151
|
Abstract
Supplemental Digital Content is Available in the Text. Pathophysiological mechanisms underlying pain associated with cancer are poorly understood. microRNAs (miRNAs) are a class of noncoding RNAs with emerging functional importance in chronic pain. In a genome-wide screen for miRNAs regulated in dorsal root ganglia (DRG) neurons in a mouse model of bone metastatic pain, we identified miR-34c-5p as a functionally important pronociceptive miRNA. Despite these functional insights and therapeutic potential for miR-34c-5p, its molecular mechanism of action in peripheral sensory neurons remains unknown. Here, we report the identification and validation of key target transcripts of miRNA-34c-5p. In-depth bioinformatics analyses revealed Cav2.3, P2rx6, Oprd1, and Oprm1 as high confidence putative targets for miRNA-34c-5p. Of these, canonical and reciprocal regulation of miR-34c-5p and Cav2.3 was observed in cultured sensory neurons as well as in DRG in vivo in mice with cancer pain. Coexpression of miR-34c-5p and Cav2.3 was observed in peptidergic and nonpeptidergic nociceptors, and luciferase reporter assays confirmed functional binding of miR-34c-5p to the 3′ UTR of Cav2.3 transcripts. Importantly, knocking down the expression of Cav2.3 specifically in DRG neurons led to hypersensitivity in mice. In summary, these results show that Cav2.3 is a novel mechanistic target for a key pronociceptive miRNA, miR-34c-5p, in the context of cancer pain and indicate an antinociceptive role for Cav2.3 in peripheral sensory neurons. The current study facilitates a deeper understanding of molecular mechanisms underlying cancer pain and suggests a potential for novel therapeutic strategies targeting miR-34c-5p and Cav2.3 in cancer pain.
Collapse
|
152
|
Kim CS, Brager DH, Johnston D. Perisomatic changes in h-channels regulate depressive behaviors following chronic unpredictable stress. Mol Psychiatry 2018; 23:892-903. [PMID: 28416809 PMCID: PMC5647208 DOI: 10.1038/mp.2017.28] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 01/10/2017] [Accepted: 01/17/2017] [Indexed: 12/23/2022]
Abstract
Chronic stress can be a precipitating factor in the onset of depression. Lentiviral-mediated knockdown of HCN1 protein expression and reduction of functional Ih produce antidepressant behavior. However, whether h-channels are altered in an animal model of depression is not known. We found that perisomatic HCN1 protein expression and Ih-sensitive physiological measurements were significantly increased in dorsal but not in ventral CA1 region/neurons following chronic unpredictable stress (CUS), a widely accepted model for major depressive disorder. Cell-attached patch clamp recordings confirmed that perisomatic Ih was increased in dorsal CA1 neurons following CUS. Furthermore, when dorsal CA1 Ih was reduced by shRNA-HCN1, the CUS-induced behavioral deficits were prevented. Finally, rats infused in the dorsal CA1 region with thapsigargin, an irreversible inhibitor of the SERCA pump, exhibited anxiogenic-like behaviors and increased Ih, similar to that observed following CUS. Our results suggest that CUS, but not acute stress, leads to an increase in perisomatic Ih in dorsal CA1 neurons and that HCN channels represent a potential target for the treatment of major depressive disorder.
Collapse
Affiliation(s)
- C S Kim
- Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, TX, USA,Neuroscience, Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, 100 East 24th St, Austin, TX 78712-0805, USA. E-mail:
| | - D H Brager
- Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, TX, USA
| | - D Johnston
- Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
153
|
|
154
|
Hummert S, Thon S, Eick T, Schmauder R, Schulz E, Benndorf K. Activation gating in HCN2 channels. PLoS Comput Biol 2018; 14:e1006045. [PMID: 29565972 PMCID: PMC5863937 DOI: 10.1371/journal.pcbi.1006045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/19/2018] [Indexed: 12/12/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-modulated (HCN) channels control electrical rhythmicity in specialized brain and heart cells. We quantitatively analysed voltage-dependent activation of homotetrameric HCN2 channels and its modulation by the second messenger cAMP using global fits of hidden Markovian models to complex experimental data. We show that voltage-dependent activation is essentially governed by two separable voltage-dependent steps followed by voltage-independent opening of the pore. According to this model analysis, the binding of cAMP to the channels exerts multiple effects on the voltage-dependent gating: It stabilizes the open pore, reduces the total gating charge from ~8 to ~5, makes an additional closed state outside the activation pathway accessible and strongly accelerates the ON-gating but not the OFF-gating. Furthermore, the open channel has a much slower computed OFF-gating current than the closed channel, in both the absence and presence of cAMP. Together, these results provide detailed new insight into the voltage- and cAMP-induced activation gating of HCN channels. Hyperpolarization-activated cyclic nucleotide-modulated (HCN) channels are tetrameric voltage-controlled ion channels in the cell membrane of specialized nerve and heart cells. Their main function is to generate a so-called pacemaker current which plays a key role in the generation of electrical rhythmicity. A special messenger molecule, cAMP, synthesized within these cells at sympathetic stimulation, can bind to these channels, thereby enhancing channel opening evoked by voltage. The mechanism of this dual activation is still poorly understood. Here we quantified this duality of activation for HCN2 channels by globally fitting hidden Markovian state models to extensive sets of data. We propose that activation of this tetrameric channel requires for a full description only two voltage-dependent steps that are followed by a voltage-independent opening step of the channel pore. According to this model analysis cAMP exerts multiple effects on channel activation: It notably accelerates the charge movement of the voltage-dependent steps and reduces the number of the involved electrical charges. Furthermore, it introduces an additional closed state and stabilizes the open pore. Together, our results provide new insight into the duality of voltage- and cAMP-induced activation of HCN channels.
Collapse
Affiliation(s)
- Sabine Hummert
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena, Germany
| | - Susanne Thon
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena, Germany
| | - Thomas Eick
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena, Germany
| | - Ralf Schmauder
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena, Germany
| | - Eckhard Schulz
- Fachhochschule Schmalkalden, Fakultät Elektrotechnik, Blechhammer, Schmalkalden, Germany
| | - Klaus Benndorf
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena, Germany
- * E-mail:
| |
Collapse
|
155
|
Riegelhaupt PM, Tibbs GR, Goldstein PA. HCN and K 2P Channels in Anesthetic Mechanisms Research. Methods Enzymol 2018; 602:391-416. [PMID: 29588040 DOI: 10.1016/bs.mie.2018.01.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The ability of a diverse group of agents to produce general anesthesia has long been an area of intense speculation and investigation. Over the past century, we have seen a paradigm shift from proposing that the anesthetized state arises from nonspecific interaction of anesthetics with the lipid membrane to the recognition that the function of distinct, and identifiable, membrane-embedded proteins is dramatically altered in the presence of intravenous and inhaled agents. Among proteinaceous targets, metabotropic and ionotropic receptors garnered much of the attention over the last 30 years, and it is only relatively recently that voltage-gated ion channels have clearly and rigorously been shown to be important molecular targets. In this review, we will consider the experimental issues relevant to two important ion channel anesthetic targets, HCN and K2P.
Collapse
Affiliation(s)
| | - Gareth R Tibbs
- Weill Cornell Medical College, New York, NY, United States
| | | |
Collapse
|
156
|
Zhu M, Idikuda VK, Wang J, Wei F, Kumar V, Shah N, Waite CB, Liu Q, Zhou L. Shank3-deficient thalamocortical neurons show HCN channelopathy and alterations in intrinsic electrical properties. J Physiol 2018; 596:1259-1276. [PMID: 29327340 DOI: 10.1113/jp275147] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 01/04/2018] [Indexed: 01/11/2023] Open
Abstract
KEY POINTS Shank3 increases the HCN channel surface expression in heterologous expression systems. Shank3Δ13-16 deficiency causes significant reduction in HCN2 expression and Ih current amplitude in thalamocortical (TC) neurons. Shank3Δ13-16 - but not Shank3Δ4-9 -deficient TC neurons share changes in basic electrical properties which are comparable to those of HCN2-/- TC neurons. HCN channelopathy may critically mediate events downstream from Shank3 deficiency. ABSTRACT SHANK3 is a scaffolding protein that is highly enriched in excitatory synapses. Mutations in the SHANK3 gene have been linked to neuropsychiatric disorders especially the autism spectrum disorders. SHANK3 deficiency is known to cause impairments in synaptic transmission, but its effects on basic neuronal electrical properties that are more localized to the soma and proximal dendrites remain unclear. Here we confirmed that in heterologous expression systems two different mouse Shank3 isoforms, Shank3A and Shank3C, significantly increase the surface expression of the mouse hyperpolarization-activated, cyclic-nucleotide-gated (HCN) channel. In Shank3Δ13-16 knockout mice, which lack exons 13-16 in the Shank3 gene (both Shank3A and Shank3C are removed) and display a severe behavioural phenotype, the expression of HCN2 is reduced to an undetectable level. The thalamocortical (TC) neurons from the ventrobasal (VB) complex of Shank3Δ13-16 mice demonstrate reduced Ih current amplitude and correspondingly increased input resistance, negatively shifted resting membrane potential, and abnormal spike firing in both tonic and burst modes. Impressively, these changes closely resemble those of HCN2-/- TC neurons but not of the TC neurons from Shank3Δ4-9 mice, which lack exons 4-9 in the Shank3 gene (Shank3C still exists) and demonstrate moderate behavioural phenotypes. Additionally, Shank3 deficiency increases the ratio of excitatory/inhibitory balance in VB neurons but has a limited impact on the electrical properties of connected thalamic reticular (RTN) neurons. These results provide new understanding about the role of HCN channelopathy in mediating detrimental effects downstream from Shank3 deficiency.
Collapse
Affiliation(s)
- Mengye Zhu
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.,Department of Pain Clinic, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Vinay Kumar Idikuda
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Jianbing Wang
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.,Department of Anesthesiology, Jiangxi Cancer Hospital, Nanchang, Jiangxi, China
| | - Fusheng Wei
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.,Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Virang Kumar
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Nikhil Shah
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Christopher B Waite
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Qinglian Liu
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Lei Zhou
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
157
|
He F, Yang Z, Dong X, Fang Z, Liu Q, Hu X, Yi S, Li L. The role of HCN channels in peristaltic dysfunction in human ureteral tuberculosis. Int Urol Nephrol 2018; 50:639-645. [PMID: 29460132 PMCID: PMC5878205 DOI: 10.1007/s11255-018-1816-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 02/02/2018] [Indexed: 11/24/2022]
Abstract
Objective To explore the role of HCN channels in ureteral peristaltic dysfunction by comparing the changes in HCN channel levels between normal and tuberculous ureters. Methods A total of 32 specimens of human upper ureters were collected by nephrectomy from patients with renal tumor (control group, n = 16) or from patients with renal tuberculosis (experimental group, n = 16); the two groups did not receive radiotherapy, chemotherapy, immunotherapy, or any other special treatment before the surgical procedure. An experimental study on smooth muscle strips of human upper ureters showed variation in contraction amplitude and frequency after adding ZD7288, a specific blocker of HCN channels. The expression of HCN channels in the ureter was confirmed by Western blot (WB) and by confocal analysis of double immunostaining for c-kit and HCN channel proteins. Results Before the addition of ZD7288, the experimental and control groups showed significant differences in the frequency and amplitude of the spontaneous contraction of isolated ureteral smooth muscle strips. After ZD7288 was added, the frequency and amplitude of the contractions of the ureteral smooth muscle strips were significantly lower in both groups. The differences observed before and after ZD7288 treatment in each group were significant (P < 0.001), and the difference in contraction amplitude observed between the two groups before ZD7288 was also significantly different (P < 0.001). By using WB technology, we showed that the expression of HCN channels was present in normal human ureters, with the expression of HCN4 and HCN1 being the highest; the expression of HCN4 and HCN1 in the control and experimental groups were both statistically significant (P < 0.001). HCN4 and HCN1 were expressed in the mucosal and smooth muscle layers of human control ureters and tuberculous ureters, as revealed by a confocal analysis of double immunostaining for c-kit and HCNs proteins; there were significant differences between the two groups (P < 0.001). Conclusion Four HCN channels are expressed in the ureter, mainly HCN4 and HCN1, suggesting that HCN channels are involved in the peristaltic contraction of ureteral ICCs, which may be an important reason for peristaltic dysfunction in ureteric tuberculosis.
Collapse
Affiliation(s)
- Fan He
- Department of Urology, Xinqiao Hospital, the Third Military Medical University, No. 183 Xinqiao Main Street, Shapinba Dist., Chongqing, 400037, People's Republic of China
| | - Zhenxing Yang
- Department of Urology, Xinqiao Hospital, the Third Military Medical University, No. 183 Xinqiao Main Street, Shapinba Dist., Chongqing, 400037, People's Republic of China
| | - Xingyou Dong
- Department of Urology, Xinqiao Hospital, the Third Military Medical University, No. 183 Xinqiao Main Street, Shapinba Dist., Chongqing, 400037, People's Republic of China
| | - Zhenqiang Fang
- Department of Urology, Xinqiao Hospital, the Third Military Medical University, No. 183 Xinqiao Main Street, Shapinba Dist., Chongqing, 400037, People's Republic of China
| | - Qian Liu
- Department of Urology, Xinqiao Hospital, the Third Military Medical University, No. 183 Xinqiao Main Street, Shapinba Dist., Chongqing, 400037, People's Republic of China
| | - Xiaoyan Hu
- Department of Urology, Xinqiao Hospital, the Third Military Medical University, No. 183 Xinqiao Main Street, Shapinba Dist., Chongqing, 400037, People's Republic of China
| | - Shanhong Yi
- Department of Urology, Xinqiao Hospital, the Third Military Medical University, No. 183 Xinqiao Main Street, Shapinba Dist., Chongqing, 400037, People's Republic of China
| | - Longkun Li
- Department of Urology, Xinqiao Hospital, the Third Military Medical University, No. 183 Xinqiao Main Street, Shapinba Dist., Chongqing, 400037, People's Republic of China.
| |
Collapse
|
158
|
Garden DLF, Oostland M, Jelitai M, Rinaldi A, Duguid I, Nolan MF. Inferior Olive HCN1 Channels Coordinate Synaptic Integration and Complex Spike Timing. Cell Rep 2018; 22:1722-1733. [PMID: 29444426 PMCID: PMC5847187 DOI: 10.1016/j.celrep.2018.01.069] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 01/03/2018] [Accepted: 01/22/2018] [Indexed: 02/07/2023] Open
Abstract
Cerebellar climbing-fiber-mediated complex spikes originate from neurons in the inferior olive (IO), are critical for motor coordination, and are central to theories of cerebellar learning. Hyperpolarization-activated cyclic-nucleotide-gated (HCN) channels expressed by IO neurons have been considered as pacemaker currents important for oscillatory and resonant dynamics. Here, we demonstrate that in vitro, network actions of HCN1 channels enable bidirectional glutamatergic synaptic responses, while local actions of HCN1 channels determine the timing and waveform of synaptically driven action potentials. These roles are distinct from, and may complement, proposed pacemaker functions of HCN channels. We find that in behaving animals HCN1 channels reduce variability in the timing of cerebellar complex spikes, which serve as a readout of IO spiking. Our results suggest that spatially distributed actions of HCN1 channels enable the IO to implement network-wide rules for synaptic integration that modulate the timing of cerebellar climbing fiber signals. HCN1 channels in IO neurons control synaptic response and spiking activity Network actions of HCN1 channels enable bidirectional synaptic responses Local actions of HCN1 channels control spike timing and spikelet number In awake mice, HCN1 channels reduce timing variability of cerebellar complex spikes
Collapse
Affiliation(s)
- Derek L F Garden
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Marlies Oostland
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Marta Jelitai
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Arianna Rinaldi
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Ian Duguid
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Matthew F Nolan
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, UK.
| |
Collapse
|
159
|
Stieglitz MS, Fenske S, Hammelmann V, Becirovic E, Schöttle V, Delorme JE, Schöll-Weidinger M, Mader R, Deussing J, Wolfer DP, Seeliger MW, Albrecht U, Wotjak CT, Biel M, Michalakis S, Wahl-Schott C. Disturbed Processing of Contextual Information in HCN3 Channel Deficient Mice. Front Mol Neurosci 2018; 10:436. [PMID: 29375299 PMCID: PMC5767300 DOI: 10.3389/fnmol.2017.00436] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 12/18/2017] [Indexed: 12/31/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated channels (HCNs) in the nervous system are implicated in a variety of neuronal functions including learning and memory, regulation of vigilance states and pain. Dysfunctions or genetic loss of these channels have been shown to cause human diseases such as epilepsy, depression, schizophrenia, and Parkinson's disease. The physiological functions of HCN1 and HCN2 channels in the nervous system have been analyzed using genetic knockout mouse models. By contrast, there are no such genetic studies for HCN3 channels so far. Here, we use a HCN3-deficient (HCN3−/−) mouse line, which has been previously generated in our group to examine the expression and function of this channel in the CNS. Specifically, we investigate the role of HCN3 channels for the regulation of circadian rhythm and for the determination of behavior. Contrary to previous suggestions we find that HCN3−/− mice show normal visual, photic, and non-photic circadian function. In addition, HCN3−/− mice are impaired in processing contextual information, which is characterized by attenuated long-term extinction of contextual fear and increased fear to a neutral context upon repeated exposure.
Collapse
Affiliation(s)
- Marc S Stieglitz
- Center for Integrated Protein Science and Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians University, Munich, Germany
| | - Stefanie Fenske
- Center for Integrated Protein Science and Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians University, Munich, Germany
| | - Verena Hammelmann
- Center for Integrated Protein Science and Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians University, Munich, Germany
| | - Elvir Becirovic
- Center for Integrated Protein Science and Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians University, Munich, Germany
| | - Verena Schöttle
- Center for Integrated Protein Science and Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians University, Munich, Germany
| | - James E Delorme
- Neurobiochemistry of Circadian Rhythms, Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Martha Schöll-Weidinger
- Center for Integrated Protein Science and Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians University, Munich, Germany
| | - Robert Mader
- Center for Integrated Protein Science and Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians University, Munich, Germany
| | - Jan Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - David P Wolfer
- Institute of Anatomy, University of Zurich, Zurich, Switzerland.,Department of Health Sciences and Technology, Institute of Human Movement Sciences and Sport, ETH Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Mathias W Seeliger
- Ocular Neurodegeneration Research Group, Centre for Ophthalmology, Institute for Ophthalmic Research, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Urs Albrecht
- Neurobiochemistry of Circadian Rhythms, Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Carsten T Wotjak
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Martin Biel
- Center for Integrated Protein Science and Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians University, Munich, Germany
| | - Stylianos Michalakis
- Center for Integrated Protein Science and Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians University, Munich, Germany
| | - Christian Wahl-Schott
- Center for Integrated Protein Science and Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians University, Munich, Germany
| |
Collapse
|
160
|
Zhong P, Vickstrom CR, Liu X, Hu Y, Yu L, Yu HG, Liu QS. HCN2 channels in the ventral tegmental area regulate behavioral responses to chronic stress. eLife 2018; 7:32420. [PMID: 29256865 PMCID: PMC5749952 DOI: 10.7554/elife.32420] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 12/18/2017] [Indexed: 12/17/2022] Open
Abstract
Dopamine neurons in the ventral tegmental area (VTA) are powerful regulators of depression-related behavior. Dopamine neuron activity is altered in chronic stress-based models of depression, but the underlying mechanisms remain incompletely understood. Here, we show that mice subject to chronic mild unpredictable stress (CMS) exhibit anxiety- and depressive-like behavior, which was associated with decreased VTA dopamine neuron firing in vivo and ex vivo. Dopamine neuron firing is governed by voltage-gated ion channels, in particular hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. Following CMS, HCN-mediated currents were decreased in nucleus accumbens-projecting VTA dopamine neurons. Furthermore, shRNA-mediated HCN2 knockdown in the VTA was sufficient to recapitulate CMS-induced depressive- and anxiety-like behavior in stress-naïve mice, whereas VTA HCN2 overexpression largely prevented CMS-induced behavioral deficits. Together, these results reveal a critical role for HCN2 in regulating VTA dopamine neuronal activity and depressive-related behaviors.
Collapse
Affiliation(s)
- Peng Zhong
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
| | - Casey R Vickstrom
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
| | - Xiaojie Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
| | - Ying Hu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
| | - Laikang Yu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
| | - Han-Gang Yu
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, United States
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
| |
Collapse
|
161
|
Hou L, Guo Y, Lian B, Wang Y, Li C, Wang G, Li Q, Pang J, Sun H, Sun L. Synaptic Ultrastructure Might Be Involved in HCN 1-Related BDNF mRNA in Withdrawal-Anxiety After Ethanol Dependence. Front Psychiatry 2018; 9:215. [PMID: 29896126 PMCID: PMC5986948 DOI: 10.3389/fpsyt.2018.00215] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 05/07/2018] [Indexed: 12/27/2022] Open
Abstract
Withdrawal from ethanol dependence has been associated with heightened anxiety and reduced expression of Brain-derived neurotropic factor which promotes the synaptic transmission and plasticity of synapses. Hyperpolarization-activated cyclic nucleotide-gated channel 1 regulates expression; however, whether Hyperpolarization-activated cyclic nucleotide-gated channel 1-related Brain-derived neurotropic factor is involved in the synaptic ultrastructure that generates withdrawal-anxiety has been poorly perceived. Sprague-Dawley rats were treated with ethanol 3-9% (v/v) for a period of 21 days. Conditioned place preference and body weight were investigated during ethanol administration. Rats were subjected to behavioral testing and biochemical assessments after ethanol withdrawal, which was induced by abrupt discontinuation of the treatment. The results showed that the ethanol administration induced severe ethanol dependence behaviors, with higher body weight and more time in the ethanol-paired compartment. After withdrawal, rats had a higher total ethanol withdrawal score and explored less. Additionally, increased Hyperpolarization-activated cyclic nucleotide-gated channel 1 protein and gene expression and decreased Brain-derived neurotropic factor protein and gene expression were detected in the Ethanol group. Eventually, there was a negative correlation between the level of Brain-derived neurotropic factor mRNA and Hyperpolarization-activated cyclic nucleotide-gated channel 1 protein. Importantly, the synaptic ultrastructure changed in the Ethanol group, including increased synaptic cleft width and reduction in postsynaptic density thickness or synaptic curvature. The synthesis of the Brain-derived neurotropic factor mRNA could be down-regulated by higher Hyperpolarization-activated cyclic nucleotide-gated channel 1 protein expression. Changes in synaptic ultrastructure may be induced by lower Brain-derived neurotropic factor protein, which could be associated with the withdrawal-anxiety that is experiences after ethanol dependence.
Collapse
Affiliation(s)
- Lanwei Hou
- Department of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Yujuan Guo
- Department of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Bo Lian
- Department of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Yanyu Wang
- Department of Psychology, Weifang Medical University, Weifang, China
| | - Changjiang Li
- Department of Psychology, Weifang Medical University, Weifang, China
| | - Gang Wang
- Laboratory for Cognitive Neuroscience, Weifang Medical University, Weifang, China
| | - Qi Li
- Department of Psychiatry and Centre for Reproduction Growth and Development, University of Hong Kong, Hong Kong, Hong Kong
| | - Jinjing Pang
- Department of Rehabilitation Medicine, Han Ting People's Hospital of Weifang, Weifang, China
| | - Hongwei Sun
- Department of Psychology, Weifang Medical University, Weifang, China
| | - Lin Sun
- Department of Clinical Medicine, Weifang Medical University, Weifang, China.,Department of Psychology, Weifang Medical University, Weifang, China
| |
Collapse
|
162
|
Modulation of Ether-à-Go-Go Related Gene (ERG) Current Governs Intrinsic Persistent Activity in Rodent Neocortical Pyramidal Cells. J Neurosci 2017; 38:423-440. [PMID: 29175952 DOI: 10.1523/jneurosci.1774-17.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 10/28/2017] [Accepted: 11/17/2017] [Indexed: 12/21/2022] Open
Abstract
While cholinergic receptor activation has long been known to dramatically enhance the excitability of cortical neurons, the cellular mechanisms responsible for this effect are not well understood. We used intracellular recordings in rat (both sexes) neocortical brain slices to assess the ionic mechanisms supporting persistent firing modes triggered by depolarizing stimuli following cholinergic receptor activation. We found multiple lines of evidence suggesting that a component of the underlying hyperexcitability associated with persistent firing reflects a reduction in the standing (leak) K+ current mediated by Ether-a-go-go-Related Gene (ERG) channels. Three chemically diverse ERG channel blockers (terfenadine, ErgToxin-1, and E-4031) abolished persistent firing and the underlying increase in input resistance in deep pyramidal cells in temporal and prefrontal association neocortex. Calcium accumulation during triggering stimuli appears to attenuate ERG currents, leading to membrane potential depolarization and increased input resistance, two critical elements generating persistent firing. Our results also suggest that ERG current normally governs cortical neuron responses to depolarizing stimuli by opposing prolonged discharges and by enhancing the poststimulus repolarization. The broad expression of ERG channels and the ability of ERG blocks to abolish persistent firing evoked by both synaptic and intracellular step stimuli suggest that modulation of ERG channels may underlie many forms of persistent activity observed in vivoSIGNIFICANCE STATEMENT Persistent activity, where spiking continues beyond the triggering stimulus, is a common phenomenon observed in many types of neurons. Identifying the mechanism underlying this elementary process of memory is a step forward in understanding higher cognitive function including short-term memory. Our results suggest that a reduction in the currents normally mediated by Ether-a-go-go-Related Gene (ERG) K+ channels contributes to persistent firing in neocortical pyramidal cells. ERG currents have been previously studied primarily in the heart; relatively little is known about ERG function in the brain, although mutations in ERG channels have recently been linked to schizophrenia. The present study is among the first to describe its role in neocortex in relation to biophysical correlates of memory function.
Collapse
|
163
|
Beining M, Mongiat LA, Schwarzacher SW, Cuntz H, Jedlicka P. T2N as a new tool for robust electrophysiological modeling demonstrated for mature and adult-born dentate granule cells. eLife 2017; 6:e26517. [PMID: 29165247 PMCID: PMC5737656 DOI: 10.7554/elife.26517] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 11/21/2017] [Indexed: 12/18/2022] Open
Abstract
Compartmental models are the theoretical tool of choice for understanding single neuron computations. However, many models are incomplete, built ad hoc and require tuning for each novel condition rendering them of limited usability. Here, we present T2N, a powerful interface to control NEURON with Matlab and TREES toolbox, which supports generating models stable over a broad range of reconstructed and synthetic morphologies. We illustrate this for a novel, highly detailed active model of dentate granule cells (GCs) replicating a wide palette of experiments from various labs. By implementing known differences in ion channel composition and morphology, our model reproduces data from mouse or rat, mature or adult-born GCs as well as pharmacological interventions and epileptic conditions. This work sets a new benchmark for detailed compartmental modeling. T2N is suitable for creating robust models useful for large-scale networks that could lead to novel predictions. We discuss possible T2N application in degeneracy studies.
Collapse
Affiliation(s)
- Marcel Beining
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck SocietyFrankfurtGermany
- Frankfurt Institute for Advanced StudiesFrankfurtGermany
- Institute of Clinical Neuroanatomy, Neuroscience CenterGoethe UniversityFrankfurtGermany
- Faculty of BiosciencesGoethe UniversityFrankfurtGermany
| | - Lucas Alberto Mongiat
- Instituto de Investigación en Biodiversidad y MedioambienteUniversidad Nacional del Comahue-CONICETSan Carlos de BarilocheArgentina
| | | | - Hermann Cuntz
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck SocietyFrankfurtGermany
- Frankfurt Institute for Advanced StudiesFrankfurtGermany
| | - Peter Jedlicka
- Institute of Clinical Neuroanatomy, Neuroscience CenterGoethe UniversityFrankfurtGermany
| |
Collapse
|
164
|
Hou G, Zhang ZW. NMDA Receptors Regulate the Development of Neuronal Intrinsic Excitability through Cell-Autonomous Mechanisms. Front Cell Neurosci 2017; 11:353. [PMID: 29163060 PMCID: PMC5674002 DOI: 10.3389/fncel.2017.00353] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 10/24/2017] [Indexed: 01/30/2023] Open
Abstract
Maturation of neuronal and synaptic functions during early life is essential for the development of neuronal circuits and behaviors. In newborns synaptic transmission at excitatory synapses is primarily mediated by N-methyl-D-aspartate receptors (NMDARs), and NMDAR-mediated signaling plays an important role in synaptic maturation. Concomitant with synapse development, the intrinsic properties of neurons undergo dramatic changes during early life. However, little is known about the role of NMDARs in the development of intrinsic excitability. By using mosaic deletion of the obligatory GluN1 subunit of NMDARs in the thalamus of newborn mice, we showed that NMDARs regulate neuronal excitability during postnatal development. Compared with neighboring control neurons, neurons lacking NMDARs exhibit hyperexcitability and this effect is present throughout early life. Morphological analyses show that thalamic neurons without NMDARs have smaller soma size and fewer dendritic branches. Deletion of NMDARs causes a reduction of hyperpolarization-activated cation (HCN) channel function in thalamic neurons, and pharmacologically blocking HCN channels in wild type neurons mimics the effects of GluN1 deletion on intrinsic excitability. Deletion of GluN1 down-regulated mechanistic target of rapamycin (mTOR) signaling in thalamic neurons, and mosaic deletion of mTOR recapitulated the effects of GluN1 deletion. Our results demonstrate that NMDARs regulate intrinsic excitability and morphology of thalamic neurons through cell autonomous mechanisms that implicate mTOR signaling.
Collapse
Affiliation(s)
| | - Zhong-Wei Zhang
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, United States
| |
Collapse
|
165
|
Wagner F, Weiss T, Veh RW. Electrophysiological properties of neurons and synapses in the lateral habenular complex (LHb). Pharmacol Biochem Behav 2017; 162:38-45. [DOI: 10.1016/j.pbb.2017.07.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 06/28/2017] [Accepted: 07/18/2017] [Indexed: 11/26/2022]
|
166
|
Notomi T, Kuno M, Hiyama A, Nozaki T, Ohura K, Ezura Y, Noda M. Role of lysosomal channel protein TPC2 in osteoclast differentiation and bone remodeling under normal and low-magnesium conditions. J Biol Chem 2017; 292:20998-21010. [PMID: 29084844 DOI: 10.1074/jbc.m117.780072] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 09/25/2017] [Indexed: 11/06/2022] Open
Abstract
The bone is the main storage site for Ca2+ and Mg2+ ions in the mammalian body. Although investigations into Ca2+ signaling have progressed rapidly and led to better understanding of bone biology, the Mg2+ signaling pathway and associated molecules remain to be elucidated. Here, we investigated the role of a potential Mg2+ signaling-related lysosomal molecule, two-pore channel subtype 2 (TPC2), in osteoclast differentiation and bone remodeling. Previously, we found that under normal Mg2+ conditions, TPC2 promotes osteoclastogenesis. We observed that under low-Mg2+ conditions, TPC2 inhibited, rather than promoted, the osteoclast differentiation and that the phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2) signaling pathway played a role in the TPC2 activation under low-Mg2+ conditions. Furthermore, PI(3,5)P2 depolarized the membrane potential by increasing the intracellular Na+ levels. To investigate how membrane depolarization affects osteoclast differentiation, we generated a light-sensitive cell line and developed a system for the light-stimulated depolarization of the membrane potential. The light-induced depolarization inhibited the osteoclast differentiation. We then tested the effect of myo-inositol supplementation, which increased the PI(3,5)P2 levels in mice fed a low-Mg2+ diet. The myo-inositol supplementation rescued the low-Mg2+ diet-induced trabecular bone loss, which was accompanied by the inhibition of osteoclastogenesis. These results indicate that low-Mg2+-induced osteoclastogenesis involves changes in the role of TPC2, which are mediated through the PI(3,5)P2 pathway. Our findings also suggest that myo-inositol consumption might provide beneficial effects in Mg2+ deficiency-induced skeletal diseases.
Collapse
Affiliation(s)
- Takuya Notomi
- From the Department of Molecular Pharmacology, Medical Research Institute and .,the Global Center of Excellence Program for Molecular Science for Tooth and Bone Diseases, Tokyo Medical and Dental University, Bunkyo 113-8510, Tokyo, Japan.,the Department of Pharmacology, Osaka Dental University, Hirakata, Osaka 573-1121, Japan
| | - Miyuki Kuno
- the Department of Physiology, Graduate School of Medicine, Osaka City University, Abeno, Osaka 545-8585, Japan, and
| | - Akiko Hiyama
- the Department of Pharmacology, Osaka Dental University, Hirakata, Osaka 573-1121, Japan
| | - Tadashige Nozaki
- the Department of Pharmacology, Osaka Dental University, Hirakata, Osaka 573-1121, Japan
| | - Kiyoshi Ohura
- the Department of Pharmacology, Osaka Dental University, Hirakata, Osaka 573-1121, Japan
| | - Yoichi Ezura
- From the Department of Molecular Pharmacology, Medical Research Institute and
| | - Masaki Noda
- From the Department of Molecular Pharmacology, Medical Research Institute and .,the Global Center of Excellence Program for Molecular Science for Tooth and Bone Diseases, Tokyo Medical and Dental University, Bunkyo 113-8510, Tokyo, Japan.,the Yokohama City Minato Red Cross Hospital, Yokohama, Kanagawa 231-8682, Japan
| |
Collapse
|
167
|
Nakashima N, Nakashima K, Nakayama T, Takaku A, Kanamori R. Dual expression of constitutively active Gα s-protein-coupled receptors differentially establishes the resting activity of the cAMP-gated HCN2 channel in a single compartment. Biochem Biophys Res Commun 2017; 494:76-81. [PMID: 29054409 DOI: 10.1016/j.bbrc.2017.10.082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 10/16/2017] [Indexed: 11/24/2022]
Abstract
The hyperpolarization-activated cyclic nucleotide-gated 2 (HCN2) channel is a major subtype of the HCN channel family expressed in the nervous system that sets the membrane potential, regulates cell excitability and senses changes in the extracellular environment. Neurons express various Gαs-protein-coupled receptors (GPCRs), many of which show ligand-independent constitutive activity. These membrane-bound proteins are expressed in various subcellular compartments of neurons. Therefore, some proportion of HCN2 channels opens in response to the basal cAMP pool size produced by constitutively active GPCRs. Here, we employed an exogenous HEK293 expression system and voltage-clamp patch-clamp recordings to investigate basal HCN2 channel activity in the presence of two GPCRs with diverse basal activities in a single compartment. We utilized the β2-adrenoceptor (β2AR) together with odorant receptors (ORs), as both GPCR families are known to show strong basal activity. Consequently, β2AR alone strongly enhanced the activity of HCN2 channels, and co-expression of ORs further diversified the HCN2 channel activity, which was totally abolished by an adenylate cyclase inhibitor. Thus, we conclude that the dual expression of constitutively active GPCRs establishes the diverse range of the basal cAMP pool size in resting cells through mutual additive or suppressive interactions, even in the absence of external stimulation.
Collapse
Affiliation(s)
- Noriyuki Nakashima
- Division of Integrated Autonomic Function, Department of Physiology, School of Medicine, Kurume University, 67 Asahi-machi, Kurume, Fukuoka 830-0011, Japan; Department of Physiology and Neurobiology, Faculty of Medicine, Kyoto University, Yoshida-Konoe, Sakyo-ku, Kyoto 606-8501, Japan.
| | - Kie Nakashima
- Department of Physiology and Neurobiology, Faculty of Medicine, Kyoto University, Yoshida-Konoe, Sakyo-ku, Kyoto 606-8501, Japan; Laboratory of Developmental Neurobiology, Graduate School of Biostudies, Kyoto University, Yoshida Hon-machi, Kyoto 606-8501, Japan
| | - Takeo Nakayama
- Department of Physiology and Neurobiology, Faculty of Medicine, Kyoto University, Yoshida-Konoe, Sakyo-ku, Kyoto 606-8501, Japan
| | - Akiko Takaku
- Department of Physiology and Neurobiology, Faculty of Medicine, Kyoto University, Yoshida-Konoe, Sakyo-ku, Kyoto 606-8501, Japan; Medical Scientist Training Program, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan
| | - Ryosuke Kanamori
- Department of Physiology and Neurobiology, Faculty of Medicine, Kyoto University, Yoshida-Konoe, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
168
|
Sartiani L, Mannaioni G, Masi A, Novella Romanelli M, Cerbai E. The Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels: from Biophysics to Pharmacology of a Unique Family of Ion Channels. Pharmacol Rev 2017; 69:354-395. [PMID: 28878030 DOI: 10.1124/pr.117.014035] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/07/2017] [Indexed: 12/22/2022] Open
Abstract
Hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels are important members of the voltage-gated pore loop channels family. They show unique features: they open at hyperpolarizing potential, carry a mixed Na/K current, and are regulated by cyclic nucleotides. Four different isoforms have been cloned (HCN1-4) that can assemble to form homo- or heterotetramers, characterized by different biophysical properties. These proteins are widely distributed throughout the body and involved in different physiologic processes, the most important being the generation of spontaneous electrical activity in the heart and the regulation of synaptic transmission in the brain. Their role in heart rate, neuronal pacemaking, dendritic integration, learning and memory, and visual and pain perceptions has been extensively studied; these channels have been found also in some peripheral tissues, where their functions still need to be fully elucidated. Genetic defects and altered expression of HCN channels are linked to several pathologies, which makes these proteins attractive targets for translational research; at the moment only one drug (ivabradine), which specifically blocks the hyperpolarization-activated current, is clinically available. This review discusses current knowledge about HCN channels, starting from their biophysical properties, origin, and developmental features, to (patho)physiologic role in different tissues and pharmacological modulation, ending with their present and future relevance as drug targets.
Collapse
Affiliation(s)
- Laura Sartiani
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| | - Guido Mannaioni
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| | - Alessio Masi
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| | - Maria Novella Romanelli
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| | - Elisabetta Cerbai
- Department of Neurosciences, Psychology, Drug Research, and Child Health, University of Florence, Firenze, Italy
| |
Collapse
|
169
|
Lyman KA, Han Y, Heuermann RJ, Cheng X, Kurz JE, Lyman RE, Van Veldhoven PP, Chetkovich DM. Allostery between two binding sites in the ion channel subunit TRIP8b confers binding specificity to HCN channels. J Biol Chem 2017; 292:17718-17730. [PMID: 28887304 DOI: 10.1074/jbc.m117.802256] [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: 06/15/2017] [Revised: 09/05/2017] [Indexed: 12/20/2022] Open
Abstract
Tetratricopeptide repeat (TPR) domains are ubiquitous structural motifs that mediate protein-protein interactions. For example, the TPR domains in the peroxisomal import receptor PEX5 enable binding to a range of type 1 peroxisomal targeting signal motifs. A homolog of PEX5, tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b), binds to and functions as an auxiliary subunit of hyperpolarization-activated cyclic nucleotide (HCN)-gated channels. Given the similarity between TRIP8b and PEX5, this difference in function raises the question of what mechanism accounts for their binding specificity. In this report, we found that the cyclic nucleotide-binding domain and the C terminus of the HCN channel are critical for conferring specificity to TRIP8b binding. We show that TRIP8b binds the HCN cyclic nucleotide-binding domain through a 37-residue domain and the HCN C terminus through the TPR domains. Using a combination of fluorescence polarization- and co-immunoprecipitation-based assays, we establish that binding at either site increases affinity at the other. Thus, allosteric coupling of the TRIP8b TPR domains both promotes binding to HCN channels and limits binding to type 1 peroxisomal targeting signal substrates. These results raise the possibility that other TPR domains may be similarly influenced by allosteric mechanisms as a general feature of protein-protein interactions.
Collapse
Affiliation(s)
- Kyle A Lyman
- From the Davee Department of Neurology and Clinical Neurosciences and
| | - Ye Han
- From the Davee Department of Neurology and Clinical Neurosciences and
| | | | - Xiangying Cheng
- From the Davee Department of Neurology and Clinical Neurosciences and
| | | | - Reagan E Lyman
- From the Davee Department of Neurology and Clinical Neurosciences and
| | - Paul P Van Veldhoven
- the Laboratory of Lipid Biochemistry and Protein Interactions, Campus Gasthuisberg, KU Leuven, 3000 Leuven, Belgium
| | - Dane M Chetkovich
- From the Davee Department of Neurology and Clinical Neurosciences and .,Physiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611 and
| |
Collapse
|
170
|
Tae HS, Smith KM, Phillips AM, Boyle KA, Li M, Forster IC, Hatch RJ, Richardson R, Hughes DI, Graham BA, Petrou S, Reid CA. Gabapentin Modulates HCN4 Channel Voltage-Dependence. Front Pharmacol 2017; 8:554. [PMID: 28871229 PMCID: PMC5566583 DOI: 10.3389/fphar.2017.00554] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 08/07/2017] [Indexed: 12/18/2022] Open
Abstract
Gabapentin (GBP) is widely used to treat epilepsy and neuropathic pain. There is evidence that GBP can act on hyperpolarization-activated cation (HCN) channel-mediated Ih in brain slice experiments. However, evidence showing that GBP directly modulates HCN channels is lacking. The effect of GBP was tested using two-electrode voltage clamp recordings from human HCN1, HCN2, and HCN4 channels expressed in Xenopus oocytes. Whole-cell recordings were also made from mouse spinal cord slices targeting either parvalbumin positive (PV+) or calretinin positive (CR+) inhibitory neurons. The effect of GBP on Ih was measured in each inhibitory neuron population. HCN4 expression was assessed in the spinal cord using immunohistochemistry. When applied to HCN4 channels, GBP (100 μM) caused a hyperpolarizing shift in the voltage of half activation (V1/2) thereby reducing the currents. Gabapentin had no impact on the V1/2 of HCN1 or HCN2 channels. There was a robust increase in the time to half activation for HCN4 channels with only a small increase noted for HCN1 channels. Gabapentin also caused a hyperpolarizing shift in the V1/2 of Ih measured from HCN4-expressing PV+ inhibitory neurons in the spinal dorsal horn. Gabapentin had minimal effect on Ih recorded from CR+ neurons. Consistent with this, immunohistochemical analysis revealed that the majority of CR+ inhibitory neurons do not express somatic HCN4 channels. In conclusion, GBP reduces HCN4 channel-mediated currents through a hyperpolarized shift in the V1/2. The HCN channel subtype selectivity of GBP provides a unique tool for investigating HCN4 channel function in the central nervous system. The HCN4 channel is a candidate molecular target for the acute analgesic and anticonvulsant actions of GBP.
Collapse
Affiliation(s)
- Han-Shen Tae
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, ParkvilleVIC, Australia
| | - Kelly M Smith
- School of Biomedical Sciences and Pharmacy, University of Newcastle, CallaghanNSW, Australia.,Hunter Medical Research Institute, New Lambton HeightsNSW, Australia
| | - A Marie Phillips
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, ParkvilleVIC, Australia.,School of BioSciences, The University of Melbourne, ParkvilleVIC, Australia
| | - Kieran A Boyle
- Institute of Neuroscience and Psychology, University of GlasgowGlasgow, United Kingdom
| | - Melody Li
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, ParkvilleVIC, Australia
| | - Ian C Forster
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, ParkvilleVIC, Australia
| | - Robert J Hatch
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, ParkvilleVIC, Australia
| | - Robert Richardson
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, ParkvilleVIC, Australia
| | - David I Hughes
- Institute of Neuroscience and Psychology, University of GlasgowGlasgow, United Kingdom
| | - Brett A Graham
- School of Biomedical Sciences and Pharmacy, University of Newcastle, CallaghanNSW, Australia.,Hunter Medical Research Institute, New Lambton HeightsNSW, Australia
| | - Steven Petrou
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, ParkvilleVIC, Australia
| | - Christopher A Reid
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, ParkvilleVIC, Australia
| |
Collapse
|
171
|
Fogerson PM, Huguenard JR. Tapping the Brakes: Cellular and Synaptic Mechanisms that Regulate Thalamic Oscillations. Neuron 2017; 92:687-704. [PMID: 27883901 DOI: 10.1016/j.neuron.2016.10.024] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 10/03/2016] [Accepted: 10/10/2016] [Indexed: 12/26/2022]
Abstract
Thalamic oscillators contribute to both normal rhythms associated with sleep and anesthesia and abnormal, hypersynchronous oscillations that manifest behaviorally as absence seizures. In this review, we highlight new findings that refine thalamic contributions to cortical rhythms and suggest that thalamic oscillators may be subject to both local and global control. We describe endogenous thalamic mechanisms that limit network synchrony and discuss how these protective brakes might be restored to prevent absence seizures. Finally, we describe how intrinsic and circuit-level specializations among thalamocortical loops may determine their involvement in widespread oscillations and render subsets of thalamic nuclei especially vulnerable to pathological synchrony.
Collapse
Affiliation(s)
- P Michelle Fogerson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - John R Huguenard
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
172
|
Nitric Oxide Modulates HCN Channels in Magnocellular Neurons of the Supraoptic Nucleus of Rats by an S-Nitrosylation-Dependent Mechanism. J Neurosci 2017; 36:11320-11330. [PMID: 27807172 DOI: 10.1523/jneurosci.1588-16.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 09/14/2016] [Indexed: 12/19/2022] Open
Abstract
The control of the excitability in magnocellular neurosecretory cells (MNCs) of the supraoptic nucleus has been attributed mainly to synaptic inputs from circunventricular organs. However, nitric oxide (NO), a gaseous messenger produced in this nucleus during isotonic and short-term hypertonic conditions, is an example of a modulator that can act directly on MNCs to modulate their firing rate. NO inhibits the electrical excitability of MNCs, leading to a decrease in the release of vasopressin and oxytocin. Although the effects of NO on MNCs are well established, the mechanism by which this gas produces its effect is, so far, unknown. Because NO acts independently of synaptic inputs, we hypothesized that ion channels present in MNCs are the targets of NO. To investigate this hypothesis, we used the patch-clamp technique in vitro and in situ to measure currents carried by hyperpolarization-activated and nucleotide-gated cation (HCN) channels and establish their role in determining the electrical excitability of MNCs in rats. Our results show that blockade of HCN channels by ZD7288 decreases MNC firing rate with significant consequences on the release of OT and VP, measured by radioimmunoassay. NO induced a significant reduction in HCN currents by binding to cysteine residues and forming S-nitrosothiol complexes. These findings shed new light on the mechanisms that control the electrical excitability of MNCs via the nitrergic system and strengthen the importance of HCN channels in the control of hydroelectrolyte homeostasis. SIGNIFICANCE STATEMENT Cells in our organism live in a liquid environment whose composition and osmolality are maintained within tight limits. Magnocellular neurons (MNCs) of the supra optic nucleus can sense osmolality and control the synthesis and secretion of vasopressin (VP) and oxytocin (OT) by the neurohypophysis. OT and VP act on the kidneys controlling the excretion of water and sodium to maintain homeostasis. Here we combined electrophysiology, molecular biology, and radioimmunoassay to show that the electrical activity of MNCs can be controlled by nitric oxide (NO), a gaseous messenger. NO reacts with cysteine residues (S-nitrosylation) on hyperpolarization-activated and nucleotide-gated cation channels decreasing the firing rate of MNCs and the consequent secretion of VP and OT.
Collapse
|
173
|
Abstract
Major depressive disorder (MDD) is a chronic and potentially life threatening illness that carries a staggering global burden. Characterized by depressed mood, MDD is often difficult to diagnose and treat owing to heterogeneity of syndrome and complex etiology. Contemporary antidepressant treatments are based on improved monoamine-based formulations from serendipitous discoveries made > 60 years ago. Novel antidepressant treatments are necessary, as roughly half of patients using available antidepressants do not see long-term remission of depressive symptoms. Current development of treatment options focuses on generating efficacious antidepressants, identifying depression-related neural substrates, and better understanding the pathophysiological mechanisms of depression. Recent insight into the brain's mesocorticolimbic circuitry from animal models of depression underscores the importance of ionic mechanisms in neuronal homeostasis and dysregulation, and substantial evidence highlights a potential role for ion channels in mediating depression-related excitability changes. In particular, hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are essential regulators of neuronal excitability. In this review, we describe seminal research on HCN channels in the prefrontal cortex and hippocampus in stress and depression-related behaviors, and highlight substantial evidence within the ventral tegmental area supporting the development of novel therapeutics targeting HCN channels in MDD. We argue that methods targeting the activity of reward-related brain areas have significant potential as superior treatments for depression.
Collapse
Affiliation(s)
- Stacy M Ku
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ming-Hu Han
- Department of Pharmacological Sciences and Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
174
|
Hyperpolarization-activated current I h in mouse trigeminal sensory neurons in a transgenic mouse model of familial hemiplegic migraine type-1. Neuroscience 2017; 351:47-64. [DOI: 10.1016/j.neuroscience.2017.03.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 03/15/2017] [Accepted: 03/20/2017] [Indexed: 12/19/2022]
|
175
|
Lee MY, Ha SE, Park C, Park PJ, Fuchs R, Wei L, Jorgensen BG, Redelman D, Ward SM, Sanders KM, Ro S. Transcriptome of interstitial cells of Cajal reveals unique and selective gene signatures. PLoS One 2017; 12:e0176031. [PMID: 28426719 PMCID: PMC5398589 DOI: 10.1371/journal.pone.0176031] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 04/04/2017] [Indexed: 01/18/2023] Open
Abstract
Transcriptome-scale data can reveal essential clues into understanding the underlying molecular mechanisms behind specific cellular functions and biological processes. Transcriptomics is a continually growing field of research utilized in biomarker discovery. The transcriptomic profile of interstitial cells of Cajal (ICC), which serve as slow-wave electrical pacemakers for gastrointestinal (GI) smooth muscle, has yet to be uncovered. Using copGFP-labeled ICC mice and flow cytometry, we isolated ICC populations from the murine small intestine and colon and obtained their transcriptomes. In analyzing the transcriptome, we identified a unique set of ICC-restricted markers including transcription factors, epigenetic enzymes/regulators, growth factors, receptors, protein kinases/phosphatases, and ion channels/transporters. This analysis provides new and unique insights into the cellular and biological functions of ICC in GI physiology. Additionally, we constructed an interactive ICC genome browser (http://med.unr.edu/physio/transcriptome) based on the UCSC genome database. To our knowledge, this is the first online resource that provides a comprehensive library of all known genetic transcripts expressed in primary ICC. Our genome browser offers a new perspective into the alternative expression of genes in ICC and provides a valuable reference for future functional studies.
Collapse
Affiliation(s)
- Moon Young Lee
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
- Department of Physiology, Wonkwang Digestive Disease Research Institute and Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan, Jeollabuk-do, Korea
| | - Se Eun Ha
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Chanjae Park
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Paul J. Park
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Robert Fuchs
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Lai Wei
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Brian G. Jorgensen
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Doug Redelman
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Sean M. Ward
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Kenton M. Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| | - Seungil Ro
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, United States of America
| |
Collapse
|
176
|
Albertson AJ, Bohannon AS, Hablitz JJ. HCN Channel Modulation of Synaptic Integration in GABAergic Interneurons in Malformed Rat Neocortex. Front Cell Neurosci 2017; 11:109. [PMID: 28469560 PMCID: PMC5396479 DOI: 10.3389/fncel.2017.00109] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 03/30/2017] [Indexed: 12/02/2022] Open
Abstract
Cortical malformations are often associated with pharmaco-resistant epilepsy. Alterations in hyperpolarization-activated, cyclic nucleotide-gated, non-specific cation (HCN) channels have been shown to contribute to malformation associated hyperexcitability. We have recently demonstrated that expression of HCN channels and Ih current amplitudes are reduced in layer (L) 5 pyramidal neurons of rats with freeze lesion induced malformations. These changes were associated with an increased EPSP temporal summation. Here, we examine the effects of HCN channel inhibition on synaptic responses in fast spiking, presumptive basket cells and accommodating, presumptive Martinotti, GABAergic interneurons in slices from freeze lesioned animals. In control animals, fast spiking cells showed small sag responses which were reduced by the HCN channel antagonist ZD7288. Fast spiking cells in lesioned animals showed absent or reduced sag responses. The amplitude of single evoked EPSPs in fast spiking cells in the control group was not affected by HCN channel inhibition with ZD7288. EPSP ratios during short stimulus trains at 25 Hz were not significantly different between control and lesion groups. ZD7288 produced an increase in EPSP ratios in the control but not lesion groups. Under voltage clamp conditions, ZD7288 did not affect EPSC ratios. In the control group, accommodating interneurons showed robust sag responses which were significantly reduced by ZD7288. HCN channel inhibition increased EPSP ratios and area in controls but not the lesioned group. The results indicate that HCN channels differentially modulate EPSPs in different classes of GABAergic interneurons and that this control is reduced in malformed rat neocortex.
Collapse
Affiliation(s)
- Asher J Albertson
- Department of Neurobiology, University of Alabama at BirminghamBirmingham, AL, USA
| | - Andrew S Bohannon
- Department of Neurobiology, University of Alabama at BirminghamBirmingham, AL, USA
| | - John J Hablitz
- Department of Neurobiology, University of Alabama at BirminghamBirmingham, AL, USA
| |
Collapse
|
177
|
The Dendrites of CA2 and CA1 Pyramidal Neurons Differentially Regulate Information Flow in the Cortico-Hippocampal Circuit. J Neurosci 2017; 37:3276-3293. [PMID: 28213444 DOI: 10.1523/jneurosci.2219-16.2017] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 12/27/2016] [Accepted: 02/07/2017] [Indexed: 12/29/2022] Open
Abstract
The impact of a given neuronal pathway depends on the number of synapses it makes with its postsynaptic target, the strength of each individual synapse, and the integrative properties of the postsynaptic dendrites. Here we explore the cellular and synaptic mechanisms responsible for the differential excitatory drive from the entorhinal cortical pathway onto mouse CA2 compared with CA1 pyramidal neurons (PNs). Although both types of neurons receive direct input from entorhinal cortex onto their distal dendrites, these inputs produce a 5- to 6-fold larger EPSP at the soma of CA2 compared with CA1 PNs, which is sufficient to drive action potential output from CA2 but not CA1. Experimental and computational approaches reveal that dendritic propagation is more efficient in CA2 than CA1 as a result of differences in dendritic morphology and dendritic expression of the hyperpolarization-activated cation current (Ih). Furthermore, there are three times as many cortical inputs onto CA2 compared with CA1 PN distal dendrites. Using a computational model, we demonstrate that the differences in dendritic properties of CA2 compared with CA1 PNs are necessary to enable the CA2 PNs to generate their characteristically large EPSPs in response to their cortical inputs; in contrast, CA1 dendritic properties limit the size of the EPSPs they generate, even to a similar number of cortical inputs. Thus, the matching of dendritic integrative properties with the density of innervation is crucial for the differential processing of information from the direct cortical inputs by CA2 compared with CA1 PNs.SIGNIFICANCE STATEMENT Recent discoveries have shown that the long-neglected hippocampal CA2 region has distinct synaptic properties and plays a prominent role in social memory and schizophrenia. This study addresses the puzzling finding that the direct entorhinal cortical inputs to hippocampus, which target the very distal pyramidal neuron dendrites, provide an unusually strong excitatory drive at the soma of CA2 pyramidal neurons, with EPSPs that are 5-6 times larger than those in CA1 pyramidal neurons. We here elucidate synaptic and dendritic mechanisms that account quantitatively for the marked difference in EPSP size. Our findings further demonstrate the general importance of fine-tuning the integrative properties of neuronal dendrites to their density of synaptic innervation.
Collapse
|
178
|
Potential synergistic action of 19 schizophrenia risk genes in the thalamus. Schizophr Res 2017; 180:64-69. [PMID: 27645107 PMCID: PMC5263182 DOI: 10.1016/j.schres.2016.09.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 08/31/2016] [Accepted: 09/03/2016] [Indexed: 11/22/2022]
Abstract
A goal of current schizophrenia (SZ) research is to understand how multiple risk genes work together with environmental factors to produce the disease. In schizophrenia, there is elevated delta frequency EEG power in the awake state, an elevation that can be mimicked in rodents by N-methyl-d-aspartate receptor (NMDAR) antagonist action in the thalamus. This thalamic delta can be blocked by dopamine D2 receptor antagonists, agents known to be therapeutic in SZ. Experiments suggest that these oscillations can interfere with brain function and may thus be causal in producing psychosis. Here we evaluate the question of whether well-established schizophrenia risk genes may interact to affect the delta generation process. We identify 19 risk genes that can plausibly work in a synergistic fashion to generate delta oscillations.
Collapse
|
179
|
Guo T, Li J, Li J, Kong D, Bi C, He Z, Tang D, Jin X, Jin L. Association between hyperpolarization-activated channel in interstitial cells of Cajal and gastrointestinal dysmotility induced by malignant ascites. Oncol Lett 2017; 13:1601-1608. [PMID: 28454297 PMCID: PMC5403200 DOI: 10.3892/ol.2017.5652] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 11/03/2016] [Indexed: 12/20/2022] Open
Abstract
Advanced malignant ascites is accompanied by gastrointestinal dysmotility, and patients often feel abdominal pain, abdominal distention, nausea and constipation. Gastrointestinal dysmotility is not only painful for the patients, but it reduces the absorption of nutrients and affects the physical recovery of patients with malignant ascites. It is reported that changes in interstitial cells of Cajal (ICCs) are responsible for the gastrointestinal dysmotility induced by malignant ascites, but the mechanism is not completely understood. The present study observed a significantly decreased expression of ion channels, including hyperpolarization-activated cyclic nucleotide-gated potassium channel 2 (HCN2) and cyclic adenosine monophosphate, in the condition of malignant ascites. Using electrophysiology, it was identified that malignant ascites led to lower amplitude and slower frequency signals in cells of the small intestine. In addition, when ICCs were cultured with malignant ascites in vitro, the expression of HCN2 of ICCs was significantly reduced, and the data of flow cytometry revealed that the Ca2+ concentration of ICCs was also decreased. The results of electron microscopy analysis demonstrated the nuclei of ICCs were pyknotic, and the processes of ICCs were reduced in malignant ascites. The present study suggests the small intestinal dysmotility caused by malignant ascites may be associated with changes in HCN2 of ICCs, which offers a potential therapeutic target for gastrointestinal dysmotility in advanced malignant ascites.
Collapse
Affiliation(s)
- Tieyun Guo
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Jiade Li
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Jing Li
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Dan Kong
- Department of Gynecology, The Third Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Chunli Bi
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Zheng He
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Dai Tang
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xiaoming Jin
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Lianhong Jin
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
180
|
Huang Z, Li G, Aguado C, Lujan R, Shah MM. HCN1 channels reduce the rate of exocytosis from a subset of cortical synaptic terminals. Sci Rep 2017; 7:40257. [PMID: 28071723 PMCID: PMC5223132 DOI: 10.1038/srep40257] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 12/02/2016] [Indexed: 12/18/2022] Open
Abstract
The hyperpolarization-activated cyclic nucleotide-gated (HCN1) channels are predominantly located in pyramidal cell dendrites within the cortex. Recent evidence suggests these channels also exist pre-synaptically in a subset of synaptic terminals within the mature entorhinal cortex (EC). Inhibition of pre-synaptic HCN channels enhances miniature excitatory post-synaptic currents (mEPSCs) onto EC layer III pyramidal neurons, suggesting that these channels decrease the release of the neurotransmitter, glutamate. Thus, do pre-synaptic HCN channels alter the rate of synaptic vesicle exocytosis and thereby enhance neurotransmitter release? To address this, we imaged the release of FM1-43, a dye that is incorporated into synaptic vesicles, from EC synaptic terminals using two photon microscopy in slices obtained from forebrain specific HCN1 deficient mice, global HCN1 knockouts and their wildtype littermates. This coupled with electrophysiology and pharmacology showed that HCN1 channels restrict the rate of exocytosis from a subset of cortical synaptic terminals within the EC and in this way, constrain non-action potential-dependent and action potential-dependent spontaneous release as well as synchronous, evoked release. Since HCN1 channels also affect post-synaptic potential kinetics and integration, our results indicate that there are diverse ways by which HCN1 channels influence synaptic strength and plasticity.
Collapse
Affiliation(s)
- Zhuo Huang
- UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Gengyu Li
- UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Carolina Aguado
- Departamento de Ciencias Medicas, Universidad de Castilla-La Mancha, 02006 Albacete, Spain
| | - Rafael Lujan
- Departamento de Ciencias Medicas, Universidad de Castilla-La Mancha, 02006 Albacete, Spain
| | - Mala M Shah
- UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
| |
Collapse
|
181
|
Cao XJ, Oertel D. Genetic perturbations suggest a role of the resting potential in regulating the expression of the ion channels of the KCNA and HCN families in octopus cells of the ventral cochlear nucleus. Hear Res 2017; 345:57-68. [PMID: 28065805 DOI: 10.1016/j.heares.2017.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 01/02/2017] [Accepted: 01/03/2017] [Indexed: 10/20/2022]
Abstract
Low-voltage-activated K+ (gKL) and hyperpolarization-activated mixed cation conductances (gh) mediate currents, IKL and Ih, through channels of the Kv1 (KCNA) and HCN families respectively and give auditory neurons the temporal precision required for signaling information about the onset, fine structure, and time of arrival of sounds. Being partially activated at rest, gKL and gh contribute to the resting potential and shape responses to even small subthreshold synaptic currents. Resting gKL and gh also affect the coupling of somatic depolarization with the generation of action potentials. To learn how these important conductances are regulated we have investigated how genetic perturbations affect their expression in octopus cells of the ventral cochlear nucleus (VCN). We report five new findings: First, the magnitude of gh and gKL varied over more than two-fold between wild type strains of mice. Second, average resting potentials are not different in different strains of mice even in the face of large differences in average gKL and gh. Third, IKL has two components, one being α-dendrotoxin (α-DTX)-sensitive and partially inactivating and the other being α-DTX-insensitive, tetraethylammonium (TEA)-sensitive, and non-inactivating. Fourth, the loss of Kv1.1 results in diminution of the α-DTX-sensitive IKL, and compensatory increased expression of an α-DTX-insensitive, tetraethylammonium (TEA)-sensitive IKL. Fifth, Ih and IKL are balanced at the resting potential in all wild type and mutant octopus cells even when resting potentials vary in individual cells over nearly 10 mV, indicating that the resting potential influences the expression of gh and gKL. The independence of resting potentials on gKL and gh shows that gKL and gh do not, over days or weeks, determine the resting potential but rather that the resting potential plays a role in regulating the magnitude of either or both gKL and gh.
Collapse
Affiliation(s)
- Xiao-Jie Cao
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA
| | - Donata Oertel
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA.
| |
Collapse
|
182
|
Zhang K, Xu T, Yuan Z, Wei Z, Yamaki VN, Huang M, Huganir RL, Cai X. Essential roles of AMPA receptor GluA1 phosphorylation and presynaptic HCN channels in fast-acting antidepressant responses of ketamine. Sci Signal 2016; 9:ra123. [PMID: 27965425 DOI: 10.1126/scisignal.aai7884] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Although the molecular mechanism is not clear, the clinically tested drug ketamine has rapid antidepressant action that does not require the multiple weeks of treatment needed for other antidepressant drugs to have an effect. We showed that ketamine potentiated Schaffer collateral-CA1 cell excitatory synaptic transmission in hippocampal slice preparations from rodents and enhanced the phosphorylation of the GluA1 subunit on Ser845 of the AMPA-type glutamate receptor in the hippocampal area CA1. These effects persisted when γ-aminobutyric acid (GABA) receptors were pharmacologically blocked. Ketamine reduced behavioral despair in wild-type mice but had no effect in GluA1 S845A knock-in mutant mice. Presynaptic (CA3 pyramidal cell), but not postsynaptic (CA1 pyramidal cell), deletion of N-methyl-d-aspartate (NMDA)-type glutamate receptors eliminated the ketamine-induced enhancement of excitatory synaptic transmission in hippocampal slices and the antidepressant actions of ketamine in mice. The synaptic and behavioral actions of ketamine were completely occluded by inhibition or deletion of the hyperpolarization-activated cyclic nucleotide-gated channel 1 (HCN1). Our results implicate presynaptic NMDA receptor inhibition followed by reduced activity of presynaptic HCN1 channels, which would result in an increase in glutamate release and postsynaptic glutamate receptor activity, as a mechanism of ketamine action. These data provide a mechanism for changes in synaptic activity that could explain the fast-acting antidepressant effects of this drug.
Collapse
Affiliation(s)
- Ke Zhang
- Department of Physiology, Southern Illinois University School of Medicine, 1135 Lincoln Drive, Carbondale, IL 62901, USA
| | - Ting Xu
- Department of Physiology, Southern Illinois University School of Medicine, 1135 Lincoln Drive, Carbondale, IL 62901, USA.,The Institute of Neuroscience, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang Road, Guangzhou, Guangdong 51030, China
| | - Zhongmin Yuan
- Department of Physiology, Southern Illinois University School of Medicine, 1135 Lincoln Drive, Carbondale, IL 62901, USA
| | - Zhisheng Wei
- The Institute of Neuroscience, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang Road, Guangzhou, Guangdong 51030, China
| | - Vitor Nagai Yamaki
- Department of Physiology, Southern Illinois University School of Medicine, 1135 Lincoln Drive, Carbondale, IL 62901, USA
| | - Mingfa Huang
- The Institute of Neuroscience, the Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang Road, Guangzhou, Guangdong 51030, China
| | - Richard L Huganir
- The Solomon H. Snyder Department of Neuroscience, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Xiang Cai
- Department of Physiology, Southern Illinois University School of Medicine, 1135 Lincoln Drive, Carbondale, IL 62901, USA.
| |
Collapse
|
183
|
Boychuk JA, Farrell JS, Palmer LA, Singleton AC, Pittman QJ, Teskey GC. HCN channels segregate stimulation-evoked movement responses in neocortex and allow for coordinated forelimb movements in rodents. J Physiol 2016; 595:247-263. [PMID: 27568501 DOI: 10.1113/jp273068] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 08/17/2016] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS The present study tested whether HCN channels contribute to the organization of motor cortex and to skilled motor behaviour during a forelimb reaching task. Experimental reductions in HCN channel signalling increase the representation of complex multiple forelimb movements in motor cortex as assessed by intracortical microstimulation. Global HCN1KO mice exhibit reduced reaching accuracy and atypical movements during a single-pellet reaching task relative to wild-type controls. Acute pharmacological inhibition of HCN channels in forelimb motor cortex decreases reaching accuracy and increases atypical movements during forelimb reaching. ABSTRACT The mechanisms by which distinct movements of a forelimb are generated from the same area of motor cortex have remained elusive. Here we examined a role for HCN channels, given their ability to alter synaptic integration, in the expression of forelimb movement responses during intracortical microstimulation (ICMS) and movements of the forelimb on a skilled reaching task. We used short-duration high-resolution ICMS to evoke forelimb movements following pharmacological (ZD7288), experimental (electrically induced cortical seizures) or genetic approaches that we confirmed with whole-cell patch clamp to substantially reduce Ih current. We observed significant increases in the number of multiple movement responses evoked at single sites in motor maps to all three experimental manipulations in rats or mice. Global HCN1 knockout mice were less successful and exhibited atypical movements on a skilled-motor learning task relative to wild-type controls. Furthermore, in reaching-proficient rats, reaching accuracy was reduced and forelimb movements were altered during infusion of ZD7288 within motor cortex. Thus, HCN channels play a critical role in the separation of overlapping movement responses and allow for successful reaching behaviours. These data provide a novel mechanism for the encoding of multiple movement responses within shared networks of motor cortex. This mechanism supports a viewpoint of primary motor cortex as a site of dynamic integration for behavioural output.
Collapse
Affiliation(s)
- Jeffery A Boychuk
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA.,Epilepsy Center, University of Kentucky, Lexington, Kentucky, USA.,Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada.,Department of Neuroscience, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Jordan S Farrell
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada.,Department of Neuroscience, University of Calgary, Calgary, Alberta, Canada
| | - Laura A Palmer
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada.,Department of Neuroscience, University of Calgary, Calgary, Alberta, Canada
| | - Anna C Singleton
- Department of Neuroscience, University of Calgary, Calgary, Alberta, Canada
| | - Quentin J Pittman
- Department of Neuroscience, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, Calgary, Alberta, Canada
| | - G Campbell Teskey
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada.,Department of Neuroscience, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, Calgary, Alberta, Canada
| |
Collapse
|
184
|
Thon S, Schulz E, Kusch J, Benndorf K. Conformational Flip of Nonactivated HCN2 Channel Subunits Evoked by Cyclic Nucleotides. Biophys J 2016; 109:2268-76. [PMID: 26636938 PMCID: PMC4675818 DOI: 10.1016/j.bpj.2015.08.054] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 08/21/2015] [Accepted: 08/24/2015] [Indexed: 12/11/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-modulated (HCN) channels are tetrameric proteins that evoke electrical rhythmicity in specialized neurons and cardiomyocytes. The channels are activated by hyperpolarizing voltage but are also receptors for the intracellular ligand adenosine-3',5'-cyclic monophosphate (cAMP) that enhances activation but is unable to activate the channels alone. Using fcAMP, a fluorescent derivative of cAMP, we analyzed the effect of ligand binding on HCN2 channels not preactivated by voltage. We identified a conformational flip of the channel as an intermediate state following the ligand binding and quantified it kinetically. Globally fitting the time courses of ligand binding and unbinding revealed modest cooperativity among the subunits in the conformational flip. The intensity of this cooperativity, however, was only moderate compared to channels preactivated by hyperpolarizing voltage. These data provide kinetic information about conformational changes proceeding in nonactivated HCN2 channels when cAMP binds. Moreover, our approach bears potential for analyzing the function of any other membrane receptor if a potent fluorescent ligand is available.
Collapse
Affiliation(s)
- Susanne Thon
- Institut für Physiologie II, Universitätsklinikum Jena, Jena, Germany
| | - Eckhard Schulz
- Fachhochschule Schmalkalden, Fakultät Elektrotechnik, Blechhammer, Schmalkalden, Germany
| | - Jana Kusch
- Institut für Physiologie II, Universitätsklinikum Jena, Jena, Germany
| | - Klaus Benndorf
- Institut für Physiologie II, Universitätsklinikum Jena, Jena, Germany.
| |
Collapse
|
185
|
HCN2 ion channels: basic science opens up possibilities for therapeutic intervention in neuropathic pain. Biochem J 2016; 473:2717-36. [DOI: 10.1042/bcj20160287] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/18/2016] [Indexed: 01/22/2023]
Abstract
Nociception — the ability to detect painful stimuli — is an invaluable sense that warns against present or imminent damage. In patients with chronic pain, however, this warning signal persists in the absence of any genuine threat and affects all aspects of everyday life. Neuropathic pain, a form of chronic pain caused by damage to sensory nerves themselves, is dishearteningly refractory to drugs that may work in other types of pain and is a major unmet medical need begging for novel analgesics. Hyperpolarisation-activated cyclic nucleotide (HCN)-modulated ion channels are best known for their fundamental pacemaker role in the heart; here, we review data demonstrating that the HCN2 isoform acts in an analogous way as a ‘pacemaker for pain’, in that its activity in nociceptive neurons is critical for the maintenance of electrical activity and for the sensation of chronic pain in pathological pain states. Pharmacological block or genetic deletion of HCN2 in sensory neurons provides robust pain relief in a variety of animal models of inflammatory and neuropathic pain, without any effect on normal sensation of acute pain. We discuss the implications of these findings for our understanding of neuropathic pain pathogenesis, and we outline possible future opportunities for the development of efficacious and safe pharmacotherapies in a range of chronic pain syndromes.
Collapse
|
186
|
Hu R, Ferguson KA, Whiteus CB, Meijer DH, Araneda RC. Hyperpolarization-Activated Currents and Subthreshold Resonance in Granule Cells of the Olfactory Bulb. eNeuro 2016; 3:ENEURO.0197-16.2016. [PMID: 27844056 PMCID: PMC5095762 DOI: 10.1523/eneuro.0197-16.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 10/19/2016] [Accepted: 10/24/2016] [Indexed: 12/22/2022] Open
Abstract
An important contribution to neural circuit oscillatory dynamics is the ongoing activation and inactivation of hyperpolarization-activated currents (Ih). Network synchrony dynamics play an important role in the initial processing of odor signals by the main olfactory bulb (MOB) and accessory olfactory bulb (AOB). In the mouse olfactory bulb, we show that Ih is present in granule cells (GCs), the most prominent inhibitory neuron in the olfactory bulb, and that Ih underlies subthreshold resonance in GCs. In accord with the properties of Ih, the currents exhibited sensitivity to changes in extracellular K+ concentration and ZD7288 (4-ethylphenylamino-1,2-dimethyl-6-methylaminopyrimidin chloride), a blocker of Ih. ZD7288 also caused GCs to hyperpolarize and increase their input resistance, suggesting that Ih is active at rest in GCs. The inclusion of cAMP in the intracellular solution shifted the activation of Ih to less negative potentials in the MOB, but not in the AOB, suggesting that channels with different subunit composition mediate Ih in these regions. Furthermore, we show that mature GCs exhibit Ih-dependent subthreshold resonance in the theta frequency range (4-12 Hz). Another inhibitory subtype in the MOB, the periglomerular cells, exhibited Ih-dependent subthreshold resonance in the delta range (1-4 Hz), while principal neurons, the mitral cells, do not exhibit Ih-dependent subthreshold resonance. Importantly, Ih size, as well as the strength and frequency of resonance in GCs, exhibited a postnatal developmental progression, suggesting that this development of Ih in GCs may differentially contribute to their integration of sensory input and contribution to oscillatory circuit dynamics.
Collapse
Affiliation(s)
- Ruilong Hu
- Department of Biology, University of Maryland, College Park, Maryland 20742
| | - Katie A. Ferguson
- Neurobiology Course, Marine Biology Laboratory, Woods Hole, Massachusetts 02543
| | | | - Dimphna H. Meijer
- Neurobiology Course, Marine Biology Laboratory, Woods Hole, Massachusetts 02543
| | - Ricardo C. Araneda
- Department of Biology, University of Maryland, College Park, Maryland 20742
| |
Collapse
|
187
|
Zúñiga R, González D, Valenzuela C, Brown N, Zúñiga L. Expression and cellular localization of HCN channels in rat cerebellar granule neurons. Biochem Biophys Res Commun 2016; 478:1429-35. [PMID: 27569278 DOI: 10.1016/j.bbrc.2016.08.141] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 08/24/2016] [Indexed: 11/17/2022]
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels belong to the superfamily of voltage-gated pore loop channels. In mammals, this family consists of four different subunits (HCN1-4) and their ion channels activity have been proposed to play an essential role in regulating the membrane potential of excitable cells. Here, we describe the expression and relative abundances of HCN channels in cerebellum and primary cultures of cerebellar granule neurons (CGN). Quantitative determination of mRNA expression levels demonstrated the existence of an accumulation pattern of transcripts in cerebellum that encode HCN2 > HCN3 = HCN4 > HCN1 subunits. Immunolocalization analyses of HCN channels in cerebella revealed positive staining in Purkinje and granule cell layers. The presence of the HCN subunits in the cerebellar granule cell layer was then confirmed in primary cultures of CGN by quantitative real-time PCR (qPCR), as well as western blot and immunofluorescence analysis, demonstrating the presence of all four channel proteins.
Collapse
Affiliation(s)
- Rafael Zúñiga
- Centro de Investigaciones Médicas CIM, Escuela de Medicina, Universidad de Talca, Talca, Chile
| | - Daniela González
- Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Claudio Valenzuela
- Centro de Investigaciones Médicas CIM, Escuela de Medicina, Universidad de Talca, Talca, Chile
| | - Nelson Brown
- Centro de Investigaciones Médicas CIM, Escuela de Medicina, Universidad de Talca, Talca, Chile
| | - Leandro Zúñiga
- Centro de Investigaciones Médicas CIM, Escuela de Medicina, Universidad de Talca, Talca, Chile.
| |
Collapse
|
188
|
Ding W, You Z, Shen S, Chen L, Zhu S, Mao J. Inhibition of HCN channel activity in the thalamus attenuates chronic pain in rats. Neurosci Lett 2016; 631:97-103. [PMID: 27542339 DOI: 10.1016/j.neulet.2016.08.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/12/2016] [Accepted: 08/13/2016] [Indexed: 10/21/2022]
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels regulate neuronal excitability in both peripheral and central nerve systems. Emerging evidence indicates that HCN channels are involved in the development and maintenance of chronic pain. However, the impact of HCN channel activity in the thalamus on chronic pain has not been examined. In this report, we evaluated the effect on nociceptive behaviors after infusion of a HCN channel blocker ZD7288 into the ventral posterolateral (VPL) nucleus of the thalamus in rats with neuropathic pain or monoarthritis. We show that ZD7288 dose-dependently attenuated mechanical allodynia and thermal hyperalgesia in rats with chronic pain. In the thalamus, immunoreactivity of both HCN1 and HCN2 subunits was increased in both rat models. These results suggest that the increased HCN channel activity in the thalamus of the ascending nociceptive pathway contributes to both chronic neuropathic and inflammatory pain conditions.
Collapse
Affiliation(s)
- Weihua Ding
- MGH Center for Translational Pain Research Department of Anesthesia, Critical Care and Pain Medicine Massachusetts General Hospital Harvard Medical School Boston, MA, 02114, United States; The First Affiliated Hospital of Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, China; Hangzhou First People's Hospital, 261 Huanshan Road, Hangzhou, 310006, China
| | - Zerong You
- MGH Center for Translational Pain Research Department of Anesthesia, Critical Care and Pain Medicine Massachusetts General Hospital Harvard Medical School Boston, MA, 02114, United States
| | - Shiqian Shen
- MGH Center for Translational Pain Research Department of Anesthesia, Critical Care and Pain Medicine Massachusetts General Hospital Harvard Medical School Boston, MA, 02114, United States
| | - Lucy Chen
- MGH Center for Translational Pain Research Department of Anesthesia, Critical Care and Pain Medicine Massachusetts General Hospital Harvard Medical School Boston, MA, 02114, United States
| | - Shengmei Zhu
- The First Affiliated Hospital of Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, China.
| | - Jianren Mao
- MGH Center for Translational Pain Research Department of Anesthesia, Critical Care and Pain Medicine Massachusetts General Hospital Harvard Medical School Boston, MA, 02114, United States.
| |
Collapse
|
189
|
Cao DN, Song R, Zhang SZ, Wu N, Li J. Nucleus accumbens hyperpolarization-activated cyclic nucleotide-gated channels modulate methamphetamine self-administration in rats. Psychopharmacology (Berl) 2016; 233:3017-29. [PMID: 27329413 DOI: 10.1007/s00213-016-4349-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 05/31/2016] [Indexed: 12/21/2022]
Abstract
RATIONALE Methamphetamine addiction is believed to primarily result from increased dopamine release and the inhibition of dopamine uptake. Some evidence suggests that hyperpolarization-activated cyclic nucleotide-gated (HCN) channels play important roles in the functional modulation of dopaminergic neurons and the pathophysiology of related diseases. However, little is known about the effects of HCN channels on methamphetamine addiction. OBJECTIVES The present study investigated the role of brain HCN channels in methamphetamine addiction. RESULTS Acute intracerebroventricular (i.c.v.) injection or bilateral intra-accumbens microinjections of non-selective HCN channel blocker ZD7288 (0.3125 and 0.625 μg) significantly reduced both methamphetamine (0.0125 or 0.05 mg/kg/infusion)-induced self-administration under fixed ratio 2 reinforcement and the breakpoint of methamphetamine (0.05 mg/kg/infusion) under progressive ratio reinforcement in rats. Moreover, compared with i.c.v. injection, bilateral intra-accumbens microinjections of ZD7288 exerted stronger inhibitory effects, suggesting that blockade of HCN channels in the nucleus accumbens reduced the reinforcing effects of and motivation for methamphetamine. We also found that ZD7288 (0.625 and 1.25 μg, i.c.v.) significantly decreased methamphetamine (1 mg/kg, intraperitoneal (i.p.))-induced hyperactivity with no effect on the spontaneous activity in rats. Finally, in vivo microdialysis experiments showed that the HCN channel blockade using ZD7288 (0.625 and 1.25 μg, i.c.v.) decreased methamphetamine (1 mg/kg, i.p.)-induced elevation of extracellular dopamine levels in the nucleus accumbens. CONCLUSIONS These results indicate that HCN channels in the nucleus accumbens are involved in the reinforcing properties of methamphetamine and highlight the importance of HCN channels in the regulation of dopamine neurotransmission underlying methamphetamine addiction.
Collapse
Affiliation(s)
- Dan-Ni Cao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China
| | - Rui Song
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China
| | - Shu-Zhuo Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China
| | - Ning Wu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China
| | - Jin Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing, 100850, China.
| |
Collapse
|
190
|
Silva FC, Paiva FA, Müller-Ribeiro FC, Caldeira HMA, Fontes MAP, de Menezes RCA, Casali KR, Fortes GH, Tobaldini E, Solbiati M, Montano N, Dias Da Silva VJ, Chianca DA. Chronic Treatment with Ivabradine Does Not Affect Cardiovascular Autonomic Control in Rats. Front Physiol 2016; 7:305. [PMID: 27507948 PMCID: PMC4960883 DOI: 10.3389/fphys.2016.00305] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/06/2016] [Indexed: 12/05/2022] Open
Abstract
A low resting heart rate (HR) would be of great benefit in cardiovascular diseases. Ivabradine—a novel selective inhibitor of hyperpolarization-activated cyclic nucleotide gated (HCN) channels- has emerged as a promising HR lowering drug. Its effects on the autonomic HR control are little known. This study assessed the effects of chronic treatment with ivabradine on the modulatory, reflex and tonic cardiovascular autonomic control and on the renal sympathetic nerve activity (RSNA). Male Wistar rats were divided in 2 groups, receiving intraperitoneal injections of vehicle (VEH) or ivabradine (IVA) during 7 or 8 consecutive days. Rats were submitted to vessels cannulation to perform arterial blood pressure (AP) and HR recordings in freely moving rats. Time series of resting pulse interval and systolic AP were used to measure cardiovascular variability parameters. We also assessed the baroreflex, chemoreflex and the Bezold-Jarish reflex sensitivities. To better evaluate the effects of ivabradine on the autonomic control of the heart, we performed sympathetic and vagal autonomic blockade. As expected, ivabradine-treated rats showed a lower resting (VEH: 362 ± 16 bpm vs. IVA: 260 ± 14 bpm, p = 0.0005) and intrinsic HR (VEH: 369 ± 9 bpm vs. IVA: 326 ± 11 bpm, p = 0.0146). However, the chronic treatment with ivabradine did not change normalized HR spectral parameters LF (nu) (VEH: 24.2 ± 4.6 vs. IVA: 29.8 ± 6.4; p > 0.05); HF (nu) (VEH: 75.1 ± 3.7 vs. IVA: 69.2 ± 5.8; p > 0.05), any cardiovascular reflexes, neither the tonic autonomic control of the HR (tonic sympathovagal index; VEH: 0.91± 0.02 vs. IVA: 0.88 ± 0.03, p = 0.3494). We performed the AP, HR and RSNA recordings in urethane-anesthetized rats. The chronic treatment with ivabradine reduced the resting HR (VEH: 364 ± 12 bpm vs. IVA: 207 ± 11 bpm, p < 0.0001), without affecting RSNA (VEH: 117 ± 16 vs. IVA: 120 ± 9 spikes/s, p = 0.9100) and mean arterial pressure (VEH: 70 ± 4 vs. IVA: 77 ± 6 mmHg, p = 0.3293). Our results suggest that, in health rats, the long-term treatment with ivabradine directly reduces the HR without changing the RSNA modulation and the reflex and tonic autonomic control of the heart.
Collapse
Affiliation(s)
- Fernanda C Silva
- Laboratory of Cardiovascular Physiology, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro PretoOuro Preto, Brazil; Graduate Program in Biological Sciences - CBIOL/NUPEB, Federal University of Ouro PretoOuro Preto, Brazil
| | - Franciny A Paiva
- Laboratory of Cardiovascular Physiology, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro PretoOuro Preto, Brazil; Graduate Program in Biological Sciences - CBIOL/NUPEB, Federal University of Ouro PretoOuro Preto, Brazil
| | - Flávia C Müller-Ribeiro
- Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais Belo Horizonte, Brazil
| | - Henrique M A Caldeira
- Laboratory of Cardiovascular Physiology, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto Ouro Preto, Brazil
| | - Marco A P Fontes
- Laboratory of Hypertension, Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais Belo Horizonte, Brazil
| | - Rodrigo C A de Menezes
- Laboratory of Cardiovascular Physiology, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro PretoOuro Preto, Brazil; Graduate Program in Biological Sciences - CBIOL/NUPEB, Federal University of Ouro PretoOuro Preto, Brazil
| | - Karina R Casali
- Laboratory of Biomedical Engineering, Institute of Science and Technology, Federal University of São Paulo São José dos Campos, Brazil
| | | | - Eleonora Tobaldini
- Department of Clinical Sciences and Community Health, IRCCS Ca' Granda Foundation, Ospedale Maggiore Policlinico, University of Milan Milan, Italy
| | - Monica Solbiati
- Department of Clinical Sciences and Community Health, IRCCS Ca' Granda Foundation, Ospedale Maggiore Policlinico, University of Milan Milan, Italy
| | - Nicola Montano
- Department of Clinical Sciences and Community Health, IRCCS Ca' Granda Foundation, Ospedale Maggiore Policlinico, University of Milan Milan, Italy
| | - Valdo J Dias Da Silva
- Department of Physiology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro Uberaba, Brazil
| | - Deoclécio A Chianca
- Laboratory of Cardiovascular Physiology, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro PretoOuro Preto, Brazil; Graduate Program in Biological Sciences - CBIOL/NUPEB, Federal University of Ouro PretoOuro Preto, Brazil
| |
Collapse
|
191
|
Matsumoto-Makidono Y, Nakayama H, Yamasaki M, Miyazaki T, Kobayashi K, Watanabe M, Kano M, Sakimura K, Hashimoto K. Ionic Basis for Membrane Potential Resonance in Neurons of the Inferior Olive. Cell Rep 2016; 16:994-1004. [PMID: 27425615 DOI: 10.1016/j.celrep.2016.06.053] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 05/04/2016] [Accepted: 06/12/2016] [Indexed: 11/25/2022] Open
Abstract
Some neurons have the ability to enhance output voltage to input current with a preferred frequency, which is called resonance. Resonance is thought to be a basis for membrane potential oscillation. Although ion channels responsible for resonance have been reported, the precise mechanisms by which these channels work remain poorly understood. We have found that resonance is reduced but clearly present in the inferior olivary neurons of Cav3.1 T-type voltage-dependent Ca(2+) channel knockout (KO) mice. The activation of Cav3.1 channels is strongly membrane potential dependent, but less frequency dependent. Residual resonance in Cav3.1 KO mice is abolished by a hyper-polarization-activated cyclic nucleotide-gated (HCN) channel blocker, ZD7288, and is partially suppressed by voltage-dependent K(+) channel blockers. Resonance is inhibited by ZD7288 in wild-type mice and impaired in HCN1 KO mice, suggesting that the HCN1 channel is essential for resonance. The ZD7288-sensitive current is nearly sinusoidal and strongly frequency dependent. These results suggest that Cav3.1 and HCN1 channels act as amplifying and resonating conductances, respectively.
Collapse
Affiliation(s)
- Yoshiko Matsumoto-Makidono
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Hiroshima 734-8551, Japan
| | - Hisako Nakayama
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Hiroshima 734-8551, Japan
| | - Miwako Yamasaki
- Department of Anatomy, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Taisuke Miyazaki
- Department of Anatomy, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Hongo, Tokyo 113-0033, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Kouichi Hashimoto
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Hiroshima 734-8551, Japan.
| |
Collapse
|
192
|
Shah MM. Hyperpolarization-Activated Cyclic Nucleotide-Gated Channel Currents in Neurons. Cold Spring Harb Protoc 2016; 2016:2016/7/pdb.top087346. [PMID: 27371604 DOI: 10.1101/pdb.top087346] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are voltage-gated ion channels that activate at potentials more negative than -50 mV and are predominantly permeable to Na(+) and K(+) ions. Four HCN subunits (HCN1-4) have been cloned. These subunits have distinct expression patterns and biophysical properties. In addition, cyclic nucleotides as well as multiple intracellular substances including various kinases and phosphatases modulate the expression and function of the subunits. Hence, the characteristics of the current, Ih, are likely to vary among neuronal subtypes. In many neuronal subtypes, Ih is present postsynaptically, where it plays a critical role in setting the resting membrane potential and the membrane resistance. By influencing these intrinsic properties, Ih will affect synaptic potential shapes and summation and thereby affect neuronal excitability. Additionally, Ih can have an effect on resonance properties and intrinsic neuronal oscillations. In some neurons, Ih may also be present presynaptically in axons and synaptic terminals, where it modulates neuronal transmitter release. Hence the effects of Ih on neuronal excitability are complex. It is, however, necessary to fully understand these as Ih has a significant impact on physiological conditions such as learning as well as pathophysiological states such as epilepsy.
Collapse
Affiliation(s)
- Mala M Shah
- Department of Pharmacology, UCL School of Pharmacy, University College London, London WC1N 1AX, United Kingdom
| |
Collapse
|
193
|
Tibbs GR, Posson DJ, Goldstein PA. Voltage-Gated Ion Channels in the PNS: Novel Therapies for Neuropathic Pain? Trends Pharmacol Sci 2016; 37:522-542. [DOI: 10.1016/j.tips.2016.05.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 03/24/2016] [Accepted: 05/03/2016] [Indexed: 12/19/2022]
|
194
|
Zhang XX, Min XC, Xu XL, Zheng M, Guo LJ. ZD7288, a selective hyperpolarization-activated cyclic nucleotide-gated channel blocker, inhibits hippocampal synaptic plasticity. Neural Regen Res 2016; 11:779-86. [PMID: 27335562 PMCID: PMC4904469 DOI: 10.4103/1673-5374.182705] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The selective hyperpolarization-activated cyclic nucleotide-gated (HCN) channel blocker 4-(N-ethyl-N-phenylamino)-1,2-dimethyl-6-(methylamino) pyrimidinium chloride (ZD7288) blocks the induction of long-term potentiation in the perforant path–CA3 region in rat hippocampus in vivo. To explore the mechanisms underlying the action of ZD7288, we recorded excitatory postsynaptic potentials in perforant path–CA3 synapses in male Sprague-Dawley rats. We measured glutamate content in the hippocampus and in cultured hippocampal neurons using high performance liquid chromatography, and determined intracellular Ca2+ concentration [Ca2+]i) using Fura-2. ZD7288 inhibited the induction and maintenance of long-term potentiation, and these effects were mirrored by the nonspecific HCN channel blocker cesium. ZD7288 also decreased glutamate release in hippocampal tissue and in cultured hippocampal neurons. Furthermore, ZD7288 attenuated glutamate-induced rises in [Ca2+]i in a concentration-dependent manner and reversed 8-Br-cAMP-mediated facilitation of these glutamate-induced [Ca2+]i rises. Our results suggest that ZD7288 inhibits hippocampal synaptic plasticity both glutamate release and resultant [Ca2+]i increases in rat hippocampal neurons.
Collapse
Affiliation(s)
- Xiao-Xue Zhang
- Department of Laboratory Medicine, Affiliated Pu'ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiao-Chun Min
- Department of Laboratory Medicine, Affiliated Pu'ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xu-Lin Xu
- Department of Pharmacology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Min Zheng
- School of Biomedical Engineering, Hubei University of Science and Technology, Xianning, Hubei Province, China
| | - Lian-Jun Guo
- Department of Pharmacology, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
195
|
Yang Z, Santamaria F. Purkinje cell intrinsic excitability increases after synaptic long term depression. J Neurophysiol 2016; 116:1208-17. [PMID: 27306677 DOI: 10.1152/jn.00369.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 06/07/2016] [Indexed: 11/22/2022] Open
Abstract
Coding in cerebellar Purkinje cells not only depends on synaptic plasticity but also on their intrinsic membrane excitability. We performed whole cell patch-clamp recordings of Purkinje cells in sagittal cerebellar slices in mice. We found that inducing long-term depression (LTD) in the parallel fiber to Purkinje cell synapses results in an increase in the gain of the firing rate response. This increase in excitability is accompanied by an increase in the input resistance and a decrease in the amplitude of the hyperpolarization-activated cyclic nucleotide-gated (HCN) channel-mediated voltage sag. Application of a HCN channel blocker prevents the increase in input resistance and excitability without blocking the expression of synaptic LTD. We conclude that the induction of parallel fiber-Purkinje cell LTD is accompanied by an increase in excitability of Purkinje cells through downregulation of the HCN-mediated h current. We suggest that HCN downregulation is linked to the biochemical pathway that sustains synaptic LTD. Given the diversity of information carried by the parallel fiber system, we suggest that changes in intrinsic excitability enhance the coding capacity of the Purkinje cell to specific input sources.
Collapse
Affiliation(s)
- Zhen Yang
- UTSA Neurosciences Institute and Department of Biology, University of Texas at San Antonio, San Antonio, Texas
| | - Fidel Santamaria
- UTSA Neurosciences Institute and Department of Biology, University of Texas at San Antonio, San Antonio, Texas
| |
Collapse
|
196
|
Knoll AT, Halladay LR, Holmes AJ, Levitt P. Quantitative Trait Loci and a Novel Genetic Candidate for Fear Learning. J Neurosci 2016; 36:6258-68. [PMID: 27277803 PMCID: PMC4899527 DOI: 10.1523/jneurosci.0177-16.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Revised: 04/15/2016] [Accepted: 05/02/2016] [Indexed: 01/10/2023] Open
Abstract
UNLABELLED Trauma- and stress-related disorders are clinically heterogeneous and associated with substantial genetic risk. Understanding the biological origins of heterogeneity of key intermediate phenotypes such as cognition and emotion can provide novel mechanistic insights into disorder pathogenesis. Performing quantitative genetics in animal models is a tractable strategy for examining both the genetic basis of intermediate phenotypes and functional testing of candidate quantitative traits genes (QTGs). Here, existing and newly collected data were used for collaborative genome-wide mapping of cued fear acquisition and expression in 65 mouse strains from the BXD genetic reference panel. For fear acquisition, we identified a significant locus on chromosome (Chr) 10 and eight suggestive loci on Chr 2, 4, 5, 11, 13, and 15. For fear expression, we identified one significant and another highly suggestive locus on Chr 13, as well as four suggestive loci on Chr 10, 11, and X. Across these loci, 60 putative QTGs were identified. The quantitative trait locus on distal Chr 13 contained a single, highly promising gene at the location of the peak likelihood ratio statistic score. The gene, hyperpolarization-activated cyclic nucleotide-gated channel 1 (Hcn1), regulates neuronal excitability. Validation experiments using behavioral pharmacology revealed that functional Hcn channels in the basolateral amygdala are necessary for conditioned fear acquisition and expression. Hcn1, together with the other candidate QTGs, thus provide new targets for neurobiological and treatment studies of fear learning and trauma- and stress-related disorders. SIGNIFICANCE STATEMENT There is a knowledge gap in understanding the genetic contributions to behavioral heterogeneity in typical and atypical populations. Mouse genetic reference panels (GRPs) provide one approach for identifying genetic sources of variation. Here, we identified three loci for conditioned fear acquisition and expression in a mouse GRP. Each locus contained candidate quantitative trait genes (QTGs). One locus had a single QTG, Hcn1 (hyperpolarization-activated cyclic nucleotide-gated channel 1), which has been implicated in neuronal excitability and learning. This discovery was validated using behavioral pharmacology, revealing that Hcn channels in the basolateral amygdala are required for fear acquisition and expression. The study thus identifies novel candidate QTGs that may contribute to variation in emotional learning and highlight the utility of mouse GRPs for the identification of genes underlying complex traits.
Collapse
Affiliation(s)
- Allison T Knoll
- Program in Developmental Neurogenetics, Institute for the Developing Mind, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California 90027
| | - Lindsay R Halladay
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland 20814
| | - Andrew J Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland 20814
| | - Pat Levitt
- Program in Developmental Neurogenetics, Institute for the Developing Mind, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California 90027, Department of Pediatrics, Keck School of Medicine of the University of Southern California, Los Angeles, California 90089, and
| |
Collapse
|
197
|
Gao SH, Wen HZ, Shen LL, Zhao YD, Ruan HZ. Activation of mGluR1 contributes to neuronal hyperexcitability in the rat anterior cingulate cortex via inhibition of HCN channels. Neuropharmacology 2016; 105:361-377. [DOI: 10.1016/j.neuropharm.2016.01.036] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/28/2016] [Accepted: 01/28/2016] [Indexed: 01/14/2023]
|
198
|
Itoh M, Ishihara K, Nakashima N, Takano M. The hyperpolarization-activated cyclic nucleotide-gated (HCN) channels contain multiple S-palmitoylation sites. J Physiol Sci 2016; 66:241-8. [PMID: 26546007 PMCID: PMC10717812 DOI: 10.1007/s12576-015-0420-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 10/18/2015] [Indexed: 11/26/2022]
Abstract
Expression of hyperpolarization-activated cyclic nucleotide-gated channels (HCN1-4) on distal dendrites of neurons is suggested to modify synaptic integration in the central nervous system. However, the mechanisms of dendritic localization are not fully understood. Recent studies have revealed that S-palmitoylation plays an important role in the enrichment of various molecules at the postsynaptic membrane. Thus, we performed an acyl-biotinyl exchange assay, and found that HCN1, HCN2, and HCN4, but not HCN3, were S-palmitoylated in HEK293 cells. Mutation of multiple intracellular cysteine residues at the N-terminus of HCN2 was required for complete inhibition of S-palmitoylation. However, this mutagenesis had a minimal effect on surface expression of HCN2 proteins or electrophysiological properties of HCN2 current when expressed in HEK293 cells or in Xenopus oocytes. These findings provide insight into the physiological roles of S-palmitoylation of HCN channels in native neurons.
Collapse
Affiliation(s)
- Masayuki Itoh
- Division of Integrated Autonomic Function, Department of Physiology, Kurume University School of Medicine, 67 Asahimachi, Kurume, Fukuoka, 830-0011, Japan.
| | - Keiko Ishihara
- Division of Integrated Autonomic Function, Department of Physiology, Kurume University School of Medicine, 67 Asahimachi, Kurume, Fukuoka, 830-0011, Japan
| | - Noriyuki Nakashima
- Department of Physiology, Faculty of Medicine, Kyoto University, Yoshida-Konoe, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Makoto Takano
- Division of Integrated Autonomic Function, Department of Physiology, Kurume University School of Medicine, 67 Asahimachi, Kurume, Fukuoka, 830-0011, Japan
| |
Collapse
|
199
|
Nishitani A, Tanaka M, Shimizu S, Kunisawa N, Yokoe M, Yoshida Y, Suzuki T, Sakuma T, Yamamoto T, Kuwamura M, Takenaka S, Ohno Y, Kuramoto T. Involvement of aspartoacylase in tremor expression in rats. Exp Anim 2016; 65:293-301. [PMID: 27026062 PMCID: PMC4976243 DOI: 10.1538/expanim.16-0007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Essential tremor (ET) is a common movement disorder with a poorly understood etiology.
The TRM/Kyo mutant rat, showing spontaneous tremor, is an animal model of ET. Recently, we
demonstrated that tremors in these rats emerge when two mutant loci, a missense mutation
in the hyperpolarization-activated cyclic nucleotide-gated potassium channel 1
(Hcn1) and the tremor (tm) deletion, are present
simultaneously. However, we did not identify which gene within the tm
deletion causes tremor expression in TRM/Kyo rats. A strong candidate among the 13 genes
within the tm deletion is aspartoacylase (Aspa), because
some Aspa-knockout mouse strains show tremor. Here, we generated
Aspa-knockout rats using transcription activator-like effector nuclease
technology and produced Aspa/Hcn1 double-mutant rats by
crossing Aspa-knockout rats with Hcn1-mutant rats. The
Aspa-knockout rats carried nonsense mutations in exon 4; and ASPA
proteins were not detectable in their brain extracts. They showed elevated levels of
N-acetyl-L-aspartate (NAA) in urine and spongy vacuolation
and abnormal myelination in the central nervous system, but no tremor. By contrast,
Aspa/Hcn1 double-mutant rats spontaneously showed
tremors resembling those in TRM/Kyo rats, and the tremor was suppressed by drugs
therapeutic for ET but not for parkinsonian tremor. These findings indicated that the lack
of the Aspa gene caused tremor expression in TRM/Kyo rats. Our animal
model suggested that the interaction of NAA accumulation due to ASPA deficiency with an
unstable neuronal membrane potential caused by HCN1 deficiency was involved in tremor
development.
Collapse
Affiliation(s)
- Ai Nishitani
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Yoshida-konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
200
|
Zhao Z, Zhang K, Liu X, Yan H, Ma X, Zhang S, Zheng J, Wang L, Wei X. Involvement of HCN Channel in Muscarinic Inhibitory Action on Tonic Firing of Dorsolateral Striatal Cholinergic Interneurons. Front Cell Neurosci 2016; 10:71. [PMID: 27047336 PMCID: PMC4801847 DOI: 10.3389/fncel.2016.00071] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 03/04/2016] [Indexed: 11/13/2022] Open
Abstract
The striatum is the most prominent nucleus in the basal ganglia and plays an important role in motor movement regulation. The cholinergic interneurons (ChIs) in striatum are involved in the motion regulation by releasing acetylcholine (ACh) and modulating the output of striatal projection neurons. Here, we report that muscarinic ACh receptor (M receptor) agonists, ACh and Oxotremorine (OXO-M), decreased the firing frequency of ChIs by blocking the hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. Scopolamine (SCO), a nonselective antagonist of M receptors, abolished the inhibition. OXO-M exerted its function by activating the Gi/o cAMP signaling cascade. The single-cell reverse transcription polymerase chain reaction (scRT-PCR) revealed that all the five subtypes of M receptors and four subtypes of HCN channels were expressed on ChIs. Among them, M2 receptors and HCN2 channels were the most dominant ones and expressed in every single studied cholinergic interneuron (ChI).Our results suggest that ACh regulates not only the output of striatal projection neurons, but also the firing activity of ChIs themselves by activating presynaptic M receptors in the dorsal striatum. The activation of M2 receptors and blockage of HCN2 channels may play an important role in ACh inhibition on the excitability of ChIs. This finding adds a new G-protein coupled receptor mediated regulation on ChIs and provides a cellular mechanism for control of cholinergic activity and ACh release in the dorsal striatum.
Collapse
Affiliation(s)
- Zhe Zhao
- State Key Laboratory of Toxicology and Medical Countermeasures, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and Toxicology Beijing, China
| | - Kang Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and Toxicology Beijing, China
| | - Xiaoyan Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and Toxicology Beijing, China
| | - Haitao Yan
- State Key Laboratory of Toxicology and Medical Countermeasures, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and Toxicology Beijing, China
| | - Xiaoyun Ma
- State Key Laboratory of Toxicology and Medical Countermeasures, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and Toxicology Beijing, China
| | - Shuzhuo Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and Toxicology Beijing, China
| | - Jianquan Zheng
- State Key Laboratory of Toxicology and Medical Countermeasures, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and Toxicology Beijing, China
| | - Liyun Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and Toxicology Beijing, China
| | - Xiaoli Wei
- State Key Laboratory of Toxicology and Medical Countermeasures, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and Toxicology Beijing, China
| |
Collapse
|