+)](o), the voltage dependence of activation was shifted to more negative potentials for erg1 (DeltaV(0.5) = -4.0 +/- 1.1 mV, n = 28) and erg3 (DeltaV(0.5) = -8.4 +/- 1.2 mV, n = 25), and was almost unchanged for erg2 (DeltaV(0.5) = -2.0 +/- 1.3 mV, n = 6). For all three erg channels, activation kinetics were independent of [K(+)](o), but the slowing of inactivation by increased [K(+)](o) was even more pronounced for erg2 and erg3 than for erg1. In addition, with increased [K(+)](o), all three erg channels exhibited significantly slower time courses of recovery from inactivation and of deactivation. Whole-cell erg-mediated conductance was determined at the end of 4 s depolarizing pulses as well as with 1 s voltage ramps starting from the fully activated state. The rise in [K(+)](o) resulted in increased conductance values for all three erg channels which were more pronounced for erg2 (factor 3-4) than for erg1 (factor 2.5-3) and erg3 (factor 2-2.5). The data demonstrate that most [K(+)](o)-dependent changes in the biophysical properties are well conserved within the erg K(+) channel family, despite gradual differences in the magnitude of the effects.
Collapse
Affiliation(s)
- Patrick Sturm
- Institut für Angewandte Physiologie, Zentrum für Experimentelle Medizin, Universitätsklinikum Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany
| | | | | | | |
Collapse
|
32
|
Hirdes W, Schweizer M, Schuricht KS, Guddat SS, Wulfsen I, Bauer CK, Schwarz JR. Fast erg K+ currents in rat embryonic serotonergic neurones. J Physiol 2005; 564:33-49. [PMID: 15677682 PMCID: PMC1456054 DOI: 10.1113/jphysiol.2004.082123] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Ether-á-go-go-related gene (erg) channels form one subfamily of the ether-á-go-go (EAG) K(+) channels and all three erg channels (erg1-3) are expressed in the brain. In the present study we characterize a fast erg current in neurones in primary culture derived from the median part of rat embryonic rhombencephala (E15-16). The relatively uniform erg current was regularly found in large multipolar serotonergic neurones, and occurred also in other less well characterized neurones. The erg current was blocked by the antiarrhythmic substance E-4031. Single-cell RT-PCR revealed the expression of erg1a, erg1b, erg2 and erg3 mRNA in different combinations in large multipolar neurones. These cells also contained neuronal tryptophan hydroxylase, a key enzyme for serotonin production. To characterize the molecular properties of the channels mediating the native erg current, we compared the voltage and time dependence of activation and deactivation of the neuronal erg current to erg1a, erg1b, erg2 and erg3 currents heterologously expressed in CHO cells. The biophysical properties of the neuronal erg current were well within the range displayed by the different heterologously expressed erg currents. Activation and deactivation kinetics of the neuronal erg current were fast and resembled those of erg3 currents. Our data suggest that the erg channels in rat embryonic rhombencephalon neurones are heteromultimers formed by different erg channel subunits.
Collapse
Affiliation(s)
- Wiebke Hirdes
- Institute of Applied Physiology, University Hospital Hamburg-Eppendorf, University of HamburgMartinistr. 52, D-20246 Hamburg, Germany
| | - Michaela Schweizer
- Center for Molecular Neurobiology, ZMNH, University of HamburgFalkenried 94, D-20251 Hamburg, Germany
| | - Kristina S Schuricht
- Institute of Applied Physiology, University Hospital Hamburg-Eppendorf, University of HamburgMartinistr. 52, D-20246 Hamburg, Germany
| | - Saskia S Guddat
- Institute of Applied Physiology, University Hospital Hamburg-Eppendorf, University of HamburgMartinistr. 52, D-20246 Hamburg, Germany
| | - Iris Wulfsen
- Institute of Applied Physiology, University Hospital Hamburg-Eppendorf, University of HamburgMartinistr. 52, D-20246 Hamburg, Germany
| | - Christiane K Bauer
- Institute of Applied Physiology, University Hospital Hamburg-Eppendorf, University of HamburgMartinistr. 52, D-20246 Hamburg, Germany
| | - Jürgen R Schwarz
- Institute of Applied Physiology, University Hospital Hamburg-Eppendorf, University of HamburgMartinistr. 52, D-20246 Hamburg, Germany
- Corresponding author J. R. Schwarz: Institute of Applied Physiology, University Hospital Hamburg-Eppendorf, Martinistr. 52, D-20246 Hamburg, Germany.
| |
Collapse
|