1
|
Gong LN, Liu HW, Lai K, Zhang Z, Mao LF, Liu ZQ, Li MX, Yin XL, Liang M, Shi HB, Wang LY, Yin SK. Selective Vulnerability of GABAergic Inhibitory Interneurons to Bilirubin Neurotoxicity in the Neonatal Brain. J Neurosci 2024; 44:e0442242024. [PMID: 39313321 PMCID: PMC11551895 DOI: 10.1523/jneurosci.0442-24.2024] [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: 03/07/2024] [Revised: 08/16/2024] [Accepted: 09/16/2024] [Indexed: 09/25/2024] Open
Abstract
Hyperbilirubinemia (HB) is a key risk factor for hearing loss in neonates, particularly premature infants. Here, we report that bilirubin (BIL)-dependent cell death in the auditory brainstem of neonatal mice of both sexes is significantly attenuated by ZD7288, a blocker for hyperpolarization-activated cyclic nucleotide-gated (HCN) channel-mediated current (I h), or by genetic deletion of HCN1. GABAergic inhibitory interneurons predominantly express HCN1, on which BIL selectively acts to increase their intrinsic excitability and mortality by enhancing HCN1 activity and Ca2+-dependent membrane targeting. Chronic BIL elevation in neonatal mice in vivo increases the fraction of spontaneously active interneurons and their firing frequency, I h, and death, compromising audition at the young adult stage in HCN1+/+, but not in HCN1-/- genotype. We conclude that HB preferentially targets HCN1 to injure inhibitory interneurons, fueling a feedforward loop in which lessening inhibition cascades hyperexcitability, Ca2+ overload, neuronal death, and auditory impairments. These findings rationalize HCN1 as a potential target for managing HB encephalopathy.
Collapse
Affiliation(s)
- Li-Na Gong
- Department of Otorhinolaryngology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Han-Wei Liu
- Department of Otorhinolaryngology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Ke Lai
- Department of Otorhinolaryngology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Programs in Neurosciences & Mental Health, SickKids Research Institute and Department of Physiology, University of Toronto, Toronto, Ontario M5G 1X8, Canada
| | - Zhen Zhang
- Department of Otorhinolaryngology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Lin-Fei Mao
- Department of Otorhinolaryngology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Zhen-Qi Liu
- Department of Otorhinolaryngology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Programs in Neurosciences & Mental Health, SickKids Research Institute and Department of Physiology, University of Toronto, Toronto, Ontario M5G 1X8, Canada
| | - Ming-Xian Li
- Department of Otorhinolaryngology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Department of Head & Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Xin-Lu Yin
- Department of Otorhinolaryngology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Department of Head & Neck Surgery, Renji Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Min Liang
- Department of Otorhinolaryngology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Department of Otorhinolaryngology Head & Neck Surgery, Xinhua Hospital and Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Hai-Bo Shi
- Department of Otorhinolaryngology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Lu-Yang Wang
- Programs in Neurosciences & Mental Health, SickKids Research Institute and Department of Physiology, University of Toronto, Toronto, Ontario M5G 1X8, Canada
| | - Shan-Kai Yin
- Department of Otorhinolaryngology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| |
Collapse
|
2
|
Herstel LJ, Wierenga CJ. Distinct Modulation of I h by Synaptic Potentiation in Excitatory and Inhibitory Neurons. eNeuro 2024; 11:ENEURO.0185-24.2024. [PMID: 39406481 PMCID: PMC11574699 DOI: 10.1523/eneuro.0185-24.2024] [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: 04/29/2024] [Revised: 09/24/2024] [Accepted: 10/02/2024] [Indexed: 11/15/2024] Open
Abstract
Selective modifications in the expression or function of dendritic ion channels regulate the propagation of synaptic inputs and determine the intrinsic excitability of a neuron. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels open upon membrane hyperpolarization and conduct a depolarizing inward current (I h). HCN channels are enriched in the dendrites of hippocampal pyramidal neurons where they regulate the integration of synaptic inputs. Synaptic plasticity can bidirectionally modify dendritic HCN channels in excitatory neurons depending on the strength of synaptic potentiation. In inhibitory neurons, however, the dendritic expression and modulation of HCN channels are largely unknown. In this study, we systematically compared the modulation of I h by synaptic potentiation in hippocampal CA1 pyramidal neurons and stratum radiatum (sRad) interneurons in mouse organotypic cultures. I h properties were similar in inhibitory and excitatory neurons and contributed to resting membrane potential and action potential firing. We found that in sRad interneurons, HCN channels were downregulated after synaptic plasticity, irrespective of the strength of synaptic potentiation. This suggests differential regulation of I h in excitatory and inhibitory neurons, possibly signifying their distinct role in network activity.
Collapse
Affiliation(s)
- Lotte J Herstel
- Biology Department, Faculty of Science, Utrecht University, Utrecht 3584 CH, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen 6525 AJ, the Netherlands
| | - Corette J Wierenga
- Biology Department, Faculty of Science, Utrecht University, Utrecht 3584 CH, the Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen 6525 AJ, the Netherlands
| |
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
|
Hornung E, Robbins S, Srivastava A, Achanta S, Chen J, Cheng ZJ, Schwaber J, Vadigepalli R. Neuromodulatory co-expression in cardiac vagal motor neurons of the dorsal motor nucleus of the vagus. iScience 2024; 27:110549. [PMID: 39171288 PMCID: PMC11338141 DOI: 10.1016/j.isci.2024.110549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/31/2024] [Accepted: 07/16/2024] [Indexed: 08/23/2024] Open
Abstract
Vagal innervation is well known to be crucial to the maintenance of cardiac health, and to protect and recover the heart from injury. Only recently has this role been shown to depend on the activity of the underappreciated dorsal motor nucleus of the vagus (DMV). By combining neural tracing, transcriptomics, and anatomical mapping in male and female Sprague-Dawley rats, we characterize cardiac-specific neuronal phenotypes in the DMV. We find that the DMV cardiac-projecting neurons differentially express pituitary adenylate cyclase-activating polypeptide (PACAP), cocaine- and amphetamine-regulated transcript (CART), and synucleins, as well as evidence that they participate in neuromodulatory co-expression involving catecholamines. The significance of these findings is enhanced by previous knowledge of the role of PACAP at the heart and of the other neuromodulators in peripheral vagal targets.
Collapse
Affiliation(s)
- Eden Hornung
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Shaina Robbins
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ankita Srivastava
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Sirisha Achanta
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jin Chen
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, BMS Building 20, Room 230, 4110 Libra Drive, Orlando, FL 32816, USA
| | - Zixi Jack Cheng
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, BMS Building 20, Room 230, 4110 Libra Drive, Orlando, FL 32816, USA
| | - James Schwaber
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
5
|
Tomasi J, Lisoway AJ, Zai CC, Zai G, Richter MA, Sanches M, Herbert D, Mohiuddin AG, Tiwari AK, Kennedy JL. Genetic and polygenic investigation of heart rate variability to identify biomarkers associated with Anxiety disorders. Psychiatry Res 2024; 338:115982. [PMID: 38850888 DOI: 10.1016/j.psychres.2024.115982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/11/2024] [Accepted: 05/26/2024] [Indexed: 06/10/2024]
Abstract
Given that anxiety disorders (AD) are associated with reduced vagally-mediated heart rate variability (HRV), genetic variants related to HRV may provide insight into anxiety etiology. This study used polygenic risk scores (PRS) to explore the genetic overlap between AD and HRV, and investigated whether HRV-related polymorphisms influence anxiety risk. Resting vagally-mediated HRV was measured using a wearable device in 188 European individuals (AD=101, healthy controls=87). AD PRS was tested for association with resting HRV, and HRV PRS for association with AD. We also investigated 15 significant hits from an HRV genome-wide association study (GWAS) for association with resting HRV and AD and if this association is mediated through resting HRV. The AD PRS and HRV PRS showed nominally significant associations with resting HRV and anxiety disorders, respectively. HRV GWAS variants associated with resting HRV were rs12980262 (NDUFA11), rs2680344 (HCN4), rs4262 and rs180238 (GNG11), and rs10842383 (LINC00477). Mediation analyses revealed that NDUFA11 rs12980262 A-carriers and GNG11 rs180238 and rs4262 C-carriers had higher anxiety risk through lower HRV. This study supports an anxiety-HRV genetic relationship, with HRV-related genetic variants translating to AD. This study encourages exploration of HRV genetics to understand mechanisms and identify novel treatment targets for anxiety.
Collapse
Affiliation(s)
- Julia Tomasi
- Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada.
| | - Amanda J Lisoway
- Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Clement C Zai
- Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Gwyneth Zai
- Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada; General Adult Psychiatry and Health Systems Division, CAMH, Toronto, ON, Canada
| | - Margaret A Richter
- Institute of Medical Science, University of Toronto, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada; Frederick W. Thompson Anxiety Disorders Centre, Department of Psychiatry, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Marcos Sanches
- Biostatistics Core, Centre for Addiction and Mental Health, Toronto, Canada
| | - Deanna Herbert
- Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada
| | - Ayeshah G Mohiuddin
- Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada
| | - Arun K Tiwari
- Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada.
| | - James L Kennedy
- Tanenbaum Centre for Pharmacogenetics, Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada
| |
Collapse
|
6
|
Hou G, Hao M, Duan J, Han MH. The Formation and Function of the VTA Dopamine System. Int J Mol Sci 2024; 25:3875. [PMID: 38612683 PMCID: PMC11011984 DOI: 10.3390/ijms25073875] [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: 10/20/2023] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 04/14/2024] Open
Abstract
The midbrain dopamine system is a sophisticated hub that integrates diverse inputs to control multiple physiological functions, including locomotion, motivation, cognition, reward, as well as maternal and reproductive behaviors. Dopamine is a neurotransmitter that binds to G-protein-coupled receptors. Dopamine also works together with other neurotransmitters and various neuropeptides to maintain the balance of synaptic functions. The dysfunction of the dopamine system leads to several conditions, including Parkinson's disease, Huntington's disease, major depression, schizophrenia, and drug addiction. The ventral tegmental area (VTA) has been identified as an important relay nucleus that modulates homeostatic plasticity in the midbrain dopamine system. Due to the complexity of synaptic transmissions and input-output connections in the VTA, the structure and function of this crucial brain region are still not fully understood. In this review article, we mainly focus on the cell types, neurotransmitters, neuropeptides, ion channels, receptors, and neural circuits of the VTA dopamine system, with the hope of obtaining new insight into the formation and function of this vital brain region.
Collapse
Affiliation(s)
- Guoqiang Hou
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China (M.H.); (J.D.)
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mei Hao
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China (M.H.); (J.D.)
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jiawen Duan
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China (M.H.); (J.D.)
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ming-Hu Han
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China (M.H.); (J.D.)
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
7
|
Yang Z, Kuang Z, Liao H, Gan S, Peng X, Yang H, Wu L. HCN1 pathogenic variants associated with childhood epilepsy in a cohort of Chinese patients. Epileptic Disord 2024; 26:90-97. [PMID: 38009841 DOI: 10.1002/epd2.20182] [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: 10/28/2022] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 11/29/2023]
Abstract
OBJECTIVE HCN ion channel family has a widespread expression in neurons, and recently, increasing studies have demonstrated their roles in epilepsies. METHODS Clinical data of the patients were gathered in a retrospective study. Exon sequencing was used for the patients with unexplained recurrent seizures and varying levels of developmental delay. RESULTS In this study, eight de novo variants of HCN1 genes were uncovered in eight patients, including six missense variants, one nonsense variant and one frameshift insertion variant; five of them were reported for the first time. The onset age for eight patients ranges from one month to one year. Their main clinical manifestations are epilepsy and varying degrees of developmental delay, and the main type of seizure is focal secondary generalized tonic-clonic seizure. Importantly, in our study, one case presented with a form of migrating focal seizure that has not been reported in the literature. Seizures from five of the eight children were effectively controlled with antiepileptic drugs including valproic acid, levetiracetam and oxcarbazepine. One child developed normally and four children developed mild delay. One child was treated with topiramate, and the convulsion was partially controlled and showed moderate to severe developmental delay. The antiepileptic treatment failed for the other two children, and the two children were treated with sodium valproate, oxcarbazepine, lamotrigine, chlorbazan, levetiracetam and nitrodiazepam successively, but their convulsions were not controlled and showed moderate to severe developmental delay. SIGNIFICANCE Our research reported eight variants in HCN1 gene causing epilepsy; among these variants, five variants were never reported before. HCN1-related epilepsy usually starts infantile period, and focal secondary generalized tonic-clonic seizure is the most common seizure type. Importantly, we reported the case with migrating focal seizure was rarely reported. Our study expanded both genotype and phenotype for HCN1-related epilepsy.
Collapse
Affiliation(s)
- Zhuanyi Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhuo Kuang
- Department of Neurology, Hunan Children's Hospital, Changsha, China
| | - Hongmei Liao
- Department of Neurology, Hunan Children's Hospital, Changsha, China
| | - Siyi Gan
- Department of Neurology, Hunan Children's Hospital, Changsha, China
| | - Xiaomei Peng
- Department of Neurology, Hunan Children's Hospital, Changsha, China
| | - Haiyan Yang
- Department of Neurology, Hunan Children's Hospital, Changsha, China
| | - Liwen Wu
- Department of Neurology, Hunan Children's Hospital, Changsha, China
| |
Collapse
|
8
|
Kazmierska-Grebowska P, Jankowski MM, MacIver MB. Missing Puzzle Pieces in Dementia Research: HCN Channels and Theta Oscillations. Aging Dis 2024; 15:22-42. [PMID: 37450922 PMCID: PMC10796085 DOI: 10.14336/ad.2023.0607] [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: 05/02/2023] [Accepted: 06/07/2023] [Indexed: 07/18/2023] Open
Abstract
Increasing evidence indicates a role of hyperpolarization activated cation (HCN) channels in controlling the resting membrane potential, pacemaker activity, memory formation, sleep, and arousal. Their disfunction may be associated with the development of epilepsy and age-related memory decline. Neuronal hyperexcitability involved in epileptogenesis and EEG desynchronization occur in the course of dementia in human Alzheimer's Disease (AD) and animal models, nevertheless the underlying ionic and cellular mechanisms of these effects are not well understood. Some suggest that theta rhythms involved in memory formation could be used as a marker of memory disturbances in the course of neurogenerative diseases, including AD. This review focusses on the interplay between hyperpolarization HCN channels, theta oscillations, memory formation and their role(s) in dementias, including AD. While individually, each of these factors have been linked to each other with strong supportive evidence, we hope here to expand this linkage to a more inclusive picture. Thus, HCN channels could provide a molecular target for developing new therapeutic agents for preventing and/or treating dementia.
Collapse
Affiliation(s)
| | - Maciej M. Jankowski
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.
- BioTechMed Center, Multimedia Systems Department, Faculty of Electronics, Telecommunications, and Informatics, Gdansk University of Technology, Gdansk, Poland.Telecommunications and Informatics, Gdansk University of Technology, Gdansk, Poland.
| | - M. Bruce MacIver
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of of Medicine, Stanford University, CA, USA.
| |
Collapse
|
9
|
Vasylyev DV, Liu S, Waxman SG. I h current stabilizes excitability in rodent DRG neurons and reverses hyperexcitability in a nociceptive neuron model of inherited neuropathic pain. J Physiol 2023; 601:5341-5366. [PMID: 37846879 PMCID: PMC10843455 DOI: 10.1113/jp284999] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 09/25/2023] [Indexed: 10/18/2023] Open
Abstract
We show here that hyperpolarization-activated current (Ih ) unexpectedly acts to inhibit the activity of dorsal root ganglion (DRG) neurons expressing WT Nav1.7, the largest inward current and primary driver of DRG neuronal firing, and hyperexcitable DRG neurons expressing a gain-of-function Nav1.7 mutation that causes inherited erythromelalgia (IEM), a human genetic model of neuropathic pain. In this study we created a kinetic model of Ih and used it, in combination with dynamic-clamp, to study Ih function in DRG neurons. We show, for the first time, that Ih increases rheobase and reduces the firing probability in small DRG neurons, and demonstrate that the amplitude of subthreshold oscillations is reduced by Ih . Our results show that Ih , due to slow gating, is not deactivated during action potentials (APs) and has a striking damping action, which reverses from depolarizing to hyperpolarizing, close to the threshold for AP generation. Moreover, we show that Ih reverses the hyperexcitability of DRG neurons expressing a gain-of-function Nav1.7 mutation that causes IEM. In the aggregate, our results show that Ih unexpectedly has strikingly different effects in DRG neurons as compared to previously- and well-studied cardiac cells. Within DRG neurons where Nav1.7 is present, Ih reduces depolarizing sodium current inflow due to enhancement of Nav1.7 channel fast inactivation and creates additional damping action by reversal of Ih direction from depolarizing to hyperpolarizing close to the threshold for AP generation. These actions of Ih limit the firing of DRG neurons expressing WT Nav1.7 and reverse the hyperexcitability of DRG neurons expressing a gain-of-function Nav1.7 mutation that causes IEM. KEY POINTS: Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, the molecular determinants of hyperpolarization-activated current (Ih ) have been characterized as a 'pain pacemaker', and thus considered to be a potential molecular target for pain therapeutics. Dorsal root ganglion (DRG) neurons express Nav1.7, a channel that is not present in central neurons or cardiac tissue. Gain-of-function mutations (GOF) of Nav1.7 identified in inherited erythromelalgia (IEM), a human genetic model of neuropathic pain, produce DRG neuron hyperexcitability, which in turn produces severe pain. We found that Ih increases rheobase and reduces firing probability in small DRG neurons expressing WT Nav1.7, and demonstrate that the amplitude of subthreshold oscillations is reduced by Ih . We also demonstrate that Ih reverses the hyperexcitability of DRG neurons expressing a GOF Nav1.7 mutation (L858H) that causes IEM. Our results show that, in contrast to cardiac cells and CNS neurons, Ih acts to stabilize DRG neuron excitability and prevents excessive firing.
Collapse
Affiliation(s)
- Dmytro V. Vasylyev
- Department of Neurology and Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT 06510
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| | - Shujun Liu
- Department of Neurology and Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT 06510
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| | - Stephen G. Waxman
- Department of Neurology and Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT 06510
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| |
Collapse
|
10
|
Gao N, Liu Z, Wang H, Shen C, Dong Z, Cui W, Xiong WC, Mei L. Deficiency of Cullin 3, a Protein Encoded by a Schizophrenia and Autism Risk Gene, Impairs Behaviors by Enhancing the Excitability of Ventral Tegmental Area (VTA) DA Neurons. J Neurosci 2023; 43:6249-6267. [PMID: 37558490 PMCID: PMC10490515 DOI: 10.1523/jneurosci.0247-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/09/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023] Open
Abstract
The dopaminergic neuromodulator system is fundamental to brain functions. Abnormal dopamine (DA) pathway is implicated in psychiatric disorders, including schizophrenia (SZ) and autism spectrum disorder (ASD). Mutations in Cullin 3 (CUL3), a core component of the Cullin-RING ubiquitin E3 ligase complex, have been associated with SZ and ASD. However, little is known about the function and mechanism of CUL3 in the DA system. Here, we show that CUL3 is critical for the function of DA neurons and DA-relevant behaviors in male mice. CUL3-deficient mice exhibited hyperactive locomotion, deficits in working memory and sensorimotor gating, and increased sensitivity to psychostimulants. In addition, enhanced DA signaling and elevated excitability of the VTA DA neurons were observed in CUL3-deficient animals. Behavioral impairments were attenuated by dopamine D2 receptor antagonist haloperidol and chemogenetic inhibition of DA neurons. Furthermore, we identified HCN2, a hyperpolarization-activated and cyclic nucleotide-gated channel, as a potential target of CUL3 in DA neurons. Our study indicates that CUL3 controls DA neuronal activity by maintaining ion channel homeostasis and provides insight into the role of CUL3 in the pathogenesis of psychiatric disorders.SIGNIFICANCE STATEMENT This study provides evidence that Cullin 3 (CUL3), a core component of the Cullin-RING ubiquitin E3 ligase complex that has been associated with autism spectrum disorder and schizophrenia, controls the excitability of dopamine (DA) neurons in mice. Its DA-specific heterozygous deficiency increased spontaneous locomotion, impaired working memory and sensorimotor gating, and elevated response to psychostimulants. We showed that CUL3 deficiency increased the excitability of VTA DA neurons, and inhibiting D2 receptor or DA neuronal activity attenuated behavioral deficits of CUL3-deficient mice. We found HCN2, a hyperpolarization-activated channel, as a target of CUL3 in DA neurons. Our findings reveal CUL3's role in DA neurons and offer insights into the pathogenic mechanisms of autism spectrum disorder and schizophrenia.
Collapse
Affiliation(s)
- Nannan Gao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Zhipeng Liu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Hongsheng Wang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Chen Shen
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Zhaoqi Dong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Wanpeng Cui
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio 44106
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio 44106
- Chinese Institutes for Medical Research, Beijing, China 100069
- Capital Medical University, Beijing, China 100069
| |
Collapse
|
11
|
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: 0] [Impact Index Per Article: 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
|
12
|
Mu L, Liu X, Yu H, Vickstrom CR, Friedman V, Kelly TJ, Hu Y, Su W, Liu S, Mantsch JR, Liu QS. cAMP-mediated upregulation of HCN channels in VTA dopamine neurons promotes cocaine reinforcement. Mol Psychiatry 2023; 28:3930-3942. [PMID: 37845497 PMCID: PMC10730389 DOI: 10.1038/s41380-023-02290-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 10/18/2023]
Abstract
Chronic cocaine exposure induces enduring neuroadaptations that facilitate motivated drug taking. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are known to modulate neuronal firing and pacemaker activity in ventral tegmental area (VTA) dopamine neurons. However, it remained unknown whether cocaine self-administration affects HCN channel function and whether HCN channel activity modulates motivated drug taking. We report that rat VTA dopamine neurons predominantly express Hcn3-4 mRNA, while VTA GABA neurons express Hcn1-4 mRNA. Both neuronal types display similar hyperpolarization-activated currents (Ih), which are facilitated by acute increases in cAMP. Acute cocaine application decreases voltage-dependent activation of Ih in VTA dopamine neurons, but not in GABA neurons. Unexpectedly, chronic cocaine self-administration results in enhanced Ih selectively in VTA dopamine neurons. This differential modulation of Ih currents is likely mediated by a D2 autoreceptor-induced decrease in cAMP as D2 (Drd2) mRNA is predominantly expressed in dopamine neurons, whereas D1 (Drd1) mRNA is barely detectable in the VTA. Moreover, chronically decreased cAMP via Gi-DREADD stimulation leads to an increase in Ih in VTA dopamine neurons and enhanced binding of HCN3/HCN4 with tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b), an auxiliary subunit that is known to facilitate HCN channel surface trafficking. Finally, we show that systemic injection and intra-VTA infusion of the HCN blocker ivabradine reduces cocaine self-administration under a progressive ratio schedule and produces a downward shift of the cocaine dose-response curve. Our results suggest that cocaine self-administration induces an upregulation of Ih in VTA dopamine neurons, while HCN inhibition reduces the motivation for cocaine intake.
Collapse
Affiliation(s)
- Lianwei Mu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Xiaojie Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Hao Yu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Casey R Vickstrom
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Vladislav Friedman
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Thomas J Kelly
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Ying Hu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Wantang Su
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Department of Exercise Physiology, Beijing Sport University, Beijing, 100084, China
| | - Shuai Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - John R Mantsch
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
13
|
Catacuzzeno L, Conti F, Franciolini F. Fifty years of gating currents and channel gating. J Gen Physiol 2023; 155:e202313380. [PMID: 37410612 PMCID: PMC10324510 DOI: 10.1085/jgp.202313380] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/12/2023] [Accepted: 06/02/2023] [Indexed: 07/08/2023] Open
Abstract
We celebrate this year the 50th anniversary of the first electrophysiological recordings of the gating currents from voltage-dependent ion channels done in 1973. This retrospective tries to illustrate the context knowledge on channel gating and the impact gating-current recording had then, and how it continued to clarify concepts, elaborate new ideas, and steer the scientific debate in these 50 years. The notion of gating particles and gating currents was first put forward by Hodgkin and Huxley in 1952 as a necessary assumption for interpreting the voltage dependence of the Na and K conductances of the action potential. 20 years later, gating currents were actually recorded, and over the following decades have represented the most direct means of tracing the movement of the gating charges and gaining insights into the mechanisms of channel gating. Most work in the early years was focused on the gating currents from the Na and K channels as found in the squid giant axon. With channel cloning and expression on heterologous systems, other channels as well as voltage-dependent enzymes were investigated. Other approaches were also introduced (cysteine mutagenesis and labeling, site-directed fluorometry, cryo-EM crystallography, and molecular dynamics [MD] modeling) to provide an integrated and coherent view of voltage-dependent gating in biological macromolecules. The layout of this retrospective reflects the past 50 years of investigations on gating currents, first addressing studies done on Na and K channels and then on other voltage-gated channels and non-channel structures. The review closes with a brief overview of how the gating-charge/voltage-sensor movements are translated into pore opening and the pathologies associated with mutations targeting the structures involved with the gating currents.
Collapse
Affiliation(s)
- Luigi Catacuzzeno
- Department of Chemistry Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Franco Conti
- Department of Physics, University of Genova, Genova, Italy
| | - Fabio Franciolini
- Department of Chemistry Biology and Biotechnology, University of Perugia, Perugia, Italy
| |
Collapse
|
14
|
Häfele M, Kreitz S, Ludwig A, Hess A, Wank I. The impact of HCN4 channels on CNS brain networks as a new target in pain development. FRONTIERS IN NETWORK PHYSIOLOGY 2023; 3:1090502. [PMID: 37496803 PMCID: PMC10368246 DOI: 10.3389/fnetp.2023.1090502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 06/22/2023] [Indexed: 07/28/2023]
Abstract
While it is well established that the isoform 2 of the hyperpolarization-activated cyclic nucleotide-gated cation channel (HCN2) plays an important role in the development and maintenance of pain, the role of the closely related HCN4 isoform in the processing of nociceptive signals is not known. HCN4 channels are highly expressed in the thalamus, a region important for stimulus transmission and information processing. We used a brain-specific HCN4-knockout mouse line (HCN4-KO) to explore the role of HCN4 channels in acute nociceptive processing using several behavioral tests as well as a multimodal magnetic resonance imaging (MRI) approach. Functional MRI (fMRI) brain responses were measured during acute peripheral thermal stimulation complemented by resting state (RS) before and after stimulation. The data were analyzed by conventional and graph-theoretical approaches. Finally, high-resolution anatomical brain data were acquired. HCN4-KO animals showed a central thermal, but not a mechanical hypersensitivity in behavioral experiments. The open field analysis showed no significant differences in motor readouts between HCN4-KO and controls but uncovered increased anxiety in the HCN4-KO mice. Thermal stimulus-driven fMRI (s-fMRI) data revealed increased response volumes and response amplitudes for HCN4-KO, most pronounced at lower stimulation temperatures in the subcortical input, the amygdala as well as in limbic/hippocampal regions, and in the cerebellum. These findings could be cross-validated by graph-theoretical analyses. Assessment of short-term RS before and after thermal stimulation revealed that stimulation-related modulations of the functional connectivity only occurred in control animals. This was consistent with the finding that the hippocampus was found to be smaller in HCN4-KO. In summary, the deletion of HCN4 channels impacts on processing of acute nociception, which is remarkably manifested as a thermal hypersensitive phenotype. This was mediated by the key regions hypothalamus, somatosensory cortex, cerebellum and the amygdala. As consequence, HCN4-KO mice were more anxious, and their brain-wide RS functional connectivity could not be modulated by thermal nociceptive stimulation.
Collapse
Affiliation(s)
- Maximilian Häfele
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Silke Kreitz
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Ludwig
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Hess
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Institute of Neuroradiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- FAU NeW—Research Center for New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Isabel Wank
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
15
|
Sun D, Wang Q. The application of SUDEP in forensic diagnosis: a mini review. Front Neurol 2023; 14:1169003. [PMID: 37181558 PMCID: PMC10169668 DOI: 10.3389/fneur.2023.1169003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 03/20/2023] [Indexed: 05/16/2023] Open
Abstract
In the epilepsy population, the risk of sudden death from epilepsy is rare but is ~24 times greater than the risk of sudden death from other causes. Sudden unexpected death in epilepsy (SUDEP) has been widely recognized in clinical studies. Despite its significance as a cause of death, SUDEP is rarely used in forensic practice. This review focuses on the forensic characteristics of SUDEP, analyzed the reasons for its underuse in forensic practice, and illustrated the prospect of establishing uniform diagnostic criteria for sudden unexpected death in epilepsy and molecular anatomy in aiding forensic diagnosis.
Collapse
Affiliation(s)
| | - Qiang Wang
- Forensic Science Center, East China University of Political Science and Law, Shanghai, China
| |
Collapse
|
16
|
Lei X, Yan Y, Zeng J, Wang R, Li S, Xiao Z, Liu X. Activation of HCN channels caused by elevated cAMP levels in periaqueductal gray promotes bone cancer pain. Neurochem Int 2023; 162:105437. [PMID: 36336089 DOI: 10.1016/j.neuint.2022.105437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 10/06/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
Abstract
The periaqueductal gray (PAG) is an important relay center for the descending pathways that regulate nociceptive information transduction. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels play critical roles in the nerve injury-induced pain hypersensitivity. Previous studies have identified that HCN1 and HCN2 channel protein located in the ventral-lateral periaqueductal gray (vlPAG), a region important for pain regulation. However, it is not clear whether the HCN channel in vlPAG is involved in bone cancer pain (BCP). In this study, we assessed the role of HCN channels in BCP by measuring changes of HCN channel expression and activity in vlPAG neurons in bone cancer rats. In the present study, the BCP model was established by injecting SHZ-88 breast cancer cells into the right tibia bone marrow in rats. The mechanical withdrawal threshold (MWT) and thermal withdrawal latency (TWL) were measured to evaluate pain behavior in rats. HCN1 and HCN2 channels expression in vlPAG were detected by using Western Blot and immunohistochemistry. In addition, the cAMP level in vlPAG neurons was detected by ELISA, and HCN channel current (Ih) of vlPAG neurons was recorded by whole cell patch-clamp to evaluate HCN channel activity. As a result, decreased MWT and TWL were observed in rats on 7d after SHZ-88 cell inoculation, and the allodynia was sustained until 21d after inoculation. At the same time, HCN1 and HCN2 channels expression and neuronal Ih in vlPAG were significantly increased in BCP rats. In addition, the level of cAMP in vlPAG also increased after SHZ-88 cell inoculation. Furthermore, intravlPAG injection of ZD7288 (HCN channels antagonist) could significantly reduce hyperalgesia and the elevation of cAMP in vlPAG in BCP rats. Our observations suggest that the elevation of cAMP may promote the activation of HCN channels in vlPAG in bone cancer rats, thereby promoting the development of bone cancer pain.
Collapse
Affiliation(s)
- Xiaolu Lei
- Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, 563000, China; Guizhou Key Laboratory of Anaesthesia and Organ Protection, Zunyi Medical University, Zunyi, 563000, China
| | - Yan Yan
- Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, 563000, China; Guizhou Key Laboratory of Anaesthesia and Organ Protection, Zunyi Medical University, Zunyi, 563000, China
| | - Junwei Zeng
- Department of Physiology, Zunyi Medical University, Zunyi, 563000, China
| | - Rong Wang
- Department of Physiology, Zunyi Medical University, Zunyi, 563000, China
| | - Song Li
- Department of Physiology, Zunyi Medical University, Zunyi, 563000, China
| | - Zhi Xiao
- Key Laboratory of Brain Science, Zunyi Medical University, Zunyi, 563000, China; Guizhou Key Laboratory of Anaesthesia and Organ Protection, Zunyi Medical University, Zunyi, 563000, China
| | - Xiaohong Liu
- Department of Physiology, Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
17
|
Analgesic effect of ivabradine against inflammatory pain mediated by hyperpolarization-activated cyclic nucleotide–gated cation channels expressed on primary afferent terminals in the spinal dorsal horn. Pain 2022; 163:1356-1369. [DOI: 10.1097/j.pain.0000000000002523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 10/14/2021] [Indexed: 11/25/2022]
|
18
|
Mecawi AS, Varanda WA, da Silva MP. Osmoregulation and the Hypothalamic Supraoptic Nucleus: From Genes to Functions. Front Physiol 2022; 13:887779. [PMID: 35685279 PMCID: PMC9171026 DOI: 10.3389/fphys.2022.887779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/18/2022] [Indexed: 11/13/2022] Open
Abstract
Due to the relatively high permeability to water of the plasma membrane, water tends to equilibrate its chemical potential gradient between the intra and extracellular compartments. Because of this, changes in osmolality of the extracellular fluid are accompanied by changes in the cell volume. Therefore, osmoregulatory mechanisms have evolved to keep the tonicity of the extracellular compartment within strict limits. This review focuses on the following aspects of osmoregulation: 1) the general problems in adjusting the "milieu interieur" to challenges imposed by water imbalance, with emphasis on conceptual aspects of osmosis and cell volume regulation; 2) osmosensation and the hypothalamic supraoptic nucleus (SON), starting with analysis of the electrophysiological responses of the magnocellular neurosecretory cells (MNCs) involved in the osmoreception phenomenon; 3) transcriptomic plasticity of SON during sustained hyperosmolality, to pinpoint the genes coding membrane channels and transporters already shown to participate in the osmosensation and new candidates that may have their role further investigated in this process, with emphasis on those expressed in the MNCs, discussing the relationships of hydration state, gene expression, and MNCs electrical activity; and 4) somatodendritic release of neuropeptides in relation to osmoregulation. Finally, we expect that by stressing the relationship between gene expression and the electrical activity of MNCs, studies about the newly discovered plastic-regulated genes that code channels and transporters in the SON may emerge.
Collapse
Affiliation(s)
- André Souza Mecawi
- Laboratory of Molecular Neuroendocrinology, Department of Biophysics, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Wamberto Antonio Varanda
- Department of Physiology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Melina Pires da Silva
- Laboratory of Cellular Neuroendocrinology, Department of Biophysics, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
19
|
Kessi M, Peng J, Duan H, He H, Chen B, Xiong J, Wang Y, Yang L, Wang G, Kiprotich K, Bamgbade OA, He F, Yin F. The Contribution of HCN Channelopathies in Different Epileptic Syndromes, Mechanisms, Modulators, and Potential Treatment Targets: A Systematic Review. Front Mol Neurosci 2022; 15:807202. [PMID: 35663267 PMCID: PMC9161305 DOI: 10.3389/fnmol.2022.807202] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 04/06/2022] [Indexed: 12/04/2022] Open
Abstract
Background Hyperpolarization-activated cyclic nucleotide-gated (HCN) current reduces dendritic summation, suppresses dendritic calcium spikes, and enables inhibitory GABA-mediated postsynaptic potentials, thereby suppressing epilepsy. However, it is unclear whether increased HCN current can produce epilepsy. We hypothesized that gain-of-function (GOF) and loss-of-function (LOF) variants of HCN channel genes may cause epilepsy. Objectives This systematic review aims to summarize the role of HCN channelopathies in epilepsy, update genetic findings in patients, create genotype–phenotype correlations, and discuss animal models, GOF and LOF mechanisms, and potential treatment targets. Methods The review was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement, for all years until August 2021. Results We identified pathogenic variants of HCN1 (n = 24), HCN2 (n = 8), HCN3 (n = 2), and HCN4 (n = 6) that were associated with epilepsy in 74 cases (43 HCN1, 20 HCN2, 2 HCN3, and 9 HCN4). Epilepsy was associated with GOF and LOF variants, and the mechanisms were indeterminate. Less than half of the cases became seizure-free and some developed drug-resistant epilepsy. Of the 74 cases, 12 (16.2%) died, comprising HCN1 (n = 4), HCN2 (n = 2), HCN3 (n = 2), and HCN4 (n = 4). Of the deceased cases, 10 (83%) had a sudden unexpected death in epilepsy (SUDEP) and 2 (16.7%) due to cardiopulmonary failure. SUDEP affected more adults (n = 10) than children (n = 2). HCN1 variants p.M234R, p.C329S, p.V414M, p.M153I, and p.M305L, as well as HCN2 variants p.S632W and delPPP (p.719–721), were associated with different phenotypes. HCN1 p.L157V and HCN4 p.R550C were associated with genetic generalized epilepsy. There are several HCN animal models, pharmacological targets, and modulators, but precise drugs have not been developed. Currently, there are no HCN channel openers. Conclusion We recommend clinicians to include HCN genes in epilepsy gene panels. Researchers should explore the possible underlying mechanisms for GOF and LOF variants by identifying the specific neuronal subtypes and neuroanatomical locations of each identified pathogenic variant. Researchers should identify specific HCN channel openers and blockers with high binding affinity. Such information will give clarity to the involvement of HCN channelopathies in epilepsy and provide the opportunity to develop targeted treatments.
Collapse
Affiliation(s)
- Miriam Kessi
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
- Department of Pediatrics, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Jing Peng
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Haolin Duan
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Hailan He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Baiyu Chen
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Juan Xiong
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Ying Wang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Lifen Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Guoli Wang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Karlmax Kiprotich
- Department of Epidemiology and Medical Statistics, School of Public Health, Moi University, Eldoret, Kenya
| | - Olumuyiwa A. Bamgbade
- Department of Anesthesiology and Pharmacology, University of British Columbia, Vancouver, BC, Canada
| | - Fang He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Fei Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
- *Correspondence: Fei Yin
| |
Collapse
|
20
|
Subramanian S, Haroutounian S, Palanca BJA, Lenze EJ. Ketamine as a therapeutic agent for depression and pain: mechanisms and evidence. J Neurol Sci 2022; 434:120152. [PMID: 35092901 DOI: 10.1016/j.jns.2022.120152] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 12/14/2022]
Abstract
Ketamine is an anesthetic drug which is now used to treat chronic pain conditions and psychiatric disorders, especially depression. It is an N-methyl-D-aspartate (NMDA) receptor antagonist with additional effects on α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, opioid receptors, and monoaminergic receptors. This article focuses on ketamine's role in treating depression and pain, two commonly comorbid challenging conditions with potentially shared neurobiologic circuitry. Many clinical trials have utilized intravenous or intranasal ketamine for treating depression and pain. Intravenous ketamine is more bioavailable than intranasal ketamine and both are effective for acute depressive episodes. Intravenous ketamine is advantageous for post-operative analgesia and is associated with a reduction in total opioid requirements. Few studies have treated chronic pain or concurrent depression and pain with ketamine. Larger, randomized control trials are needed to examine the safety and efficacy of intravenous vs. intranasal ketamine, ideal target populations, and optimal dosing to treat both depression and pain.
Collapse
Affiliation(s)
- Subha Subramanian
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| | - Simon Haroutounian
- Department of Anesthesiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Ben Julian A Palanca
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Department of Anesthesiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Eric J Lenze
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
21
|
Salinas-Luypaert C, Sáez-Cortez F, Quintanilla ME, Herrera-Marschitz M, Rivera-Meza M. Gene knockdown of HCN2 ion channels in the ventral tegmental area reduces ethanol consumption in alcohol preferring rats. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2022; 48:165-175. [PMID: 35377277 DOI: 10.1080/00952990.2022.2033759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/04/2022] [Accepted: 01/15/2022] [Indexed: 06/14/2023]
Abstract
Background: Hyperpolarization-Activated Cyclic Nucleotide-Gated (HCN) ionic channels are known to play a key role in the control of neuron excitability and have been proposed as a molecular target of ethanol. Previous studies in rats have shown that gene-induced overexpression of the HCN2 channel in the ventral tegmental area (VTA) increases the rewarding effects of ethanol and its intake by the animals.Objective: The aim of this work was to study the effects of VTA HCN2 gene knockdown in the voluntary ethanol consumption of alcohol-preferring UChB rats.Methods: Two lentiviral vectors were generated; LV-siRNA-HCN2, coding for a siRNA that elicited >95% reduction of HCN2 protein levels in vitro, and a control vector coding for a scrambled siRNA sequence. Female UChB naïve rats (n = 14) were microinjected into the VTA with LV-siRNA-HCN2 or the scrambled control vector (n = 11). Four days after, animals were given a daily free access to 10% ethanol and water for 10 days.Results: Rats treated with the LV-siRNA-HCN2 vector showed a ~ 70% reduction (p < .001) in their ethanol preference and ethanol intake compared to control animals. No changes were observed in the total fluid intake of both groups. HCN2 levels in the VTA were measured by Western blot showing a reduction of 40% (p < .05) in the rats injected with LV-siRNA-HCN2, compared to control animals.Conclusion: These results show that knockdown of HCN2 ionic channels in the VTA of UChB rats markedly reduces their voluntary ethanol intake, supporting the idea that HCN2 channels may constitute a therapeutic target for alcohol use disorders.
Collapse
Affiliation(s)
- Catalina Salinas-Luypaert
- Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences Santiago, Chile
| | - Felipe Sáez-Cortez
- Program of Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - María Elena Quintanilla
- Program of Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Mario Herrera-Marschitz
- Program of Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Mario Rivera-Meza
- Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences Santiago, Chile
- Research Center for the Development of Novel Therapeutic Alternatives for Alcohol Use Disorders, Santiago, Chile
| |
Collapse
|
22
|
Glucocorticoid-glucocorticoid receptor-HCN1 channels reduce neuronal excitability in dorsal hippocampal CA1 neurons. Mol Psychiatry 2022; 27:4035-4049. [PMID: 35840797 PMCID: PMC9718682 DOI: 10.1038/s41380-022-01682-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/15/2022] [Accepted: 06/27/2022] [Indexed: 02/07/2023]
Abstract
While chronic stress increases hyperpolarization-activated current (Ih) in dorsal hippocampal CA1 neurons, the underlying molecular mechanisms are entirely unknown. Following chronic social defeat stress (CSDS), susceptible mice displayed social avoidance and impaired spatial working memory, which were linked to decreased neuronal excitability, increased perisomatic hyperpolarization-activated cyclic nucleotide-gated (HCN) 1 protein expression, and elevated Ih in dorsal but not ventral CA1 neurons. In control mice, bath application of corticosterone reduced neuronal excitability, increased tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b) and HCN1 protein expression, and elevated Ih in dorsal but not ventral CA1 region/neurons. Corticosterone-induced upregulation of functional Ih was mediated by the glucocorticoid receptor (GR), HCN channels, and the protein kinase A (PKA) but not the calcium/calmodulin-dependent protein kinase II (CaMKII) pathway. Three months after the end of CSDS, susceptible mice displayed persistent social avoidance when exposed to a novel aggressor. The sustained behavioral deficit was associated with lower neuronal excitability and higher functional Ih in dorsal CA1 neurons, both of which were unaffected by corticosterone treatment. Our findings show that corticosterone treatment mimics the pathophysiological effects of dorsal CA1 neurons/region found in susceptible mice. The aberrant expression of HCN1 protein along the somatodendritic axis of the dorsal hippocampal CA1 region might be the molecular mechanism driving susceptibility to social avoidance.
Collapse
|
23
|
Wills L, Ables JL, Braunscheidel KM, Caligiuri SPB, Elayouby KS, Fillinger C, Ishikawa M, Moen JK, Kenny PJ. Neurobiological Mechanisms of Nicotine Reward and Aversion. Pharmacol Rev 2022; 74:271-310. [PMID: 35017179 PMCID: PMC11060337 DOI: 10.1124/pharmrev.121.000299] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 08/24/2021] [Indexed: 12/27/2022] Open
Abstract
Neuronal nicotinic acetylcholine receptors (nAChRs) regulate the rewarding actions of nicotine contained in tobacco that establish and maintain the smoking habit. nAChRs also regulate the aversive properties of nicotine, sensitivity to which decreases tobacco use and protects against tobacco use disorder. These opposing behavioral actions of nicotine reflect nAChR expression in brain reward and aversion circuits. nAChRs containing α4 and β2 subunits are responsible for the high-affinity nicotine binding sites in the brain and are densely expressed by reward-relevant neurons, most notably dopaminergic, GABAergic, and glutamatergic neurons in the ventral tegmental area. High-affinity nAChRs can incorporate additional subunits, including β3, α6, or α5 subunits, with the resulting nAChR subtypes playing discrete and dissociable roles in the stimulatory actions of nicotine on brain dopamine transmission. nAChRs in brain dopamine circuits also participate in aversive reactions to nicotine and the negative affective state experienced during nicotine withdrawal. nAChRs containing α3 and β4 subunits are responsible for the low-affinity nicotine binding sites in the brain and are enriched in brain sites involved in aversion, including the medial habenula, interpeduncular nucleus, and nucleus of the solitary tract, brain sites in which α5 nAChR subunits are also expressed. These aversion-related brain sites regulate nicotine avoidance behaviors, and genetic variation that modifies the function of nAChRs in these sites increases vulnerability to tobacco dependence and smoking-related diseases. Here, we review the molecular, cellular, and circuit-level mechanisms through which nicotine elicits reward and aversion and the adaptations in these processes that drive the development of nicotine dependence. SIGNIFICANCE STATEMENT: Tobacco use disorder in the form of habitual cigarette smoking or regular use of other tobacco-related products is a major cause of death and disease worldwide. This article reviews the actions of nicotine in the brain that contribute to tobacco use disorder.
Collapse
Affiliation(s)
- Lauren Wills
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York
| | - Jessica L Ables
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York
| | - Kevin M Braunscheidel
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York
| | - Stephanie P B Caligiuri
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York
| | - Karim S Elayouby
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York
| | - Clementine Fillinger
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York
| | - Masago Ishikawa
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York
| | - Janna K Moen
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York
| | - Paul J Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, New York
| |
Collapse
|
24
|
Combe CL, Gasparini S. I h from synapses to networks: HCN channel functions and modulation in neurons. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 166:119-132. [PMID: 34181891 DOI: 10.1016/j.pbiomolbio.2021.06.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/25/2021] [Accepted: 06/03/2021] [Indexed: 01/16/2023]
Abstract
Hyperpolarization-activated cyclic nucleotide gated (HCN) channels and the current they carry, Ih, are widely and diversely distributed in the central nervous system (CNS). The distribution of the four subunits of HCN channels is variable within the CNS, within brain regions, and often within subcellular compartments. The precise function of Ih can depend heavily on what other channels are co-expressed. In this review, we give an overview of HCN channel structure, distribution, and modulation by cyclic adenosine monophosphate (cAMP). We then discuss HCN channel and Ih functions, where we have parsed the roles into two main effects: a steady effect on maintaining the resting membrane potential at relatively depolarized values, and slow channel dynamics. Within this framework, we discuss Ih involvement in resonance, synaptic integration, transmitter release, plasticity, and point out a special case, where the effects of Ih on the membrane potential and its slow channel dynamics have dual roles in thalamic neurons.
Collapse
Affiliation(s)
- Crescent L Combe
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Sonia Gasparini
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA.
| |
Collapse
|
25
|
Loss of HCN2 in Dorsal Hippocampus of Young Adult Mice Induces Specific Apoptosis of the CA1 Pyramidal Neuron Layer. Int J Mol Sci 2021; 22:ijms22136699. [PMID: 34206649 PMCID: PMC8269412 DOI: 10.3390/ijms22136699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/08/2021] [Accepted: 06/18/2021] [Indexed: 11/20/2022] Open
Abstract
Neurons inevitably rely on a proper repertoire and distribution of membrane-bound ion-conducting channels. Among these proteins, the family of hyperpolarization-activated and cyclic nucleotide-gated (HCN) channels possesses unique properties giving rise to the corresponding Ih-current that contributes to various aspects of neural signaling. In mammals, four genes (hcn1-4) encode subunits of HCN channels. These subunits can assemble as hetero- or homotetrameric ion-conducting channels. In order to elaborate on the specific role of the HCN2 subunit in shaping electrical properties of neurons, we applied an Adeno-associated virus (AAV)-mediated, RNAi-based knock-down strategy of hcn2 gene expression both in vitro and in vivo. Electrophysiological measurements showed that HCN2 subunit knock-down resulted in specific yet anticipated changes in Ih-current properties in primary hippocampal neurons and, in addition, corroborated that the HCN2 subunit participates in postsynaptic signal integration. To further address the role of the HCN2 subunit in vivo, we injected recombinant (r)AAVs into the dorsal hippocampus of young adult male mice. Behavioral and biochemical analyses were conducted to assess the contribution of HCN2-containing channels in shaping hippocampal network properties. Surprisingly, knock-down of hcn2 expression resulted in a severe degeneration of the CA1 pyramidal cell layer, which did not occur in mice injected with control rAAV constructs. This finding might pinpoint to a vital and yet unknown contribution of HCN2 channels in establishing or maintaining the proper function of CA1 pyramidal neurons of the dorsal hippocampus.
Collapse
|
26
|
Wills L, Kenny PJ. Addiction-related neuroadaptations following chronic nicotine exposure. J Neurochem 2021; 157:1652-1673. [PMID: 33742685 DOI: 10.1111/jnc.15356] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/14/2021] [Accepted: 03/15/2021] [Indexed: 12/16/2022]
Abstract
The addiction-relevant molecular, cellular, and behavioral actions of nicotine are derived from its stimulatory effects on neuronal nicotinic acetylcholine receptors (nAChRs) in the central nervous system. nAChRs expressed by dopamine-containing neurons in the ventral midbrain, most notably in the ventral tegmental area (VTA), contribute to the reward-enhancing properties of nicotine that motivate the use of tobacco products. nAChRs are also expressed by neurons in brain circuits that regulate aversion. In particular, nAChRs expressed by neurons in the medial habenula (mHb) and the interpeduncular nucleus (IPn) to which the mHb almost exclusively projects regulate the "set-point" for nicotine aversion and control nicotine intake. Different nAChR subtypes are expressed in brain reward and aversion circuits and nicotine intake is titrated to maximally engage reward-enhancing nAChRs while minimizing the recruitment of aversion-promoting nAChRs. With repeated exposure to nicotine, reward- and aversion-related nAChRs and the brain circuits in which they are expressed undergo adaptations that influence whether tobacco use will transition from occasional to habitual. Genetic variation that influences the sensitivity of addiction-relevant brain circuits to the actions of nicotine also influence the propensity to develop habitual tobacco use. Here, we review some of the key advances in our understanding of the mechanisms by which nicotine acts on brain reward and aversion circuits and the adaptations that occur in these circuits that may drive addiction to nicotine-containing tobacco products.
Collapse
Affiliation(s)
- Lauren Wills
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Paul J Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| |
Collapse
|
27
|
Zhong W, Darmani NA. The HCN Channel Blocker ZD7288 Induces Emesis in the Least Shrew ( Cryptotis parva). Front Pharmacol 2021; 12:647021. [PMID: 33995059 PMCID: PMC8117105 DOI: 10.3389/fphar.2021.647021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/12/2021] [Indexed: 12/02/2022] Open
Abstract
Subtypes (1-4) of the hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are widely expressed in the central and peripheral nervous systems, as well as the cells of smooth muscles in many organs. They mainly serve to regulate cellular excitability in these tissues. The HCN channel blocker ZD7288 has been shown to reduce apomorphine-induced conditioned taste aversion on saccharin preference in rats suggesting potential antinausea/antiemetic effects. Currently, in the least shew model of emesis we find that ZD7288 induces vomiting in a dose-dependent manner, with maximal efficacies of 100% at 1 mg/kg (i.p.) and 83.3% at 10 µg (i.c.v.). HCN channel subtype (1-4) expression was assessed using immunohistochemistry in the least shrew brainstem dorsal vagal complex (DVC) containing the emetic nuclei (area postrema (AP), nucleus tractus solitarius and dorsal motor nucleus of the vagus). Highly enriched HCN1 and HCN4 subtypes are present in the AP. A 1 mg/kg (i.p.) dose of ZD7288 strongly evoked c-Fos expression and ERK1/2 phosphorylation in the shrew brainstem DVC, but not in the in the enteric nervous system in the jejunum, suggesting a central contribution to the evoked vomiting. The ZD7288-evoked c-Fos expression exclusively occurred in tryptophan hydroxylase 2-positive serotonin neurons of the dorsal vagal complex, indicating activation of serotonin neurons may contribute to ZD7288-induced vomiting. To reveal its mechanism(s) of emetic action, we evaluated the efficacy of diverse antiemetics against ZD7288-evoked vomiting including the antagonists/inhibitors of: ERK1/2 (U0126), L-type Ca2+ channel (nifedipine); store-operated Ca2+ entry (MRS 1845); T-type Ca2+ channel (Z944), IP3R (2-APB), RyR receptor (dantrolene); the serotoninergic type 3 receptor (palonosetron); neurokinin 1 receptor (netupitant), dopamine type 2 receptor (sulpride), and the transient receptor potential vanilloid 1 receptor agonist, resiniferatoxin. All tested antiemetics except sulpride attenuated ZD7288-evoked vomiting to varying degrees. In sum, ZD7288 has emetic potential mainly via central mechanisms, a process which involves Ca2+ signaling and several emetic receptors. HCN channel blockers have been reported to have emetic potential in the clinic since they are currently used/investigated as therapeutic candidates for cancer therapy related- or unrelated-heart failure, pain, and cognitive impairment.
Collapse
Affiliation(s)
| | - N. A. Darmani
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| |
Collapse
|
28
|
Miranda-Barrientos J, Chambers I, Mongia S, Liu B, Wang HL, Mateo-Semidey GE, Margolis EB, Zhang S, Morales M. Ventral tegmental area GABA, glutamate, and glutamate-GABA neurons are heterogeneous in their electrophysiological and pharmacological properties. Eur J Neurosci 2021; 54:10.1111/ejn.15156. [PMID: 33619763 PMCID: PMC8380271 DOI: 10.1111/ejn.15156] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 02/16/2021] [Accepted: 02/16/2021] [Indexed: 01/05/2023]
Abstract
The ventral tegmental area (VTA) contains dopamine neurons intermixed with GABA-releasing (expressing vesicular GABA transporter, VGaT), glutamate-releasing (expressing vesicular glutamate transporter 2, VGluT2), and glutamate-GABA co-releasing (co-expressing VGluT2 and VGaT) neurons. By delivering INTRSECT viral vectors into the VTA of double vglut2-Cre/vgat-Flp transgenic mice, we targeted specific VTA cell populations for ex vivo recordings. We found that VGluT2+ VGaT- and VGluT2+ VGaT+ neurons on average had relatively hyperpolarized resting membrane potential, greater rheobase, and lower spontaneous firing frequency compared to VGluT2- VGaT+ neurons, suggesting that VTA glutamate-releasing and glutamate-GABA co-releasing neurons require stronger excitatory drive to fire than GABA-releasing neurons. In addition, we detected expression of Oprm1mRNA (encoding µ opioid receptors, MOR) in VGluT2+ VGaT- and VGluT2- VGaT+ neurons, and that the MOR agonist DAMGO hyperpolarized neurons with these phenotypes. Collectively, we demonstrate the utility of the double transgenic mouse to access VTA glutamate, glutamate-GABA, and GABA neurons to determine their electrophysiological properties. SIGNIFICANT STATEMENT: Some physiological properties of VTA glutamate-releasing and glutamate-GABA co-releasing neurons are distinct from those of VTA GABA-releasing neurons. µ-opioid receptor activation hyperpolarizes some VTA glutamate-releasing and some GABA-releasing neurons.
Collapse
Affiliation(s)
| | - Ian Chambers
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Smriti Mongia
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Bing Liu
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Hui-Ling Wang
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Baltimore, MD, USA
| | | | - Elyssa B. Margolis
- UCSF Weill Institute of Neurosciences|Department of Neurology, University of California, San Francisco, CA, USA
| | - Shiliang Zhang
- Confocal and Electron Microscopy Core, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Marisela Morales
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Baltimore, MD, USA
| |
Collapse
|
29
|
Concepcion FA, Khan MN, Ju Wang JD, Wei AD, Ojemann JG, Ko AL, Shi Y, Eng JK, Ramirez JM, Poolos NP. HCN Channel Phosphorylation Sites Mapped by Mass Spectrometry in Human Epilepsy Patients and in an Animal Model of Temporal Lobe Epilepsy. Neuroscience 2021; 460:13-30. [PMID: 33571596 DOI: 10.1016/j.neuroscience.2021.01.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/07/2021] [Accepted: 01/26/2021] [Indexed: 10/22/2022]
Abstract
Because hyperpolarization-activated cyclic nucleotide-gated (HCN) ion channels modulate the excitability of cortical and hippocampal principal neurons, these channels play a key role in the hyperexcitability that occurs during the development of epilepsy after a brain insult, or epileptogenesis. In epileptic rats generated by pilocarpine-induced status epilepticus, HCN channel activity is downregulated by two main mechanisms: a hyperpolarizing shift in gating and a decrease in amplitude of the current mediated by HCN channels, Ih. Because these mechanisms are modulated by various phosphorylation signaling pathways, we hypothesized that phosphorylation changes occur at individual HCN channel amino acid residues (phosphosites) during epileptogenesis. We collected CA1 hippocampal tissue from male Sprague Dawley rats made epileptic by pilocarpine-induced status epilepticus, and age-matched naïve controls. We also included resected human brain tissue containing epileptogenic zones (EZs) where seizures arise for comparison to our chronically epileptic rats. After enrichment for HCN1 and HCN2 isoforms by immunoprecipitation and trypsin in-gel digestion, the samples were analyzed by mass spectrometry. We identified numerous phosphosites from HCN1 and HCN2 channels, representing a novel survey of phosphorylation sites within HCN channels. We found high levels of HCN channel phosphosite homology between humans and rats. We also identified a novel HCN1 channel phosphosite S791, which underwent significantly increased phosphorylation during the chronic epilepsy stage. Heterologous expression of a phosphomimetic mutant, S791D, replicated a hyperpolarizing shift in Ih gating seen in neurons from chronically epileptic rats. These results show that HCN1 channel phosphorylation is altered in epilepsy and may be of pathogenic importance.
Collapse
Affiliation(s)
- F A Concepcion
- Department of Neurology and Regional Epilepsy Center, University of Washington, Seattle, WA, United States
| | - M N Khan
- Department of Neurology and Regional Epilepsy Center, University of Washington, Seattle, WA, United States
| | - J-D Ju Wang
- Seattle Children's Research Institute, Center for Integrative Brain Research, Seattle, WA, United States
| | - A D Wei
- Seattle Children's Research Institute, Center for Integrative Brain Research, Seattle, WA, United States
| | - J G Ojemann
- Seattle Children's Research Institute, Center for Integrative Brain Research, Seattle, WA, United States; Department of Neurological Surgery, University of Washington, Seattle, WA, United States
| | - A L Ko
- Department of Neurological Surgery, University of Washington, Seattle, WA, United States
| | - Y Shi
- Department of Electrical and Computer Engineering, University of Washington, Seattle, WA, United States
| | - J K Eng
- Proteomics Resource, University of Washington, Seattle, WA, United States
| | - J-M Ramirez
- Seattle Children's Research Institute, Center for Integrative Brain Research, Seattle, WA, United States; Department of Neurological Surgery, University of Washington, Seattle, WA, United States
| | - N P Poolos
- Department of Neurology and Regional Epilepsy Center, University of Washington, Seattle, WA, United States.
| |
Collapse
|
30
|
Hemizygous FLNA variant in West syndrome without periventricular nodular heterotopia. Hum Genome Var 2020; 7:43. [PMID: 33298907 PMCID: PMC7713383 DOI: 10.1038/s41439-020-00131-9] [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: 09/08/2020] [Revised: 10/29/2020] [Accepted: 10/29/2020] [Indexed: 11/21/2022] Open
Abstract
Pathogenic FLNA variants can be identified in patients with seizures accompanied by periventricular nodular heterotopia (PVNH). It is unusual to find FLNA aberrations in epileptic patients without PVNH on brain imaging. We report a boy with cryptogenic West syndrome followed by refractory seizures and psychomotor delay. We performed whole-exome sequencing and identified a de novo missense variant in FLNA. It is noteworthy that this patient showed no PVNH. As no other pathogenic variants were found in epilepsy-related genes, this FLNA variant likely caused West syndrome but with no PVNH.
Collapse
|
31
|
Hsieh LS, Wen JH, Nguyen LH, Zhang L, Getz S, Torres-Reveron J, Wang Y, Spencer DD, Bordey A. Ectopic HCN4 expression drives mTOR-dependent epilepsy in mice. Sci Transl Med 2020; 12:12/570/eabc1492. [PMID: 33208499 PMCID: PMC9888000 DOI: 10.1126/scitranslmed.abc1492] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 07/31/2020] [Indexed: 02/03/2023]
Abstract
The causative link between focal cortical malformations (FCMs) and epilepsy is well accepted, especially among patients with focal cortical dysplasia type II (FCDII) and tuberous sclerosis complex (TSC). However, the mechanisms underlying seizures remain unclear. Using a mouse model of TSC- and FCDII-associated FCM, we showed that FCM neurons were responsible for seizure activity via their unexpected abnormal expression of the hyperpolarization-activated cyclic nucleotide-gated potassium channel isoform 4 (HCN4), which is normally not present in cortical pyramidal neurons after birth. Increasing intracellular cAMP concentrations, which preferentially affects HCN4 gating relative to the other isoforms, drove repetitive firing of FCM neurons but not control pyramidal neurons. Ectopic HCN4 expression was dependent on the mechanistic target of rapamycin (mTOR), preceded the onset of seizures, and was also found in diseased neurons in tissue resected from patients with TSC and FCDII. Last, blocking HCN4 channel activity in FCM neurons prevented epilepsy in the mouse model. These findings suggest that HCN4 play a main role in seizure and identify a cAMP-dependent seizure mechanism in TSC and FCDII. Furthermore, the unique expression of HCN4 exclusively in FCM neurons suggests that gene therapy targeting HCN4 might be effective in reducing seizures in FCDII or TSC.
Collapse
Affiliation(s)
- Lawrence S. Hsieh
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - John H. Wen
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Lena H. Nguyen
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Longbo Zhang
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Stephanie Getz
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Juan Torres-Reveron
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Ying Wang
- Emergency Department, Xiangya Hospital, Central South University, 87 Xiangya Street, Changsha, Hunan 410008, China
| | - Dennis D. Spencer
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Angélique Bordey
- Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA,Department of Cellular & Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA,To whom correspondence should be addressed: Angélique Bordey, Ph.D., Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, FMB 422, New Haven, CT 06520-8082, Phone: 203-737-2515, Fax: 203-737-2159,
| |
Collapse
|
32
|
Li D, Liu X, Liu H, Li T, Jia S, Wang X, Wang P, Qin D, Wang YF. Key Roles of Cyclooxygenase 2-Protein Kinase A-Hyperpolarization-activated Cyclic Nucleotide-gated Channel 3 Pathway in the Regulation of Oxytocin Neuronal Activity in Lactating Rats with Intermittent Pup-Deprivation. Neuroscience 2020; 452:13-25. [PMID: 33137408 DOI: 10.1016/j.neuroscience.2020.10.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/09/2020] [Indexed: 12/14/2022]
Abstract
Suckling-evoked pulsatile release of oxytocin (OT) from the posterior pituitary plays a key role in breastfeeding, which relies on burst-like discharges of OT neurons. To explore cellular mechanisms regulating OT neuronal activity, using lactating rats with pup-deprivation (PD) during postpartum day 1-5, we observed the involvement of prostaglandin, cyclic AMP/protein kinase A (PKA) and hyperpolarization-activated cyclic nucleotide-gated channel 3 (HCN3) signaling pathway in OT neuronal activity. PD gradually reduced lactation efficiency. Intermittent PD (IPD) was largely reversed by intranasally-applied OT (IAO) but not by hypodermically-applied OT. IPD caused involution-like histological changes in the mammary glands, increased hypothalamic OT release but did not influence plasma OT concentrations. In the supraoptic nucleus, IPD increased OT receptor (OTR) expressions in OT neurons as well as Gαq subunit, Gβ subunit and cyclooxygenase 2 (Cox-2). These effects except that on Gβ subunit were reversed by IAO. Notably, IPD increased the expression of catalytic subunit of PKA in the SON, specifically in vasopressin neurons but not in OT neurons. In addition, IPD increased the expression of HCN3. IAO partially reversed these changes in the SON. Lastly, blocking HCN3 blocked excitation and burst firing in OT neurons-evoked by prostaglandin E2, a key mediator of OT-evoked burst firing; blocking Cox-2 or PKA reduced the molecular association between OTR and HCN3. Thus, there is a prostaglandin-cAMP/PKA-HCN3 pathway in the regulation of OT neuronal activity. PD disrupts lactation performance through uncoupling OTR and PKA-HCN3 signaling. The reversal effect of IAO highlights its therapeutic potential in PD-evoked hypogalactia.
Collapse
Affiliation(s)
- Dongyang Li
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Xiaoyu Liu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Haitao Liu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Tong Li
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Shuwei Jia
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Xiaoran Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Ping Wang
- Department of Genetics, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Danian Qin
- Department of Physiology, Shantou University of Medical College, Shantou, China
| | - Yu-Feng Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China.
| |
Collapse
|
33
|
T-Type Calcium Channels Contribute to Burst Firing in a Subpopulation of Medial Habenula Neurons. eNeuro 2020; 7:ENEURO.0201-20.2020. [PMID: 32719103 PMCID: PMC7433892 DOI: 10.1523/eneuro.0201-20.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 11/24/2022] Open
Abstract
Action potential (AP) burst firing caused by the activation of low-voltage-activated T-type Ca2+ channels is a unique mode of neuronal firing. T-type channels have been implicated in diverse physiological and pathophysiological processes, including epilepsy, autism, and mood regulation, but the brain structures involved remain incompletely understood. The medial habenula (MHb) is an epithalamic structure implicated in anxiety-like and withdrawal behavior. Previous studies have shown that MHb neurons fire tonic APs at a frequency of ∼2–10 Hz or display depolarized low-amplitude membrane oscillations. Here, we report in C57BL/6J mice that a subpopulation of MHb neurons are capable of firing transient, high-frequency AP bursts mediated by T-type channels. Burst firing was observed following rebounding from hyperpolarizing current injections or during depolarization from hyperpolarized membrane potentials in ∼20% of MHb neurons. It was rarely observed at baseline but could be evoked in MHb neurons displaying different initial activity states. Further, we show that T-type channel mRNA, in particular Cav3.1, is expressed in the MHb in both cholinergic and substance P-ergic neurons. Pharmacological Cav3 antagonism blocked both burst firing and evoked Ca2+ currents in MHb neurons. Additionally, we observed high-frequency AP doublet firing at sustained depolarized membrane potentials that was independent of T-type channels. Thus, there is a greater diversity of AP firing patterns in MHb neurons than previously identified, including T-type channel-mediated burst firing, which may uniquely contribute to behaviors with relevance to neuropsychiatric disease.
Collapse
|
34
|
Trompoukis G, Rigas P, Leontiadis LJ, Papatheodoropoulos C. I h, GIRK, and KCNQ/Kv7 channels differently modulate sharp wave - ripples in the dorsal and ventral hippocampus. Mol Cell Neurosci 2020; 107:103531. [PMID: 32711112 DOI: 10.1016/j.mcn.2020.103531] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 07/11/2020] [Accepted: 07/16/2020] [Indexed: 12/14/2022] Open
Abstract
Sharp waves and ripples (SPW-Rs) are endogenous transient patterns of hippocampus local network activity implicated in several functions including memory consolidation, and they are diversified between the dorsal and the ventral hippocampus. Ion channels in the neuronal membrane play important roles in cell and local network function. In this study, using transverse slices and field potential recordings from the CA1 field of rat hippocampus we show that GIRK and KCNQ2/3 potassium channels play a higher role in modulating SPW-Rs in the dorsal hippocampus, while Ih and other KCNQ (presumably KCNQ5) channels, contribute to shaping SPW-R activity more in the ventral than in dorsal hippocampus. Specifically, blockade of Ih channels by ZD 7288 reduced the rate of occurrence of SPW-Rs and increased the generation of SPW-Rs in the form of clusters in both hippocampal segments, while enhanced the amplitude of SPW-Rs only in the ventral hippocampus. Most effects of ZD 7288 appeared to be independent of NMDA receptors' activity. However, the effects of blockade of NMDA receptors depended on the functional state of Ih channels in both hippocampal segments. Blockade of GIRK channels by Tertiapin-Q increased the rate of occurrence of SPW-Rs only in the dorsal hippocampus and the probability of clusters in both segments of the hippocampus. Blockade of KCNQ2/3 channels by XE 991 increased the rate of occurrence of SPW-Rs and the probability of clusters in the dorsal hippocampus, and only reduced the clustered generation of SPW-Rs in the ventral hippocampus. The blocker of KCNQ1/2 channels, that also enhances KCNQ5 channels, UCL 2077, increased the probability of clusters and the power of the ripple oscillation in the ventral hippocampus only. These results suggest that GIRK, KCNQ and Ih channels represent a key mechanism for modulation of SPW-R activity which act differently in the dorsal and ventral hippocampus, fundamentally supporting functional diversification along the dorsal-ventral axis of the hippocampus.
Collapse
Affiliation(s)
- George Trompoukis
- Laboratory of Physiology, Department of Medicine, University of Patras, Rion, Greece
| | - Pavlos Rigas
- Laboratory of Physiology, Department of Medicine, University of Patras, Rion, Greece
| | - Leonidas J Leontiadis
- Laboratory of Physiology, Department of Medicine, University of Patras, Rion, Greece
| | | |
Collapse
|
35
|
Kim CS, Johnston D. Antidepressant Effects of (S)-Ketamine through a Reduction of Hyperpolarization-Activated Current I h. iScience 2020; 23:101239. [PMID: 32629607 PMCID: PMC7322259 DOI: 10.1016/j.isci.2020.101239] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 11/11/2019] [Accepted: 06/02/2020] [Indexed: 11/24/2022] Open
Abstract
Compelling evidence suggests that a single sub-anesthetic dose of (R,S)-ketamine exerts rapid and robust antidepressant effects. However, the cellular mechanisms underlying the antidepressant effects of (R,S)-ketamine remain unclear. Here, we show that (S)-ketamine reduced dendritic but not somatic hyperpolarization-activated current Ih of dorsal CA1 neurons in unstressed rats, whereas (S)-ketamine decreased both somatic and dendritic Ih in chronic unpredictable stress (CUS) rats. The reduction of Ih by (S)-ketamine was independent of NMDA receptors, barium-sensitive conductances, and cAMP-dependent signaling pathways in both unstressed and CUS groups. (S)-ketamine pretreatment before the onset of depression prevented CUS-induced behavioral phenotypes and neuropathological changes of dorsal CA1 neurons. Finally, in vivo infusion of thapsigargin-induced anxiogenic- and anhedonic-like behaviors and upregulation of functional Ih, but these were reversed by (S)-ketamine. Our results suggest that (S)-ketamine reduces or prevents Ih from being increased following CUS, which contributes to the rapid antidepressant effects and resiliency to CUS. (S)-ketamine reduced the CUS-induced upregulation of somatic Ih This was independent of NMDAR, Ba2+-sensitive conductances, and cAMP signaling (S)-ketamine pretreatment before the onset of depression provided resiliency to CUS In vivo thapsigargin-induced changes in behaviors were reversed by (S)-ketamine
Collapse
Affiliation(s)
- Chung Sub Kim
- Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, 1 University Station Stop, C7000, Austin, TX 78712, USA.
| | - Daniel Johnston
- Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, 1 University Station Stop, C7000, Austin, TX 78712, USA
| |
Collapse
|
36
|
Liang H, Paxinos G. Afferents of the mouse linear nucleus. Mol Brain 2020; 13:67. [PMID: 32370769 PMCID: PMC7201812 DOI: 10.1186/s13041-020-00602-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/08/2020] [Indexed: 11/10/2022] Open
Abstract
The linear nucleus (Li) was identified in 1978 from its projections to the cerebellum. However, there is no systematic study of its connections with other areas of the central nervous system possibly due to the challenge of injecting retrograde tracers into this nucleus. The present study examines its afferents from some nuclei involved in motor and cardiovascular control with anterograde tracer injections. BDA injections into the central amygdaloid nucleus result in labeled fibers to the ipsilateral Li. Bilateral projections with an ipsilateral dominance were observed after injections in a) jointly the paralemniscal nucleus, the noradrenergic group 7/ Köllike -Fuse nucleus/subcoeruleus nucleus, b) the gigantocellular reticular nucleus, c) and the solitary nucleus/the parvicellular/intermediate reticular nucleus. Retrogradely labeled neurons were observed in Li after BDA injections into all these nuclei except the central amygdaloid and the paralemniscal nuclei. Our results suggest that Li is involved in a variety of physiological functions apart from motor and balance control it may exert via its cerebellar projections.
Collapse
Affiliation(s)
- Huazheng Liang
- Department of Neurology, Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, 1878 North Sichuan Road, Hongkou District, Shanghai, 200081, China. .,Neuroscience Research Australia, Sydney, NSW, 2031, Australia.
| | - George Paxinos
- Neuroscience Research Australia, Sydney, NSW, 2031, Australia
| |
Collapse
|
37
|
Lee SY, Vuong TA, So HK, Kim HJ, Kim YB, Kang JS, Kwon I, Cho H. PRMT7 deficiency causes dysregulation of the HCN channels in the CA1 pyramidal cells and impairment of social behaviors. Exp Mol Med 2020; 52:604-614. [PMID: 32269286 PMCID: PMC7210990 DOI: 10.1038/s12276-020-0417-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/14/2020] [Accepted: 03/05/2020] [Indexed: 12/21/2022] Open
Abstract
HCN channels regulate excitability and rhythmicity in the hippocampal CA1 pyramidal cells. Perturbation in the HCN channel current (Ih) is associated with neuropsychiatric disorders, such as autism spectrum disorders. Recently, protein arginine methyltransferase 7 (PRMT7) was shown to be highly expressed in the hippocampus, including the CA1 region. However, the physiological function of PRMT7 in the CA1 neurons and the relationship to psychiatric disorders are unclear. Here we showed that PRMT7 knockout (KO) mice exhibit hyperactivity and deficits in social interaction. The firing frequency of the CA1 neurons in the PRMT7 KO mice was significantly higher than that in the wild-type (WT) mice. Compared with the WT CA1 neurons, the PRMT7 KO CA1 neurons showed a more hyperpolarized resting potential and a higher input resistance, which were occluded by the Ih-current inhibitor ZD7288; these findings were consistent with the decreased Ih and suggested the contribution of Ih-channel dysfunction to the PRMT7 KO phenotypes. The HCN1 protein level was decreased in the CA1 region of the PRMT7 KO mice in conjunction with a decrease in the expression of Shank3, which encodes a core scaffolding protein for HCN channel proteins. A brief application of the PRMT7 inhibitor DS437 did not reproduce the phenotype of the PRMT7 KO neurons, further indicating that PRMT7 regulates Ih by controlling the channel number rather than the open probability. Moreover, shRNA-mediated PRMT7 suppression reduced both the mRNA and protein levels of SHANK3, implying that PRMT7 deficiency might be responsible for the decrease in the HCN protein levels by altering Shank3 expression. These findings reveal a key role for PRMT7 in the regulation of HCN channel density in the CA1 pyramidal cells that may be amenable to pharmacological intervention for neuropsychiatric disorders. Disrupted expression of an ion channel that helps stabilize brain cell activity contributes to behavioral symptoms in mice resembling those seen in autism spectrum disorders (ASDs). Nerve cell firing depends on the right balance of ions inside and outside cells, and a channel protein called HCN helps establish ionic conditions that prevent excessive activity. Researchers led by Hana Cho and Ilmin Kwon of the Sungkyunkwan University School of Medicine, Suwon, South Korea have demonstrated that mice lacking another protein called PRMT7 exhibit reduced numbers of HCN channels in brain structures known to be affected in animal models of ASDs. These mice exhibit hyperactivity and social anxiety, presumably as a consequence of poor regulation of nerve cell firing. The authors propose that this PRMT7-HCN pathway may offer a fruitful target for the development of neuropsychiatric therapies.
Collapse
Affiliation(s)
- Seul-Yi Lee
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Tuan Anh Vuong
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Korea.,Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Hyun-Kyung So
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Korea.,Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Hyun-Ji Kim
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Yoo Bin Kim
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Jong-Sun Kang
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Korea.,Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Ilmin Kwon
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea.
| | - Hana Cho
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea. .,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Korea.
| |
Collapse
|
38
|
Wei F, Wang Q, Han J, Goswamee P, Gupta A, McQuiston AR, Liu Q, Zhou L. Photodynamic Modification of Native HCN Channels Expressed in Thalamocortical Neurons. ACS Chem Neurosci 2020; 11:851-863. [PMID: 32078767 DOI: 10.1021/acschemneuro.9b00475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The photodynamic process requires three elements: light, oxygen, and photosensitizer, and involves the formation of singlet oxygen, the molecular oxygen in excited electronic states. Previously, we reported that heterologously expressed hyperpolarization-activated cAMP-gated (HCN) channels in excised membrane patches are sensitive to photodynamic modification (PDM). Here we extend this study to native HCN channels expressed in thalamocortical (TC) neurons in the ventrobasal (VB) complex of the thalamus and dopaminergic neurons (DA) of the ventral tegmental area (VTA). To do this, we introduced the photosensitizer FITC-cAMP into TCs or DAs of rodent brain slices via a whole-cell patch-clamp recording pipette. After illumination with blue light pulses, we observed an increase in the voltage-insensitive, instantaneous Iinst component, accompanied by a long-lasting decrease in the hyperpolarization-dependent Ih component. Both Ih and the increased Iinst after PDM could be blocked by the HCN blockers Cs+ and ZD7288. When FITC and cAMP were dissociated and loaded into neurons as two separate chemicals, light application did not result in any long-lasting changes of the HCN currents. In contrast, light pulses applied to HCN2-/- neurons loaded with FITC-cAMP generated a much greater reduction in the Iinst component compared to that of WT neurons. Next, we investigated the impact of the long-lasting increases in Iinst after PDM on the cellular physiology of VB neurons. Consistent with an upregulation of HCN channel function, PDM elicited a depolarization of the resting membrane potential (RMP). Importantly, Trolox-C, an effective quencher for singlet oxygen, could block the PDM-dependent increase in Iinst and depolarization of the RMP. We propose that PDM of native HCN channels under physiological conditions may provide a photodynamic approach to alleviate HCN channelopathy in certain pathological conditions.
Collapse
Affiliation(s)
- Fusheng Wei
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330031, Jiangxi, China
| | - Qiang Wang
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Jizhong Han
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Priyodarshan Goswamee
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Ankush Gupta
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Adam Rory McQuiston
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Qinglian Liu
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Lei Zhou
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| |
Collapse
|
39
|
Lee CH, Park JH, Won MH. Protein expression changes of HCN1 and HCN2 in hippocampal subregions of gerbils during the normal aging process. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:1308-1313. [PMID: 32128096 PMCID: PMC7038419 DOI: 10.22038/ijbms.2019.35760.8520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 05/14/2019] [Indexed: 01/05/2023]
Abstract
OBJECTIVES Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels play essential roles in various hippocampal functions, including regulation of long-term potentiation, synaptic plasticity, and hippocampal-dependent cognitive process. The objective of this study was to investigate age-related changes in HCN1 and HCN2 protein expressions in gerbil hippocampus at various ages. MATERIALS AND METHODS In this study, the protein expressions of HCN1 and HCN2 were compared in the hippocampus at the ages of 1, 3, 12, and 24 months using Western blot analysis and immunohistochemistry. RESULTS Immunoreactivity of both HCN1 and HCN2 was shown primarily in cells of the pyramidal cell layer in the hippocampus proper and in cells of the granule cell layer in the dentate gyrus. HCN1 and HCN2 protein expression levels and immunoreactivity were significantly increased at three months (3 M) of age compared with those at 1 M of age. After that, both HCN1 and HCN2 expression levels in the hippocampus were gradually decreased with age. CONCLUSION Our results show that the normal aging process affects the expression levels of HCN1 and HCN2 in hippocampal cells in gerbils. There are marked reductions in HCN1 and HCN2 expressions in the aged hippocampus compared to the young hippocampus. Such reductions might be related to aging in the hippocampus.
Collapse
Affiliation(s)
- Choong-Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, Chungnam 31116, Republic of Korea
| | - Joon Ha Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongju, Gyeongbuk 38066, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| |
Collapse
|
40
|
Park JH, Kim DW, Lee TK, Park CW, Park YE, Ahn JH, Lee HA, Won MH, Lee CH. Improved HCN channels in pyramidal neurons and their new expression levels in pericytes and astrocytes in the gerbil hippocampal CA1 subfield following transient ischemia. Int J Mol Med 2019; 44:1801-1810. [PMID: 31573045 PMCID: PMC6777693 DOI: 10.3892/ijmm.2019.4353] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 09/04/2019] [Indexed: 11/30/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels have been known to participate in the regulation of neuronal excitability, synaptic transmission and long-term potentiation in the hippocampus. The present study investigated transient ischemia-induced changes of HCN1 and HCN2 expressions in the Cornu Ammonis 1 (CA1) subfield of the hippocampus in gerbils subjected to 5 min transient global cerebral ischemia (tgCI). Neuronal death was exhibited in pyramidal neurons of the striatum pyramidale in the CA1 subfield 4 days after tgCI. HCN1 and HCN2 immunoreactivities were demonstrated in intact CA1 pyramidal neurons, and were transiently and markedly increased in the CA pyramidal neurons at 6 h after ischemia. Thereafter, they gradually decreased in a time-dependent manner. A total of 4 days after ischemia, HCN1 and HCN2 immunoreactivities were barely detected in the CA1 pyramidal neurons; however, HCN1 and HCN2 were began to be expressed in pericytes and astrocytes at 4 days after ischemia. The results indicated that HCN1 and HCN2 expression levels were apparently changed in the gerbil hippocampal CA1 subfield following tgCI and suggested that ischemia-induced alterations in HCN1 and HCN2 expression levels may be closely associated with the death of CA1 pyramidal neurons following 5 min of tgCI.
Collapse
Affiliation(s)
- Joon Ha Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongju, Gyeongbuk 38066, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung‑Wonju National University, Gangneung, Gangwon 25457, Republic of Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Cheol Woo Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Young Eun Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Hyang-Ah Lee
- Department of Obstetrics and Gynecology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Choong-Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, Chungcheongnam 31116, Republic of Korea
| |
Collapse
|
41
|
Marini C, Porro A, Rastetter A, Dalle C, Rivolta I, Bauer D, Oegema R, Nava C, Parrini E, Mei D, Mercer C, Dhamija R, Chambers C, Coubes C, Thévenon J, Kuentz P, Julia S, Pasquier L, Dubourg C, Carré W, Rosati A, Melani F, Pisano T, Giardino M, Innes AM, Alembik Y, Scheidecker S, Santos M, Figueiroa S, Garrido C, Fusco C, Frattini D, Spagnoli C, Binda A, Granata T, Ragona F, Freri E, Franceschetti S, Canafoglia L, Castellotti B, Gellera C, Milanesi R, Mancardi MM, Clark DR, Kok F, Helbig KL, Ichikawa S, Sadler L, Neupauerová J, Laššuthova P, Šterbová K, Laridon A, Brilstra E, Koeleman B, Lemke JR, Zara F, Striano P, Soblet J, Smits G, Deconinck N, Barbuti A, DiFrancesco D, LeGuern E, Guerrini R, Santoro B, Hamacher K, Thiel G, Moroni A, DiFrancesco JC, Depienne C. HCN1 mutation spectrum: from neonatal epileptic encephalopathy to benign generalized epilepsy and beyond. Brain 2019; 141:3160-3178. [PMID: 30351409 DOI: 10.1093/brain/awy263] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 08/09/2018] [Indexed: 12/15/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels control neuronal excitability and their dysfunction has been linked to epileptogenesis but few individuals with neurological disorders related to variants altering HCN channels have been reported so far. In 2014, we described five individuals with epileptic encephalopathy due to de novo HCN1 variants. To delineate HCN1-related disorders and investigate genotype-phenotype correlations further, we assembled a cohort of 33 unpublished patients with novel pathogenic or likely pathogenic variants: 19 probands carrying 14 different de novo mutations and four families with dominantly inherited variants segregating with epilepsy in 14 individuals, but not penetrant in six additional individuals. Sporadic patients had epilepsy with median onset at age 7 months and in 36% the first seizure occurred during a febrile illness. Overall, considering familial and sporadic patients, the predominant phenotypes were mild, including genetic generalized epilepsies and genetic epilepsy with febrile seizures plus (GEFS+) spectrum. About 20% manifested neonatal/infantile onset otherwise unclassified epileptic encephalopathy. The study also included eight patients with variants of unknown significance: one adopted patient had two HCN1 variants, four probands had intellectual disability without seizures, and three individuals had missense variants inherited from an asymptomatic parent. Of the 18 novel pathogenic missense variants identified, 12 were associated with severe phenotypes and clustered within or close to transmembrane domains, while variants segregating with milder phenotypes were located outside transmembrane domains, in the intracellular N- and C-terminal parts of the channel. Five recurrent variants were associated with similar phenotypes. Using whole-cell patch-clamp, we showed that the impact of 12 selected variants ranged from complete loss-of-function to significant shifts in activation kinetics and/or voltage dependence. Functional analysis of three different substitutions altering Gly391 revealed that these variants had different consequences on channel biophysical properties. The Gly391Asp variant, associated with the most severe, neonatal phenotype, also had the most severe impact on channel function. Molecular dynamics simulation on channel structure showed that homotetramers were not conducting ions because the permeation path was blocked by cation(s) strongly complexed to the Asp residue, whereas heterotetramers showed an instantaneous current component possibly linked to deformation of the channel pore. In conclusion, our results considerably expand the clinical spectrum related to HCN1 variants to include common generalized epilepsy phenotypes and further illustrate how HCN1 has a pivotal function in brain development and control of neuronal excitability.
Collapse
Affiliation(s)
- Carla Marini
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Department, A Meyer Children's Hospital, University of Florence, Viale Pieraccini 24, Florence, Italy.,EuroEPINOMICS RES Consortium
| | | | - Agnès Rastetter
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Carine Dalle
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Ilaria Rivolta
- School of Medicine and Surgery, University Milano-Bicocca, Monza, Italy
| | - Daniel Bauer
- Computational Biology and Simulation Group, Department of Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Renske Oegema
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Caroline Nava
- EuroEPINOMICS RES Consortium.,Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Département de Génétique, Paris, France
| | - Elena Parrini
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Department, A Meyer Children's Hospital, University of Florence, Viale Pieraccini 24, Florence, Italy
| | - Davide Mei
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Department, A Meyer Children's Hospital, University of Florence, Viale Pieraccini 24, Florence, Italy
| | - Catherine Mercer
- Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, UK
| | - Radhika Dhamija
- Department of Clinical Genomics and Neurology, Mayo Clinic, Phoenix, AZ, USA
| | - Chelsea Chambers
- Department of Neurosciences, University of Virginia, Charlottesville, VA, USA
| | - Christine Coubes
- Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Hôpital Arnaud de Villeneuve, Montpellier, France
| | - Julien Thévenon
- FHU-TRANSLAD, Université de Bourgogne/CHU Dijon and INSERM UMR 1231 GAD team, Genetics of Developmental Anomalies, Université de Bourgogne-Franche Comté, Dijon, France
| | - Paul Kuentz
- FHU-TRANSLAD, Université de Bourgogne/CHU Dijon and INSERM UMR 1231 GAD team, Genetics of Developmental Anomalies, Université de Bourgogne-Franche Comté, Dijon, France.,Génétique Biologique Histologie, CHRU de Besançon, Besançon, France
| | - Sophie Julia
- Service de génétique médicale, Pôle de biologie, CHU de Toulouse - Hôpital Purpan, Toulouse, France
| | - Laurent Pasquier
- Service de Génétique Clinique, Centre Référence Déficiences Intellectuelles de causes rares (CRDI), CHU Rennes, Rennes, France
| | - Christèle Dubourg
- Laboratoire de Génétique Moléculaire et Génomique, CHU de Rennes, Rennes, France
| | - Wilfrid Carré
- Laboratoire de Génétique Moléculaire et Génomique, CHU de Rennes, Rennes, France
| | - Anna Rosati
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Department, A Meyer Children's Hospital, University of Florence, Viale Pieraccini 24, Florence, Italy
| | - Federico Melani
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Department, A Meyer Children's Hospital, University of Florence, Viale Pieraccini 24, Florence, Italy
| | - Tiziana Pisano
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Department, A Meyer Children's Hospital, University of Florence, Viale Pieraccini 24, Florence, Italy
| | - Maria Giardino
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Department, A Meyer Children's Hospital, University of Florence, Viale Pieraccini 24, Florence, Italy
| | - A Micheil Innes
- Department of Medical Genetics and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Yves Alembik
- Laboratoires de génétique, Institut de génétique médicale d'Alsace, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Sophie Scheidecker
- Laboratoires de génétique, Institut de génétique médicale d'Alsace, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Manuela Santos
- Neuropediatric Department, Centro Hospitalar do Porto, Porto, Portugal
| | - Sonia Figueiroa
- Neuropediatric Department, Centro Hospitalar do Porto, Porto, Portugal
| | - Cristina Garrido
- Neuropediatric Department, Centro Hospitalar do Porto, Porto, Portugal
| | - Carlo Fusco
- Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Daniele Frattini
- Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Carlotta Spagnoli
- Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Anna Binda
- School of Medicine and Surgery, University Milano-Bicocca, Monza, Italy
| | - Tiziana Granata
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | - Elena Freri
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | | | | | - Cinzia Gellera
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Raffaella Milanesi
- Department of Biosciences, The PaceLab, Università degli Studi di Milano, Milan, Italy
| | - Maria Margherita Mancardi
- Child Neuropsychiatry Unit, Department of Medical and Surgical Neurosciences and Rehabilitation, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | | | - Fernando Kok
- Mendelics Genomic Analysis, Sao Paulo, SP, Brazil
| | - Katherine L Helbig
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Shoji Ichikawa
- Department of Clinical Diagnostics, Ambry Genetics, Aliso Viejo, CA, USA
| | - Laurie Sadler
- Division of Genetics, Department of Pediatrics, Oishei Children's Hospital, Jacobs School of Medicine and Biomedical Sciences, University of Buffalo, State University of New York, Buffalo, NY, USA
| | - Jana Neupauerová
- Department of Child Neurology, Charles University 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Petra Laššuthova
- Department of Child Neurology, Charles University 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Katalin Šterbová
- EuroEPINOMICS RES Consortium.,Department of Child Neurology, Charles University 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Annick Laridon
- Department of Neurology, Academic Center for Epileptology, Kempenhaeghe/Maastricht University Medical Center, Heeze, The Netherlands
| | - Eva Brilstra
- EuroEPINOMICS RES Consortium.,Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bobby Koeleman
- EuroEPINOMICS RES Consortium.,Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Johannes R Lemke
- EuroEPINOMICS RES Consortium.,Institute of Human Genetics, University of Leipzig Hospitals and Clinics, Leipzig, Germany
| | - Federico Zara
- Laboratory of Neurogenetics and Neuroscience, Institute G Gaslini, Genova, Italy
| | - Pasquale Striano
- EuroEPINOMICS RES Consortium.,Pediatric Neurology and Muscular Diseases Unit, Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 'G Gaslini' Institute, Genova, Italy
| | - Julie Soblet
- Department of Genetics, Hôpital Universitaire des Enfants Reine Fabiola, ULB Center of Human Genetics, Université Libre de Bruxelles, Brussels, Belgium.,Department of Genetics, Hôpital Erasme ULB Center of Human Genetics, Université Libre de Bruxelles, Brussels, Belgium.,Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles, Brussels, Belgium
| | - Guillaume Smits
- Department of Genetics, Hôpital Universitaire des Enfants Reine Fabiola, ULB Center of Human Genetics, Université Libre de Bruxelles, Brussels, Belgium.,Department of Genetics, Hôpital Erasme ULB Center of Human Genetics, Université Libre de Bruxelles, Brussels, Belgium.,Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles, Brussels, Belgium
| | - Nicolas Deconinck
- Department of Pediatric Neurology, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles, ULB, Brussels, Belgium
| | - Andrea Barbuti
- Department of Biosciences, The PaceLab, Università degli Studi di Milano, Milan, Italy
| | - Dario DiFrancesco
- Department of Biosciences, The PaceLab, Università degli Studi di Milano, Milan, Italy
| | - Eric LeGuern
- EuroEPINOMICS RES Consortium.,Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Département de Génétique, Paris, France
| | - Renzo Guerrini
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Department, A Meyer Children's Hospital, University of Florence, Viale Pieraccini 24, Florence, Italy.,EuroEPINOMICS RES Consortium
| | - Bina Santoro
- Department of Neuroscience, Columbia University, New York, NY, USA
| | - Kay Hamacher
- Computational Biology and Simulation Group, Department of Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Gerhard Thiel
- Membrane Biophysics, Deparment of Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Anna Moroni
- Department of Biosciences, University of Milan, Milan, Italy
| | - Jacopo C DiFrancesco
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Department of Neurology, San Gerardo Hospital, University Milano-Bicocca, Monza, Italy
| | - Christel Depienne
- EuroEPINOMICS RES Consortium.,Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,IGBMC, CNRS UMR 7104/INSERM U964/Université de Strasbourg, Illkirch, France.,Institute of Human Genetics, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
42
|
Lussier Y, Fürst O, Fortea E, Leclerc M, Priolo D, Moeller L, Bichet DG, Blunck R, D'Avanzo N. Disease-linked mutations alter the stoichiometries of HCN-KCNE2 complexes. Sci Rep 2019; 9:9113. [PMID: 31235733 PMCID: PMC6591248 DOI: 10.1038/s41598-019-45592-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 06/11/2019] [Indexed: 12/21/2022] Open
Abstract
The four hyperpolarization-activated cylic-nucleotide gated (HCN) channel isoforms and their auxiliary subunit KCNE2 are important in the regulation of peripheral and central neuronal firing and the heartbeat. Disruption of their normal function has been implicated in cardiac arrhythmias, peripheral pain, and epilepsy. However, molecular details of the HCN-KCNE2 complexes are unknown. Using single-molecule subunit counting, we determined that the number of KCNE2 subunits in complex with the pore-forming subunits of human HCN channels differs with each HCN isoform and is dynamic with respect to concentration. These interactions can be altered by KCNE2 gene-variants with functional implications. The results provide an additional consideration necessary to understand heart rhythm, pain, and epileptic disorders.
Collapse
Affiliation(s)
- Yoann Lussier
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Canada
| | - Oliver Fürst
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Canada
| | - Eva Fortea
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Canada
| | - Marc Leclerc
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Canada
| | - Dimitri Priolo
- Department of Physics, Université de Montréal, Montréal, Canada
| | - Lena Moeller
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Canada
| | - Daniel G Bichet
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Canada
| | - Rikard Blunck
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Canada.,Department of Physics, Université de Montréal, Montréal, Canada
| | - Nazzareno D'Avanzo
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Canada. .,Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Canada.
| |
Collapse
|
43
|
Zobeiri M, Chaudhary R, Blaich A, Rottmann M, Herrmann S, Meuth P, Bista P, Kanyshkova T, Lüttjohann A, Narayanan V, Hundehege P, Meuth SG, Romanelli MN, Urbano FJ, Pape HC, Budde T, Ludwig A. The Hyperpolarization-Activated HCN4 Channel is Important for Proper Maintenance of Oscillatory Activity in the Thalamocortical System. Cereb Cortex 2019; 29:2291-2304. [PMID: 30877792 PMCID: PMC6458902 DOI: 10.1093/cercor/bhz047] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/15/2019] [Accepted: 02/21/2019] [Indexed: 11/14/2022] Open
Abstract
Hyperpolarization-activated cation channels are involved, among other functions, in learning and memory, control of synaptic transmission and epileptogenesis. The importance of the HCN1 and HCN2 isoforms for brain function has been demonstrated, while the role of HCN4, the third major neuronal HCN subunit, is not known. Here we show that HCN4 is essential for oscillatory activity in the thalamocortical (TC) network. HCN4 is selectively expressed in various thalamic nuclei, excluding the thalamic reticular nucleus. HCN4-deficient TC neurons revealed a massive reduction of Ih and strongly reduced intrinsic burst firing, whereas the current was normal in cortical pyramidal neurons. In addition, evoked bursting in a thalamic slice preparation was strongly reduced in the mutant mice probes. HCN4-deficiency also significantly slowed down thalamic and cortical oscillations during active wakefulness. Taken together, these results establish that thalamic HCN4 channels are essential for the production of rhythmic intrathalamic oscillations and determine regular TC oscillatory activity during alert states.
Collapse
Affiliation(s)
- Mehrnoush Zobeiri
- Institut für Physiologie I, Westfälische Wilhelms-Universität, Münster, Germany
| | - Rahul Chaudhary
- Institut für Physiologie I, Westfälische Wilhelms-Universität, Münster, Germany
| | - Anne Blaich
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Rottmann
- Institut für Physiologie I, Westfälische Wilhelms-Universität, Münster, Germany
| | - Stefan Herrmann
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Patrick Meuth
- Klinik für Neurologie mit Institut für Translationale Neurologie, Westfälische Wilhelms-Universität, Münster, Germany
| | - Pawan Bista
- Institut für Physiologie I, Westfälische Wilhelms-Universität, Münster, Germany
| | - Tatyana Kanyshkova
- Institut für Physiologie I, Westfälische Wilhelms-Universität, Münster, Germany
| | - Annika Lüttjohann
- Institut für Physiologie I, Westfälische Wilhelms-Universität, Münster, Germany
| | - Venu Narayanan
- Klinik für Neurologie mit Institut für Translationale Neurologie, Westfälische Wilhelms-Universität, Münster, Germany
| | - Petra Hundehege
- Klinik für Neurologie mit Institut für Translationale Neurologie, Westfälische Wilhelms-Universität, Münster, Germany
| | - Sven G Meuth
- Klinik für Neurologie mit Institut für Translationale Neurologie, Westfälische Wilhelms-Universität, Münster, Germany
| | - Maria Novella Romanelli
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Via Ugo Schiff 6, Sesto Fiorentino, Italy
| | | | - Hans-Christian Pape
- Institut für Physiologie I, Westfälische Wilhelms-Universität, Münster, Germany
| | - Thomas Budde
- Institut für Physiologie I, Westfälische Wilhelms-Universität, Münster, Germany
| | - Andreas Ludwig
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
44
|
Santos-Vera B, Vaquer-Alicea ADC, Maria-Rios CE, Montiel-Ramos A, Ramos-Cardona A, Vázquez-Torres R, Sanabria P, Jiménez-Rivera CA. Protein and surface expression of HCN2 and HCN4 subunits in mesocorticolimbic areas after cocaine sensitization. Neurochem Int 2019; 125:91-98. [PMID: 30794847 DOI: 10.1016/j.neuint.2019.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/05/2019] [Accepted: 02/06/2019] [Indexed: 01/20/2023]
Abstract
The Ih is a mixed depolarizing current present in neurons which, upon activation by hyperpolarization, modulates neuronal excitability in the mesocorticolimbic (MCL) system, an area which regulates emotions such as pleasure, reward, and motivation. Its biophysical properties are determined by HCN protein expression profiles, specifically HCN subunits 1-4. Previously, we reported that cocaine-induced behavioral sensitization increases HCN2 protein expression in all MCL areas with the Ventral Tegmental Area (VTA) showing the most significant increase. Recent evidence suggests that HCN4 also has an important expression in the MCL system. Although there is a significant expression of HCN channels in the MCL system their role in addictive processes is largely unknown. Thus, in this study we aim to compare HCN2 and HCN4 expression profiles and their cellular compartmental distribution in the MCL system, before and after cocaine sensitization. Surface/intracellular (S/I) ratio analysis indicates that VTA HCN2 subunits are mostly expressed in the cell surface in contrast to other areas tested. Our findings demonstrate that after cocaine sensitization, the HCN2 S/I ratio in the VTA was decreased whereas in the Prefrontal Cortex it was increased. In addition, HCN4 total expression in the VTA was decreased after cocaine sensitization, although the S/I ratio was not altered. Together, these results demonstrate differential cocaine effects on HCN2 and HCN4 protein expression profiles and therefore suggest a diverse Ih modulation of cellular activity during cocaine addictive processes.
Collapse
Affiliation(s)
- Bermary Santos-Vera
- Physiology Department, University of Puerto Rico, School of Medicine, Medical Sciences Campus, San Juan, 00936-5067, Puerto Rico
| | - Ana Del C Vaquer-Alicea
- Physiology Department, University of Puerto Rico, School of Medicine, Medical Sciences Campus, San Juan, 00936-5067, Puerto Rico
| | - Cristina E Maria-Rios
- Biology Department, University of Puerto Rico, Rio Piedras Campus, San Juan, 00936-5067, Puerto Rico
| | - Alan Montiel-Ramos
- Biology Department, University of Puerto Rico, Rio Piedras Campus, San Juan, 00936-5067, Puerto Rico
| | - Aynette Ramos-Cardona
- Psychology Department, University of Puerto Rico, Rio Piedras Campus, San Juan, 00936-5067, Puerto Rico
| | - Rafael Vázquez-Torres
- Physiology Department, University of Puerto Rico, School of Medicine, Medical Sciences Campus, San Juan, 00936-5067, Puerto Rico
| | - Priscila Sanabria
- Physiology Department, Universidad Central del Caribe, Bayamon, 00960, Puerto Rico
| | - Carlos A Jiménez-Rivera
- Physiology Department, University of Puerto Rico, School of Medicine, Medical Sciences Campus, San Juan, 00936-5067, Puerto Rico.
| |
Collapse
|
45
|
Rivera-Meza M. The Hyperpolarization-Activated Cyclic Nucleotide-Gated Ion Channels in the Rewarding Effects of Ethanol. NEUROSCIENCE OF ALCOHOL 2019:171-178. [DOI: 10.1016/b978-0-12-813125-1.00018-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
46
|
Feetham CH, O'Brien F, Barrett-Jolley R. Ion Channels in the Paraventricular Hypothalamic Nucleus (PVN); Emerging Diversity and Functional Roles. Front Physiol 2018; 9:760. [PMID: 30034342 PMCID: PMC6043726 DOI: 10.3389/fphys.2018.00760] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 05/30/2018] [Indexed: 12/11/2022] Open
Abstract
The paraventricular nucleus of the hypothalamus (PVN) is critical for the regulation of homeostatic function. Although also important for endocrine regulation, it has been referred to as the "autonomic master controller." The emerging consensus is that the PVN is a multifunctional nucleus, with autonomic roles including (but not limited to) coordination of cardiovascular, thermoregulatory, metabolic, circadian and stress responses. However, the cellular mechanisms underlying these multifunctional roles remain poorly understood. Neurones from the PVN project to and can alter the function of sympathetic control regions in the medulla and spinal cord. Dysfunction of sympathetic pre-autonomic neurones (typically hyperactivity) is linked to several diseases including hypertension and heart failure and targeting this region with specific pharmacological or biological agents is a promising area of medical research. However, to facilitate future medical exploitation of the PVN, more detailed models of its neuronal control are required; populated by a greater compliment of constituent ion channels. Whilst the cytoarchitecture, projections and neurotransmitters present in the PVN are reasonably well documented, there have been fewer studies on the expression and interplay of ion channels. In this review we bring together an up to date analysis of PVN ion channel studies and discuss how these channels may interact to control, in particular, the activity of the sympathetic system.
Collapse
Affiliation(s)
- Claire H Feetham
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Fiona O'Brien
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Richard Barrett-Jolley
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
47
|
Yin XL, Jie HQ, Liang M, Gong LN, Liu HW, Pan HL, Xing YZ, Shi HB, Li CY, Wang LY, Yin SK. Accelerated Development of the First-Order Central Auditory Neurons With Spontaneous Activity. Front Mol Neurosci 2018; 11:183. [PMID: 29904342 PMCID: PMC5990604 DOI: 10.3389/fnmol.2018.00183] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 05/11/2018] [Indexed: 11/24/2022] Open
Abstract
In developing sensory systems, elaborate morphological connectivity between peripheral cells and first-order central neurons emerges via genetic programming before the onset of sensory activities. However, how the first-order central neurons acquire the capacity to interface with peripheral cells remains elusive. By making patch-clamp recordings from mouse brainstem slices, we found that a subset of neurons in the cochlear nuclei, the first central station to receive peripheral acoustic impulses, exhibits spontaneous firings (SFs) as early as at birth, and the fraction of such neurons increases during the prehearing period. SFs are reduced but not eliminated by a cocktail of blockers for excitatory and inhibitory synaptic inputs, implicating the involvement of intrinsic pacemaker channels. Furthermore, we demonstrate that these intrinsic firings (IFs) are largely driven by hyperpolarization- and cyclic nucleotide-gated channel (HCN) mediated currents (Ih), as evidenced by their attenuation in the presence of HCN blockers or in neurons from HCN1 knockout mice. Interestingly, genetic deletion of HCN1 cannot be fully compensated by other pacemaker conductances and precludes age-dependent up regulation in the fraction of spontaneous active neurons and their firing rate. Surprisingly, neurons with SFs show accelerated development in excitability, spike waveform and firing pattern as well as synaptic pruning towards mature phenotypes compared to those without SFs. Our results imply that SFs of the first-order central neurons may reciprocally promote their wiring and firing with peripheral inputs, potentially enabling the correlated activity and crosstalk between the developing brain and external environment.
Collapse
Affiliation(s)
- Xin-Lu Yin
- Department of Otorhinolaryngology, The Sixth People's Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Hui-Qun Jie
- Department of Otorhinolaryngology, The Sixth People's Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Min Liang
- Department of Otorhinolaryngology, The Sixth People's Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Li-Na Gong
- Department of Otorhinolaryngology, The Sixth People's Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Han-Wei Liu
- Department of Otorhinolaryngology, The Sixth People's Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Hao-Lai Pan
- Department of Otorhinolaryngology, The Sixth People's Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Ya-Zhi Xing
- Department of Otorhinolaryngology, The Sixth People's Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Hai-Bo Shi
- Department of Otorhinolaryngology, The Sixth People's Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Chun-Yan Li
- Department of Otorhinolaryngology, The Sixth People's Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Lu-Yang Wang
- Programs in Neurosciences & Mental Health, Department of Physiology, Sick Kids Research Institute, Toronto, ON, Canada
| | - Shan-Kai Yin
- Department of Otorhinolaryngology, The Sixth People's Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
48
|
Kim CS, Brager DH, Johnston D. Perisomatic changes in h-channels regulate depressive behaviors following chronic unpredictable stress. Mol Psychiatry 2018; 23:892-903. [PMID: 28416809 PMCID: PMC5647208 DOI: 10.1038/mp.2017.28] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 01/10/2017] [Accepted: 01/17/2017] [Indexed: 12/23/2022]
Abstract
Chronic stress can be a precipitating factor in the onset of depression. Lentiviral-mediated knockdown of HCN1 protein expression and reduction of functional Ih produce antidepressant behavior. However, whether h-channels are altered in an animal model of depression is not known. We found that perisomatic HCN1 protein expression and Ih-sensitive physiological measurements were significantly increased in dorsal but not in ventral CA1 region/neurons following chronic unpredictable stress (CUS), a widely accepted model for major depressive disorder. Cell-attached patch clamp recordings confirmed that perisomatic Ih was increased in dorsal CA1 neurons following CUS. Furthermore, when dorsal CA1 Ih was reduced by shRNA-HCN1, the CUS-induced behavioral deficits were prevented. Finally, rats infused in the dorsal CA1 region with thapsigargin, an irreversible inhibitor of the SERCA pump, exhibited anxiogenic-like behaviors and increased Ih, similar to that observed following CUS. Our results suggest that CUS, but not acute stress, leads to an increase in perisomatic Ih in dorsal CA1 neurons and that HCN channels represent a potential target for the treatment of major depressive disorder.
Collapse
Affiliation(s)
- C S Kim
- Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, TX, USA,Neuroscience, Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, 100 East 24th St, Austin, TX 78712-0805, USA. E-mail:
| | - D H Brager
- Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, TX, USA
| | - D Johnston
- Center for Learning and Memory and Department of Neuroscience, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
49
|
Zhong P, Vickstrom CR, Liu X, Hu Y, Yu L, Yu HG, Liu QS. HCN2 channels in the ventral tegmental area regulate behavioral responses to chronic stress. eLife 2018; 7:32420. [PMID: 29256865 PMCID: PMC5749952 DOI: 10.7554/elife.32420] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 12/18/2017] [Indexed: 12/17/2022] Open
Abstract
Dopamine neurons in the ventral tegmental area (VTA) are powerful regulators of depression-related behavior. Dopamine neuron activity is altered in chronic stress-based models of depression, but the underlying mechanisms remain incompletely understood. Here, we show that mice subject to chronic mild unpredictable stress (CMS) exhibit anxiety- and depressive-like behavior, which was associated with decreased VTA dopamine neuron firing in vivo and ex vivo. Dopamine neuron firing is governed by voltage-gated ion channels, in particular hyperpolarization-activated cyclic nucleotide-gated (HCN) channels. Following CMS, HCN-mediated currents were decreased in nucleus accumbens-projecting VTA dopamine neurons. Furthermore, shRNA-mediated HCN2 knockdown in the VTA was sufficient to recapitulate CMS-induced depressive- and anxiety-like behavior in stress-naïve mice, whereas VTA HCN2 overexpression largely prevented CMS-induced behavioral deficits. Together, these results reveal a critical role for HCN2 in regulating VTA dopamine neuronal activity and depressive-related behaviors.
Collapse
Affiliation(s)
- Peng Zhong
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
| | - Casey R Vickstrom
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
| | - Xiaojie Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
| | - Ying Hu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
| | - Laikang Yu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
| | - Han-Gang Yu
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, United States
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, United States
| |
Collapse
|
50
|
Hou L, Guo Y, Lian B, Wang Y, Li C, Wang G, Li Q, Pang J, Sun H, Sun L. Synaptic Ultrastructure Might Be Involved in HCN 1-Related BDNF mRNA in Withdrawal-Anxiety After Ethanol Dependence. Front Psychiatry 2018; 9:215. [PMID: 29896126 PMCID: PMC5986948 DOI: 10.3389/fpsyt.2018.00215] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 05/07/2018] [Indexed: 12/27/2022] Open
Abstract
Withdrawal from ethanol dependence has been associated with heightened anxiety and reduced expression of Brain-derived neurotropic factor which promotes the synaptic transmission and plasticity of synapses. Hyperpolarization-activated cyclic nucleotide-gated channel 1 regulates expression; however, whether Hyperpolarization-activated cyclic nucleotide-gated channel 1-related Brain-derived neurotropic factor is involved in the synaptic ultrastructure that generates withdrawal-anxiety has been poorly perceived. Sprague-Dawley rats were treated with ethanol 3-9% (v/v) for a period of 21 days. Conditioned place preference and body weight were investigated during ethanol administration. Rats were subjected to behavioral testing and biochemical assessments after ethanol withdrawal, which was induced by abrupt discontinuation of the treatment. The results showed that the ethanol administration induced severe ethanol dependence behaviors, with higher body weight and more time in the ethanol-paired compartment. After withdrawal, rats had a higher total ethanol withdrawal score and explored less. Additionally, increased Hyperpolarization-activated cyclic nucleotide-gated channel 1 protein and gene expression and decreased Brain-derived neurotropic factor protein and gene expression were detected in the Ethanol group. Eventually, there was a negative correlation between the level of Brain-derived neurotropic factor mRNA and Hyperpolarization-activated cyclic nucleotide-gated channel 1 protein. Importantly, the synaptic ultrastructure changed in the Ethanol group, including increased synaptic cleft width and reduction in postsynaptic density thickness or synaptic curvature. The synthesis of the Brain-derived neurotropic factor mRNA could be down-regulated by higher Hyperpolarization-activated cyclic nucleotide-gated channel 1 protein expression. Changes in synaptic ultrastructure may be induced by lower Brain-derived neurotropic factor protein, which could be associated with the withdrawal-anxiety that is experiences after ethanol dependence.
Collapse
Affiliation(s)
- Lanwei Hou
- Department of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Yujuan Guo
- Department of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Bo Lian
- Department of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Yanyu Wang
- Department of Psychology, Weifang Medical University, Weifang, China
| | - Changjiang Li
- Department of Psychology, Weifang Medical University, Weifang, China
| | - Gang Wang
- Laboratory for Cognitive Neuroscience, Weifang Medical University, Weifang, China
| | - Qi Li
- Department of Psychiatry and Centre for Reproduction Growth and Development, University of Hong Kong, Hong Kong, Hong Kong
| | - Jinjing Pang
- Department of Rehabilitation Medicine, Han Ting People's Hospital of Weifang, Weifang, China
| | - Hongwei Sun
- Department of Psychology, Weifang Medical University, Weifang, China
| | - Lin Sun
- Department of Clinical Medicine, Weifang Medical University, Weifang, China.,Department of Psychology, Weifang Medical University, Weifang, China
| |
Collapse
|