1
|
Wu X, Hao J, Jiang K, Wu M, Zhao X, Zhang X. Neuroinflammation and pathways that contribute to tourette syndrome. Ital J Pediatr 2025; 51:63. [PMID: 40022157 PMCID: PMC11871796 DOI: 10.1186/s13052-025-01874-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/26/2025] [Indexed: 03/03/2025] Open
Abstract
Tourette syndrome (TS), a neurological and psychological disease, typically exhibit motor and phonic tics. The pathophysiology of TS remains controversial. Currently, the recognized pathogenesis of TS is the imbalance of neurotransmitters, involving abnormality of the cortex-striatum-thalamus-cortex circuit. Recently, clinical researches demonstrate that triggers such as infection and allergic reaction could lead to the onset or exacerbation of tic symptoms. Current studies have also suggested that neural-immune crosstalk caused by inflammation is also associated with TS, potentially leading to the occurrence of tics by inducing neurotransmitter abnormalities. Herein, we review inflammation-related factors contributing to the occurrence of TS as well as the mechanisms by which immune-inflammatory pathways mediate the onset of TS. This aims to clarify the pathogenesis of TS and provide a theoretical basis for the treatment of TS.
Collapse
Affiliation(s)
- Xinnan Wu
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juanjuan Hao
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Keyu Jiang
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Wu
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Zhao
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Zhang
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
Buss EW, Lofaro OM, Barnett A, Leroy F, Santoro B, Siegelbaum SA, Bock T. HCN1 hyperpolarization-activated cyclic nucleotide-gated channels enhance evoked GABA release from parvalbumin-positive interneurons. Proc Natl Acad Sci U S A 2024; 121:e2319246121. [PMID: 39378096 PMCID: PMC11494348 DOI: 10.1073/pnas.2319246121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 08/27/2024] [Indexed: 10/10/2024] Open
Abstract
Hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels generate the cationic Ih current in neurons and regulate the excitability of neuronal networks. The function of HCN channels depends, in part, on their subcellular localization. Of the four HCN isoforms (HCN1-4), HCN1 is strongly expressed in the dendrites of pyramidal neurons (PNs) in hippocampal area CA1 but also in presynaptic terminals of parvalbumin-positive interneurons (PV+ INs), which provide strong inhibitory control over hippocampal activity. Yet, little is known about how HCN1 channels in these cells regulate the evoked release of the inhibitory transmitter GABA from their axon terminals. Here, we used genetic, optogenetic, electrophysiological, and imaging techniques to investigate how the electrophysiological properties of PV+ INs are regulated by HCN1, including how HCN1 activity at presynaptic terminals regulates the release of GABA onto PNs in CA1. We found that application of HCN1 pharmacological blockers reduced the amplitude of the inhibitory postsynaptic potential recorded from CA1 PNs in response to selective optogenetic stimulation of PV+ INs. Homozygous HCN1 knockout mice also show reduced IPSCs in postsynaptic cells. Finally, two-photon imaging using genetically encoded fluorescent calcium indicators revealed that HCN1 blockers reduced the probability that an extracellular electrical stimulating pulse evoked a Ca2+ response in individual PV+ IN presynaptic boutons. Taken together, our results show that HCN1 channels in the axon terminals of PV+ interneurons facilitate GABAergic transmission in the hippocampal CA1 region.
Collapse
Affiliation(s)
- Eric W. Buss
- Departments of Neuroscience and Pharmacology, Kavli Institute for Brain Science, Zuckerman Mind Brain Behavior Institute, Columbia University Medical Center, New York, NY10027
| | - Olivia M. Lofaro
- Departments of Neuroscience and Pharmacology, Kavli Institute for Brain Science, Zuckerman Mind Brain Behavior Institute, Columbia University Medical Center, New York, NY10027
| | - Anastasia Barnett
- Departments of Neuroscience and Pharmacology, Kavli Institute for Brain Science, Zuckerman Mind Brain Behavior Institute, Columbia University Medical Center, New York, NY10027
| | - Felix Leroy
- Departments of Neuroscience and Pharmacology, Kavli Institute for Brain Science, Zuckerman Mind Brain Behavior Institute, Columbia University Medical Center, New York, NY10027
| | - Bina Santoro
- Departments of Neuroscience and Pharmacology, Kavli Institute for Brain Science, Zuckerman Mind Brain Behavior Institute, Columbia University Medical Center, New York, NY10027
| | - Steven A. Siegelbaum
- Departments of Neuroscience and Pharmacology, Kavli Institute for Brain Science, Zuckerman Mind Brain Behavior Institute, Columbia University Medical Center, New York, NY10027
| | - Tobias Bock
- Departments of Neuroscience and Pharmacology, Kavli Institute for Brain Science, Zuckerman Mind Brain Behavior Institute, Columbia University Medical Center, New York, NY10027
- Department of Systems Neurophysiology, Institute for Zoology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen52074, Germany
| |
Collapse
|
3
|
Zhang Z, Luo X, Jiang L, Wu H, Tan Z. How do HCN channels play a part in Alzheimer's and Parkinson's disease? Ageing Res Rev 2024; 100:102436. [PMID: 39047878 DOI: 10.1016/j.arr.2024.102436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/08/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
Neurodegenerative diseases like Alzheimer's and Parkinson's disease (AD and PD) are well-known, yet their underlying causes remain unclear. Recent studies have suggested that disruption of ion channels contribute to their pathogenesis. Among these channels, the hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, encoded by HCN1-4 genes, are of particular interest due to their role in generating hyperpolarization-activated current (Ih), which is crucial in various neural activities impacting memory and motor functions. A growing body of evidence underscores the pivotal role of HCN in Aβ generation, glial cell function, and ischemia-induced dementia; while HCN is expressed in various regions of the basal ganglia, modulating their functions and influencing motor disorders in PD; neuroinflammation triggered by microglial activation represents a shared pathological mechanism in both AD and PD, in which HCN also plays a significant part. This review delves into the neuronal functions governed by HCN, its roles in the aforementioned pathogenesis, its expression patterns in AD and PD, and discusses potential therapeutic drugs targeting HCN for the treatment of these diseases, aiming to offer a novel perspective and inspire future research endeavors.
Collapse
Affiliation(s)
- Zhuo Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Changsha 410008, PR China; Changsha Taihe Hospital, Changsha 410000, PR China; Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha 410205, PR China
| | - Xin Luo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Changsha 410008, PR China; Changsha Taihe Hospital, Changsha 410000, PR China; Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha 410205, PR China
| | - Liping Jiang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Changsha 410008, PR China; Department of Physiology, Basic Medical School, Hengyang Medical College, The Neuroscience Institute, University of South China, Hengyang 421001, PR China; Changsha Taihe Hospital, Changsha 410000, PR China; Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha 410205, PR China
| | - Huilan Wu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Changsha 410008, PR China; Changsha Taihe Hospital, Changsha 410000, PR China; Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha 410205, PR China
| | - Zhirong Tan
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Changsha 410008, PR China; Changsha Taihe Hospital, Changsha 410000, PR China; Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha 410205, PR China.
| |
Collapse
|
4
|
Guo B, Liu T, Choi S, Mao H, Wang W, Xi K, Jones C, Hartley ND, Feng D, Chen Q, Liu Y, Wimmer RD, Xie Y, Zhao N, Ou J, Arias-Garcia MA, Malhotra D, Liu Y, Lee S, Pasqualoni S, Kast RJ, Fleishman M, Halassa MM, Wu S, Fu Z. Restoring thalamocortical circuit dysfunction by correcting HCN channelopathy in Shank3 mutant mice. Cell Rep Med 2024; 5:101534. [PMID: 38670100 PMCID: PMC11149412 DOI: 10.1016/j.xcrm.2024.101534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 12/11/2023] [Accepted: 04/04/2024] [Indexed: 04/28/2024]
Abstract
Thalamocortical (TC) circuits are essential for sensory information processing. Clinical and preclinical studies of autism spectrum disorders (ASDs) have highlighted abnormal thalamic development and TC circuit dysfunction. However, mechanistic understanding of how TC dysfunction contributes to behavioral abnormalities in ASDs is limited. Here, our study on a Shank3 mouse model of ASD reveals TC neuron hyperexcitability with excessive burst firing and a temporal mismatch relationship with slow cortical rhythms during sleep. These TC electrophysiological alterations and the consequent sensory hypersensitivity and sleep fragmentation in Shank3 mutant mice are causally linked to HCN2 channelopathy. Restoring HCN2 function early in postnatal development via a viral approach or lamotrigine (LTG) ameliorates sensory and sleep problems. A retrospective case series also supports beneficial effects of LTG treatment on sensory behavior in ASD patients. Our study identifies a clinically relevant circuit mechanism and proposes a targeted molecular intervention for ASD-related behavioral impairments.
Collapse
Affiliation(s)
- Baolin Guo
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Tiaotiao Liu
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin 300070, China
| | - Soonwook Choi
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Honghui Mao
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Wenting Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Kaiwen Xi
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Carter Jones
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Nolan D Hartley
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Dayun Feng
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Qian Chen
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Yingying Liu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Ralf D Wimmer
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Yuqiao Xie
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Ningxia Zhao
- Xi'an TCM Hospital of Encephalopathy, Shaanxi University of Chinese Medicine, Xi'an 710032, China
| | - Jianjun Ou
- Department of Psychiatry, The Second Xiangya Hospital of Central South University, National Clinical Research Center for Mental Disorders, Changsha 410011, China
| | - Mario A Arias-Garcia
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Diya Malhotra
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Yang Liu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Sihak Lee
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sammuel Pasqualoni
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ryan J Kast
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Morgan Fleishman
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Michael M Halassa
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Zhanyan Fu
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA.
| |
Collapse
|
5
|
Qi ZX, Yan Q, Fan XJ, Peng JY, Zhu HX, Jiang YM, Chen L, Zhuang QX. Role of HCN channels in the functions of basal ganglia and Parkinson's disease. Cell Mol Life Sci 2024; 81:135. [PMID: 38478096 PMCID: PMC10937777 DOI: 10.1007/s00018-024-05163-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/19/2024] [Accepted: 02/06/2024] [Indexed: 03/17/2024]
Abstract
Parkinson's disease (PD) is a motor disorder resulting from dopaminergic neuron degeneration in the substantia nigra caused by age, genetics, and environment. The disease severely impacts a patient's quality of life and can even be life-threatening. The hyperpolarization-activated cyclic nucleotide-gated (HCN) channel is a member of the HCN1-4 gene family and is widely expressed in basal ganglia nuclei. The hyperpolarization-activated current mediated by the HCN channel has a distinct impact on neuronal excitability and rhythmic activity associated with PD pathogenesis, as it affects the firing activity, including both firing rate and firing pattern, of neurons in the basal ganglia nuclei. This review aims to comprehensively understand the characteristics of HCN channels by summarizing their regulatory role in neuronal firing activity of the basal ganglia nuclei. Furthermore, the distribution and characteristics of HCN channels in each nucleus of the basal ganglia group and their effect on PD symptoms through modulating neuronal electrical activity are discussed. Since the roles of the substantia nigra pars compacta and reticulata, as well as globus pallidus externus and internus, are distinct in the basal ganglia circuit, they are individually described. Lastly, this investigation briefly highlights that the HCN channel expressed on microglia plays a role in the pathological process of PD by affecting the neuroinflammatory response.
Collapse
Affiliation(s)
- Zeng-Xin Qi
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200030, China
- National Center for Neurological Disorders, Shanghai, 200030, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200030, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200030, China
| | - Qi Yan
- Department of Physiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Xiu-Juan Fan
- Department of Physiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Jian-Ya Peng
- Department of Physiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Hui-Xian Zhu
- Department of Physiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Yi-Miao Jiang
- Department of Physiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Liang Chen
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200030, China.
- National Center for Neurological Disorders, Shanghai, 200030, China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200030, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200030, China.
| | - Qian-Xing Zhuang
- Department of Physiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
6
|
Cai M, Zhu Y, Shanley MR, Morel C, Ku SM, Zhang H, Shen Y, Friedman AK, Han MH. HCN channel inhibitor induces ketamine-like rapid and sustained antidepressant effects in chronic social defeat stress model. Neurobiol Stress 2023; 26:100565. [PMID: 37664876 PMCID: PMC10468802 DOI: 10.1016/j.ynstr.2023.100565] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 09/05/2023] Open
Abstract
Repeated, long-term (weeks to months) exposure to standard antidepressant medications is required to achieve treatment efficacy. In contrast, acute ketamine quickly improves mood for an extended time. Recent work implicates that hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are involved in mediating ketamine's antidepressant effects. In this study, we directly targeted HCN channels and achieved ketamine-like rapid and sustained antidepressant efficacy. Our in vitro electrophysiological recordings first showed that HCN inhibitor DK-AH 269 (also called cilobradine) decreased the pathological HCN-mediated current (Ih) and abnormal hyperactivity of ventral tegmental area (VTA) dopamine (DA) neurons in a depressive-like model produced by chronic social defeat stress (CSDS). Our in vivo studies further showed that acute intra-VTA or acute systemic administration of DK-AH 269 normalized social behavior and rescued sucrose preference in CSDS-susceptible mice. The single-dose of DK-AH 269, both by intra-VTA microinfusion and intraperitoneal (ip) approaches, could produce an extended 13-day duration of antidepressant-like efficacy. Animals treated with acute DK-AH 269 spent less time immobile than vehicle-treated mice during forced swim test. A social behavioral reversal lasted up to 13 days following the acute DK-AH 269 ip injection, and this rapid and sustained antidepressant-like response is paralleled with a single-dose treatment of ketamine. This study provides a novel ion channel target for acutely acting, long-lasting antidepressant-like effects.
Collapse
Affiliation(s)
- Min Cai
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yingbo Zhu
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- China Shenzhen Naowunao Network Technology Co.,Ltd., Shenzhen, Guangdong, China
| | - Mary Regis Shanley
- Department of Biological Sciences, Hunter College, Biology and Biochemistry PhD Program, Graduate Center, The City University of New York, New York, NY, USA
| | - Carole Morel
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stacy M. Ku
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hongxing Zhang
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yuan Shen
- Anesthesia and Brain Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Allyson K. Friedman
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ming-Hu Han
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong, China
| |
Collapse
|
7
|
Presynaptic HCN channel activity is required for the expression of long-term potentiation at lateral amygdala to basal amygdala synapses. Biochem Biophys Res Commun 2022; 637:100-107. [DOI: 10.1016/j.bbrc.2022.11.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 11/09/2022] [Indexed: 11/11/2022]
|
8
|
Munoz B, Fritz BM, Yin F, Atwood BK. HCN1 channels mediate mu opioid receptor long-term depression at insular cortex inputs to the dorsal striatum. J Physiol 2022; 600:4917-4938. [PMID: 36181477 PMCID: PMC11457701 DOI: 10.1113/jp283513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/26/2022] [Indexed: 12/24/2022] Open
Abstract
Mu opioid receptors (MORs) are expressed in the dorsal striatum, a brain region that mediates goal-directed (via the dorsomedial striatum) and habitual (via the dorsolateral striatum, DLS) behaviours. Our previous work indicates that glutamate transmission is depressed when MORs are activated in the dorsal striatum, inducing MOR-mediated long-term synaptic depression (MOR-LTD) or short-term depression (MOR-STD), depending on the input. In the DLS, MOR-LTD is produced by MORs on anterior insular cortex (AIC) inputs and MOR-STD occurs at thalamic inputs, suggesting input-specific MOR plasticity mechanisms. Here, we evaluated the mechanisms of induction of MOR-LTD and MOR-STD in the DLS using pharmacology and optogenetics combined with patch-clamp electrophysiology. We found that cAMP/PKA signalling and protein synthesis are necessary for MOR-LTD expression, similar to previous studies of cannabinoid-mediated LTD in DLS. MOR-STD does not utilize these same mechanisms. We also demonstrated that cannabinoid-LTD occurs at AIC inputs to DLS. However, while cannabinoid-LTD requires mTOR signalling in DLS, MOR-LTD does not. We characterized the role of presynaptic HCN1 channels in MOR-LTD induction as HCN1 channels expressed in AIC are necessary for MOR-LTD expression in the DLS. These results suggest a mechanism in which MOR activation requires HCN1 to induce MOR-LTD, suggesting a new target for pharmacological modulation of synaptic plasticity, providing new opportunities to develop novel drugs to treat alcohol and opioid use disorders. KEY POINTS: Mu opioid receptor-mediated long-term depression at anterior insular cortex inputs to dorsolateral striatum involves presynaptic cAMP/PKA signalling and protein translation, similar to known mechanisms of cannabinoid long-term depression. Dorsal striatal cannabinoid long-term depression also occurs at anterior insular cortex inputs to the dorsolateral striatum. Dorsal striatal cannabinoid long-term depression requires mTOR signalling, similar to hippocampal cannabinoid long-term depression, but dorsal striatal mu opioid long-term depression does not require mTOR signalling. Mu opioid long-term depression requires presynaptic HCN1 channels at anterior insular cortex inputs to dorsolateral striatum.
Collapse
Affiliation(s)
- Braulio Munoz
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Brandon M. Fritz
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Fuqin Yin
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Brady K. Atwood
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
9
|
Willis DE, Goldstein PA. Targeting Affective Mood Disorders With Ketamine to Prevent Chronic Postsurgical Pain. FRONTIERS IN PAIN RESEARCH 2022; 3:872696. [PMID: 35832728 PMCID: PMC9271565 DOI: 10.3389/fpain.2022.872696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 06/06/2022] [Indexed: 12/02/2022] Open
Abstract
The phencyclidine-derivative ketamine [2-(2-chlorophenyl)-2-(methylamino)cyclohexan-1-one] was added to the World Health Organization's Model List of Essential Medicines in 1985 and is also on the Model List of Essential Medicines for Children due to its efficacy and safety as an intravenous anesthetic. In sub-anesthetic doses, ketamine is an effective analgesic for the treatment of acute pain (such as may occur in the perioperative setting). Additionally, ketamine may have efficacy in relieving some forms of chronic pain. In 2019, Janssen Pharmaceuticals received regulatory-approval in both the United States and Europe for use of the S-enantiomer of ketamine in adults living with treatment-resistant major depressive disorder. Pre-existing anxiety/depression and the severity of postoperative pain are risk factors for development of chronic postsurgical pain. An important question is whether short-term administration of ketamine can prevent the conversion of acute postsurgical pain to chronic postsurgical pain. Here, we have reviewed ketamine's effects on the biopsychological processes underlying pain perception and affective mood disorders, focusing on non-NMDA receptor-mediated effects, with an emphasis on results from human trials where available.
Collapse
Affiliation(s)
- Dianna E. Willis
- Burke Neurological Institute, White Plains, NY, United States
- Feil Family Brain and Mind Institute, Weill Cornell Medicine, New York, NY, United States
| | - Peter A. Goldstein
- Feil Family Brain and Mind Institute, Weill Cornell Medicine, New York, NY, United States
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States
- Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- *Correspondence: Peter A. Goldstein
| |
Collapse
|
10
|
Cai W, Liu SS, Li BM, Zhang XH. Presynaptic HCN channels constrain GABAergic synaptic transmission in pyramidal cells of the medial prefrontal cortex. Biol Open 2021; 11:272636. [PMID: 34709375 PMCID: PMC8966777 DOI: 10.1242/bio.058840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/26/2021] [Indexed: 11/20/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are widely expressed in neurons in the central nervous system. It has been documented that HCN channels regulate the intrinsic excitability of pyramidal cells in the medial prefrontal cortex (mPFC) of rodents. Here, we report that HCN channels limited GABAergic transmission onto pyramidal cells in rat mPFC. The pharmacological blockade of HCN channels resulted in a significant increase in the frequency of both spontaneous and miniature inhibitory postsynaptic currents (IPSCs) in mPFC pyramidal cells, whereas potentiation of HCN channels reversely decreases the frequency of mIPSCs. Furthermore, such facilitation effect on mIPSC frequency required presynaptic Ca2+ influx. Immunofluorescence staining showed that HCN channels expressed in presynaptic GABAergic terminals, as well as in both soma and neurite of parvalbumin-expressing (PV-expressing) basket cells in mPFC. The present results indicate that HCN channels in GABAergic interneurons, most likely PV-expressing basket cells, constrain inhibitory control over layer 5-6 pyramidal cells by restricting presynaptic Ca2+ entry.
Collapse
Affiliation(s)
- Wei Cai
- Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Shu-Su Liu
- Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Bao-Ming Li
- Center for Neuropsychiatric Diseases, Institute of Life Science, Nanchang University, Nanchang 330031, China
| | - Xue-Han Zhang
- Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| |
Collapse
|
11
|
Daniel NH, Aravind A, Thakur P. Are ion channels potential therapeutic targets for Parkinson's disease? Neurotoxicology 2021; 87:243-257. [PMID: 34699791 DOI: 10.1016/j.neuro.2021.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/15/2021] [Accepted: 10/21/2021] [Indexed: 01/31/2023]
Abstract
Parkinson's disease (PD) is primarily associated with the progressive neurodegeneration of the dopaminergic neurons in the substantia nigra region of the brain. The resulting motor symptoms are managed with the help of dopamine replacement therapies. However, these therapeutics do not prevent the neurodegeneration underlying the disease and therefore lose their effectiveness in managing disease symptoms over time. Thus, there is an urgent need to develop newer therapeutics for the benefit of patients. The release of dopamine and the firing activity of substantia nigra neurons is regulated by several ion channels that act in concert. Dysregulations of these channels cause the aberrant movement of various ions in the intracellular milieu. This eventually leads to disruption of intracellular signalling cascades, alterations in cellular homeostasis, and bioenergetic deficits. Therefore, ion channels play a central role in driving the high vulnerability of dopaminergic neurons to degenerate during PD. Targeting ion channels offers an attractive mechanistic strategy to combat the process of neurodegeneration. In this review, we highlight the evidence pointing to the role of various ion channels in driving the PD processes. In addition, we also discuss the various drugs or compounds that target the ion channels and have shown neuroprotective potential in the in-vitro and in-vivo models of PD. We also discuss the current clinical status of various drugs targeting the ion channels in the context of PD.
Collapse
Affiliation(s)
- Neha Hanna Daniel
- School of Biology, Indian Institute of Science Education and Research (IISER)-Thiruvananthapuram, Kerala, 695551, India
| | - Ananya Aravind
- School of Biology, Indian Institute of Science Education and Research (IISER)-Thiruvananthapuram, Kerala, 695551, India
| | - Poonam Thakur
- School of Biology, Indian Institute of Science Education and Research (IISER)-Thiruvananthapuram, Kerala, 695551, India.
| |
Collapse
|
12
|
Han Y, Lyman KA, Foote KM, Chetkovich DM. The structure and function of TRIP8b, an auxiliary subunit of hyperpolarization-activated cyclic-nucleotide gated channels. Channels (Austin) 2020; 14:110-122. [PMID: 32189562 PMCID: PMC7153792 DOI: 10.1080/19336950.2020.1740501] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/18/2020] [Accepted: 02/21/2020] [Indexed: 02/08/2023] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are expressed throughout the mammalian central nervous system (CNS). These channels have been implicated in a wide range of diseases, including Major Depressive Disorder and multiple subtypes of epilepsy. The diversity of functions that HCN channels perform is in part attributable to differences in their subcellular localization. To facilitate a broad range of subcellular distributions, HCN channels are bound by auxiliary subunits that regulate surface trafficking and channel function. One of the best studied auxiliary subunits is tetratricopeptide-repeat containing, Rab8b-interacting protein (TRIP8b). TRIP8b is an extensively alternatively spliced protein whose only known function is to regulate HCN channels. TRIP8b binds to HCN pore-forming subunits at multiple interaction sites that differentially regulate HCN channel function and subcellular distribution. In this review, we summarize what is currently known about the structure and function of TRIP8b isoforms with an emphasis on the role of this auxiliary subunit in health and disease.
Collapse
Affiliation(s)
- Ye Han
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kyle A. Lyman
- Department of Neurology, Stanford University, Palo Alto, CA, USA
| | - Kendall M. Foote
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Dane M. Chetkovich
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
13
|
Pinner JFL, Coffman BA, Stephen JM. Covariation Between Brain Function (MEG) and Structure (DTI) Differentiates Adolescents with Fetal Alcohol Spectrum Disorder from Typically Developing Controls. Neuroscience 2020; 449:74-87. [PMID: 33010344 DOI: 10.1016/j.neuroscience.2020.09.053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 07/29/2020] [Accepted: 09/24/2020] [Indexed: 01/22/2023]
Abstract
The behavioral, cognitive, and sensory difficulties experienced by individuals exposed to alcohol prenatally currently fail to provide early identification for fetal alcohol spectrum disorder (FASD). Attempting to advance this pursuit through a multivariate analysis, we collected magnetoencephalography (MEG) data during auditory, somatosensory, visual paradigms, DTI, and behavior in adolescents ages 12-21 years (FASD: N = 13; HC: N = 20). We assessed the relationship between brain function (MEG) and structure (fractional anisotropy (FA)) utilizing joint independent component analysis (jICA), and examined how this measure relates to behavior. We identified 5 components that reveal group differences in co-variation between MEG and FA. For example, component 5 (t = 3.162, p = 0.003, Hedges' g = 1.13) contained MEG activity corresponding to all three sensory modalities, most robustly in occipital lobes, and DTI-derived cerebellar FA, underlying the role of the cerebellum in sensory processing. Further, in HCs component 5's loading factor was positively correlated with verbal ability (r = 0.646, p = 0.002), indicating higher covariation was associated with better verbal performance. Interestingly, this relationship is lacking in FASD (r = 0.009, p = 0.979). Also, component 5 loading factor negatively correlated with impulsivity (r = -0.527, p = 0.002), indicating that stronger function-structure associations were associated with individuals with lower impulsivity. These findings suggest that multimodal integration of MEG and FA provides novel associations between structure and function that may help differentiate adolescents with FASD from HC.
Collapse
Affiliation(s)
- John F L Pinner
- The Mind Research Network, Albuquerque, NM, United States; Department of Psychology, The University of New Mexico, Albuquerque, NM, United States.
| | - Brian A Coffman
- The Mind Research Network, Albuquerque, NM, United States; The University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | | |
Collapse
|
14
|
Hao XM, Xu R, Chen AQ, Sun FJ, Wang Y, Liu HX, Chen H, Xue Y, Chen L. Endogenous HCN Channels Modulate the Firing Activity of Globus Pallidus Neurons in Parkinsonian Animals. Front Aging Neurosci 2019; 11:190. [PMID: 31402860 PMCID: PMC6670024 DOI: 10.3389/fnagi.2019.00190] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 07/11/2019] [Indexed: 11/13/2022] Open
Abstract
The globus pallidus occupies a critical position in the indirect pathway of the basal ganglia motor control system. Hyperpolarization-activated cyclic-nucleotide gated (HCN) channels play an important role in the modulation of neuronal excitability. In vivo extracellular single unit recording, behavioral test and immunohistochemistry were performed to explore the possible modulation of endogenous HCN channels in the globus pallidus under parkinsonian states. In MPTP parkinsonian mice, micro-pressure application of the selective HCN channel antagonist, ZD7288, decreased the firing rate in 10 out of the 28 pallidal neurons, while increased the firing rate in another 15 out of the 28 neurons. In 6-OHDA parkinsonian rats, ZD7288 also bidirectionally regulated the spontaneous firing activity of the globus pallidus neurons. The proportion of pallidal neurons with ZD7288-induced slowing of firing rate tended to reduce in both parkinsonian animals. Morphological studies revealed a weaker staining of HCN channels in the globus pallidus under parkinsonian state. Finally, behavioral study demonstrated that intrapallidal microinjection of ZD7288 alleviated locomotor deficits in MPTP parkinsonian mice. These results suggest that endogenous HCN channels modulate the activities of pallidal neurons under parkinsonian states.
Collapse
Affiliation(s)
- Xiao-Meng Hao
- Department of Physiology, Qingdao University, Qingdao, China
| | - Rong Xu
- Department of Physiology, Qingdao University, Qingdao, China
| | - An-Qi Chen
- Department of Physiology, Qingdao University, Qingdao, China
| | - Feng-Jiao Sun
- Department of Physiology, Qingdao University, Qingdao, China
| | - Ying Wang
- Department of Physiology, Qingdao University, Qingdao, China
| | - Hong-Xia Liu
- Department of Physiology, Qingdao University, Qingdao, China
| | - Hua Chen
- Department of Pathology, Qingdao Municipal Hospital, Qingdao, China
| | - Yan Xue
- Department of Physiology, Qingdao University, Qingdao, China
| | - Lei Chen
- Department of Physiology, Qingdao University, Qingdao, China
| |
Collapse
|
15
|
|
16
|
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
|
17
|
Mukunda CL, Narayanan R. Degeneracy in the regulation of short-term plasticity and synaptic filtering by presynaptic mechanisms. J Physiol 2017; 595:2611-2637. [PMID: 28026868 DOI: 10.1113/jp273482] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/13/2016] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS We develop a new biophysically rooted, physiologically constrained conductance-based synaptic model to mechanistically account for short-term facilitation and depression, respectively through residual calcium and transmitter depletion kinetics. We address the specific question of how presynaptic components (including voltage-gated ion channels, pumps, buffers and release-handling mechanisms) and interactions among them define synaptic filtering and short-term plasticity profiles. Employing global sensitivity analyses (GSAs), we show that near-identical synaptic filters and short-term plasticity profiles could emerge from disparate presynaptic parametric combinations with weak pairwise correlations. Using virtual knockout models, a technique to address the question of channel-specific contributions within the GSA framework, we unveil the differential and variable impact of each ion channel on synaptic physiology. Our conclusions strengthen the argument that parametric and interactional complexity in biological systems should not be viewed from the limited curse-of-dimensionality standpoint, but from the evolutionarily advantageous perspective of providing functional robustness through degeneracy. ABSTRACT Information processing in neurons is known to emerge as a gestalt of pre- and post-synaptic filtering. However, the impact of presynaptic mechanisms on synaptic filters has not been quantitatively assessed. Here, we developed a biophysically rooted, conductance-based model synapse that was endowed with six different voltage-gated ion channels, calcium pumps, calcium buffer and neurotransmitter-replenishment mechanisms in the presynaptic terminal. We tuned our model to match the short-term plasticity profile and band-pass structure of Schaffer collateral synapses, and performed sensitivity analyses to demonstrate that presynaptic voltage-gated ion channels regulated synaptic filters through changes in excitability and associated calcium influx. These sensitivity analyses also revealed that calcium- and release-control mechanisms were effective regulators of synaptic filters, but accomplished this without changes in terminal excitability or calcium influx. Next, to perform global sensitivity analysis, we generated 7000 randomized models spanning 15 presynaptic parameters, and computed eight different physiological measurements in each of these models. We validated these models by applying experimentally obtained bounds on their measurements, and found 104 (∼1.5%) models to match the validation criteria for all eight measurements. Analysing these valid models, we demonstrate that analogous synaptic filters emerge from disparate combinations of presynaptic parameters exhibiting weak pairwise correlations. Finally, using virtual knockout models, we establish the variable and differential impact of different presynaptic channels on synaptic filters, underlining the critical importance of interactions among different presynaptic components in defining synaptic physiology. Our results have significant implications for protein-localization strategies required for physiological robustness and for degeneracy in long-term synaptic plasticity profiles.
Collapse
Affiliation(s)
- Chinmayee L Mukunda
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, India
| | - Rishikesh Narayanan
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
18
|
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
|
19
|
Goitia B, Rivero-Echeto MC, Weisstaub NV, Gingrich JA, Garcia-Rill E, Bisagno V, Urbano FJ. Modulation of GABA release from the thalamic reticular nucleus by cocaine and caffeine: role of serotonin receptors. J Neurochem 2015; 136:526-35. [PMID: 26484945 DOI: 10.1111/jnc.13398] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 09/23/2015] [Accepted: 10/02/2015] [Indexed: 11/30/2022]
Abstract
Serotonin receptors are targets of drug therapies for a variety of neuropsychiatric and neurodegenerative disorders. Cocaine inhibits the re-uptake of serotonin (5-HT), dopamine, and noradrenaline, whereas caffeine blocks adenosine receptors and opens ryanodine receptors in the endoplasmic reticulum. We studied how 5-HT and adenosine affected spontaneous GABAergic transmission from thalamic reticular nucleus. We combined whole-cell patch clamp recordings of miniature inhibitory post-synaptic currents (mIPSCs) in ventrobasal thalamic neurons during local (puff) application of 5-HT in wild type (WT) or knockout mice lacking 5-HT2A receptors (5-HT2A -/-). Inhibition of mIPSCs frequency by low (10 μM) and high (100 μM) 5-HT concentrations was observed in ventrobasal neurons from 5-HT2A -/- mice. In WT mice, only 100 μM 5-HT significantly reduced mIPSCs frequency. In 5-HT2A -/- mice, NAN-190, a specific 5-HT1A antagonist, prevented the 100 μM 5-HT inhibition while blocking H-currents that prolonged inhibition during post-puff periods. The inhibitory effects of 100 μM 5-HT were enhanced in cocaine binge-treated 5-HT2A -/- mice. Caffeine binge treatment did not affect 5-HT-mediated inhibition. Our findings suggest that both 5-HT1A and 5-HT2A receptors are present in pre-synaptic thalamic reticular nucleus terminals. Serotonergic-mediated inhibition of GABA release could underlie aberrant thalamocortical physiology described after repetitive consumption of cocaine. Our findings suggest that both 5-HT1A , 5-HT2A and A1 receptors are present in pre-synaptic TRN terminals. 5-HT1A and A1 receptors would down-regulate adenylate cyclase, whereas 5-HT1A would also increase the probability of the opening of G-protein-activated inwardly rectifying K(+) channels (GIRK). Sustained opening of GIRK channels would hyperpolarize pre-synaptic terminals activating H-currents, resulting in less GABA release. 5-HT2A -would activate PLC and IP3 , increasing intracellular [Ca(2+) ] and thus facilitating GABA release.
Collapse
Affiliation(s)
- Belén Goitia
- Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado" (DFBMC) Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-CONICET-UBA), Universidad de Buenos Aires, Ciudad Universitaria, Ciudad de Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - María Celeste Rivero-Echeto
- Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado" (DFBMC) Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-CONICET-UBA), Universidad de Buenos Aires, Ciudad Universitaria, Ciudad de Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Noelia V Weisstaub
- Grupo de Neurociencia de Sistemas, Departamento de Fisiología, Facultad de Medicina, Instituto de Fisiología y Biofísica (IFIBIO), UBA, Ciudad de Buenos Aires, Argentina
| | - Jay A Gingrich
- Division of Developmental Neuroscience, Columbia University and the NYSPI, Sackler Institute for Developmental Psychobiology, New York City, New York, USA
| | - Edgar Garcia-Rill
- Department of Neurobiology and Developmental Sciences, Center for Translational Neuroscience, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Verónica Bisagno
- Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Francisco J Urbano
- Departamento de Fisiología, Biología Molecular y Celular "Dr. Héctor Maldonado" (DFBMC) Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-CONICET-UBA), Universidad de Buenos Aires, Ciudad Universitaria, Ciudad de Buenos Aires, Argentina
| |
Collapse
|
20
|
Impact of Hyperpolarization-activated, Cyclic Nucleotide-gated Cation Channel Type 2 for the Xenon-mediated Anesthetic Effect: Evidence from In Vitro and In Vivo Experiments. Anesthesiology 2015; 122:1047-59. [PMID: 25782754 DOI: 10.1097/aln.0000000000000635] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND The thalamus is thought to be crucially involved in the anesthetic state. Here, we investigated the effect of the inhaled anesthetic xenon on stimulus-evoked thalamocortical network activity and on excitability of thalamocortical neurons. Because hyperpolarization-activated, cyclic nucleotide-gated cation (HCN) channels are key regulators of neuronal excitability in the thalamus, the effect of xenon on HCN channels was examined. METHODS The effects of xenon on thalamocortical network activity were investigated in acutely prepared brain slices from adult wild-type and HCN2 knockout mice by means of voltage-sensitive dye imaging. The influence of xenon on single-cell excitability in brain slices was investigated using the whole-cell patch-clamp technique. Effects of xenon on HCN channels were verified in human embryonic kidney cells expressing HCN2 channels. RESULTS Xenon concentration-dependently diminished thalamocortical signal propagation. In neurons, xenon reduced HCN channel-mediated Ih current amplitude by 33.4 ± 12.2% (at -133 mV; n = 7; P = 0.041) and caused a left-shift in the voltage of half-maximum activation (V1/2) from -98.8 ± 1.6 to -108.0 ± 4.2 mV (n = 8; P = 0.035). Similar effects were seen in human embryonic kidney cells. The impairment of HCN channel function was negligible when intracellular cyclic adenosine monophosphate level was increased. Using HCN2 mice, we could demonstrate that xenon did neither attenuate in vitro thalamocortical signal propagation nor did it show sedating effects in vivo. CONCLUSIONS Here, we clearly showed that xenon impairs HCN2 channel function, and this impairment is dependent on intracellular cyclic adenosine monophosphate levels. We provide evidence that this effect reduces thalamocortical signal propagation and probably contributes to the hypnotic properties of xenon.
Collapse
|
21
|
Chen L, Xu R, Sun FJ, Xue Y, Hao XM, Liu HX, Wang H, Chen XY, Liu ZR, Deng WS, Han XH, Xie JX, Yung WH. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels regulate firing of globus pallidus neurons in vivo. Mol Cell Neurosci 2015; 68:46-55. [PMID: 25858108 DOI: 10.1016/j.mcn.2015.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 03/26/2015] [Accepted: 04/03/2015] [Indexed: 01/27/2023] Open
Abstract
The globus pallidus plays a significant role in motor control under both health and pathological states. Recent studies have revealed that hyperpolarization-activated cyclic nucleotide-gated (HCN) channels occupy a critical position in globus pallidus pacemaking activity. Morphological studies have shown the expression of HCN channels in the globus pallidus. To investigate the in vivo effects of HCN channels in the globus pallidus, extracellular recordings and behavioral tests were performed in the present study. In normal rats, micro-pressure ejection of 0.05mM ZD7288, the selective HCN channel blocker, decreased the frequency of spontaneous firing in 21 out of the 40 pallidal neurons. The average decrease was 50.4±5.4%. Interestingly, in another 18 out of the 40 pallidal neurons, ZD7288 increased the firing rate by 137.1±27.6%. Similar bidirectional modulation on the firing rate was observed by a higher concentration of ZD7288 (0.5mM) as well as another HCN channel blocker, CsCl. Furthermore, activation of HCN channels by 8-Br-cAMP increased the firing rate by 63.0±9.3% in 15 out of the 25 pallidal neurons and decreased the firing rate by 46.9±9.4% in another 8 out of the 25 pallidal neurons. Further experiments revealed that modulation of glutamatergic but not GABAergic transmission may be involved in ZD7288-induced increase in firing rate. Consistent with electrophysiological results, further studies revealed that modulation of HCN channels also had bidirectional effects on behavior. Taken together, the present studies suggest that HCN channels may modulate the activity of pallidal neurons by different pathways in vivo.
Collapse
Affiliation(s)
- Lei Chen
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao 266071, China.
| | - Rong Xu
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao 266071, China
| | - Feng-Jiao Sun
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao 266071, China
| | - Yan Xue
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao 266071, China
| | - Xiao-Meng Hao
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao 266071, China
| | - Hong-Xia Liu
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao 266071, China
| | - Hua Wang
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao 266071, China
| | - Xin-Yi Chen
- Department of Neurology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Zi-Ran Liu
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao 266071, China
| | - Wen-Shuai Deng
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao 266071, China
| | - Xiao-Hua Han
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao 266071, China
| | - Jun-Xia Xie
- Department of Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao 266071, China
| | - Wing-Ho Yung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| |
Collapse
|
22
|
Williams SB, Hablitz JJ. Differential modulation of repetitive firing and synchronous network activity in neocortical interneurons by inhibition of A-type K(+) channels and Ih. Front Cell Neurosci 2015; 9:89. [PMID: 25852481 PMCID: PMC4364302 DOI: 10.3389/fncel.2015.00089] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 02/26/2015] [Indexed: 12/04/2022] Open
Abstract
GABAergic interneurons provide the main source of inhibition in the neocortex and are important in regulating neocortical network activity. In the presence 4-aminopyridine (4-AP), CNQX, and D-APV, large amplitude GABAA-receptor mediated depolarizing responses were observed in the neocortex. GABAergic networks are comprised of several types of interneurons, each with its own protein expression pattern, firing properties, and inhibitory role in network activity. Voltage-gated ion channels, especially A-type K(+) channels, differentially regulate passive membrane properties, action potential (AP) waveform, and repetitive firing properties in interneurons depending on their composition and localization. HCN channels are known modulators of pyramidal cell intrinsic excitability and excitatory network activity. Little information is available regarding how HCN channels functionally modulate excitability of individual interneurons and inhibitory networks. In this study, we examined the effect of 4-AP on intrinsic excitability of fast-spiking basket cells (FS-BCs) and Martinotti cells (MCs). 4-AP increased the duration of APs in both FS-BCs and MCs. The repetitive firing properties of MCs were differentially affected compared to FS-BCs. We also examined the effect of Ih inhibition on synchronous GABAergic depolarizations and synaptic integration of depolarizing IPSPs. ZD 7288 enhanced the amplitude and area of evoked GABAergic responses in both cell types. Similarly, the frequency and area of spontaneous GABAergic depolarizations in both FS-BCs and MCs were increased in presence of ZD 7288. Synaptic integration of IPSPs in MCs was significantly enhanced, but remained unaltered in FS-BCs. These results indicate that 4-AP differentially alters the firing properties of interneurons, suggesting MCs and FS-BCs may have unique roles in GABAergic network synchronization. Enhancement of GABAergic network synchronization by ZD 7288 suggests that HCN channels attenuate inhibitory network activity.
Collapse
Affiliation(s)
| | - John J. Hablitz
- Department of Neurobiology, Civitan International Research Center and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, ALUSA
| |
Collapse
|
23
|
Godar SC, Mosher LJ, Di Giovanni G, Bortolato M. Animal models of tic disorders: a translational perspective. J Neurosci Methods 2014; 238:54-69. [PMID: 25244952 DOI: 10.1016/j.jneumeth.2014.09.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 09/09/2014] [Accepted: 09/11/2014] [Indexed: 12/30/2022]
Abstract
Tics are repetitive, sudden movements and/or vocalizations, typically enacted as maladaptive responses to intrusive premonitory urges. The most severe tic disorder, Tourette syndrome (TS), is a childhood-onset condition featuring multiple motor and at least one phonic tic for a duration longer than 1 year. The pharmacological treatment of TS is mainly based on antipsychotic agents; while these drugs are often effective in reducing tic severity and frequency, their therapeutic compliance is limited by serious motor and cognitive side effects. The identification of novel therapeutic targets and development of better treatments for tic disorders is conditional on the development of animal models with high translational validity. In addition, these experimental tools can prove extremely useful to test hypotheses on the etiology and neurobiological bases of TS and related conditions. In recent years, the translational value of these animal models has been enhanced, thanks to a significant re-organization of our conceptual framework of neuropsychiatric disorders, with a greater focus on endophenotypes and quantitative indices, rather than qualitative descriptors. Given the complex and multifactorial nature of TS and other tic disorders, the selection of animal models that can appropriately capture specific symptomatic aspects of these conditions can pose significant theoretical and methodological challenges. In this article, we will review the state of the art on the available animal models of tic disorders, based on genetic mutations, environmental interventions as well as pharmacological manipulations. Furthermore, we will outline emerging lines of translational research showing how some of these experimental preparations have led to significant progress in the identification of novel therapeutic targets for tic disorders.
Collapse
Affiliation(s)
- Sean C Godar
- Department of Pharmacology and Toxicology, School of Pharmacy; University of Kansas, Lawrence, KS, USA
| | - Laura J Mosher
- Department of Pharmacology and Toxicology, School of Pharmacy; University of Kansas, Lawrence, KS, USA
| | - Giuseppe Di Giovanni
- Department of Physiology and Biochemistry, University of Malta, Msida, Malta; School of Biosciences, Cardiff University, Cardiff, UK
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, School of Pharmacy; University of Kansas, Lawrence, KS, USA; Consortium for Translational Research on Aggression and Drug Abuse (ConTRADA), University of Kansas, Lawrence, KS, USA.
| |
Collapse
|
24
|
Shah MM. Cortical HCN channels: function, trafficking and plasticity. J Physiol 2014; 592:2711-9. [PMID: 24756635 PMCID: PMC4104471 DOI: 10.1113/jphysiol.2013.270058] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 04/15/2014] [Indexed: 12/26/2022] Open
Abstract
The hyperpolarization-activated cyclic nucleotide-gated (HCN) channels belong to the superfamily of voltage-gated potassium ion channels. They are, however, activated by hyperpolarizing potentials and are permeable to cations. Four HCN subunits have been cloned, of which HCN1 and HCN2 subunits are predominantly expressed in the cortex. These subunits are principally located in pyramidal cell dendrites, although they are also found at lower concentrations in the somata of pyramidal neurons as well as other neuron subtypes. HCN channels are actively trafficked to dendrites by binding to the chaperone protein TRIP8b. Somato-dendritic HCN channels in pyramidal neurons modulate spike firing and synaptic potential integration by influencing the membrane resistance and resting membrane potential. Intriguingly, HCN channels are present in certain cortical axons and synaptic terminals too. Here, they regulate synaptic transmission but the underlying mechanisms appear to vary considerably amongst different synaptic terminals. In conclusion, HCN channels are expressed in multiple neuronal subcellular compartments in the cortex, where they have a diverse and complex effect on neuronal excitability.
Collapse
Affiliation(s)
- Mala M Shah
- Department of Pharmacology, UCL School of Pharmacy, London, UK
| |
Collapse
|
25
|
He C, Chen F, Li B, Hu Z. Neurophysiology of HCN channels: From cellular functions to multiple regulations. Prog Neurobiol 2014; 112:1-23. [DOI: 10.1016/j.pneurobio.2013.10.001] [Citation(s) in RCA: 230] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Revised: 10/01/2013] [Accepted: 10/07/2013] [Indexed: 12/18/2022]
|
26
|
Merrison-Hort R, Borisyuk R. The emergence of two anti-phase oscillatory neural populations in a computational model of the Parkinsonian globus pallidus. Front Comput Neurosci 2013; 7:173. [PMID: 24348374 PMCID: PMC3844854 DOI: 10.3389/fncom.2013.00173] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 11/12/2013] [Indexed: 01/28/2023] Open
Abstract
Experiments in rodent models of Parkinson's disease have demonstrated a prominent increase of oscillatory firing patterns in neurons within the Parkinsonian globus pallidus (GP) which may underlie some of the motor symptoms of the disease. There are two main pathways from the cortex to GP: via the striatum and via the subthalamic nucleus (STN), but it is not known how these inputs sculpt the pathological pallidal firing patterns. To study this we developed a novel neural network model of conductance-based spiking pallidal neurons with cortex-modulated input from STN neurons. Our results support the hypothesis that entrainment occurs primarily via the subthalamic pathway. We find that as a result of the interplay between excitatory input from the STN and mutual inhibitory coupling between GP neurons, a homogeneous population of GP neurons demonstrates a self-organizing dynamical behavior where two groups of neurons emerge: one spiking in-phase with the cortical rhythm and the other in anti-phase. This finding mirrors what is seen in recordings from the GP of rodents that have had Parkinsonism induced via brain lesions. Our model also includes downregulation of Hyperpolarization-activated Cyclic Nucleotide-gated (HCN) channels in response to burst firing of GP neurons, since this has been suggested as a possible mechanism for the emergence of Parkinsonian activity. We found that the downregulation of HCN channels provides even better correspondence with experimental data but that it is not essential in order for the two groups of oscillatory neurons to appear. We discuss how the influence of inhibitory striatal input will strengthen our results.
Collapse
Affiliation(s)
- Robert Merrison-Hort
- Centre for Robotics and Neural Systems, School of Computing and Mathematics, The University of Plymouth Plymouth, UK
| | - Roman Borisyuk
- Centre for Robotics and Neural Systems, School of Computing and Mathematics, The University of Plymouth Plymouth, UK ; Neural Networks Laboratory, Institute of Mathematical Problems in Biology, Russian Academy of Sciences Pushchino, Russia
| |
Collapse
|
27
|
Rivera-Arconada I, Roza C, Lopez-Garcia JA. Characterization of hyperpolarization-activated currents in deep dorsal horn neurons of neonate mouse spinal cord in vitro. Neuropharmacology 2013; 70:148-55. [PMID: 23376246 DOI: 10.1016/j.neuropharm.2013.01.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Revised: 12/13/2012] [Accepted: 01/17/2013] [Indexed: 10/27/2022]
Abstract
Emerging evidence suggests that blockade of hyperpolarization-activated current (Ih) produces analgesia acting at peripheral sites. However, little is known about the role of this current in central pain-processing structures. The aim of the present work was to characterize the Ih in deep dorsal horn neurons and to assess the role of the current in the transmission of somatosensory signals across spinal circuits. To these purpose in vitro preparations of the spinal cord from mice pups were used in combination with whole cell recordings to characterize the current in native neurons. Extracellular recordings from sensory and motor pathways were performed to assess the role of the current in spinal somatosensory processing. Cesium chloride and ZD7288 were used as current blockers. Most deep dorsal horn neurons showed a functional Ih that was blocked by ZD7288 and cesium. Ih blockade caused hyperpolarization, increased input resistance and potentiation of synaptic responses. Excitatory effects of Ih blockade on synaptic transmission were confirmed in projecting anterolateral axons and ventral roots. Ih modulation by cAMP produced a rightward shift in the voltage dependency curve and blocked excitatory effects of ZD7288 on sensory pathways. Results indicate that Ih currents play a stabilizing role in the spinal cord controlling transmission across sensory and motor spinal pathways via cellular effects on input resistance and excitability. In addition, results suggest that current modulation may alter significantly the role of the current in somatosensory processing.
Collapse
Affiliation(s)
- Ivan Rivera-Arconada
- Departamento de Fisiología, Edificio de Medicina, Universidad de Alcala, Alcala de Henares, 28871 Madrid, Spain
| | | | | |
Collapse
|
28
|
TRIP8b-independent trafficking and plasticity of adult cortical presynaptic HCN1 channels. J Neurosci 2013; 32:14835-48. [PMID: 23077068 DOI: 10.1523/jneurosci.1544-12.2012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are subthreshold activated voltage-gated ion channels. In the cortex, these channels are predominantly expressed in dendrites where they significantly modify dendritic intrinsic excitability as well synaptic potential shapes and integration. HCN channel trafficking to dendrites is regulated by the protein, TRIP8b. Additionally, altered TRIP8b expression may be one mechanism underlying seizure-induced dendritic HCN channel plasticity. HCN channels, though, are also located in certain mature cortical synaptic terminals, where they play a vital role in modulating synaptic transmission. In this study, using electrophysiological recordings as well as electron microscopy we show that presynaptic, but not dendritic, cortical HCN channel expression and function is comparable in adult TRIP8b-null mice and wild-type littermates. We further investigated whether presynaptic HCN channels undergo seizure-dependent plasticity. We found that, like dendritic channels, wild-type presynaptic HCN channel function was persistently decreased following induction of kainic acid-induced seizures. Since TRIP8b does not affect presynaptic HCN subunit trafficking, seizure-dependent plasticity of these cortical HCN channels is not conditional upon TRIP8b. Our results, thus, suggest that the molecular mechanisms underlying HCN subunit targeting, expression and plasticity in adult neurons is compartment selective, providing a means by which pre- and postsynaptic processes that are critically dependent upon HCN channel function may be distinctly influenced.
Collapse
|
29
|
Yeh CB, Shui HA, Chu TH, Chen YA, Tsung HC, Shyu JF. Hyperpolarisation-activated cyclic nucleotide channel 4 (HCN4) involvement in Tourette's syndrome autoimmunity. J Neuroimmunol 2012; 250:18-26. [PMID: 22683190 DOI: 10.1016/j.jneuroim.2012.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 05/08/2012] [Accepted: 05/12/2012] [Indexed: 11/17/2022]
Abstract
OBJECTIVES We previously found that antibodies in Tourette's syndrome (TS) patients' sera reacted with a 120 kDa protein from rat brain tissue. Here, we sought to identify this protein and determine if it was involved in TS pathogenesis. METHODS The 120 kDa protein was identified using immunoprecipitation, Western blotting, and mass spectrometry. ELISAs were used to quantify anti-120 kDa protein antibodies in serum of interest using samples from 32 TS patients, 47 patients with attention deficit hyperactivity disorder (ADHD) and 14 healthy controls. Involvement of the 120 kDa protein in TS was confirmed using co-localisation assays with GH3 cells. TS sera were micro-infused into SD rats' brain striatum and their stereotypical behaviours were monitored. RESULTS The brain protein was identified as hyperpolarisation-activated cyclic nucleotide channel 4 (HCN4). TS patients' sera contained significantly more anti-HCN4 antibodies than ADHD patient and control sera. After microinfusing TS serum, SD rats exhibited increased stereotyped tic behaviours, which were correlated with the amount of infused anti-HCN4 antibody. CONCLUSIONS Anti-HCN4 antibodies in the brain might contribute to the pathogenesis of tic symptoms in TS patients. However, further studies are needed to investigate the validity of this animal model of TS induced by microinfusing anti-HCN4 antibody.
Collapse
Affiliation(s)
- Chin-Bin Yeh
- Department of Psychiatry, Tri-Service General Hospital, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
30
|
The Role of HCN Channels on Membrane Excitability in the Nervous System. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:619747. [PMID: 22934165 PMCID: PMC3425855 DOI: 10.1155/2012/619747] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 06/19/2012] [Indexed: 01/07/2023]
Abstract
Hyperpolarization-activated and cyclic nucleotide-gated (HCN) channels were first reported in heart cells and are recently known to be involved in a variety of neural functions in healthy and diseased brains. HCN channels generate inward currents when the membrane potential is hyperpolarized. Voltage dependence of HCN channels is regulated by intracellular signaling cascades, which contain cyclic AMP, PIP(2), and TRIP8b. In addition, voltage-gated potassium channels have a strong influence on HCN channel activity. Because of these funny features, HCN channel currents, previously called funny currents, can have a wide range of functions that are determined by a delicate balance of modulatory factors. These multifaceted features also make it difficult to predict and elucidate the functional role of HCN channels in actual neurons. In this paper, we focus on the impacts of HCN channels on neural activity. The functions of HCN channels reported previously will be summarized, and their mechanisms will be explained by using numerical simulation of simplified model neurons.
Collapse
|
31
|
Modulation of in vivo GABA-evoked responses by nitric oxide-active compounds in the globus pallidus of rat. J Neural Transm (Vienna) 2012; 119:911-21. [DOI: 10.1007/s00702-011-0760-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 12/25/2011] [Indexed: 10/14/2022]
|
32
|
Goeritz ML, Ouyang Q, Harris-Warrick RM. Localization and function of Ih channels in a small neural network. J Neurophysiol 2011; 106:44-58. [PMID: 21490285 PMCID: PMC3129722 DOI: 10.1152/jn.00897.2010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Accepted: 04/07/2011] [Indexed: 11/22/2022] Open
Abstract
Subthreshold ionic currents, which activate below the firing threshold and shape the cell's firing properties, play important roles in shaping neural network activity. We examined the distribution and synaptic roles of the hyperpolarization-activated inward current (I(h)) in the pyloric network of the lobster stomatogastric ganglion (STG). I(h) channels are expressed throughout the STG in a patchy distribution and are highly expressed in the fine neuropil, an area that is rich in synaptic contacts. We performed double labeling for I(h) protein and for the presynaptic marker synaptotagmin. The large majority of labeling in the fine neuropil was adjacent but nonoverlapping, suggesting that I(h) is localized in close proximity to synapses but not in the presynaptic terminals. We compared the pattern of I(h) localization with Shal transient potassium channels, whose expression is coregulated with I(h) in many STG neurons. Unlike I(h), we found significant levels of Shal protein in the soma membrane and the primary neurite. Both proteins were found in the synaptic fine neuropil, but with little evidence of colocalization in individual neurites. We performed electrophysiological experiments to study a potential role for I(h) in regulating synaptic transmission. At a synapse between two identified pyloric neurons, the amplitude of inhibitory postsynaptic potentials (IPSPs) decreased with increasing postsynaptic activation of I(h). Pharmacological block of I(h) restored IPSP amplitudes to levels seen when I(h) was not activated. These experiments suggest that modulation of postsynaptic I(h) might play an important role in the control of synaptic strength in this rhythmogenic neural network.
Collapse
Affiliation(s)
- Marie L Goeritz
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA.
| | | | | |
Collapse
|
33
|
Presynaptic HCN1 channels regulate Cav3.2 activity and neurotransmission at select cortical synapses. Nat Neurosci 2011; 14:478-86. [PMID: 21358644 PMCID: PMC3068302 DOI: 10.1038/nn.2757] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Accepted: 01/13/2011] [Indexed: 11/09/2022]
Abstract
The hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are subthreshold, voltage-gated ion channels that are highly expressed in hippocampal and cortical pyramidal cell dendrites, where they are important for regulating synaptic potential integration and plasticity. We found that HCN1 subunits are also localized to the active zone of mature asymmetric synaptic terminals targeting mouse entorhinal cortical layer III pyramidal neurons. HCN channels inhibited glutamate synaptic release by suppressing the activity of low-threshold voltage-gated T-type (Ca(V)3.2) Ca²(+) channels. Consistent with this, electron microscopy revealed colocalization of presynaptic HCN1 and Ca(V)3.2 subunit. This represents a previously unknown mechanism by which HCN channels regulate synaptic strength and thereby neural information processing and network excitability.
Collapse
|
34
|
Selective participation of somatodendritic HCN channels in inhibitory but not excitatory synaptic integration in neurons of the subthalamic nucleus. J Neurosci 2010; 30:16025-40. [PMID: 21106841 DOI: 10.1523/jneurosci.3898-10.2010] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The activity patterns of subthalamic nucleus (STN) neurons are intimately linked to motor function and dysfunction and arise through the complex interaction of intrinsic properties and inhibitory and excitatory synaptic inputs. In many neurons, hyperpolarization-activated cyclic nucleotide-gated (HCN) channels play key roles in intrinsic excitability and synaptic integration both under normal conditions and in disease states. However, in STN neurons, which strongly express HCN channels, their roles remain relatively obscure. To address this deficit, complementary molecular and cellular electrophysiological, imaging, and computational approaches were applied to the rat STN. Molecular profiling demonstrated that individual STN neurons express mRNA encoding several HCN subunits, with HCN2 and 3 being the most abundant. Light and electron microscopic analysis showed that HCN2 subunits are strongly expressed and distributed throughout the somatodendritic plasma membrane. Voltage-, current-, and dynamic-clamp analysis, two-photon Ca(2+) imaging, and computational modeling revealed that HCN channels are activated by GABA(A) receptor-mediated inputs and thus limit synaptic hyperpolarization and deinactivation of low-voltage-activated Ca(2+) channels. Although HCN channels also limited the temporal summation of EPSPs, generated through two-photon uncaging of glutamate, this action was largely shunted by GABAergic inhibition that was necessary for HCN channel activation. Together the data demonstrate that HCN channels in STN neurons selectively counteract GABA(A) receptor-mediated inhibition arising from the globus pallidus and thus promote single-spike activity rather than rebound burst firing.
Collapse
|
35
|
HCN channel activity-dependent modulation of inhibitory synaptic transmission in the rat basolateral amygdala. Biochem Biophys Res Commun 2010; 404:952-7. [PMID: 21185265 DOI: 10.1016/j.bbrc.2010.12.087] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2010] [Accepted: 12/18/2010] [Indexed: 11/23/2022]
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are expressed in the central nervous system and play a regulatory role in neuronal excitability. In the present study, we examined a physiological role of HCN channels in the rat basolateral amygdala (BLA). In vitro electrophysiological studies showed that ZD7288 decreased spontaneous inhibitory postsynaptic current (sIPSC) without changing miniature IPSC (mIPSC). HCN channel blockade also attenuated feedback inhibitions in BLA principal neurons. However, blockade of HCN channel had little effects on spontaneous excitatory postsynaptic current (sEPSC) and mEPSC. Therefore, HCN channel appeared to decrease BLA excitability by increasing the action potential-dependent inhibitory control over the BLA principal neurons. Anxiety is reported to be influenced by neuronal excitability in the BLA and inhibitory synaptic transmission is thought to play a pivotal role in regulating overall excitability of the amygdala. As expected, blockade of HCN channels by targeted injection of ZD7288 to the BLA increased anxiety-like behavior under elevated plus maze test. Our results suggest that HCN channel activity can modulate the GABAergic synaptic transmission in the BLA, which in turn control the amygdala-related emotional behaviors such as anxiety.
Collapse
|
36
|
Massella A, Gusciglio M, D'Intino G, Sivilia S, Ferraro L, Calzà L, Giardino L. Gabapentin treatment improves motor coordination in a mice model of progressive ataxia. Brain Res 2009; 1301:135-42. [DOI: 10.1016/j.brainres.2009.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 08/27/2009] [Accepted: 09/01/2009] [Indexed: 02/07/2023]
|
37
|
Complex intrinsic membrane properties and dopamine shape spiking activity in a motor axon. J Neurosci 2009; 29:5062-74. [PMID: 19386902 DOI: 10.1523/jneurosci.0716-09.2009] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We studied the peripheral motor axons of the two pyloric dilator (PD) neurons of the stomatogastric ganglion in the lobster, Homarus americanus. Intracellular recordings from the motor nerve showed both fast and slow voltage- and activity-dependent dynamics. During rhythmic bursts, the PD axons displayed changes in spike amplitude and duration. Pharmacological experiments and the voltage dependence of these phenomena suggest that inactivation of sodium and A-type potassium channels are responsible. In addition, the "resting" membrane potential was dependent on ongoing spike or burst activity, with more hyperpolarized values when activity was strong. Nerve stimulations, pharmacological block and current clamp experiments suggest that this is due to a functional antagonism between a slow after-hyperpolarization (sAHP) and inward rectification through hyperpolarization-activated current (IH). Dopamine application resulted in modest depolarization and "ectopic" peripheral spike initiation in the absence of centrally generated activity. This effect was blocked by CsCl and ZD7288, consistent with a role of IH. High frequency nerve stimulation inhibited peripheral spike initiation for several seconds, presumably due to the sAHP. Both during normal bursting activity and antidromic nerve stimulation, the conduction delay over the length of the peripheral nerve changed in a complex manner. This suggests that axonal membrane dynamics can have a substantial effect on the temporal fidelity of spike patterns propagated from a spike initiation site to a synaptic target, and that neuromodulators can influence the extent to which spike patterns are modified.
Collapse
|
38
|
Sims RE, Woodhall GL, Wilson CL, Stanford IM. Functional characterization of GABAergic pallidopallidal and striatopallidal synapses in the rat globus pallidus in vitro. Eur J Neurosci 2009; 28:2401-8. [PMID: 19087170 DOI: 10.1111/j.1460-9568.2008.06546.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
As a central integrator of basal ganglia function, the external segment of the globus pallidus (GP) plays a critical role in the control of voluntary movement. Driven by intrinsic mechanisms and excitatory glutamatergic inputs from the subthalamic nucleus, GP neurons receive GABAergic inhibitory input from the striatum (Str-GP) and from local collaterals of neighbouring pallidal neurons (GP-GP). Here we provide electrophysiological evidence for functional differences between these two inhibitory inputs. The basic synaptic characteristics of GP-GP and Str-GP GABAergic synapses were studied using whole-cell recordings with paired-pulse and train stimulation protocols and variance-mean (VM) analysis. We found (i) IPSC kinetics are consistent with local collaterals innervating the soma and proximal dendrites of GP neurons whereas striatal inputs innervate more distal regions. (ii) Compared to GP-GP synapses Str-GP synapses have a greater paired-pulse ratio, indicative of a lower probability of release. This was confirmed using VM analysis. (iii) In response to 20 and 50 Hz train stimulation, GP-GP synapses are weakly facilitatory in 1 mM external calcium and depressant in 2.4 mM calcium. This is in contrast to Str-GP synapses which display facilitation under both conditions. This is the first quantitative study comparing the properties of GP-GP and Str-GP synapses. The results are consistent with the differential location of these inhibitory synapses and subtle differences in their release probability which underpin stable GP-GP responses and robust short-term facilitation of Str-GP responses. These fundamental differences may provide the physiological basis for functional specialization.
Collapse
Affiliation(s)
- Robert E Sims
- Biomedical Sciences, School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | | | | | | |
Collapse
|
39
|
Constantin S, Wray S. Gonadotropin-releasing hormone-1 neuronal activity is independent of hyperpolarization-activated cyclic nucleotide-modulated channels but is sensitive to protein kinase a-dependent phosphorylation. Endocrinology 2008; 149:3500-11. [PMID: 18372334 PMCID: PMC2453097 DOI: 10.1210/en.2007-1508] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Pulsatile release of GnRH-1 stimulates the anterior pituitary and induces secretion of gonadotropin hormones. GnRH-1 release is modulated by many neurotransmitters that act via G protein-coupled membrane receptors. cAMP is the most ubiquitous effector for these receptors. GnRH-1 neurons express hyperpolarization-activated cyclic nucleotide-modulated (HCN) channel protein in vivo. HCN channels are involved in neuronal pacemaking and can integrate cAMP signals. cAMP-dependent protein kinase (PKA) is also activated by cAMP signals, and PKA-dependent phosphorylation modulates voltage-activated channels. In this report, these two pathways were examined in GnRH-1 neurons as integrators of forskolin (FSK)-induced stimulation. The HCN3 isoform was detected in GnRH-1 neurons obtained from mouse nasal explants. ZD7288, a HCN channel blocker, significantly reduced the efficiency of FSK to stimulate GnRH-1 neurons, whereas blockade of PKA with Rp-adenosine-3',5'-cyclic monophosphorothioate triethylammonium did not attenuate the FSK-induced stimulation. To ensure that disruption of HCN channels on GnRH-1 neurons was responsible for reduction of FSK stimulation, experiments were performed removing gamma-aminobutyric acid (GABA), the major excitatory input to GnRH-1 neurons in nasal explants. Under these conditions, Rp-adenosine-3',5'-cyclic monophosphorothioate triethylammonium, but not ZD7288, altered the FSK-induced response of GnRH-1 neurons. These studies indicate that PKA-dependent phosphorylation is involved in the FSK-induced stimulation of GnRH-1 neurons rather than HCN channels, and HCN channels integrate the FSK-induced stimulation on GABAergic neurons. In addition, blockade of HCN channels did not modify basal GnRH-1 neuronal activity when GABAergic input was intact or removed, negating a role for these channels in basal GABAergic or GnRH-1 neuronal activity.
Collapse
Affiliation(s)
- Stephanie Constantin
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
40
|
Sadek AR, Magill PJ, Bolam JP. A single-cell analysis of intrinsic connectivity in the rat globus pallidus. J Neurosci 2007; 27:6352-62. [PMID: 17567796 PMCID: PMC6672453 DOI: 10.1523/jneurosci.0953-07.2007] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2007] [Revised: 04/09/2007] [Accepted: 04/27/2007] [Indexed: 11/21/2022] Open
Abstract
GABAergic neurons of the globus pallidus (GP) play critical roles in basal ganglia function by virtue of their widespread axonal projections to all parts of the basal ganglia. They also possess local axon collaterals. In view of the importance of GABAergic inputs in sculpting neuronal activity, we quantitatively characterized the local axon collaterals of individual GP neurons by in vivo recording, juxtacellular labeling, reconstruction, and light and electron microscopic analysis in the rat. All labeled GP neurons had similar firing properties and gave rise to local axon collaterals, the main synaptic targets of which were perikarya and primary dendrites. The neurons could be divided into two populations; neurons located within approximately 100 microm of the striatopallidal border ("outer" neurons), which possess a mean of 264 local axonal boutons, and those located approximately 100 microm or more from the striatopallidal border ("inner" neurons), which possess a mean of 581 local axonal boutons. The local axon collaterals gave rise to arborizations close to, or within, the parent dendritic field and arborizations located caudal, medial, and ventral to the parent neuron. The qualitative and quantitative differences in the connectivity of neurons located in the outer and inner regions of the GP underlie a complex microcircuitry that follows an asymmetric rostral to caudal organization. These data suggest that the GP should no longer be considered as an homogeneous relay nucleus that simply transmits striatal information to the subthalamic nucleus and basal ganglia output nuclei, but rather as a structure that can perform complex computations within its borders.
Collapse
Affiliation(s)
- Ahmed R. Sadek
- Medical Research Council Anatomical Neuropharmacology Unit, University of Oxford, Oxford OX1 3TH, United Kingdom
| | - Peter J. Magill
- Medical Research Council Anatomical Neuropharmacology Unit, University of Oxford, Oxford OX1 3TH, United Kingdom
| | - J. Paul Bolam
- Medical Research Council Anatomical Neuropharmacology Unit, University of Oxford, Oxford OX1 3TH, United Kingdom
| |
Collapse
|