1
|
So TW, Choi HY, Xu H, Zhu J, Shi L, Ip JPK. Hippocampal dorsal CA1: Functional connectivity and role in HCN channelopathies in affective diseases and epilepsy. IBRO Neurosci Rep 2025; 18:644-656. [PMID: 40292082 PMCID: PMC12023878 DOI: 10.1016/j.ibneur.2025.03.008] [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: 09/14/2024] [Revised: 01/31/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
The hippocampus is a complex structure consisting of the dentate gyrus (DG), cornu ammonis (CA) and the subiculum. CA1 is further subdivided into the ventral (vCA1) and dorsal (dCA1) compartments, with dCA1 believed to be crucial in spatial learning and memory as well as cognitive processing. Although dCA1 was traditionally thought to be not likely relevant to affective diseases, recent studies suggest otherwise. In fact, it has been found that diseases including certain types of post-traumatic stress disorder (PTSD), depression and epilepsy may be attributed to channelopathies in dCA1, particularly that of hyperpolarization-activated cyclic nucleotide gated (HCN) channels. However, it remains unclear how disruptions of HCN transcription, post-transcriptional modification and activation kinetics are related to changes of downstream structures along neural circuits. Their effect on behavioural changes and disease development, as well as the corresponding potential therapeutic strategies implicated in the findings have not been extensively studied as well. With the existing research gap and the significant clinical implications of dCA1 HCN channelopathies, the mechanisms of how defects of these channels result in brain disorders including PTSD, depression and temporal lobe epilepsy are worthy of further investigation. Therefore, in this review, we summarize the recent findings on the involvement of dCA1 HCN channelopathies in brain disorders after providing an outline on the neuroanatomy and functional connectivity of dCA1, and the features of HCN channels in that region. We also propose future directions of molecular and systems neuroscience studies, as well as the translational research on potential therapeutics that address the brain disorders related to dCA1 HCN channelopathies.
Collapse
Affiliation(s)
- Tsz Wei So
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Hoi Yi Choi
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Haoyu Xu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jinwei Zhu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lei Shi
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Pharmacy, Jinan University, Guangzhou, Guangdong 510632, China
| | - Jacque Pak Kan Ip
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
2
|
Benndorf K, Enke U, Tewari D, Kusch J, Liu H, Sun H, Schmauder R, Sattler C. Subunit-specific conductance of single homomeric and heteromeric HCN pacemaker channels at femtosiemens resolution. Proc Natl Acad Sci U S A 2025; 122:e2422533122. [PMID: 39879240 PMCID: PMC11804576 DOI: 10.1073/pnas.2422533122] [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: 10/30/2024] [Accepted: 01/03/2025] [Indexed: 01/31/2025] Open
Abstract
In mammals, the four subunit isoforms HCN1-4 assemble to form functional homotetrameric and heterotetrameric hyperpolarization-activated cyclic nucleotide-modulated (HCN) ion channels. Despite the outstanding relevance of HCN channels for organisms, including generating electrical rhythmicity in cardiac pacemaker cells and diverse types of brain neurons, key channel properties are still elusive. In particular, the unitary conductance, γ, of HCN channels is highly controversial. We analyzed the unitary conductance at femtosiemens resolution of all four homotetrameric channels of the mouse, mHCN1-4. All conductance values are in the range of 1 pS which is exceptionally small compared to most other ion channels. Surprisingly, the conductance among the isoforms differs up to threefold (γmHCN2 = 1.54 pS > γmHCN1 = 0.84 pS > γmHCN3 = 0.54 pS ≈ γmHCN4 = 0.51 pS) though the residues in the two narrow parts of the pore, the selectivity filter and the inner gate, are conserved. Mutagenesis and all-atom molecular dynamics simulations demonstrate that the differences in the conductance are generated by different amounts of negative charges in the outer channel vestibule, which control ion accumulation. In line with these results, heterotetrameric channels exhibit intermediate unitary conductance values with respect to the homotetrameric channels. Our approach demonstrates how HCN channels can be functionally differentiated at the single-channel level, paving the way to target specific channels with selective drugs.
Collapse
Affiliation(s)
- Klaus Benndorf
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena07740, Germany
| | - Uta Enke
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena07740, Germany
| | - Debanjan Tewari
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena07740, Germany
| | - Jana Kusch
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena07740, Germany
| | - Haoran Liu
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin13125, Germany
- Institute of Chemistry, Technical University of Berlin, Berlin10623, Germany
| | - Han Sun
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin13125, Germany
- Institute of Chemistry, Technical University of Berlin, Berlin10623, Germany
| | - Ralf Schmauder
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena07740, Germany
| | - Christian Sattler
- Institut für Physiologie II, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena07740, Germany
| |
Collapse
|
3
|
Mishra P, Narayanan R. The enigmatic HCN channels: A cellular neurophysiology perspective. Proteins 2025; 93:72-92. [PMID: 37982354 PMCID: PMC7616572 DOI: 10.1002/prot.26643] [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: 08/03/2023] [Revised: 10/24/2023] [Accepted: 11/09/2023] [Indexed: 11/21/2023]
Abstract
What physiological role does a slow hyperpolarization-activated ion channel with mixed cation selectivity play in the fast world of neuronal action potentials that are driven by depolarization? That puzzling question has piqued the curiosity of physiology enthusiasts about the hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, which are widely expressed across the body and especially in neurons. In this review, we emphasize the need to assess HCN channels from the perspective of how they respond to time-varying signals, while also accounting for their interactions with other co-expressing channels and receptors. First, we illustrate how the unique structural and functional characteristics of HCN channels allow them to mediate a slow negative feedback loop in the neurons that they express in. We present the several physiological implications of this negative feedback loop to neuronal response characteristics including neuronal gain, voltage sag and rebound, temporal summation, membrane potential resonance, inductive phase lead, spike triggered average, and coincidence detection. Next, we argue that the overall impact of HCN channels on neuronal physiology critically relies on their interactions with other co-expressing channels and receptors. Interactions with other channels allow HCN channels to mediate intrinsic oscillations, earning them the "pacemaker channel" moniker, and to regulate spike frequency adaptation, plateau potentials, neurotransmitter release from presynaptic terminals, and spike initiation at the axonal initial segment. We also explore the impact of spatially non-homogeneous subcellular distributions of HCN channels in different neuronal subtypes and their interactions with other channels and receptors. Finally, we discuss how plasticity in HCN channels is widely prevalent and can mediate different encoding, homeostatic, and neuroprotective functions in a neuron. In summary, we argue that HCN channels form an important class of channels that mediate a diversity of neuronal functions owing to their unique gating kinetics that made them a puzzle in the first place.
Collapse
Affiliation(s)
- Poonam Mishra
- Department of Neuroscience, Yale School of MedicineYale UniversityNew HavenConnecticutUSA
| | - Rishikesh Narayanan
- Cellular Neurophysiology Laboratory, Molecular Biophysics UnitIndian Institute of ScienceBangaloreIndia
| |
Collapse
|
4
|
Wojciechowski M, Jokiel J, Kuss H, Bermúdez M, Jose J. Combination of Autodisplay and Dynamic Pharmacophore Modeling Reveals New Insights into Cyclic Nucleotide Binding in Hyperpolarization-Activated and Cyclic Nucleotide-Gated Ion Channel 4 (HCN4). ACS Pharmacol Transl Sci 2024; 7:4010-4020. [PMID: 39698292 PMCID: PMC11651207 DOI: 10.1021/acsptsci.4c00497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 12/20/2024]
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) ion channels play a critical role in regulating neuronal and cardiac rhythmicity, with their function being modulated by cyclic nucleotide binding. Dysfunction of HCN ion channels leads to the genesis of several diseases such as arrhythmia, bradycardia, or epilepsy. This study employs a multidisciplinary approach integrating mutagenesis, ligand binding assays, and molecular dynamics (MD) simulations combined with dynamic pharmacophore studies to investigate the impact of single residue mutations within the cyclic nucleotide-binding domain (CNBD) of HCN4 channels. Utilizing an autodisplay-based ligand binding assay, surface-displayed HCN4 CNBD mutants were evaluated for their interaction with 8-Fluo-cAMP, providing insights into the ligand binding properties. While some known mutational effects could be confirmed (R669, T670), we identified L652 to be crucial for successful ligand binding. Surprisingly, C662, located in the center of the binding pocket, was discovered to play a negligible role in cAMP-binding. Both E660 and R710 were shown to substantially affect 8-Fluo-cAMP-binding, uncovering the direct ligand binding capability of the R710A mutant for the first time. Furthermore, MD simulations coupled with dynamic pharmacophore analysis offered detailed insights into dynamic ligand-protein interactions, elucidating the structural basis of ligand binding and modulation induced by single residue mutations. Here, a novel bypass mechanism of R713 that interacts with cAMP in the absence of R710 was demonstrated. These findings unveil new perspectives on cyclic nucleotide binding in HCN4 channels, providing a foundation for future studies of pathogenic HCN4 ion channel mutations.
Collapse
Affiliation(s)
- Magdalena
N. Wojciechowski
- University of Münster, Institute
of Pharmaceutical and Medicinal Chemistry, Pharma Campus, Corrensstr. 48, 48149 Münster, Germany
| | - Johannes Jokiel
- University of Münster, Institute
of Pharmaceutical and Medicinal Chemistry, Pharma Campus, Corrensstr. 48, 48149 Münster, Germany
| | - Hanna Kuss
- University of Münster, Institute
of Pharmaceutical and Medicinal Chemistry, Pharma Campus, Corrensstr. 48, 48149 Münster, Germany
| | - Marcel Bermúdez
- University of Münster, Institute
of Pharmaceutical and Medicinal Chemistry, Pharma Campus, Corrensstr. 48, 48149 Münster, Germany
| | - Joachim Jose
- University of Münster, Institute
of Pharmaceutical and Medicinal Chemistry, Pharma Campus, Corrensstr. 48, 48149 Münster, Germany
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Lin JL, Chang Y, Tewari D, Cowgill J, Chanda B. Mapping the contribution of the C-linker domain to gating polarity in CNBD channels. Biophys J 2024; 123:2176-2184. [PMID: 38678368 PMCID: PMC11309966 DOI: 10.1016/j.bpj.2024.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/23/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024] Open
Abstract
Ion channels of the cyclic nucleotide-binding domain (CNBD) family play a crucial role in the regulation of key biological processes, such as photoreception and pacemaking activity in the heart. These channels exhibit high sequence and structural similarity but differ greatly in their functional responses to membrane potential. The CNBD family includes hyperpolarization-activated ion channels and depolarization-activated ether-à-go-go channels. Structural and functional studies show that the differences in the coupling interface between these two subfamilies' voltage-sensing domain and pore domain may underlie their differential response to membrane polarity. However, other structural components may also contribute to defining the polarity differences in activation. Here, we focus on the role of the C-terminal domain, which interacts with elements in both the pore and voltage-sensing domains. By generating a series of chimeras involving the C-terminal domain derived from distant members of the CNBD family, we find that the nature of the C-termini profoundly influences the gating polarity of these ion channels. Scanning mutagenesis of the C-linker region, a helix-turn-helix motif connecting the pore helix to the CNBD, reveals that residues at the intersubunit interface between the C-linkers are crucial for hyperpolarization-dependent activation. These findings highlight the unique and unexpected role of the intersubunit interface of the C-linker region in regulating the gating polarity of voltage-gated ion channels.
Collapse
Affiliation(s)
- Jenna L Lin
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri; Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, Missouri; Graduate Program in Biochemistry, Biophysics, & Structural Biology, Washington University School of Medicine, St. Louis, Missouri
| | - Yongchang Chang
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri; Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, Missouri
| | - Debanjan Tewari
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri; Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, Missouri
| | - John Cowgill
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri; Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, Missouri
| | - Baron Chanda
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri; Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
8
|
Wu J, El-Hassar L, Datta D, Thomas M, Zhang Y, Jenkins DP, DeLuca NJ, Chatterjee M, Gribkoff VK, Arnsten AFT, Kaczmarek LK. Interaction Between HCN and Slack Channels Regulates mPFC Pyramidal Cell Excitability in Working Memory Circuits. Mol Neurobiol 2024; 61:2430-2445. [PMID: 37889366 DOI: 10.1007/s12035-023-03719-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 10/17/2023] [Indexed: 10/28/2023]
Abstract
The ability of monkeys and rats to carry out spatial working memory tasks has been shown to depend on the persistent firing of pyramidal cells in the prefrontal cortex (PFC), arising from recurrent excitatory connections on dendritic spines. These spines express hyperpolarization-activated cyclic nucleotide-gated (HCN) channels whose open state is increased by cAMP signaling, and which markedly alter PFC network connectivity and neuronal firing. In traditional neural circuits, activation of these non-selective cation channels leads to neuronal depolarization and increased firing rate. Paradoxically, cAMP activation of HCN channels in PFC pyramidal cells reduces working memory-related neuronal firing. This suggests that activation of HCN channels may hyperpolarize rather than depolarize these neurons. The current study tested the hypothesis that Na+ influx through HCN channels activates Slack Na+-activated K+ (KNa) channels to hyperpolarize the membrane. We have found that HCN and Slack KNa channels co-immunoprecipitate in cortical extracts and that, by immunoelectron microscopy, they colocalize at postsynaptic spines of PFC pyramidal neurons. A specific blocker of HCN channels, ZD7288, reduces KNa current in pyramidal cells that express both HCN and Slack channels, but has no effect on KNa currents in an HEK cell line expressing Slack without HCN channels, indicating that blockade of HCN channels in neurons reduces K+ current indirectly by lowering Na+ influx. Activation of HCN channels by cAMP in a cell line expressing a Ca2+ reporter results in elevation of cytoplasmic Ca2+, but the effect of cAMP is reversed if the HCN channels are co-expressed with Slack channels. Finally, we used a novel pharmacological blocker of Slack channels to show that inhibition of Slack in rat PFC improves working memory performance, an effect previously demonstrated for blockers of HCN channels. Our results suggest that the regulation of working memory by HCN channels in PFC pyramidal neurons is mediated by an HCN-Slack channel complex that links activation HCN channels to suppression of neuronal excitability.
Collapse
Affiliation(s)
- Jing Wu
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Lynda El-Hassar
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Dibyadeep Datta
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Merrilee Thomas
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Yalan Zhang
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - David P Jenkins
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Nicholas J DeLuca
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Manavi Chatterjee
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Valentin K Gribkoff
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Amy F T Arnsten
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Leonard K Kaczmarek
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, 06520, USA.
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
9
|
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
|
10
|
Porro A, Saponaro A, Castelli R, Introini B, Hafez Alkotob A, Ranjbari G, Enke U, Kusch J, Benndorf K, Santoro B, DiFrancesco D, Thiel G, Moroni A. A high affinity switch for cAMP in the HCN pacemaker channels. Nat Commun 2024; 15:843. [PMID: 38287019 PMCID: PMC10825183 DOI: 10.1038/s41467-024-45136-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 01/16/2024] [Indexed: 01/31/2024] Open
Abstract
Binding of cAMP to Hyperpolarization activated cyclic nucleotide gated (HCN) channels facilitates pore opening. It is unclear why the isolated cyclic nucleotide binding domain (CNBD) displays in vitro lower affinity for cAMP than the full-length channel in patch experiments. Here we show that HCN are endowed with an affinity switch for cAMP. Alpha helices D and E, downstream of the cyclic nucleotide binding domain (CNBD), bind to and stabilize the holo CNBD in a high affinity state. These helices increase by 30-fold cAMP efficacy and affinity measured in patch clamp and ITC, respectively. We further show that helices D and E regulate affinity by interacting with helix C of the CNBD, similarly to the regulatory protein TRIP8b. Our results uncover an intramolecular mechanism whereby changes in binding affinity, rather than changes in cAMP concentration, can modulate HCN channels, adding another layer to the complex regulation of their activity.
Collapse
Affiliation(s)
| | - Andrea Saponaro
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milano, Italy
| | | | - Bianca Introini
- Department of Biosciences, University of Milan, Milano, Italy
| | | | - Golnaz Ranjbari
- Department of Biosciences, University of Milan, Milano, Italy
| | - Uta Enke
- Institut für Physiologie II, Universitätsklinikum Jena, Jena, Germany
| | - Jana Kusch
- Institut für Physiologie II, Universitätsklinikum Jena, Jena, Germany
| | - Klaus Benndorf
- Institut für Physiologie II, Universitätsklinikum Jena, Jena, Germany
| | - Bina Santoro
- Department of Neuroscience, Zuckerman Institute, Columbia University, New York, NY, USA
| | | | - Gerhard Thiel
- Department of Biology, TU-Darmstadt, Darmstadt, Germany
| | - Anna Moroni
- Department of Biosciences, University of Milan, Milano, Italy.
- Institute of Biophysics Milan, Consiglio Nazionale delle Ricerche, Milano, Italy.
| |
Collapse
|
11
|
Al-attar R, Jargstorf J, Romagnuolo R, Jouni M, Alibhai FJ, Lampe PD, Solan JL, Laflamme MA. Casein Kinase 1 Phosphomimetic Mutations Negatively Impact Connexin-43 Gap Junctions in Human Pluripotent Stem Cell-Derived Cardiomyocytes. Biomolecules 2024; 14:61. [PMID: 38254663 PMCID: PMC10813327 DOI: 10.3390/biom14010061] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/14/2023] [Accepted: 12/25/2023] [Indexed: 01/24/2024] Open
Abstract
The transplantation of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) has shown promise in preclinical models of myocardial infarction, but graft myocardium exhibits incomplete host-graft electromechanical integration and a propensity for pro-arrhythmic behavior. Perhaps contributing to this situation, hPSC-CM grafts show low expression of connexin 43 (Cx43), the major gap junction (GJ) protein, in ventricular myocardia. We hypothesized that Cx43 expression and function could be rescued by engineering Cx43 in hPSC-CMs with a series of phosphatase-resistant mutations at three casein kinase 1 phosphorylation sites (Cx43-S3E) that have been previously reported to stabilize Cx43 GJs and reduce arrhythmias in transgenic mice. However, contrary to our predictions, transgenic Cx43-S3E hPSC-CMs exhibited reduced Cx43 expression relative to wild-type cells, both at baseline and following ischemic challenge. Cx43-S3E hPSC-CMs showed correspondingly slower conduction velocities, increased automaticity, and differential expression of other connexin isoforms and various genes involved in cardiac excitation-contraction coupling. Cx43-S3E hPSC-CMs also had phosphorylation marks associated with Cx43 GJ internalization, a finding that may account for their impaired GJ localization. Taken collectively, our data indicate that the Cx43-S3E mutation behaves differently in hPSC-CMs than in adult mouse ventricular myocytes and that multiple biological factors likely need to be addressed synchronously to ensure proper Cx43 expression, localization, and function.
Collapse
Affiliation(s)
- Rasha Al-attar
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (R.A.-a.); (J.J.); (R.R.); (M.J.); (F.J.A.)
| | - Joseph Jargstorf
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (R.A.-a.); (J.J.); (R.R.); (M.J.); (F.J.A.)
| | - Rocco Romagnuolo
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (R.A.-a.); (J.J.); (R.R.); (M.J.); (F.J.A.)
| | - Mariam Jouni
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (R.A.-a.); (J.J.); (R.R.); (M.J.); (F.J.A.)
| | - Faisal J. Alibhai
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (R.A.-a.); (J.J.); (R.R.); (M.J.); (F.J.A.)
| | - Paul D. Lampe
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (P.D.L.); (J.L.S.)
| | - Joell L. Solan
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (P.D.L.); (J.L.S.)
| | - Michael A. Laflamme
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (R.A.-a.); (J.J.); (R.R.); (M.J.); (F.J.A.)
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5G 1L7, Canada
| |
Collapse
|
12
|
Tibbs GR, Uprety R, Warren JD, Beyer NP, Joyce RL, Ferrer MA, Mellado W, Wong VSC, Goldberg DC, Cohen MW, Costa CJ, Li Z, Zhang G, Dephoure NE, Barman DN, Sun D, Ingólfsson HI, Sauve AA, Willis DE, Goldstein PA. An anchor-tether 'hindered' HCN1 inhibitor is antihyperalgesic in a rat spared nerve injury neuropathic pain model. Br J Anaesth 2023; 131:745-763. [PMID: 37567808 PMCID: PMC10541997 DOI: 10.1016/j.bja.2023.06.067] [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: 04/13/2023] [Revised: 06/20/2023] [Accepted: 06/29/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Neuropathic pain impairs quality of life, is widely prevalent, and incurs significant costs. Current pharmacological therapies have poor/no efficacy and significant adverse effects; safe and effective alternatives are needed. Hyperpolarisation-activated cyclic nucleotide-regulated (HCN) channels are causally implicated in some forms of peripherally mediated neuropathic pain. Whilst 2,6-substituted phenols, such as 2,6-di-tert-butylphenol (26DTB-P), selectively inhibit HCN1 gating and are antihyperalgesic, the development of therapeutically tolerable, HCN-selective antihyperalgesics based on their inverse agonist activity requires that such drugs spare the cardiac isoforms and do not cross the blood-brain barrier. METHODS In silico molecular dynamics simulation, in vitro electrophysiology, and in vivo rat spared nerve injury methods were used to test whether 'hindered' variants of 26DTB-P (wherein a hydrophilic 'anchor' is attached in the para-position of 26DTB-P via an acyl chain 'tether') had the desired properties. RESULTS Molecular dynamics simulation showed that membrane penetration of hindered 26DTB-Ps is controlled by a tethered diol anchor without elimination of head group rotational freedom. In vitro and in vivo analysis showed that BP4L-18:1:1, a variant wherein a diol anchor is attached to 26DTB-P via an 18-carbon tether, is an HCN1 inverse agonist and an orally available antihyperalgesic. With a CNS multiparameter optimisation score of 2.25, a >100-fold lower drug load in the brain vs blood, and an absence of adverse cardiovascular or CNS effects, BP4L-18:1:1 was shown to be poorly CNS penetrant and cardiac sparing. CONCLUSIONS These findings provide a proof-of-concept demonstration that anchor-tethered drugs are a new chemotype for treatment of disorders involving membrane targets.
Collapse
Affiliation(s)
- Gareth R Tibbs
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Rajendra Uprety
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - J David Warren
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Nicole P Beyer
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Rebecca L Joyce
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Matthew A Ferrer
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | | | | | | | | | | | - Zhucui Li
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Guoan Zhang
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Noah E Dephoure
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Dipti N Barman
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA
| | - Delin Sun
- Lawrence Livermore National Laboratory, Livermore, CA, USA
| | | | - Anthony A Sauve
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Dianna E Willis
- Burke Neurological Institute, White Plains, NY, USA; Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| | - Peter A Goldstein
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA; Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, New York, NY, USA; Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
13
|
Ślęczkowska M, Misra K, Santoro S, Gerrits MM, Hoeijmakers JGJ. Ion Channel Genes in Painful Neuropathies. Biomedicines 2023; 11:2680. [PMID: 37893054 PMCID: PMC10604193 DOI: 10.3390/biomedicines11102680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Neuropathic pain (NP) is a typical symptom of peripheral nerve disorders, including painful neuropathy. The biological mechanisms that control ion channels are important for many cell activities and are also therapeutic targets. Disruption of the cellular mechanisms that govern ion channel activity can contribute to pain pathophysiology. The voltage-gated sodium channel (VGSC) is the most researched ion channel in terms of NP; however, VGSC impairment is detected in only <20% of painful neuropathy patients. Here, we discuss the potential role of the other peripheral ion channels involved in sensory signaling (transient receptor potential cation channels), neuronal excitation regulation (potassium channels), involuntary action potential generation (hyperpolarization-activated cyclic nucleotide-gated channels), thermal pain (anoctamins), pH modulation (acid sensing ion channels), and neurotransmitter release (calcium channels) related to pain and their prospective role as therapeutic targets for painful neuropathy.
Collapse
Affiliation(s)
- Milena Ślęczkowska
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, The Netherlands;
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Kaalindi Misra
- Laboratory of Human Genetics of Neurological Disorders, IRCCS San Raffaele Scientific Institute, INSPE, 20132 Milan, Italy; (K.M.); (S.S.)
| | - Silvia Santoro
- Laboratory of Human Genetics of Neurological Disorders, IRCCS San Raffaele Scientific Institute, INSPE, 20132 Milan, Italy; (K.M.); (S.S.)
| | - Monique M. Gerrits
- Department of Clinical Genetics, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands;
| | - Janneke G. J. Hoeijmakers
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
14
|
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
|
15
|
McKenzie CE, Hung A, Phillips AM, Soh MS, Reid CA, Forster IC. The Potential Antidepressant Compound Org 34167 Modulates HCN Channels Via a Novel Mode of Action. Mol Pharmacol 2023; 104:62-72. [PMID: 37280099 DOI: 10.1124/molpharm.123.000676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/19/2023] [Accepted: 05/19/2023] [Indexed: 06/08/2023] Open
Abstract
Org 34167 is a small molecule hyperpolarization-activated cyclic nucleotide-gated (HCN) channel modulator that has been trialed in humans for its potential antidepressant activity. The precise action of Org 34167 is not fully understood. Here we use two-electrode voltage clamp recordings and an allosteric model to explore the interaction of Org 34167 with human HCN1 channels. The impact of Org 34167 on channel function included a hyperpolarizing shift in activation voltage dependence and a slowing of activation kinetics. Furthermore, a reduction in the maximum open probability at extreme hyperpolarization argued for an additional voltage-independent mechanism. Org 34167 had a similar impact on a truncated HCN1 channel lacking the C-terminal nucleotide binding domain, thus ruling out an interaction with this domain. Fitting a gating model, derived from a 10-state allosteric scheme, predicted that Org 34167 strongly reduced the equilibrium constant for the voltage-independent pore domain to favor a closed pore, as well as reducing the voltage sensing domain-pore domain coupling and shifting the zero voltage equilibrium constant of the voltage sensing domain to favor the inactive state. SIGNIFICANCE STATEMENT: The brain penetrant small molecule Org 34167 has been reported to have an antidepressant action by targeting HCN channels; however, its mode of action is unknown. We used heterologously expressed human HCN1 channels to show that Org 34167 inhibits channel activity by modulating kinetic parameters associated with the channel pore domain, voltage sensing domain, and interdomain coupling.
Collapse
Affiliation(s)
- Chaseley E McKenzie
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia (C.E.M., A.M.P., M.S.S., C.A.R, I.C.F.); School of Science, STEM College, RMIT University, Melbourne, VIC, Australia (A.H.); and School of Biosciences, The University of Melbourne, Parkville, VIC, Australia (A.M.P.)
| | - Andrew Hung
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia (C.E.M., A.M.P., M.S.S., C.A.R, I.C.F.); School of Science, STEM College, RMIT University, Melbourne, VIC, Australia (A.H.); and School of Biosciences, The University of Melbourne, Parkville, VIC, Australia (A.M.P.)
| | - A Marie Phillips
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia (C.E.M., A.M.P., M.S.S., C.A.R, I.C.F.); School of Science, STEM College, RMIT University, Melbourne, VIC, Australia (A.H.); and School of Biosciences, The University of Melbourne, Parkville, VIC, Australia (A.M.P.)
| | - Ming S Soh
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia (C.E.M., A.M.P., M.S.S., C.A.R, I.C.F.); School of Science, STEM College, RMIT University, Melbourne, VIC, Australia (A.H.); and School of Biosciences, The University of Melbourne, Parkville, VIC, Australia (A.M.P.)
| | - Christopher A Reid
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia (C.E.M., A.M.P., M.S.S., C.A.R, I.C.F.); School of Science, STEM College, RMIT University, Melbourne, VIC, Australia (A.H.); and School of Biosciences, The University of Melbourne, Parkville, VIC, Australia (A.M.P.)
| | - Ian C Forster
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia (C.E.M., A.M.P., M.S.S., C.A.R, I.C.F.); School of Science, STEM College, RMIT University, Melbourne, VIC, Australia (A.H.); and School of Biosciences, The University of Melbourne, Parkville, VIC, Australia (A.M.P.)
| |
Collapse
|
16
|
Wojciechowski MN, Schreiber S, Jose J. A Novel Flow Cytometry-Based Assay for the Identification of HCN4 CNBD Ligands. Pharmaceuticals (Basel) 2023; 16:ph16050710. [PMID: 37242492 DOI: 10.3390/ph16050710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/25/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Hyperpolarization-activated and cyclic nucleotide-gated (HCN) channels are promising therapeutic targets because of their association with the genesis of several diseases. The identification of selective compounds that alter cAMP-induced ion channel modulation by binding to the cyclic nucleotide-binding domain (CNBD) will facilitate HCN channel-specific drug development. In this study, a fast and protein purification-free ligand-binding approach with a surface-displayed HCN4 C-Linker-CNBD on E. coli is presented. 8-Fluo-cAMP ligand binding was monitored by single-cell analysis via flow cytometry, and a Kd-value of 173 ± 46 nM was determined. The Kd value was confirmed by ligand depletion analysis and equilibrium state measurements. Applying increasing concentrations of cAMP led to a concentration-dependent decrease in fluorescence intensity, indicating a displacement of 8-Fluo-cAMP. A Ki-value of 8.5 ± 2 µM was determined. The linear relationship of IC50 values obtained for cAMP as a function of ligand concentration confirmed the competitive binding mode: IC50: 13 ± 2 µM/16 ± 3 µM/23 ± 1 µM/27 ± 1 µM for 50 nM/150 nM/250 nM/500 nM 8-Fluo-cAMP. A similar competitive mode of binding was confirmed for 7-CH-cAMP, and an IC50 value of 230 ± 41 nM and a Ki of 159 ± 29 nM were determined. Two established drugs were tested in the assay. Ivabradine, an approved HCN channel pore blocker and gabapentin, is known to bind to HCN4 channels in preference to other isoforms with an unknown mode of action. As expected, ivabradine had no impact on ligand binding. In addition, gabapentin had no influence on 8-Fluo-cAMP's binding to HCN4-CNBD. This is the first indication that gabapentin is not interacting with this part of the HCN4 channel. The ligand-binding assay as described can be used to determine binding constants for ligands such as cAMP and derivatives. It could also be applied for the identification of new ligands binding to the HCN4-CNBD.
Collapse
Affiliation(s)
- Magdalena N Wojciechowski
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, Pharmacampus, 48149 Münster, Germany
| | - Sebastian Schreiber
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, Pharmacampus, 48149 Münster, Germany
| | - Joachim Jose
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, Pharmacampus, 48149 Münster, Germany
| |
Collapse
|
17
|
Wu J, El-Hassar L, Datta D, Thomas M, Zhang Y, Jenkins DP, DeLuca NJ, Chatterjee M, Gribkoff VK, Arnsten AFT, Kaczmarek LK. Interaction Between HCN and Slack Channels Regulates mPFC Pyramidal Cell Excitability and Working Memory. RESEARCH SQUARE 2023:rs.3.rs-2870277. [PMID: 37205397 PMCID: PMC10187370 DOI: 10.21203/rs.3.rs-2870277/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The ability of monkeys and rats to carry out spatial working memory tasks has been shown to depend on the persistent firing of pyramidal cells in the prefrontal cortex (PFC), arising from recurrent excitatory connections on dendritic spines. These spines express hyperpolarization-activated cyclic nucleotide-gated (HCN) channels whose open state is increased by cAMP signaling, and which markedly alter PFC network connectivity and neuronal firing. In traditional neural circuits, activation of these non-selective cation channels leads to neuronal depolarization and increased firing rate. Paradoxically, cAMP activation of HCN channels in PFC pyramidal cells reduces working memory-related neuronal firing. This suggests that activation of HCN channels may hyperpolarize rather than depolarize these neurons. The current study tested the hypothesis that Na+ influx through HCN channels activates Slack Na+-activated K+ (KNa) channels to hyperpolarize the membrane. We have found that HCN and Slack KNa channels coimmunoprecipitate in cortical extracts and that, by immunoelectron microscopy, they colocalize at postsynaptic spines of PFC pyramidal neurons. A specific blocker of HCN channels, ZD7288, reduces KNa current in pyramidal cells that express both HCN and Slack channels, but has no effect on KNa currents in an HEK cell line expressing Slack without HCN channels, indicating that blockade of HCN channels in neurons reduces K+ +current indirectly by lowering Na+ influx. Activation of HCN channels by cAMP in a cell line expressing a Ca2+ reporter results in elevation of cytoplasmic Ca2+, but the effect of cAMP is reversed if the HCN channels are co-expressed with Slack channels. Finally, we used a novel pharmacological blocker of Slack channels to show that inhibition of Slack in rat PFC improves working memory performance, an effect previously demonstrated for blockers of HCN channels. Our results suggest that the regulation of working memory by HCN channels in PFC pyramidal neurons is mediated by an HCN-Slack channel complex that links activation HCN channels to suppression of neuronal excitability.
Collapse
Affiliation(s)
- Jing Wu
- Yale University School of Medicine
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Wu X, Cunningham KP, Ramentol R, Perez ME, Larsson HP. Similar voltage-sensor movement in spHCN channels can cause closing, opening, or inactivation. J Gen Physiol 2023; 155:e202213170. [PMID: 36752823 PMCID: PMC9948645 DOI: 10.1085/jgp.202213170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 11/22/2022] [Accepted: 01/27/2023] [Indexed: 02/09/2023] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels contribute to the rhythmic firing of pacemaker neurons and cardiomyocytes. Mutations in HCN channels are associated with cardiac arrhythmia and epilepsy. HCN channels belong to the superfamily of voltage-gated K+ channels, most of which are activated by depolarization. HCN channels, however, are activated by hyperpolarization. The mechanism behind this reversed gating polarity of HCN channels is not clear. We here show that sea urchin HCN (spHCN) channels with mutations in the C-terminal part of the voltage sensor use the same voltage-sensor movement to either close or open in response to hyperpolarizations depending on the absence or presence of cAMP. Our results support that non-covalent interactions at the C-terminal end of the voltage sensor are critical for HCN gating polarity. These interactions are also critical for the proper closing of the channels because these mutations exhibit large constitutive currents. Since a similar voltage-sensor movement can cause both depolarization- and hyperpolarization-activation in the same channel, this suggests that the coupling between the voltage sensor and the pore is changed to create channels opened by different polarities. We also show an identical voltage-sensor movement in activated and inactivated spHCN channels and suggest a model for spHCN activation and inactivation. Our results suggest the possibility that channels open by opposite voltage dependence, such as HCN and the related EAG channels, use the same voltage-sensor movement but different coupling mechanisms between the voltage sensor and the gate.
Collapse
Affiliation(s)
- Xiaoan Wu
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kevin P. Cunningham
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rosamary Ramentol
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Marta E. Perez
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - H. Peter Larsson
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
19
|
Bi-directional modulation of hyperpolarization-activated cation currents (I h) by ethanol in rat hippocampal CA3 pyramidal neurons. Neuropharmacology 2023; 227:109423. [PMID: 36690323 DOI: 10.1016/j.neuropharm.2023.109423] [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: 09/09/2022] [Revised: 01/12/2023] [Accepted: 01/15/2023] [Indexed: 01/22/2023]
Abstract
It is widely acknowledged that ethanol (EtOH) can alter many neuronal functions, including synaptic signaling, firing discharge, and membrane excitability, through its interaction with multiple membrane proteins and intracellular pathways. Previous work has demonstrated that EtOH enhances the firing rate of hippocampal GABAergic interneurons and thus the presynaptic GABA release at CA1 and CA3 inhibitory synapses through a positive modulation of the hyperpolarization-activated cyclic nucleotide-gated cation (HCN) channels. Activation of HCN channels produce an inward current, commonly called Ih, which plays an essential role in generating/regulating specific neuronal activities in GABAergic interneurons and principal glutamatergic pyramidal neurons such as those in the CA3 subregion. Since the direct effect of EtOH on HCN channels expressed in CA3 pyramidal neurons was not thoroughly elucidated, we investigated the possible interaction between EtOH and HCN channels and the impact on excitability and postsynaptic integration of these neurons. Patch-clamp recordings were performed in single CA3 pyramidal neurons from acute male rat coronal hippocampal slices. Our results show that EtOH modulates HCN-mediated Ih in a concentration-dependent and bi-directional manner, with a positive modulation at lower (20 mM) and an inhibitory action at higher (60-80 mM) concentrations. The modulation of Ih by EtOH was mimicked by forskolin, antagonized by different drugs that selectively interfere with the AC/cAMP/PKA intracellular pathway, as well as by the selective HCN inhibitor ZD7288. Altogether, these data further support the evidence that HCN channels may represent an important molecular target through which EtOH may regulate neuronal activity.
Collapse
|
20
|
Sitnikova E, Rutskova E, Smirnov K. Alpha2-Adrenergic Receptors as a Pharmacological Target for Spike-Wave Epilepsy. Int J Mol Sci 2023; 24:1477. [PMID: 36674992 PMCID: PMC9862736 DOI: 10.3390/ijms24021477] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/30/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Spike-wave discharges are the hallmark of idiopathic generalized epilepsy. They are caused by a disorder in the thalamocortical network. Commercially available anti-epileptic drugs have pronounced side effects (i.e., sedation and gastroenterological concerns), which might result from a low selectivity to molecular targets. We suggest a specific subtype of adrenergic receptors (ARs) as a promising anti-epileptic molecular target. In rats with a predisposition to absence epilepsy, alpha2 ARs agonists provoke sedation and enhance spike-wave activity during transitions from awake/sedation. A number of studies together with our own observations bring evidence that the sedative and proepileptic effects require different alpha2 ARs subtypes activation. Here we introduce a new concept on target pharmacotherapy of absence epilepsy via alpha2B ARs which are presented almost exclusively in the thalamus. We discuss HCN and calcium channels as the most relevant cellular targets of alpha2 ARs involved in spike-wave activity generation.
Collapse
Affiliation(s)
- Evgenia Sitnikova
- Institute of the Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Butlerova Str., 5A, Moscow 117485, Russia
| | - Elizaveta Rutskova
- Institute of the Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Butlerova Str., 5A, Moscow 117485, Russia
| | - Kirill Smirnov
- Institute of the Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Butlerova Str., 5A, Moscow 117485, Russia
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Bld. 1, Moscow 121205, Russia
| |
Collapse
|
21
|
Mäki-Marttunen T, Mäki-Marttunen V. Excitatory and inhibitory effects of HCN channel modulation on excitability of layer V pyramidal cells. PLoS Comput Biol 2022; 18:e1010506. [PMID: 36099307 PMCID: PMC9506642 DOI: 10.1371/journal.pcbi.1010506] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 09/23/2022] [Accepted: 08/19/2022] [Indexed: 11/19/2022] Open
Abstract
Dendrites of cortical pyramidal cells are densely populated by hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, a.k.a. Ih channels. Ih channels are targeted by multiple neuromodulatory pathways, and thus are one of the key ion-channel populations regulating the pyramidal cell activity. Previous observations and theories attribute opposing effects of the Ih channels on neuronal excitability due to their mildly hyperpolarized reversal potential. These effects are difficult to measure experimentally due to the fine spatiotemporal landscape of the Ih activity in the dendrites, but computational models provide an efficient tool for studying this question in a reduced but generalizable setting. In this work, we build upon existing biophysically detailed models of thick-tufted layer V pyramidal cells and model the effects of over- and under-expression of Ih channels as well as their neuromodulation. We show that Ih channels facilitate the action potentials of layer V pyramidal cells in response to proximal dendritic stimulus while they hinder the action potentials in response to distal dendritic stimulus at the apical dendrite. We also show that the inhibitory action of the Ih channels in layer V pyramidal cells is due to the interactions between Ih channels and a hot zone of low voltage-activated Ca2+ channels at the apical dendrite. Our simulations suggest that a combination of Ih-enhancing neuromodulation at the proximal part of the apical dendrite and Ih-inhibiting modulation at the distal part of the apical dendrite can increase the layer V pyramidal excitability more than either of the two alone. Our analyses uncover the effects of Ih-channel neuromodulation of layer V pyramidal cells at a single-cell level and shed light on how these neurons integrate information and enable higher-order functions of the brain.
Collapse
Affiliation(s)
- Tuomo Mäki-Marttunen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Biosciences, University of Oslo, Oslo, Norway
- Simula Research Laboratory, Oslo, Norway
- * E-mail:
| | - Verónica Mäki-Marttunen
- Cognitive Psychology Unit, Faculty of Social Sciences, University of Leiden, Leiden, Netherlands
| |
Collapse
|
22
|
Nordström T, Andersson LC, Åkerman KEO. Role of hyperpolarization-activated cyclic nucleotide-gated channel HCN2 in embryonic neural stem cell proliferation and differentiation. Neurochem Int 2022; 159:105387. [PMID: 35835292 DOI: 10.1016/j.neuint.2022.105387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/06/2022] [Accepted: 07/03/2022] [Indexed: 10/17/2022]
Abstract
Hyperpolarization-activated cyclic nucleotide-gated channels (HCN channels) are involved in spontaneous activity in many electrically active cell types such as cardiomyocytes and neurons. In this study, the role of HCN channels in proliferation and migration of Nestin and Sox2 expressing embryonic neural progenitor cells (NPC) originating from the subventricular zone (SVZ) was examined. Immunostaining and PCR data showed that the HCN2 subtype was highly expressed in these cells. Patch clamp recordings revealed a hyperpolarization-activated current, which was sensitive to inhibitors of HCN channels. Using the fluorescence dye bis-(1,3-dibutylbarbituric acid)-trimethineoxonol (DiBAC(4)(3)) we found that a prompt reduction of the extracellular K+ concentration, or exposing the cells to acute hypoxia, induced an instant hyperpolarization in the whole cell population. Recovery from low K+ induced hyperpolarization after extracellular calcium removal, or by re-oxygenation of hypoxic cells, was sensitive to ZD7288, a HCN channel inhibitor. Treatment of neurosphere cultures from the SVZ with ZD7288 caused a significant and reversible inhibition of neurosphere formation from single cells indicating that proliferation of progenitor cells was reduced. Furthermore, the migration of neuronal cells from neurospheres was considerably retarded in the presence of ZD7288. The results suggest that HCN2 channels are involved in controlling the proliferation of NPC and that HCN2 channel-induced spontaneous electrical activity may trigger the motility response of neurosphere-derived neurons in concert with other ion channels. Furthermore, the response to hypoxia suggests that HCN2 channels may trigger the chemotactic response of NPC to ischemic brain regions seen in many studies.
Collapse
Affiliation(s)
- Tommy Nordström
- Faculty of Medicine, Medicum, Division of Physiology, P.O. Box 63, University of Helsinki, FIN-00014, Helsinki, Finland.
| | - Leif C Andersson
- Department of Pathology, Haartmaninkatu 3 (PB 21), 00014 University of Helsinki, Helsinki, Finland
| | - Karl E O Åkerman
- Faculty of Medicine, Medicum, Division of Physiology, P.O. Box 63, University of Helsinki, FIN-00014, Helsinki, Finland
| |
Collapse
|
23
|
Lemtiri-Chlieh F, Baker DS, Al-Naggar IM, Ramasamy R, Kuchel GA, Levine ES, Robson P, Smith PP. The hyperpolarization-activated, cyclic nucleotide-gated channel resides on myocytes in mouse bladders and contributes to adrenergic-induced detrusor relaxation. Am J Physiol Regul Integr Comp Physiol 2022; 323:R110-R122. [PMID: 35503519 PMCID: PMC9236879 DOI: 10.1152/ajpregu.00277.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Control of urinary continence is predicated on sensory signaling about bladder volume. Bladder sensory nerve activity is dependent on tension, implicating autonomic control over detrusor myocyte activity during bladder filling. Hyperpolarization-activated cyclic nucleotide-gated (HCN) ion channels are known contributors to bladder control, but their mechanism of action is not well understood. The lack of a definitive identification of cell type(s) expressing HCN in the bladder presents a significant knowledge gap. We recently reported a complete transcriptomic atlas of the C57BL/6 mouse bladder showing the dominant HCN paralog in mouse bladder, Hcn1, is limited to a subpopulation of detrusor smooth myocytes (DSMs). Here, we report details of these findings, along with results of patch-clamp experiments, immunohistochemistry, and functional myobath/tension experiments in bladder strips. With the use of a transgenic mouse expressing fluorescence-tagged α-smooth muscle actin, our data confirmed location and function of DSM HCN channels. Despite previous associations of HCN with postulated bladder interstitial cells, neither evidence of specific interstitial cell types nor an association of nonmyocytes with HCN was discovered. We confirm that HCN activation participates in reducing sustained (tonic) detrusor tension via cAMP, with no effect on intermittent (phasic) detrusor activity. In contrast, blockade of HCN increases phasic activity induced by a protein kinase A (PKA) blocker or a large-conductance Ca2+-activated K+ (BK) channel opener. Our findings, therefore, suggest a central role for detrusor myocyte HCN in regulating and constraining detrusor myocyte activity during bladder filling.
Collapse
Affiliation(s)
- Fouad Lemtiri-Chlieh
- 1University of Connecticut Center on Aging, University of Connecticut Health, Farmington, Connecticut,5Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut
| | - Dylan S. Baker
- 1University of Connecticut Center on Aging, University of Connecticut Health, Farmington, Connecticut,4Department of Genetics and Genome Sciences, Institute for Systems Genomics, University of Connecticut School of Medicine, Farmington, Connecticut,7The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut
| | - Iman M. Al-Naggar
- 1University of Connecticut Center on Aging, University of Connecticut Health, Farmington, Connecticut,6Department of Cell Biology, University of Connecticut School of Medicine, Farmington, Connecticut
| | - Ramalakshmi Ramasamy
- 1University of Connecticut Center on Aging, University of Connecticut Health, Farmington, Connecticut,5Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut
| | - George A. Kuchel
- 1University of Connecticut Center on Aging, University of Connecticut Health, Farmington, Connecticut
| | - Eric S. Levine
- 2Connecticut Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, Connecticut,5Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut
| | - Paul Robson
- 4Department of Genetics and Genome Sciences, Institute for Systems Genomics, University of Connecticut School of Medicine, Farmington, Connecticut,7The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut
| | - Phillip P. Smith
- 1University of Connecticut Center on Aging, University of Connecticut Health, Farmington, Connecticut,2Connecticut Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, Connecticut,3Department of Surgery, University of Connecticut School of Medicine, Farmington, Connecticut
| |
Collapse
|
24
|
Liu J, Kasuya G, Zempo B, Nakajo K. Two HCN4 Channels Play Functional Roles in the Zebrafish Heart. Front Physiol 2022; 13:901571. [PMID: 35846012 PMCID: PMC9281569 DOI: 10.3389/fphys.2022.901571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
The HCN4 channel is essential for heart rate regulation in vertebrates by generating pacemaker potentials in the sinoatrial node. HCN4 channel abnormality may cause bradycardia and sick sinus syndrome, making it an important target for clinical research and drug discovery. The zebrafish is a popular animal model for cardiovascular research. They are potentially suitable for studying inherited heart diseases, including cardiac arrhythmia. However, it has not been determined how similar the ion channels that underlie cardiac automaticity are in zebrafish and humans. In the case of HCN4, humans have one gene, whereas zebrafish have two ortholog genes (DrHCN4 and DrHCN4L; ‘Dr’ referring to Danio rerio). However, it is not known whether the two HCN4 channels have different physiological functions and roles in heart rate regulation. In this study, we characterized the biophysical properties of the two zebrafish HCN4 channels in Xenopus oocytes and compared them to those of the human HCN4 channel. We found that they showed different gating properties: DrHCN4L currents showed faster activation kinetics and a more positively shifted G-V curve than did DrHCN4 and human HCN4 currents. We made chimeric channels of DrHCN4 and DrHCN4L and found that cytoplasmic domains were determinants for the faster activation and the positively shifted G-V relationship in DrHCN4L. The use of a dominant-negative HCN4 mutant confirmed that DrHCN4 and DrHCN4L can form a heteromultimeric channel in Xenopus oocytes. Next, we confirmed that both are sensitive to common HCN channel inhibitors/blockers including Cs+, ivabradine, and ZD7288. These HCN inhibitors successfully lowered zebrafish heart rate during early embryonic stages. Finally, we knocked down the HCN4 genes using antisense morpholino and found that knocking down either or both of the HCN4 channels caused a temporal decrease in heart rate and tended to cause pericardial edema. These findings suggest that both DrHCN4 and DrHCN4L play a significant role in zebrafish heart rate regulation.
Collapse
|
25
|
Willis DE, Goldstein PA. Targeting Affective Mood Disorders With Ketamine to Prevent Chronic Postsurgical Pain. FRONTIERS IN PAIN RESEARCH 2022; 3:872696. [PMID: 35832728 PMCID: PMC9271565 DOI: 10.3389/fpain.2022.872696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 06/06/2022] [Indexed: 12/02/2022] Open
Abstract
The phencyclidine-derivative ketamine [2-(2-chlorophenyl)-2-(methylamino)cyclohexan-1-one] was added to the World Health Organization's Model List of Essential Medicines in 1985 and is also on the Model List of Essential Medicines for Children due to its efficacy and safety as an intravenous anesthetic. In sub-anesthetic doses, ketamine is an effective analgesic for the treatment of acute pain (such as may occur in the perioperative setting). Additionally, ketamine may have efficacy in relieving some forms of chronic pain. In 2019, Janssen Pharmaceuticals received regulatory-approval in both the United States and Europe for use of the S-enantiomer of ketamine in adults living with treatment-resistant major depressive disorder. Pre-existing anxiety/depression and the severity of postoperative pain are risk factors for development of chronic postsurgical pain. An important question is whether short-term administration of ketamine can prevent the conversion of acute postsurgical pain to chronic postsurgical pain. Here, we have reviewed ketamine's effects on the biopsychological processes underlying pain perception and affective mood disorders, focusing on non-NMDA receptor-mediated effects, with an emphasis on results from human trials where available.
Collapse
Affiliation(s)
- Dianna E. Willis
- Burke Neurological Institute, White Plains, NY, United States
- Feil Family Brain and Mind Institute, Weill Cornell Medicine, New York, NY, United States
| | - Peter A. Goldstein
- Feil Family Brain and Mind Institute, Weill Cornell Medicine, New York, NY, United States
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States
- Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- *Correspondence: Peter A. Goldstein
| |
Collapse
|
26
|
Toyoda H, Won J, Kim W, Kim H, Davy O, Saito M, Kim D, Tanaka T, Kang Y, Oh SB. The Nature of Noradrenergic Volume Transmission From Locus Coeruleus to Brainstem Mesencephalic Trigeminal Sensory Neurons. Front Cell Neurosci 2022; 16:841239. [PMID: 35558874 PMCID: PMC9087804 DOI: 10.3389/fncel.2022.841239] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
Noradrenergic neurons in the locus coeruleus (LC) release noradrenaline (NA) that acts via volume transmission to activate extrasynaptic G-protein coupled receptors (GPCRs) in target cells throughout the brain. As the closest projection, the dorsal LC laterally adjoins the mesencephalic trigeminal nucleus (MTN), in which proprioceptive primary sensory neurons innervating muscle spindles of jaw-closing muscles are exceptionally located. MTN neurons express α2-adrenergic receptors (α2-ARs) and display hyperpolarization-activated cyclic nucleotide-gated (HCN) currents (Ihs), which is downregulated by α2-AR activation. To quantify the activity-dependent outcome of volume transmission of NA from LC to MTN, we investigated how direct LC activation inhibits Ih in MTN neurons by performing dual whole-cell recordings from LC and MTN neurons. Repetition of 20 Hz spike-train evoked with 1-s current-pulse in LC neurons every 30 s resulted in a gradual decrease in Ih evoked every 30 s, revealing a Hill-type relationship between the number of spike-trains in LC neurons and the degree of Ih inhibition in MTN neurons. On the other hand, when microstimulation was applied in LC every 30 s, an LC neuron repeatedly displayed a transient higher-frequency firing followed by a tonic firing at 5–10 Hz for 30 s. This subsequently caused a similar Hill-type inhibition of Ih in the simultaneously recorded MTN neuron, but with a smaller Hill coefficient, suggesting a lower signal transduction efficacy. In contrast, 20 Hz activity induced by a 1-s pulse applied every 5–10 s caused only a transient facilitation of Ih inhibition followed by a forced termination of Ih inhibition. Thus, the three modes of LC activities modulated the volume transmission to activate α2-adrenergic GPCR to differentially inhibit Ih in MTN neurons.
Collapse
Affiliation(s)
- Hiroki Toyoda
- Department of Neuroscience and Oral Physiology, Graduate School of Dentistry, Osaka University, Suita, Japan
| | - Jonghwa Won
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Department of Neurobiology and Physiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Wheedong Kim
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Hayun Kim
- Interdisciplinary Program for Brain Science, College of Natural Sciences, Seoul National University, Seoul, South Korea
| | - Oscar Davy
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Mitsuru Saito
- Department of Oral Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Doyun Kim
- Department of Neurobiology and Physiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Takuma Tanaka
- Graduate School of Data Science, Shiga University, Hikone, Japan
| | - Youngnam Kang
- Department of Neuroscience and Oral Physiology, Graduate School of Dentistry, Osaka University, Suita, Japan
- Department of Neurobiology and Physiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, South Korea
- Department of Behavioral Sciences, Graduate School of Human Sciences, Osaka University, Suita, Japan
| | - Seog Bae Oh
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Department of Neurobiology and Physiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, South Korea
- Interdisciplinary Program for Brain Science, College of Natural Sciences, Seoul National University, Seoul, South Korea
| |
Collapse
|
27
|
Claveras Cabezudo A, Feriel Khoualdi A, D’Avanzo N. Computational Prediction of Phosphoinositide Binding to Hyperpolarization-Activated Cyclic-Nucleotide Gated Channels. Front Physiol 2022; 13:859087. [PMID: 35399260 PMCID: PMC8990809 DOI: 10.3389/fphys.2022.859087] [Citation(s) in RCA: 3] [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: 01/20/2022] [Accepted: 02/25/2022] [Indexed: 12/31/2022] Open
Abstract
Protein-lipid interactions are key regulators of ion channel function. Numerous ion channels, including hyperpolarization-activated cyclic-nucleotide gated (HCN) channels have been shown to be regulated by phosphoinositides (PIPs), with important implications in cardiac and neuronal function. Specifically, PIPs have been shown to enhance HCN activation. Using computational approaches, we aim to identify potential binding sites for HCN1-PIP interactions. Computational docking and coarse-grained simulations indicate that PIP binding to HCN1 channels is not well coordinated, but rather occurs over a broad surface of charged residues primarily in the HCN-domain, S2 and S3 helices that can be loosely organized in 2 or 3 overlapping clusters. Thus, PIP-HCN1 interactions are more resembling of electrostatic interactions that occur in myristoylated alanine-rich C kinase substrate (MARCKS) proteins, than the specifically coordinated interactions that occur in pleckstrin homology domains (PH domains) or ion channels such as inward rectifier potassium (Kir) channels. Our results also indicate that phosphatidylinositol (PI) interactions with HCN1 are even lower affinity, explaining why unphosphorylated PI have no effect on HCN1 activation unlike phosphorylated PIPs.
Collapse
Affiliation(s)
- Ainara Claveras Cabezudo
- Institute of Pharmacy and Molecular Biotechnology (IPMB), Heidelberg University, Heidelberg, Germany
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC, Canada
| | - Asma Feriel Khoualdi
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC, Canada
| | - Nazzareno D’Avanzo
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
28
|
Roza C, Bernal L. Electrophysiological characterization of ectopic spontaneous discharge in axotomized and intact fibers upon nerve transection: a role in spontaneous pain? Pflugers Arch 2022; 474:387-396. [PMID: 35088129 DOI: 10.1007/s00424-021-02655-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/14/2022]
Abstract
Many patients experience positive symptoms after traumatic nerve injury. Despite the increasing number of experimental studies in models of peripheral neuropathy and the knowledge acquired, most of these patients lack an effective treatment for their chronic pain. One possible explanation might be that most of the preclinical studies focused on the development of mechanical or thermal allodynia/hyperalgesia, neglecting that most of the patients with peripheral neuropathies complain mostly about spontaneous forms of pains. Here, we summarize the aberrant electrophysiological behavior of peripheral nerve fibers recorded in experimental models, the underlying pathophysiological mechanisms, and their relationship with the symptoms reported by patients. Upon nerve section, axotomized but also intact fibers develop ectopic spontaneous activity. Most interestingly, a proportion of axotomized fibers might present receptive fields in the skin far beyond the site of damage, indicative of a functional cross talk between neuromatose and intact fibers. All these features can be linked with some of the symptoms that neuropathic patients experience. Furthermore, we spotlight the consequence of primary afferents with different patterns of spontaneous discharge on the neural code and its relationship with chronic pain states. With this article, readers will be able to understand the pathophysiological mechanisms that might underlie some of the symptoms that experience neuropathic patients, with a special focus on spontaneous pain.
Collapse
Affiliation(s)
- Carolina Roza
- Dpto. Biología de Sistemas, Edificio de Medicina Universidad de Alcalá, 28871, Alcalá de Henares, Madrid, Spain.
| | | |
Collapse
|
29
|
OUP accepted manuscript. Nutr Rev 2022; 80:2002-2016. [DOI: 10.1093/nutrit/nuac019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
30
|
Dixon RE, Navedo MF, Binder MD, Santana LF. Mechanisms and Physiological Implications of Cooperative Gating of Ion Channels Clusters. Physiol Rev 2021; 102:1159-1210. [PMID: 34927454 DOI: 10.1152/physrev.00022.2021] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ion channels play a central role in the regulation of nearly every cellular process. Dating back to the classic 1952 Hodgkin-Huxley model of the generation of the action potential, ion channels have always been thought of as independent agents. A myriad of recent experimental findings exploiting advances in electrophysiology, structural biology, and imaging techniques, however, have posed a serious challenge to this long-held axiom as several classes of ion channels appear to open and close in a coordinated, cooperative manner. Ion channel cooperativity ranges from variable-sized oligomeric cooperative gating in voltage-gated, dihydropyridine-sensitive Cav1.2 and Cav1.3 channels to obligatory dimeric assembly and gating of voltage-gated Nav1.5 channels. Potassium channels, transient receptor potential channels, hyperpolarization cyclic nucleotide-activated channels, ryanodine receptors (RyRs), and inositol trisphosphate receptors (IP3Rs) have also been shown to gate cooperatively. The implications of cooperative gating of these ion channels range from fine tuning excitation-contraction coupling in muscle cells to regulating cardiac function and vascular tone, to modulation of action potential and conduction velocity in neurons and cardiac cells, and to control of pace-making activity in the heart. In this review, we discuss the mechanisms leading to cooperative gating of ion channels, their physiological consequences and how alterations in cooperative gating of ion channels may induce a range of clinically significant pathologies.
Collapse
Affiliation(s)
- Rose Ellen Dixon
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| | - Manuel F Navedo
- Department of Pharmacology, University of California, Davis, CA, United States
| | - Marc D Binder
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States
| | - L Fernando Santana
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| |
Collapse
|
31
|
Lyman KA, Han Y, Zhou C, Renteria I, Besing GL, Kurz JE, Chetkovich DM. Hippocampal cAMP regulates HCN channel function on two time scales with differential effects on animal behavior. Sci Transl Med 2021; 13:eabl4580. [PMID: 34818058 DOI: 10.1126/scitranslmed.abl4580] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Kyle A Lyman
- Department of Neurology, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave., Chicago, IL 60611, USA.,Department of Neurology, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, USA.,Department of Neurology, Stanford University, 453 Quarry Road, Palo Alto, CA 94304, USA
| | - Ye Han
- Department of Neurology, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, USA
| | - Chengwen Zhou
- Department of Neurology, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, USA
| | - Isabelle Renteria
- Department of Neurology, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, USA
| | - Gai-Linn Besing
- Department of Neurology, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, USA
| | - Jonathan E Kurz
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, 225 E. Chicago Ave., Chicago, IL 60611, USA
| | - Dane M Chetkovich
- Department of Neurology, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, USA
| |
Collapse
|
32
|
Cai W, Liu SS, Li BM, Zhang XH. Presynaptic HCN channels constrain GABAergic synaptic transmission in pyramidal cells of the medial prefrontal cortex. Biol Open 2021; 11:272636. [PMID: 34709375 PMCID: PMC8966777 DOI: 10.1242/bio.058840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/26/2021] [Indexed: 11/20/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are widely expressed in neurons in the central nervous system. It has been documented that HCN channels regulate the intrinsic excitability of pyramidal cells in the medial prefrontal cortex (mPFC) of rodents. Here, we report that HCN channels limited GABAergic transmission onto pyramidal cells in rat mPFC. The pharmacological blockade of HCN channels resulted in a significant increase in the frequency of both spontaneous and miniature inhibitory postsynaptic currents (IPSCs) in mPFC pyramidal cells, whereas potentiation of HCN channels reversely decreases the frequency of mIPSCs. Furthermore, such facilitation effect on mIPSC frequency required presynaptic Ca2+ influx. Immunofluorescence staining showed that HCN channels expressed in presynaptic GABAergic terminals, as well as in both soma and neurite of parvalbumin-expressing (PV-expressing) basket cells in mPFC. The present results indicate that HCN channels in GABAergic interneurons, most likely PV-expressing basket cells, constrain inhibitory control over layer 5-6 pyramidal cells by restricting presynaptic Ca2+ entry.
Collapse
Affiliation(s)
- Wei Cai
- Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Shu-Su Liu
- Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | - Bao-Ming Li
- Center for Neuropsychiatric Diseases, Institute of Life Science, Nanchang University, Nanchang 330031, China
| | - Xue-Han Zhang
- Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| |
Collapse
|
33
|
Biophysical analysis of an HCN1 epilepsy variant suggests a critical role for S5 helix Met-305 in voltage sensor to pore domain coupling. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 166:156-172. [PMID: 34298002 DOI: 10.1016/j.pbiomolbio.2021.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 06/30/2021] [Accepted: 07/13/2021] [Indexed: 12/22/2022]
Abstract
Hyperpolarization-gated, cyclic nucleotide-activated (HCN1-4) channels are inwardly rectifying cation channels that display voltage dependent activation and de-activation. Pathogenic variants in HCN1 are associated with severe developmental and epileptic encephalopathies including the de novo HCN1 M305L variant. M305 is located in the S5 domain that is implicated in coupling voltage sensor domain movement to pore opening. This variant lacks voltage-dependent activation and de-activation and displays normal cation selectivity. To elucidate the impact of the mutation on the channel structure-function relations, molecular dynamics simulations of the wild type and mutant homotetramers were compared and identified a sulphur-aromatic interaction between M305 and F389 that contributes to the coupling of the voltage-sensing domain to the pore domain. To mimic the heterozygous condition as a heterotetrameric channel assembly, Xenopus oocytes were co-injected with various ratios of wild-type and mutant subunit cRNAs and the biophysical properties of channels with different subunit stoichiometries were determined. The results showed that a single mutated subunit was sufficient to significantly disrupt the voltage dependence of activation. The functional data were qualitatively consistent with predictions of a model that assumes independent activation of the voltage sensing domains allosterically controlling the closed to open transition of the pore. Overall, the M305L mutation results in an HCN1 channel that lacks voltage dependence and facilitates excitatory cation flow at membrane potentials that would normally close the channel. Our findings provide molecular insights into HCN1 channels and reveal the structural and biophysical basis of the severe epilepsy phenotype associated with the M305L mutation.
Collapse
|
34
|
Saponaro A, Bauer D, Giese MH, Swuec P, Porro A, Gasparri F, Sharifzadeh AS, Chaves-Sanjuan A, Alberio L, Parisi G, Cerutti G, Clarke OB, Hamacher K, Colecraft HM, Mancia F, Hendrickson WA, Siegelbaum SA, DiFrancesco D, Bolognesi M, Thiel G, Santoro B, Moroni A. Gating movements and ion permeation in HCN4 pacemaker channels. Mol Cell 2021; 81:2929-2943.e6. [PMID: 34166608 PMCID: PMC8294335 DOI: 10.1016/j.molcel.2021.05.033] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/12/2021] [Accepted: 05/27/2021] [Indexed: 10/31/2022]
Abstract
The HCN1-4 channel family is responsible for the hyperpolarization-activated cation current If/Ih that controls automaticity in cardiac and neuronal pacemaker cells. We present cryoelectron microscopy (cryo-EM) structures of HCN4 in the presence or absence of bound cAMP, displaying the pore domain in closed and open conformations. Analysis of cAMP-bound and -unbound structures sheds light on how ligand-induced transitions in the channel cytosolic portion mediate the effect of cAMP on channel gating and highlights the regulatory role of a Mg2+ coordination site formed between the C-linker and the S4-S5 linker. Comparison of open/closed pore states shows that the cytosolic gate opens through concerted movements of the S5 and S6 transmembrane helices. Furthermore, in combination with molecular dynamics analyses, the open pore structures provide insights into the mechanisms of K+/Na+ permeation. Our results contribute mechanistic understanding on HCN channel gating, cyclic nucleotide-dependent modulation, and ion permeation.
Collapse
Affiliation(s)
- Andrea Saponaro
- Department of Biosciences, University of Milan, Milan, Italy
| | - Daniel Bauer
- Department of Biology, TU-Darmstadt, Darmstadt, Germany
| | - M Hunter Giese
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Paolo Swuec
- Department of Biosciences, University of Milan, Milan, Italy; Pediatric Research Center "Romeo ed Enrica Invernizzi," University of Milan, Milan, Italy
| | | | | | | | - Antonio Chaves-Sanjuan
- Department of Biosciences, University of Milan, Milan, Italy; Pediatric Research Center "Romeo ed Enrica Invernizzi," University of Milan, Milan, Italy
| | - Laura Alberio
- Department of Biosciences, University of Milan, Milan, Italy; Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Giacomo Parisi
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Gabriele Cerutti
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - Oliver B Clarke
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA; Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Kay Hamacher
- Department of Biology, TU-Darmstadt, Darmstadt, Germany
| | - Henry M Colecraft
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Filippo Mancia
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Wayne A Hendrickson
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Steven A Siegelbaum
- Department of Neuroscience, Zuckerman Institute, Columbia University, New York, NY, USA
| | - Dario DiFrancesco
- Department of Biosciences, University of Milan, Milan, Italy; Institute of Biophysics-Milano, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Martino Bolognesi
- Department of Biosciences, University of Milan, Milan, Italy; Pediatric Research Center "Romeo ed Enrica Invernizzi," University of Milan, Milan, Italy
| | - Gerhard Thiel
- Department of Biology, TU-Darmstadt, Darmstadt, Germany
| | - Bina Santoro
- Department of Neuroscience, Zuckerman Institute, Columbia University, New York, NY, USA.
| | - Anna Moroni
- Department of Biosciences, University of Milan, Milan, Italy; Institute of Biophysics-Milano, Consiglio Nazionale delle Ricerche, Rome, Italy.
| |
Collapse
|
35
|
Regulation of sinoatrial funny channels by cyclic nucleotides: From adrenaline and I K2 to direct binding of ligands to protein subunits. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 166:12-21. [PMID: 34237319 DOI: 10.1016/j.pbiomolbio.2021.06.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 06/13/2021] [Accepted: 06/25/2021] [Indexed: 12/27/2022]
Abstract
The funny current, and the HCN channels that form it, are affected by the direct binding of cyclic nucleotides. Binding of these second messengers causes a depolarizing shift of the activation curve, which leads to greater availability of current at physiological membrane voltages. This review outlines a brief history on this regulation and provides some evidence that other cyclic nucleotides, especially cGMP, may be important for the regulation of the funny channel in the heart. Current understanding of the molecular mechanism of cyclic nucleotide regulation is also presented, which includes the notions that full and partial agonism occur as a consequence of negatively cooperative binding. Knowledge gaps, including a potential role of cyclic nucleotide-regulation of the funny current under pathophysiological conditions, are included. The work highlighted here is in dedication to Dario DiFrancesco on his retirement.
Collapse
|
36
|
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: 36] [Impact Index Per Article: 9.0] [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
|
37
|
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: 3] [Impact Index Per Article: 0.8] [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
|
38
|
Tanwar M, Kateriya S, Nair D, Jose M. Optogenetic modulation of real-time nanoscale dynamics of HCN channels using photoactivated adenylyl cyclases. RSC Chem Biol 2021; 2:863-875. [PMID: 34458814 PMCID: PMC8341789 DOI: 10.1039/d0cb00124d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 02/12/2021] [Indexed: 12/12/2022] Open
Abstract
Adenosine 3',5'-cyclic monophosphate (cAMP) is a key second messenger that activates several signal transduction pathways in eukaryotic cells. Alteration of basal levels of cAMP is known to activate protein kinases, regulate phosphodiesterases and modulate the activity of ion channels such as Hyper polarization-activated cyclic nucleotide gated channels (HCN). Recent advances in optogenetics have resulted in the availability of novel genetically encoded molecules with the capability to alter cytoplasmic profiles of cAMP with unprecedented spatial and temporal precision. Using single molecule based super-resolution microscopy and different optogenetic modulators of cellular cAMP in both live and fixed cells, we illustrate a novel paradigm to report alteration in nanoscale confinement of ectopically expressed HCN channels. We characterized the efficacy of cAMP generation using ensemble photoactivation of different optogenetic modulators. Then we demonstrate that local modulation of cAMP alters the exchange of membrane bound HCN channels with its nanoenvironment. Additionally, using high density single particle tracking in combination with both acute and chronic optogenetic elevation of cAMP in the cytoplasm, we show that HCN channels are confined to sub 100 nm sized functional domains on the plasma membrane. The nanoscale properties of these domains along with the exchange kinetics of HCN channels in and out of these molecular zones are altered upon temporal changes in the cytoplasmic cAMP. Using HCN2 point mutants and a truncated construct of HCN2 with altered sensitivity to cAMP, we confirmed these alterations in lateral organization of HCN2 to be specific to cAMP binding. Thus, combining these advanced non-invasive paradigms, we report a cAMP dependent ensemble and single particle behavior of HCN channels mediated by its cyclic nucleotide binding domain, opening innovative ways to dissect biochemical pathways at the nanoscale and real-time in living cells.
Collapse
Affiliation(s)
- Meenakshi Tanwar
- Centre for Neuroscience, Indian Institute of Science Bangalore-560012 India
| | - Suneel Kateriya
- Laboratory of Optobiology, School of Biotechnology, Jawaharlal Nehru University New Delhi-110067 India
| | - Deepak Nair
- Centre for Neuroscience, Indian Institute of Science Bangalore-560012 India
| | - Mini Jose
- Centre for Neuroscience, Indian Institute of Science Bangalore-560012 India
| |
Collapse
|
39
|
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
|
40
|
Pfleger C, Kusch J, Kondapuram M, Schwabe T, Sattler C, Benndorf K, Gohlke H. Allosteric signaling in C-linker and cyclic nucleotide-binding domain of HCN2 channels. Biophys J 2021; 120:950-963. [PMID: 33515603 DOI: 10.1016/j.bpj.2021.01.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 01/04/2021] [Accepted: 01/19/2021] [Indexed: 12/22/2022] Open
Abstract
Opening of hyperpolarization-activated cyclic nucleotide-modulated (HCN) channels is controlled by membrane hyperpolarization and binding of cyclic nucleotides to the tetrameric cyclic nucleotide-binding domain (CNBD), attached to the C-linker (CL) disk. Confocal patch-clamp fluorometry revealed pronounced cooperativity of ligand binding among protomers. However, by which pathways allosteric signal transmission occurs remained elusive. Here, we investigate how changes in the structural dynamics of the CL-CNBD of mouse HCN2 upon cAMP binding relate to inter- and intrasubunit signal transmission. Applying a rigidity-theory-based approach, we identify two intersubunit and one intrasubunit pathways that differ in allosteric coupling strength between cAMP-binding sites or toward the CL. These predictions agree with results from electrophysiological and patch-clamp fluorometry experiments. Our results map out distinct routes within the CL-CNBD that modulate different cAMP-binding responses in HCN2 channels. They signify that functionally relevant submodules may exist within and across structurally discernable subunits in HCN channels.
Collapse
Affiliation(s)
- Christopher Pfleger
- Mathematisch-Naturwissenschaftliche Fakultät, Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Jana Kusch
- Institute of Physiology II, Jena University Hospital, Jena, Germany
| | | | - Tina Schwabe
- Institute of Physiology II, Jena University Hospital, Jena, Germany
| | | | - Klaus Benndorf
- Institute of Physiology II, Jena University Hospital, Jena, Germany
| | - Holger Gohlke
- Mathematisch-Naturwissenschaftliche Fakultät, Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany; John von Neumann Institute for Computing, Jülich Supercomputing Centre, and Institute of Biological Information Processing (IBI-7, Structural Biochemistry), Forschungszentrum Jülich, Jülich, Germany.
| |
Collapse
|
41
|
Rich S, Moradi Chameh H, Sekulic V, Valiante TA, Skinner FK. Modeling Reveals Human-Rodent Differences in H-Current Kinetics Influencing Resonance in Cortical Layer 5 Neurons. Cereb Cortex 2021; 31:845-872. [PMID: 33068000 PMCID: PMC7906797 DOI: 10.1093/cercor/bhaa261] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/18/2020] [Accepted: 08/18/2020] [Indexed: 01/01/2023] Open
Abstract
While our understanding of human neurons is often inferred from rodent data, inter-species differences between neurons can be captured by building cellular models specifically from human data. This includes understanding differences at the level of ion channels and their implications for human brain function. Thus, we here present a full spiking, biophysically detailed multi-compartment model of a human layer 5 (L5) cortical pyramidal cell. Model development was primarily based on morphological and electrophysiological data from the same human L5 neuron, avoiding confounds of experimental variability. Focus was placed on describing the behavior of the hyperpolarization-activated cation (h-) channel, given increasing interest in this channel due to its role in pacemaking and differentiating cell types. We ensured that the model exhibited post-inhibitory rebound spiking considering its relationship with the h-current, along with other general spiking characteristics. The model was validated against data not used in its development, which highlighted distinctly slower kinetics of the human h-current relative to the rodent setting. We linked the lack of subthreshold resonance observed in human L5 neurons to these human-specific h-current kinetics. This work shows that it is possible and necessary to build human-specific biophysical neuron models in order to understand human brain dynamics.
Collapse
Affiliation(s)
- Scott Rich
- Division of Clinical and Computational Neuroscience, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Homeira Moradi Chameh
- Division of Clinical and Computational Neuroscience, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Vladislav Sekulic
- Division of Clinical and Computational Neuroscience, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
| | - Taufik A Valiante
- Division of Clinical and Computational Neuroscience, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 1A1, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A1, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON M5S 1A1, Canada
- Electrical and Computer Engineering, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Frances K Skinner
- Division of Clinical and Computational Neuroscience, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada
- Departments of Medicine (Neurology) and Physiology, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
42
|
Cook DC, Goldstein PA. Non-canonical Molecular Targets for Novel Analgesics: Intracellular Calcium and HCN Channels. Curr Neuropharmacol 2021; 19:1937-1951. [PMID: 33463473 PMCID: PMC9185781 DOI: 10.2174/1570159x19666210119153047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/04/2021] [Accepted: 01/17/2021] [Indexed: 11/22/2022] Open
Abstract
Pain is a prevalent biopsychosocial condition that poses a significant challenge to healthcare providers, contributes substantially to a disability, and is a major economic burden worldwide. An overreliance on opioid analgesics, which primarily target the μ-opioid receptor, has caused devastating morbidity and mortality in the form of misuse and overdose-related death. Thus, novel analgesic medications are needed that can effectively treat pain and provide an alternative to opioids. A variety of cellular ion channels contribute to nociception, the response of the sensory nervous system to a noxious stimulus that commonly leads to pain. Ion channels involved in nociception may provide a suitable target for pharmacologic modulation to achieve pain relief. This narrative review summarizes the evidence for two ion channels that merit consideration as targets for non-opioid pain medications: ryanodine receptors (RyRs), which are intracellular calcium channels, and hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, which belong to the superfamily of voltage-gated K+ channels. The role of these channels in nociception and neuropathic pain is discussed and suitability as targets for novel analgesics and antihyperalgesics is considered.
Collapse
Affiliation(s)
- Daniel C. Cook
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Peter A. Goldstein
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY 10065, USA
- Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
43
|
Liang Y, Xu Z, Wu X, Pang J, Zhou P, Cao Y. Inhibition of hyperpolarization-activated cyclic nucleotide-gated channels with natural flavonoid quercetin. Biochem Biophys Res Commun 2020; 533:952-957. [PMID: 33008592 DOI: 10.1016/j.bbrc.2020.09.102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 09/22/2020] [Indexed: 11/18/2022]
Abstract
Quercetin is a natural flavonoid which has been reported to be analgesic in different animal models of pain. However, the mechanism underlying the pain-relieving effects is still unclear. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels play critical roles in controlling pacemaker activity in cardiac and nervous systems, making the channel a new target for therapeutic exploration. In this study, we explored a series of flavonoids for their modulation on HCN channels. Among all tested flavonoids, quercetin was the most potent inhibitor for HCN channels with an IC50 value of 27.32 ± 1.19 μM for HCN2. Furthermore, quercetin prominently left shifted the voltage-dependent activation curves of HCN channels and decelerated deactivation process. The results presented herein firstly characterize quercetin as a novel and potent inhibitor for HCN channels, which represents a novel structure for future drug design of HCN channel inhibitors.
Collapse
Affiliation(s)
- Yemei Liang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Ziwei Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaoyan Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jianxin Pang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Pingzheng Zhou
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| | - Ying Cao
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
44
|
Han Y, Lyman KA, Foote KM, Chetkovich DM. The structure and function of TRIP8b, an auxiliary subunit of hyperpolarization-activated cyclic-nucleotide gated channels. Channels (Austin) 2020; 14:110-122. [PMID: 32189562 PMCID: PMC7153792 DOI: 10.1080/19336950.2020.1740501] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/18/2020] [Accepted: 02/21/2020] [Indexed: 02/08/2023] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are expressed throughout the mammalian central nervous system (CNS). These channels have been implicated in a wide range of diseases, including Major Depressive Disorder and multiple subtypes of epilepsy. The diversity of functions that HCN channels perform is in part attributable to differences in their subcellular localization. To facilitate a broad range of subcellular distributions, HCN channels are bound by auxiliary subunits that regulate surface trafficking and channel function. One of the best studied auxiliary subunits is tetratricopeptide-repeat containing, Rab8b-interacting protein (TRIP8b). TRIP8b is an extensively alternatively spliced protein whose only known function is to regulate HCN channels. TRIP8b binds to HCN pore-forming subunits at multiple interaction sites that differentially regulate HCN channel function and subcellular distribution. In this review, we summarize what is currently known about the structure and function of TRIP8b isoforms with an emphasis on the role of this auxiliary subunit in health and disease.
Collapse
Affiliation(s)
- Ye Han
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kyle A. Lyman
- Department of Neurology, Stanford University, Palo Alto, CA, USA
| | - Kendall M. Foote
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Dane M. Chetkovich
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
45
|
Fang Y, Ren R, Shi H, Huang L, Lenahan C, Lu Q, Tang L, Huang Y, Tang J, Zhang J, Zhang JH. Pituitary Adenylate Cyclase-Activating Polypeptide: A Promising Neuroprotective Peptide in Stroke. Aging Dis 2020; 11:1496-1512. [PMID: 33269103 PMCID: PMC7673855 DOI: 10.14336/ad.2020.0626] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 06/26/2020] [Indexed: 12/11/2022] Open
Abstract
The search for viable, effective treatments for acute stroke continues to be a global priority due to the high mortality and morbidity. Current therapeutic treatments have limited effects, making the search for new treatments imperative. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a well-established cytoprotective neuropeptide that participates in diverse neural physiological and pathological activities, such as neuronal proliferation, differentiation, and migration, as well as neuroprotection. It is considered a promising treatment in numerous neurological diseases. Thus, PACAP bears potential as a new therapeutic strategy for stroke treatment. Herein, we provide an overview pertaining to the current knowledge of PACAP, its receptors, and its potential neuroprotective role in the setting of stroke, as well as various mechanisms of neuroprotection involving ionic homeostasis, excitotoxicity, cell edema, oxidative stress, inflammation, and cell death, as well as the route of PACAP administration.
Collapse
Affiliation(s)
- Yuanjian Fang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Reng Ren
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hui Shi
- 2Department of Neurosurgery, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Lei Huang
- 3Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA.,4Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Cameron Lenahan
- 3Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA.,4Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA.,5Burrell College of Osteopathic Medicine, Las Cruces, NM, USA
| | - Qin Lu
- 6Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China
| | - Lihui Tang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yi Huang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiping Tang
- 3Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA.,4Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA.,7Department of Anesthesiology, Loma Linda University, Loma Linda, CA, USA
| | - Jianmin Zhang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - John H Zhang
- 3Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA.,4Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA.,7Department of Anesthesiology, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
46
|
Santoro B, Shah MM. Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels as Drug Targets for Neurological Disorders. Annu Rev Pharmacol Toxicol 2020; 60:109-131. [PMID: 31914897 DOI: 10.1146/annurev-pharmtox-010919-023356] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are voltage-gated ion channels that critically modulate neuronal activity. Four HCN subunits (HCN1-4) have been cloned, each having a unique expression profile and distinctive effects on neuronal excitability within the brain. Consistent with this, the expression and function of these subunits are altered in diverse ways in neurological disorders. Here, we review current knowledge on the structure and distribution of the individual HCN channel isoforms, their effects on neuronal activity under physiological conditions, and how their expression and function are altered in neurological disorders, particularly epilepsy, neuropathic pain, and affective disorders. We discuss the suitability of HCN channels as therapeutic targets and how drugs might be strategically designed to specifically act on particular isoforms. We conclude that medicines that target individual HCN isoforms and/or their auxiliary subunit, TRIP8b, may provide valuable means of treating distinct neurological conditions.
Collapse
Affiliation(s)
- Bina Santoro
- Department of Neuroscience, Columbia University, New York, NY 10027, USA
| | - Mala M Shah
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, United Kingdom;
| |
Collapse
|
47
|
cAMP-dependent regulation of HCN4 controls the tonic entrainment process in sinoatrial node pacemaker cells. Nat Commun 2020; 11:5555. [PMID: 33144559 PMCID: PMC7641277 DOI: 10.1038/s41467-020-19304-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 10/08/2020] [Indexed: 11/13/2022] Open
Abstract
It is highly debated how cyclic adenosine monophosphate-dependent regulation (CDR) of the major pacemaker channel HCN4 in the sinoatrial node (SAN) is involved in heart rate regulation by the autonomic nervous system. We addressed this question using a knockin mouse line expressing cyclic adenosine monophosphate-insensitive HCN4 channels. This mouse line displayed a complex cardiac phenotype characterized by sinus dysrhythmia, severe sinus bradycardia, sinus pauses and chronotropic incompetence. Furthermore, the absence of CDR leads to inappropriately enhanced heart rate responses of the SAN to vagal nerve activity in vivo. The mechanism underlying these symptoms can be explained by the presence of nonfiring pacemaker cells. We provide evidence that a tonic and mutual interaction process (tonic entrainment) between firing and nonfiring cells slows down the overall rhythm of the SAN. Most importantly, we show that the proportion of firing cells can be increased by CDR of HCN4 to efficiently oppose enhanced responses to vagal activity. In conclusion, we provide evidence for a novel role of CDR of HCN4 for the central pacemaker process in the sinoatrial node. The involvement of cAMP-dependent regulation of HCN4 in the chronotropic heart rate response is a matter of debate. Here the authors use a knockin mouse model expressing cAMP-insensitive HCN4 channels to discover an inhibitory nonfiring cell pool in the sinoatrial node and a tonic and mutual interaction between firing and nonfiring pacemaker cells that is controlled by cAMP-dependent regulation of HCN4, with implications in chronotropic heart rate responses.
Collapse
|
48
|
Sherif MA, Neymotin SA, Lytton WW. In silico hippocampal modeling for multi-target pharmacotherapy in schizophrenia. NPJ SCHIZOPHRENIA 2020; 6:25. [PMID: 32958782 PMCID: PMC7506542 DOI: 10.1038/s41537-020-00109-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 06/23/2020] [Indexed: 02/08/2023]
Abstract
Treatment of schizophrenia has had limited success in treating core cognitive symptoms. The evidence of multi-gene involvement suggests that multi-target therapy may be needed. Meanwhile, the complexity of schizophrenia pathophysiology and psychopathology, coupled with the species-specificity of much of the symptomatology, places limits on analysis via animal models, in vitro assays, and patient assessment. Multiscale computer modeling complements these traditional modes of study. Using a hippocampal CA3 computer model with 1200 neurons, we examined the effects of alterations in NMDAR, HCN (Ih current), and GABAAR on information flow (measured with normalized transfer entropy), and in gamma activity in local field potential (LFP). We found that altering NMDARs, GABAAR, Ih, individually or in combination, modified information flow in an inverted-U shape manner, with information flow reduced at low and high levels of these parameters. Theta-gamma phase-amplitude coupling also had an inverted-U shape relationship with NMDAR augmentation. The strong information flow was associated with an intermediate level of synchrony, seen as an intermediate level of gamma activity in the LFP, and an intermediate level of pyramidal cell excitability. Our results are consistent with the idea that overly low or high gamma power is associated with pathological information flow and information processing. These data suggest the need for careful titration of schizophrenia pharmacotherapy to avoid extremes that alter information flow in different ways. These results also identify gamma power as a potential biomarker for monitoring pathology and multi-target pharmacotherapy.
Collapse
Affiliation(s)
- Mohamed A Sherif
- Department of Psychiatry, VA Connecticut Healthcare System, 950 Campbell Avenue, West Haven, CT, USA.
- Department of Psychiatry, Yale University, New Haven, CT, USA.
- Biomedical Engineering Graduate Program, SUNY Downstate Medical Center/NYU Tandon School of Engineering, Brooklyn, NY, USA.
| | - Samuel A Neymotin
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - William W Lytton
- Biomedical Engineering Graduate Program, SUNY Downstate Medical Center/NYU Tandon School of Engineering, Brooklyn, NY, USA
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY, USA
- Department of Neurology, SUNY Downstate Medical Center, Brooklyn, NY, USA
- Department of Neurology, Kings County Hospital Center, Brooklyn, NY, USA
| |
Collapse
|
49
|
Porro A, Binda A, Pisoni M, Donadoni C, Rivolta I, Saponaro A. Rational design of a mutation to investigate the role of the brain protein TRIP8b in limiting the cAMP response of HCN channels in neurons. J Gen Physiol 2020; 152:e202012596. [PMID: 32633755 PMCID: PMC7478871 DOI: 10.1085/jgp.202012596] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/02/2020] [Accepted: 06/08/2020] [Indexed: 01/22/2023] Open
Abstract
TRIP8b (tetratricopeptide repeat-containing Rab8b-interacting protein) is the neuronal regulatory subunit of HCN channels, a family of voltage-dependent cation channels also modulated by direct cAMP binding. TRIP8b interacts with the C-terminal region of HCN channels and controls both channel trafficking and gating. The association of HCN channels with TRIP8b is required for the correct expression and subcellular targeting of the channel protein in vivo. TRIP8b controls HCN gating by interacting with the cyclic nucleotide-binding domain (CNBD) and competing for cAMP binding. Detailed structural knowledge of the complex between TRIP8b and CNBD was used as a starting point to engineer a mutant channel, whose gating is controlled by cAMP, but not by TRIP8b, while leaving TRIP8b-dependent regulation of channel trafficking unaltered. We found two-point mutations (N/A and C/D) in the loop connecting the CNBD to the C-linker (N-bundle loop) that, when combined, strongly reduce the binding of TRIP8b to CNBD, leaving cAMP affinity unaltered both in isolated CNBD and in the full-length protein. Proof-of-principle experiments performed in cultured cortical neurons confirm that the mutant channel provides a genetic tool for dissecting the two effects of TRIP8b (gating versus trafficking). This will allow the study of the functional role of the TRIP8b antagonism of cAMP binding, a thus far poorly investigated aspect of HCN physiology in neurons.
Collapse
Affiliation(s)
| | - Anna Binda
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | | - Chiara Donadoni
- Department of Biosciences, University of Milano, Milano, Italy
| | - Ilaria Rivolta
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Andrea Saponaro
- Department of Biosciences, University of Milano, Milano, Italy
| |
Collapse
|
50
|
HCN2 activation modulation: An electrophysiological and molecular study of the well-preserved LCI sequence in the pore channel. Arch Biochem Biophys 2020; 689:108436. [DOI: 10.1016/j.abb.2020.108436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/21/2020] [Accepted: 05/25/2020] [Indexed: 11/17/2022]
|