1
|
Dalla Porta L, Barbero-Castillo A, Sanchez-Sanchez JM, Cancino N, Sanchez-Vives MV. H-current modulation of cortical Up and Down states. J Physiol 2025; 603:2409-2424. [PMID: 40153850 DOI: 10.1113/jp287616] [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: 09/05/2024] [Accepted: 02/11/2025] [Indexed: 04/01/2025] Open
Abstract
Understanding the link between cellular processes and brain function remains a key challenge in neuroscience. One crucial aspect is the interplay between specific ion channels and network dynamics. This work reveals a role for h-current, a hyperpolarization-activated cationic current, in shaping cortical slow oscillations. Cortical slow oscillations are generated not only during slow wave sleep and deep anaesthesia, but also in association with disorders of consciousness and brain lesions. Cortical slow oscillations exhibit rhythmic periods of activity (Up states) alternating with silent periods (Down states). By progressively reducing h-current in both cortical slices and in a computational model, we observed Up states transformed into prolonged plateaus of sustained firing, while Down states were also significantly extended. This transformation led to a fivefold reduction in oscillation frequency. In a biophysical recurrent network model, we identified the cellular mechanisms underlying this transformation of network dynamics: an increased neuronal input resistance and membrane time constant, increasing neuronal responsiveness to even weak inputs. A partial block of h-current therefore resulted in a change in brain state. HCN (hyperpolarization-activated cyclic nucleotide-gated) channels, which generate h-current, are known targets for neuromodulation, suggesting potential pathways for dynamic control of brain rhythms. KEY POINTS: We investigated the role of h-current in shaping emergent cortical slow oscillation dynamics, specifically Up and Down states, in cortical slices. Blocking h-current transformed Up states into prolonged plateaus of sustained firing, lasting up to 4 s. Down states were also significantly elongated and the oscillatory frequency decreased. A biophysical model of the cortical network replicated these findings and allowed us to explore the underlying mechanisms. An increase in cellular input resistance and time constant led to a rise in network excitability, synaptic responsiveness and firing rates. Our results highlight the significant role of h-current in controlling cortical slow rhythmic patterns, making it a relevant target for neuromodulators regulating brain states.
Collapse
Affiliation(s)
- Leonardo Dalla Porta
- Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Roselló, Barcelona, Spain
| | | | | | - Nathalia Cancino
- Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Roselló, Barcelona, Spain
| | - Maria V Sanchez-Vives
- Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Roselló, Barcelona, Spain
- ICREA, Passeig Lluís Companys 23, Barcelona, Spain
| |
Collapse
|
2
|
María-Ríos CE, Murphy GG, Morrow JD. Individual Variation in Intrinsic Neuronal Properties of Nucleus Accumbens Core and Shell Medium Spiny Neurons in Animals Prone to Sign- or Goal-Track. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.644332. [PMID: 40236090 PMCID: PMC11996421 DOI: 10.1101/2025.03.24.644332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
The "sign-tracking" and "goal-tracking" model of individual variation in associative learning permits the identification of rats with different cue-reactivity and predisposition to addiction-like behaviors. Certainly, compared to "goal-trackers" (GTs), "sign-trackers" (STs) show more susceptibility traits such as increased cue-induced 'relapse' of drugs of abuse. Different cue- and reward-evoked patterns of activity in the nucleus accumbens (NAc) have been a hallmark of the ST/GT phenotype. However, it is unknown whether differences in the intrinsic neuronal properties of NAc medium spiny neurons (MSNs) in the core and shell subregions are also a physiological correlate of these phenotypes. We performed whole-cell slice electrophysiology in outbred male rats and found that STs exhibited the lowest excitability in the NAc core, with lower number of action potentials and firing frequency as well as a blunted voltage/current relationship curve in response to hyperpolarized potentials in both the NAc core and shell. Although firing properties of shell MSNs did not differ between STs and GTs, intermediate responders that engage in both behaviors showed greater excitability compared to both STs and GTs. These findings suggest that intrinsic excitability in the NAc may contribute to individual differences in the attribution of incentive salience. Significance Statement During associative learning, cues acquire predictive value, but in some instances, they also acquire incentive salience, meaning they take on some of the motivational properties of the reward. The propensity to attribute cues with incentive salience varies between individuals, and excessive attribution can lead to maladaptive behaviors. The "sign-and goal-tracking" model allows us to isolate these two properties and disambiguate the neurobiological processes that govern them. To our knowledge this is the first study characterizing passive and active membrane properties of MSNs in the NAc core and shell of STs and GTs, as well as IRs. These findings are meant to better inform investigations of the distinct role of the NAc in reward learning, particularly in the attribution of incentive salience and addiction predisposition.
Collapse
|
3
|
Idikuda V, Roy Chowdhury S, Chang Y, Ren Q, Bao H, Goldsmith R, Chanda B. Lipid bilayers determine the allostery but not intrinsic affinity of cAMP binding to pacemaker channels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.23.630133. [PMID: 39763952 PMCID: PMC11703277 DOI: 10.1101/2024.12.23.630133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2025]
Abstract
Cyclic adenosine monophosphate (cAMP), a second messenger, binds to hyperpolarization and cyclic nucleotide-gated (HCN) ion channels and regulates the automaticity of pacemaking activity. While cellular studies suggest that cAMP binding to HCN channels exhibits unusual cooperativity, recent findings using purified detergent-solubilized channels indicate independent binding to each subunit. This discrepancy raises the question of whether the lipid environment or endogenous cellular cofactors influence cAMP-dependent gating. To address this, we reconstituted purified human HCN channels in nanodiscs and resolved cAMP binding energetics at single-molecule resolution using nanophotonic waveguides. Our measurements reveal that, in contrast to detergent-solubilized channels, cAMP binds cooperatively to HCN channels reconstituted in a variety of lipid nanodiscs. Remarkably, the presence of lipid bilayer promotes ligand-binding allostery but not intrinsic binding affinity. To explore the molecular basis of bilayer-induced allostery, we determine the cryo-EM structure of HCN1 in soy polar lipid nanodiscs at a nominal resolution of 3.77 angstrom resolution. Although the overall architecture is conserved, the average interfacial distance between the transmembrane domain and C-terminal domain of neighboring subunits are shorter in lipid nanodiscs. These findings indicate that the lipid bilayer regulates the function of pacemaker ion channels by enhancing inter-subunit interactions and underscore the fundamental role of membranes in amplifying the gating sensitivity of ion channels by promoting long-range cooperative interactions.
Collapse
|
4
|
Wu X, Kobeissi AM, Phillips HL, Dai H, Yao WD. A Prefrontal Cortex-Nucleus Accumbens Circuit Attenuates Cocaine-conditioned Place Preference Memories. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.21.644656. [PMID: 40196555 PMCID: PMC11974754 DOI: 10.1101/2025.03.21.644656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
The infralimbic (IL) subregion of the prefrontal cortex (PFC), via its descending projection to the nucleus accumbens (NAc), inhibits cue-induced drug seeking and reinstatement, but the underlying mechanisms are not fully understood. Here we show that the intrinsic membrane excitability of IL layer 5 pyramidal neurons projecting to the NAc shell (IL-NAcSh neurons) suppresses cocaine-associated memories. Following repeated cocaine exposures in a conditioned place preference paradigm, IL-NAcSh neurons anatomically traced by fluorescent retrobeads undergo prolonged decrease of membrane excitability, lasting for at least 15 days after cocaine withdrawal. This persistent IL-NAcSh neuron hypoexcitability was accompanied by an increase in the rheobase, an increase in the afterhyperpolarization potential, and a decrease in the membrane input resistance. This cocaine induced neuroadapation in intrinsic excitability was not observed in prelimibic cortex neurons projecting to the NAc core (PL-NAcCo neurons), a separate descending circuit thought to promote cue-triggered drug seeking. Chemogenetic restoration of IL-NAcSh neuron activity extinguishes both the acquisition and retention of cocaine conditioned place preference memories. Our results provide direct support for the notion that the IL-NAcSh circuit serves to extinct drug associated memories and restoring the drug impaired excitability of IL-NAcSh neurons has the potential to mitigate drug-cue association memories and drug seeking.
Collapse
Affiliation(s)
- Xiaobo Wu
- Departments of Psychiatry & Behavioral Sciences and of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13210
- Institute of Pain Medicine and Special Environmental Medicine, Nantong University, Jiangsu, China, 226019
| | - Aya M. Kobeissi
- Departments of Psychiatry & Behavioral Sciences and of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13210
| | - Hannah L. Phillips
- Departments of Psychiatry & Behavioral Sciences and of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13210
| | - Huihui Dai
- Departments of Psychiatry & Behavioral Sciences and of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13210
| | - Wei-Dong Yao
- Departments of Psychiatry & Behavioral Sciences and of Neuroscience and Physiology, State University of New York, Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
5
|
Swanson RA, Chinigò E, Levenstein D, Vöröslakos M, Mousavi N, Wang XJ, Basu J, Buzsáki G. Topography of putative bi-directional interaction between hippocampal sharp-wave ripples and neocortical slow oscillations. Neuron 2025; 113:754-768.e9. [PMID: 39874961 DOI: 10.1016/j.neuron.2024.12.019] [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: 03/15/2024] [Revised: 10/26/2024] [Accepted: 12/18/2024] [Indexed: 01/30/2025]
Abstract
Systems consolidation relies on coordination between hippocampal sharp-wave ripples (SWRs) and neocortical UP/DOWN states during sleep. However, whether this coupling exists across the neocortex and the mechanisms enabling it remains unknown. By combining electrophysiology in mouse hippocampus (HPC) and retrosplenial cortex (RSC) with wide-field imaging of the dorsal neocortex, we found spatially and temporally precise bi-directional hippocampo-neocortical interaction. HPC multi-unit activity and SWR probability were correlated with UP/DOWN states in the default mode network (DMN), with the highest modulation by the RSC in deep sleep. Further, some SWRs were preceded by the high rebound excitation accompanying DMN DOWN → UP transitions, whereas large-amplitude SWRs were often followed by DOWN states originating in the RSC. We explain these electrophysiological results with a model in which the HPC and RSC are weakly coupled excitable systems capable of bi-directional perturbation and suggest that the RSC may act as a gateway through which SWRs can perturb downstream cortical regions via cortico-cortical propagation of DOWN states.
Collapse
Affiliation(s)
- Rachel A Swanson
- Neuroscience Institute, Langone Medical Center, New York University, New York, NY, USA
| | - Elisa Chinigò
- Center for Neural Science, New York University, New York, NY, USA
| | - Daniel Levenstein
- Department of Neurology and Neurosurgery, McGill University Montreal, QC, Canada; Mila - The Quebec AI Institute, Montreal, QC, Canada
| | - Mihály Vöröslakos
- Neuroscience Institute, Langone Medical Center, New York University, New York, NY, USA
| | - Navid Mousavi
- Neuroscience Institute, Langone Medical Center, New York University, New York, NY, USA
| | - Xiao-Jing Wang
- Center for Neural Science, New York University, New York, NY, USA
| | - Jayeeta Basu
- Neuroscience Institute, Langone Medical Center, New York University, New York, NY, USA; Department of Physiology and Neuroscience, Langone Medical Center, New York University, New York, NY, USA; Department of Psychiatry, Langone Medical Center, New York University, New York, NY, USA.
| | - György Buzsáki
- Neuroscience Institute, Langone Medical Center, New York University, New York, NY, USA; Department of Physiology and Neuroscience, Langone Medical Center, New York University, New York, NY, USA; Department of Neurology, Langone Medical Center, New York University, New York, NY, USA.
| |
Collapse
|
6
|
Perucca E, Taglialatela M. Targeting Kv7 Potassium Channels for Epilepsy. CNS Drugs 2025; 39:263-288. [PMID: 39853501 PMCID: PMC11850491 DOI: 10.1007/s40263-024-01155-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/22/2024] [Indexed: 01/26/2025]
Abstract
Voltage-gated Kv7 potassium channels, particularly Kv7.2 and Kv.7.3 channels, play a critical role in modulating susceptibility to seizures, and mutations in genes that encode these channels cause heterogeneous epilepsy phenotypes. On the basis of this evidence, activation of Kv7.2 and Kv.7.3 channels has long been considered an attractive target in the search for novel antiseizure medications. Ezogabine (retigabine), the first Kv7.2/3 activator introduced in 2011 for the treatment of focal seizures, was withdrawn from the market in 2017 due to declining use after discovery of its association with pigmentation changes in the retina, skin, and mucosae. A novel formulation of ezogabine for pediatric use (XEN496) has been recently investigated in children with KCNQ2-related developmental and epileptic encephalopathy, but the trial was terminated prematurely for reasons unrelated to safety. Among novel Kv7.2/3 openers in clinical development, azetukalner has shown dose-dependent efficacy against drug-resistant focal seizures with a good tolerability profile and no evidence of pigmentation-related adverse effects in early clinical studies, and it is now under investigation in phase III trials for the treatment of focal seizures, generalized tonic-clonic seizures, and major depressive disorder. Another Kv7.2/3 activator, BHV-7000, has completed phase I studies in healthy subjects, with excellent tolerability at plasma drug concentrations that exceed the median effective concentrations in a preclinical model of anticonvulsant activity, but no efficacy data in patients with epilepsy are available to date. Among other Kv7.2/3 activators in clinical development as potential antiseizure medications, pynegabine and CB-003 have completed phase I safety and pharmacokinetic studies, but results have not been yet reported. Overall, interest in targeting Kv7 channels for the treatment of epilepsy and for other indications remains strong. Future breakthroughs in this area could come from exploitation of mechanistic differences in the action of Kv7 activators, and from the development of molecules that combine Kv7 activation with other mechanisms of action.
Collapse
Affiliation(s)
- Emilio Perucca
- Department of Medicine (Austin Health), Melbourne Brain Center, The University of Melbourne, 245 Burgundy St., Heidelberg, VIC, 3084, Australia.
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia.
| | - Maurizio Taglialatela
- Division of Pharmacology, Department of Neuroscience, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
7
|
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
|
8
|
Hashemizadeh S, Alaee E, Aghajani N, Azizi H, Semnanian S. Atorvastatin facilitates extinction and prevents reinstatement of morphine-induced conditioned place preference in rats. Biomed Pharmacother 2024; 181:117639. [PMID: 39520913 DOI: 10.1016/j.biopha.2024.117639] [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: 08/10/2024] [Revised: 10/19/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Opioid addiction is known as a chronic relapsing disorder associated with long-lasting molecular and cellular neuroadaptations that lead to compulsive behavior. Current pharmacotherapies target the modulation of mu-opioid receptors (MOR); however, the relapse rate remains high. In this study, we evaluated the potential effect of atorvastatin, a blood-brain barrier-permeable statin, on preventing morphine relapse through both extinction-reinstatement and abstinence-reinstatement models using conditioned place preference (CPP). Adult male Wistar rats were used to establish morphine-induced CPP (5 mg/kg), followed by extinction training and subsequent priming injection of morphine (2 mg/kg, i.p.) to induce relapse-like behavior. Extinguished rats significantly reinstated their morphine-seeking behavior. In contrast, rats that received different doses of atorvastatin (0.1, 0.5, 1 mg/kg) 1 hour before each extinction training session did not show a preference for the morphine-paired chamber. Moreover, acute atorvastatin injection (1 mg/kg, i.p.) 1 h before the reinstatement test significantly prevented reinstated morphine-seeking behavior. We found that atorvastatin 1 mg/kg attenuated morphine-seeking behaviors, and this attenuation of reinstatement was partly mediated by the upregulation of brain-derived neurotrophic factor (BDNF) in the prefrontal cortex (PFC) and hippocampus (Hipp). Furthermore, atorvastatin reversed Oprm1 upregulation (mu-opioid receptor gene) induced by relapse in the nucleus accumbens and Hipp. Moreover, treatment with atorvastatin during the extinction period alters the electrophysiological properties of the mPFC neurons following morphine priming and enhances neuronal excitability. We conclude that atorvastatin was effective in decreasing reinstatement.
Collapse
Affiliation(s)
- Shiva Hashemizadeh
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences, IPM, Tehran, Iran; Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Elham Alaee
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences, IPM, Tehran, Iran; Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Niloofar Aghajani
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Azizi
- Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran; Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Saeed Semnanian
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences, IPM, Tehran, Iran; Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
9
|
Handlin LJ, Macchi NL, Dumaire NLA, Salih L, Lessie EN, McCommis KS, Moutal A, Dai G. Membrane lipid nanodomains modulate HCN pacemaker channels in nociceptor DRG neurons. Nat Commun 2024; 15:9898. [PMID: 39548079 PMCID: PMC11568329 DOI: 10.1038/s41467-024-54053-z] [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: 03/13/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
Cell membranes consist of heterogeneous lipid nanodomains that influence key cellular processes. Using FRET-based fluorescent assays and fluorescence lifetime imaging microscopy (FLIM), we find that the dimension of cholesterol-enriched ordered membrane domains (OMD) varies considerably, depending on specific cell types. Particularly, nociceptor dorsal root ganglion (DRG) neurons exhibit large OMDs. Disruption of OMDs potentiated action potential firing in nociceptor DRG neurons and facilitated the opening of native hyperpolarization-activated cyclic nucleotide-gated (HCN) pacemaker channels. This increased neuronal firing is partially due to an increased open probability and altered gating kinetics of HCN channels. The gating effect on HCN channels is likely due to a direct modulation of their voltage sensors by OMDs. In animal models of neuropathic pain, we observe reduced OMD size and a loss of HCN channel localization within OMDs. Additionally, cholesterol supplementation inhibited HCN channels and reduced neuronal hyperexcitability in pain models. These findings suggest that disturbances in lipid nanodomains play a critical role in regulating HCN channels within nociceptor DRG neurons, influencing pain modulation.
Collapse
Affiliation(s)
- Lucas J Handlin
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Natalie L Macchi
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Nicolas L A Dumaire
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Lyuba Salih
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Erin N Lessie
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Kyle S McCommis
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Aubin Moutal
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Gucan Dai
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA.
| |
Collapse
|
10
|
Swanson R, Chinigò E, Levenstein D, Vöröslakos M, Mousavi N, Wang XJ, Basu J, Buzsáki G. Topography of putative bidirectional interaction between hippocampal sharp wave ripples and neocortical slow oscillations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619879. [PMID: 39484611 PMCID: PMC11526890 DOI: 10.1101/2024.10.23.619879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Systems consolidation relies on coordination between hippocampal sharp-wave ripples (SWRs) and neocortical UP/DOWN states during sleep. However, whether this coupling exists across neocortex and the mechanisms enabling it remain unknown. By combining electrophysiology in mouse hippocampus (HPC) and retrosplenial cortex (RSC) with widefield imaging of dorsal neocortex, we found spatially and temporally precise bidirectional hippocampo-neocortical interaction. HPC multi-unit activity and SWR probability was correlated with UP/DOWN states in mouse default mode network, with highest modulation by RSC in deep sleep. Further, some SWRs were preceded by the high rebound excitation accompanying DMN DOWN→UP transitions, while large-amplitude SWRs were often followed by DOWN states originating in RSC. We explain these electrophysiological results with a model in which HPC and RSC are weakly coupled excitable systems capable of bi-directional perturbation and suggest RSC may act as a gateway through which SWRs can perturb downstream cortical regions via cortico-cortical propagation of DOWN states.
Collapse
Affiliation(s)
- Rachel Swanson
- Neuroscience Institute, Langone Medical Center, New York University, New York, NY, USA
| | - Elisa Chinigò
- Center for Neural Science, New York University, New York, NY, USA
| | - Daniel Levenstein
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Mila – The Quebec AI Institute, Montreal, QC, Canada
| | - Mihály Vöröslakos
- Neuroscience Institute, Langone Medical Center, New York University, New York, NY, USA
| | - Navid Mousavi
- Neuroscience Institute, Langone Medical Center, New York University, New York, NY, USA
| | - Xiao-Jing Wang
- Center for Neural Science, New York University, New York, NY, USA
| | - Jayeeta Basu
- Neuroscience Institute, Langone Medical Center, New York University, New York, NY, USA
- Department of Physiology and Neuroscience, Langone Medical Center, New York University, New York, NY, USA
- Department of Psychiatry, Langone Medical Center, New York University, New York, NY, USA
| | - György Buzsáki
- Neuroscience Institute, Langone Medical Center, New York University, New York, NY, USA
- Department of Physiology and Neuroscience, Langone Medical Center, New York University, New York, NY, USA
- Department of Neurology, Langone Medical Center, New York University, New York, NY, USA
| |
Collapse
|
11
|
Schapiro K, Rittenberg JD, Kenngott M, Marder E. I h block reveals separation of timescales in pyloric rhythm response to temperature changes in Cancer borealis. eLife 2024; 13:RP98844. [PMID: 39404608 PMCID: PMC11479588 DOI: 10.7554/elife.98844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
Motor systems operate over a range of frequencies and relative timing (phase). We studied the role of the hyperpolarization-activated inward current (Ih) in regulating these features in the pyloric rhythm of the stomatogastric ganglion (STG) of the crab, Cancer borealis, as temperature was altered from 11°C to 21°C. Under control conditions, rhythm frequency increased monotonically with temperature, while the phases of the pyloric dilator (PD), lateral pyloric (LP), and pyloric (PY) neurons remained constant. Blocking Ih with cesium (Cs+) phase advanced PD offset, LP onset, and LP offset at 11°C, and the latter two further advanced as temperature increased. In Cs+ the frequency increase with temperature diminished and the Q10 of the frequency dropped from ~1.75 to ~1.35. Unexpectedly in Cs+, the frequency dynamics became non-monotonic during temperature transitions; frequency initially dropped as temperature increased, then rose once temperature stabilized, creating a characteristic 'jag'. Interestingly, these jags persisted during temperature transitions in Cs+ when the pacemaker was isolated by picrotoxin, although the temperature-induced change in frequency recovered to control levels. Overall, these data suggest that Ih plays an important role in maintaining smooth transitory responses and persistent frequency increases by different mechanisms in the pyloric circuitry during temperature fluctuations.
Collapse
Affiliation(s)
- Kyra Schapiro
- Biology Department, Brandeis UniversityWalthamUnited States
| | - JD Rittenberg
- Biology Department, Brandeis UniversityWalthamUnited States
| | - Max Kenngott
- Biology Department, Brandeis UniversityWalthamUnited States
| | - Eve Marder
- Biology Department, Brandeis UniversityWalthamUnited States
- Volen Center and Biology Department, Brandeis UniversityWalthamUnited States
| |
Collapse
|
12
|
Buss EW, Lofaro OM, Barnett A, Leroy F, Santoro B, Siegelbaum SA, Bock T. HCN1 hyperpolarization-activated cyclic nucleotide-gated channels enhance evoked GABA release from parvalbumin-positive interneurons. Proc Natl Acad Sci U S A 2024; 121:e2319246121. [PMID: 39378096 PMCID: PMC11494348 DOI: 10.1073/pnas.2319246121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 08/27/2024] [Indexed: 10/10/2024] Open
Abstract
Hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels generate the cationic Ih current in neurons and regulate the excitability of neuronal networks. The function of HCN channels depends, in part, on their subcellular localization. Of the four HCN isoforms (HCN1-4), HCN1 is strongly expressed in the dendrites of pyramidal neurons (PNs) in hippocampal area CA1 but also in presynaptic terminals of parvalbumin-positive interneurons (PV+ INs), which provide strong inhibitory control over hippocampal activity. Yet, little is known about how HCN1 channels in these cells regulate the evoked release of the inhibitory transmitter GABA from their axon terminals. Here, we used genetic, optogenetic, electrophysiological, and imaging techniques to investigate how the electrophysiological properties of PV+ INs are regulated by HCN1, including how HCN1 activity at presynaptic terminals regulates the release of GABA onto PNs in CA1. We found that application of HCN1 pharmacological blockers reduced the amplitude of the inhibitory postsynaptic potential recorded from CA1 PNs in response to selective optogenetic stimulation of PV+ INs. Homozygous HCN1 knockout mice also show reduced IPSCs in postsynaptic cells. Finally, two-photon imaging using genetically encoded fluorescent calcium indicators revealed that HCN1 blockers reduced the probability that an extracellular electrical stimulating pulse evoked a Ca2+ response in individual PV+ IN presynaptic boutons. Taken together, our results show that HCN1 channels in the axon terminals of PV+ interneurons facilitate GABAergic transmission in the hippocampal CA1 region.
Collapse
Affiliation(s)
- Eric W. Buss
- Departments of Neuroscience and Pharmacology, Kavli Institute for Brain Science, Zuckerman Mind Brain Behavior Institute, Columbia University Medical Center, New York, NY10027
| | - Olivia M. Lofaro
- Departments of Neuroscience and Pharmacology, Kavli Institute for Brain Science, Zuckerman Mind Brain Behavior Institute, Columbia University Medical Center, New York, NY10027
| | - Anastasia Barnett
- Departments of Neuroscience and Pharmacology, Kavli Institute for Brain Science, Zuckerman Mind Brain Behavior Institute, Columbia University Medical Center, New York, NY10027
| | - Felix Leroy
- Departments of Neuroscience and Pharmacology, Kavli Institute for Brain Science, Zuckerman Mind Brain Behavior Institute, Columbia University Medical Center, New York, NY10027
| | - Bina Santoro
- Departments of Neuroscience and Pharmacology, Kavli Institute for Brain Science, Zuckerman Mind Brain Behavior Institute, Columbia University Medical Center, New York, NY10027
| | - Steven A. Siegelbaum
- Departments of Neuroscience and Pharmacology, Kavli Institute for Brain Science, Zuckerman Mind Brain Behavior Institute, Columbia University Medical Center, New York, NY10027
| | - Tobias Bock
- Departments of Neuroscience and Pharmacology, Kavli Institute for Brain Science, Zuckerman Mind Brain Behavior Institute, Columbia University Medical Center, New York, NY10027
- Department of Systems Neurophysiology, Institute for Zoology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen52074, Germany
| |
Collapse
|
13
|
Rahdar M, Davoudi S, Dehghan S, Javan M, Hosseinmardi N, Behzadi G, Janahmadi M. Reversal of electrophysiological and behavioral deficits mediated by 5-HT7 receptor upregulation following LP-211 treatment in an autistic-like rat model induced by prenatal valproic acid exposure. Neuropharmacology 2024; 257:110057. [PMID: 38964596 DOI: 10.1016/j.neuropharm.2024.110057] [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: 03/10/2024] [Revised: 06/11/2024] [Accepted: 06/27/2024] [Indexed: 07/06/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by alterations and imbalances in multiple brain neurochemical systems, particularly the serotonergic neurotransmission. This includes changes in serotonin (5-HT) levels, aberrations in 5-HT transporter activity, and decreased synthesis and expression of 5-HT receptors (5-HT7Rs). The exact role of the brain 5-HT system in the development of ASD remains unclear, with conflicting evidence on its involvement. Recently, we have reported research has shown a significant decrease in serotonergic neurons originating from the raphe nuclei and projecting to the CA1 region of the dorsal hippocampus in autistic-like rats. Additionally, we have shown that chronic activation of 5-HT7Rs reverses the effects of autism induction on synaptic plasticity. However, the functional significance of 5-HT7Rs at the cellular level is still not fully understood. This study presents new evidence indicating an upregulation of 5-HT7R in the CA1 subregion of the hippocampus following the induction of autism. The present account also demonstrates that activation of 5-HT7R with its agonist LP-211 can reverse electrophysiological abnormalities in hippocampal pyramidal neurons in a rat model of autism induced by prenatal exposure to VPA. Additionally, in vivo administration of LP-211 resulted in improvements in motor coordination, novel object recognition, and a reduction in stereotypic behaviors in autistic-like offspring. The findings suggest that dysregulated expression of 5-HT7Rs may play a role in the pathophysiology of ASD, and that agonists like LP-211 could potentially be explored as a pharmacological treatment for autism spectrum disorder.
Collapse
Affiliation(s)
- Mona Rahdar
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shima Davoudi
- Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samaneh Dehghan
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Narges Hosseinmardi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gila Behzadi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahyar Janahmadi
- Neuroscience Research Center and Dep. of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Walters JM, Noblet HA, Chung HJ. An emerging role of STriatal-Enriched protein tyrosine Phosphatase in hyperexcitability-associated brain disorders. Neurobiol Dis 2024; 200:106641. [PMID: 39159894 DOI: 10.1016/j.nbd.2024.106641] [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: 04/27/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024] Open
Abstract
STriatal-Enriched protein tyrosine Phosphatase (STEP) is a brain-specific tyrosine phosphatase that is associated with numerous neurological and neuropsychiatric disorders. STEP dephosphorylates and inactivates various kinases and phosphatases critical for neuronal function and health including Fyn, Pyk2, ERK1/2, p38, and PTPα. Importantly, STEP dephosphorylates NMDA and AMPA receptors, two major glutamate receptors that mediate fast excitatory synaptic transmission. This STEP-mediated dephosphorylation leads to their internalization and inhibits both Hebbian synaptic potentiation and homeostatic synaptic scaling. Hence, STEP has been widely accepted to weaken excitatory synaptic strength. However, emerging evidence implicates a novel role of STEP in neuronal hyperexcitability and seizure disorders. Genetic deletion and pharmacological blockade of STEP reduces seizure susceptibility in acute seizure mouse models and audiogenic seizures in a mouse model of Fragile X syndrome. Pharmacologic inhibition of STEP also decreases hippocampal activity and neuronal intrinsic excitability. Here, we will highlight the divergent roles of STEP in excitatory synaptic transmission and neuronal intrinsic excitability, present the potential underlying mechanisms, and discuss their impact on STEP-associated neurologic and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jennifer M Walters
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hayden A Noblet
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hee Jung Chung
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Institute of Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
15
|
Porro A, Armano E, Brandalise F, Appiani R, Beltrame M, Saponaro A, Dallanoce C, Nakajo K, Ryu K, Leone R, Thiel G, Pallavicini M, Moroni A, Bolchi C. A Photoactivatable Version of Ivabradine Enables Light-Induced Block of HCN Current In Vivo. J Med Chem 2024; 67:16209-16221. [PMID: 39238314 DOI: 10.1021/acs.jmedchem.4c01047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Therapeutic drugs, whose bioactivity is hindered by a photoremovable cage, offer the advantage of spatiotemporal confinement of their action to the target diseased tissue with improved bioavailability and efficacy. Here, we have applied such an approach to ivabradine (IVA), a bradycardic agent indicated for angina pectoris and heart failure, acting as a specific HCN channel blocker. To overcome the side effects due to its poor discrimination among HCN channel subtypes (HCN1-4), we prepared a caged version of IVA linked to a photocleavable bromoquinolinylmethyl group (BHQ-IVA). We show that upon illumination with blue light (440 nm), BHQ-IVA releases active IVA that blocks HCN channel currents in vitro and exerts a bradycardic effect in vivo. Both BHQ-IVA and the cage are inactive. Caging is stable in aqueous medium and in the dark, and it does not impair aqueous solubility and cell permeation, indispensable for IVA activity. This approach allows for bypassing the poor subtype-specificity of IVA, expanding its prescription to HCN-related diseases besides cardiac.
Collapse
Affiliation(s)
- Alessandro Porro
- Department of Biosciences, University of Milan, Milano 20133, Italy
| | - Edoardo Armano
- Department of Pharmaceutical Sciences, University of Milan, Milano 20133, Italy
| | | | - Rebecca Appiani
- Department of Pharmaceutical Sciences, University of Milan, Milano 20133, Italy
| | - Monica Beltrame
- Department of Biosciences, University of Milan, Milano 20133, Italy
| | - Andrea Saponaro
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milano 20133, Italy
| | - Clelia Dallanoce
- Department of Pharmaceutical Sciences, University of Milan, Milano 20133, Italy
| | - Koichi Nakajo
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University, Kawachi District, Shimotsuke 329-0498, Japan
| | - Kaei Ryu
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University, Kawachi District, Shimotsuke 329-0498, Japan
| | - Roberta Leone
- Department of Biosciences, University of Milan, Milano 20133, Italy
| | - Gerhard Thiel
- Department of Biology, TU-Darmstadt, Darmstadt 64287, Germany
| | - Marco Pallavicini
- Department of Pharmaceutical Sciences, University of Milan, Milano 20133, Italy
| | - Anna Moroni
- Department of Biosciences, University of Milan, Milano 20133, Italy
| | - Cristiano Bolchi
- Department of Pharmaceutical Sciences, University of Milan, Milano 20133, Italy
| |
Collapse
|
16
|
Dembrow NC, Sawchuk S, Dalley R, Opitz-Araya X, Hudson M, Radaelli C, Alfiler L, Walling-Bell S, Bertagnolli D, Goldy J, Johansen N, Miller JA, Nasirova K, Owen SF, Parga-Becerra A, Taskin N, Tieu M, Vumbaco D, Weed N, Wilson J, Lee BR, Smith KA, Sorensen SA, Spain WJ, Lein ES, Perlmutter SI, Ting JT, Kalmbach BE. Areal specializations in the morpho-electric and transcriptomic properties of primate layer 5 extratelencephalic projection neurons. Cell Rep 2024; 43:114718. [PMID: 39277859 PMCID: PMC11488157 DOI: 10.1016/j.celrep.2024.114718] [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: 03/15/2024] [Revised: 07/22/2024] [Accepted: 08/20/2024] [Indexed: 09/17/2024] Open
Abstract
Large-scale analysis of single-cell gene expression has revealed transcriptomically defined cell subclasses present throughout the primate neocortex with gene expression profiles that differ depending upon neocortical region. Here, we test whether the interareal differences in gene expression translate to regional specializations in the physiology and morphology of infragranular glutamatergic neurons by performing Patch-seq experiments in brain slices from the temporal cortex (TCx) and motor cortex (MCx) of the macaque. We confirm that transcriptomically defined extratelencephalically projecting neurons of layer 5 (L5 ET neurons) include retrogradely labeled corticospinal neurons in the MCx and find multiple physiological properties and ion channel genes that distinguish L5 ET from non-ET neurons in both areas. Additionally, while infragranular ET and non-ET neurons retain distinct neuronal properties across multiple regions, there are regional morpho-electric and gene expression specializations in the L5 ET subclass, providing mechanistic insights into the specialized functional architecture of the primate neocortex.
Collapse
Affiliation(s)
- Nikolai C Dembrow
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA; Epilepsy Center of Excellence, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA.
| | - Scott Sawchuk
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Rachel Dalley
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Mark Hudson
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | | | - Lauren Alfiler
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | | | - Scott F Owen
- Allen Institute for Brain Science, Seattle, WA 98109, USA; Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Alejandro Parga-Becerra
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA; Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Naz Taskin
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Michael Tieu
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - David Vumbaco
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Natalie Weed
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Julia Wilson
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Brian R Lee
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - William J Spain
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA; Epilepsy Center of Excellence, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Steve I Perlmutter
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA; Washington National Primate Research Center, Seattle, WA 98195, USA
| | - Jonathan T Ting
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA; Allen Institute for Brain Science, Seattle, WA 98109, USA; Washington National Primate Research Center, Seattle, WA 98195, USA
| | - Brian E Kalmbach
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA; Allen Institute for Brain Science, Seattle, WA 98109, USA.
| |
Collapse
|
17
|
Handlin LJ, Macchi NL, Dumaire NLA, Salih L, Lessie EN, McCommis KS, Moutal A, Dai G. Membrane Lipid Nanodomains Modulate HCN Pacemaker Channels in Nociceptor DRG Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.02.556056. [PMID: 37732182 PMCID: PMC10508734 DOI: 10.1101/2023.09.02.556056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Cell membranes consist of heterogeneous lipid nanodomains that influence key cellular processes. Using FRET-based fluorescent assays and fluorescence lifetime imaging microscopy (FLIM), we found that the dimension of cholesterol-enriched ordered membrane domains (OMD) varies considerably, depending on specific cell types. Particularly, nociceptor dorsal root ganglion (DRG) neurons exhibit large OMDs. Disruption of OMDs potentiated action potential firing in nociceptor DRG neurons and facilitated the opening of native hyperpolarization-activated cyclic nucleotide-gated (HCN) pacemaker channels. This increased neuronal firing is partially due to an increased open probability and altered gating kinetics of HCN channels. The gating effect on HCN channels was likely due to a direct modulation of their voltage sensors by OMDs. In animal models of neuropathic pain, we observed reduced OMD size and a loss of HCN channel localization within OMDs. Additionally, cholesterol supplementation inhibited HCN channels and reduced neuronal hyperexcitability in pain models. These findings suggest that disturbances in lipid nanodomains play a critical role in regulating HCN channels within nociceptor DRG neurons, influencing pain modulation.
Collapse
|
18
|
Basso V, Döbrössy MD, Thompson LH, Kirik D, Fuller HR, Gates MA. State of the Art in Sub-Phenotyping Midbrain Dopamine Neurons. BIOLOGY 2024; 13:690. [PMID: 39336117 PMCID: PMC11428604 DOI: 10.3390/biology13090690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024]
Abstract
Dopaminergic neurons in the ventral tegmental area (VTA) and the substantia nigra pars compacta (SNpc) comprise around 75% of all dopaminergic neurons in the human brain. While both groups of dopaminergic neurons are in close proximity in the midbrain and partially overlap, development, function, and impairments in these two classes of neurons are highly diverse. The molecular and cellular mechanisms underlying these differences are not yet fully understood, but research over the past decade has highlighted the need to differentiate between these two classes of dopaminergic neurons during their development and in the mature brain. This differentiation is crucial not only for understanding fundamental circuitry formation in the brain but also for developing therapies targeted to specific dopaminergic neuron classes without affecting others. In this review, we summarize the state of the art in our understanding of the differences between the dopaminergic neurons of the VTA and the SNpc, such as anatomy, structure, morphology, output and input, electrophysiology, development, and disorders, and discuss the current technologies and methods available for studying these two classes of dopaminergic neurons, highlighting their advantages, limitations, and the necessary improvements required to achieve more-precise therapeutic interventions.
Collapse
Affiliation(s)
- Valentina Basso
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK
| | - Máté D Döbrössy
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional, Neurosurgery, Medical Center, University of Freiburg, 79106 Freiburg im Breisgau, Germany
- Department of Stereotactic and Functional Neurosurgery, Medical Center, University of Freiburg, 79106 Freiburg im Breisgau, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Lachlan H Thompson
- Charles Perkins Centre, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Deniz Kirik
- Brain Repair and Imaging in Neural Systems (B.R.A.I.N.S) Unit, Department of Experimental Medical Science, Lund University, BMC D11, 22184 Lund, Sweden
| | - Heidi R Fuller
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | - Monte A Gates
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK
| |
Collapse
|
19
|
Nissenkorn A, Bar L, Ben-Bassat A, Rothstein L, Abdelrahim H, Sokol R, Gabis LV, Attali B. Donepezil as a new therapeutic potential in KCNQ2- and KCNQ3-related autism. Front Cell Neurosci 2024; 18:1380442. [PMID: 39175503 PMCID: PMC11338814 DOI: 10.3389/fncel.2024.1380442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 07/29/2024] [Indexed: 08/24/2024] Open
Abstract
Introduction The KCNQ2/KCNQ3 genes encode the voltage-gated K channel underlying the neuronal M-current, regulating neuronal excitability. Loss-of-function (LoF) variants cause neonatal epilepsy, treatable with the M-current-opener retigabine, which is no longer marketed due to side effects. Gain-of-function (GoF) variants cause developmental encephalopathy and autism that could be amenable to M-current, but such therapies are not clinically available. In this translational project, we investigated whether donepezil, a cholinergic drug used in Alzheimer's, suppresses M currents in vitro and improves cognitive symptoms in patients with GoF variants. Methods (1) The effect of 1 μM donepezil on the amplitude of the M-current was measured in excitatory and inhibitory neurons of mouse primary cultured hippocampal cells. M-current was measured using the standard deactivation protocol (holding at 0 mV and deactivation at -60 mV) in the voltage-clamp configuration of the whole-cell patch clamp technique. The impact of donepezil was also examined on the spontaneous firing activity of hippocampal neurons in the current-clamp configuration. (2) Four children with autism, aged 2.5-8 years, with the following GoF variants were enrolled: KCNQ2 (p. Arg144Gln) and KCNQ 3 (p.Arg227Gln, p.Arg230Cys). Patients were treated off-label with donepezil 2.5-5 mg/d for 12 months and assessed with: clinical Global Impression of Change (CGI-c), Childhood Autism Rating Scale 2 (CARS-2), Adaptive Behavior Assessment System-II (ABAS-II), and Child Development Inventory (CDI). Results (1) Application of donepezil for at least 6 min produced a significant inhibition of the M-current with an IC50 of 0.4 μM. At 1 μM, donepezil reduced by 67% the M-current density of excitatory neurons (2.4 ± 0.46 vs. 0.89 ± 0.15 pA/pF, p < 0.05*). In inhibitory neurons, application of 1 μM donepezil produced a lesser inhibition of 59% of the M-current density (1.39 ± 0.43 vs. 0.57 ± 0.21, p > 0.05). Donepezil (1 μM) potently increased by 2.6-fold the spontaneous firing frequency, which was prevented by the muscarinic receptor antagonist atropine (10 μM). (2) The CARS-2 decreased by 3.8 ± 4.9 points (p > 0.05), but in two patients with KCNQ3 variants, the improvement was over the 4.5 clinically relevant threshold. The global clinical change was also clinically significant in these patients (CGI-c = 1). The CDI increased by 65% (p < 0.05*), while the ABAS-II remained unchanged. Discussion Donepezil should be repurposed as a novel alternative treatment for GoF variants in KCNQ2/KCNQ3 encephalopathy.
Collapse
Affiliation(s)
- Andreea Nissenkorn
- Pediatric Neurology Unit, Edith Wolfson Medical Center, Holon, Israel
- Magen National Center for Rare Disorders, Edith Wolfson Medical Center, Holon, Israel
- Department of Pediatric, School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Lior Bar
- Department of Electrophysiology, School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ariel Ben-Bassat
- Department of Electrophysiology, School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lynn Rothstein
- Pediatric Neurology Unit, Edith Wolfson Medical Center, Holon, Israel
- Magen National Center for Rare Disorders, Edith Wolfson Medical Center, Holon, Israel
| | - Hoda Abdelrahim
- Pediatric Neurology Unit, Edith Wolfson Medical Center, Holon, Israel
- Magen National Center for Rare Disorders, Edith Wolfson Medical Center, Holon, Israel
| | - Riki Sokol
- Pediatric Neurology Unit, Edith Wolfson Medical Center, Holon, Israel
| | - Lidia V. Gabis
- Magen National Center for Rare Disorders, Edith Wolfson Medical Center, Holon, Israel
- Department of Pediatric, School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Bernard Attali
- Department of Electrophysiology, School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
20
|
Schapiro KA, Rittenberg JD, Kenngott M, Marder E. I h Block Reveals Separation of Timescales in Pyloric Rhythm Response to Temperature Changes in Cancer borealis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.04.592541. [PMID: 38766157 PMCID: PMC11100622 DOI: 10.1101/2024.05.04.592541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Motor systems operate over a range of frequencies and relative timing (phase). We studied the contribution of the hyperpolarization-activated inward current (Ih) to frequency and phase in the pyloric rhythm of the stomatogastric ganglion (STG) of the crab, Cancer borealis as temperature was altered from 11°C to 21°C. Under control conditions, the frequency of the rhythm increased monotonically with temperature, while the phases of the pyloric dilator (PD), lateral pyloric (LP), and pyloric (PY) neurons remained constant. When we blocked Ih with cesium (Cs+) PD offset, LP onset, and LP offset were all phase advanced in Cs+ at 11°C, and the latter two further advanced as temperature increased. In Cs+ the steady state increase in pyloric frequency with temperature diminished and the Q10 of the pyloric frequency dropped from ~1.75 to ~1.35. Unexpectedly in Cs+, the frequency displayed non-monotonic dynamics during temperature transitions; the frequency initially dropped as temperature increased, then rose once temperature stabilized, creating a characteristic "jag". Interestingly, these jags were still present during temperature transitions in Cs+ when the pacemaker was isolated by picrotoxin, although the temperature-induced change in frequency recovered to control levels. Overall, these data suggest that Ih plays an important role in the ability of this circuit to produce smooth transitory responses and persistent frequency increases by different mechanisms during temperature fluctuations.
Collapse
Affiliation(s)
- Kyra A Schapiro
- Volen Center and Biology Department, Brandeis University, Waltham, MA 02454 USA
| | - J D Rittenberg
- Volen Center and Biology Department, Brandeis University, Waltham, MA 02454 USA
| | - Max Kenngott
- Volen Center and Biology Department, Brandeis University, Waltham, MA 02454 USA
| | - Eve Marder
- Volen Center and Biology Department, Brandeis University, Waltham, MA 02454 USA
| |
Collapse
|
21
|
Romano EJ, Zhang DQ. Dopaminergic amacrine cells express HCN channels in the developing and adult mouse retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.20.604440. [PMID: 39091772 PMCID: PMC11291019 DOI: 10.1101/2024.07.20.604440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Purpose To determine the molecular and functional expression of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels in developing and mature dopaminergic amacrine cells (DACs), the sole source of ocular dopamine that plays a vital role in visual function and eye development. Methods HCN channels are encoded by isoforms 1-4. HCN1, HCN2, and HCN4 were immunostained in retinal slices obtained from mice at postnatal day 4 (P4), P8, and P12 as well as in adults. Each HCN channel isoform was also immunostained with tyrosine hydroxylase, a marker for DACs, at P12 and adult retinas. Genetically-marked DACs were recorded in flat-mount retina preparation using a whole-cell current-clamp technique. Results HCN1 was expressed in rods/cones, amacrine cells, and retinal ganglion cells (RGCs) at P4, along with bipolar cells by P12. Different from HCN1, HCN2 and HCN4 were each expressed in amacrine cells and RGCs at P4, along with bipolar cells by P8, and in rods/cones by P12. Double immunostaining shows that each of the three isoforms was expressed in approximately half of DACs at P12 but in almost all DACs in adults. Electrophysiology results demonstrate that HCN channel isoforms form functional HCN channels, and the pharmacological blockade of HCN channels reduced the spontaneous firing frequency in most DACs. Conclusions Each class of retinal neurons may use different isoforms of HCN channels to function during development. HCN1, HCN2, and HCN4 form functional HCN channels in DACs, which appears to modulate their spontaneous firing activity.
Collapse
Affiliation(s)
- Emilio J Romano
- Eye Research Institute, Oakland University, Rochester, Michigan
| | - Dao-Qi Zhang
- Eye Research Institute, Oakland University, Rochester, Michigan
- Eye Research Center, Oakland University William Beaumont School of Medicine, Rochester, Michigan
| |
Collapse
|
22
|
Mehrotra D, Levenstein D, Duszkiewicz AJ, Carrasco SS, Booker SA, Kwiatkowska A, Peyrache A. Hyperpolarization-activated currents drive neuronal activation sequences in sleep. Curr Biol 2024; 34:3043-3054.e8. [PMID: 38901427 DOI: 10.1016/j.cub.2024.05.048] [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: 10/24/2023] [Revised: 04/03/2024] [Accepted: 05/23/2024] [Indexed: 06/22/2024]
Abstract
Sequential neuronal patterns are believed to support information processing in the cortex, yet their origin is still a matter of debate. We report that neuronal activity in the mouse postsubiculum (PoSub), where a majority of neurons are modulated by the animal's head direction, was sequentially activated along the dorsoventral axis during sleep at the transition from hyperpolarized "DOWN" to activated "UP" states, while representing a stable direction. Computational modeling suggested that these dynamics could be attributed to a spatial gradient of hyperpolarization-activated currents (Ih), which we confirmed in ex vivo slice experiments and corroborated in other cortical structures. These findings open up the possibility that varying amounts of Ih across cortical neurons could result in sequential neuronal patterns and that traveling activity upstream of the entorhinal-hippocampal circuit organizes large-scale neuronal activity supporting learning and memory during sleep.
Collapse
Affiliation(s)
- Dhruv Mehrotra
- Montréal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, 3801 Rue University, Montréal, QC H3A 2B4, Canada; Integrated Program in Neuroscience, McGill University, 3801 Rue University, Montréal, QC H3A 2B4, Canada
| | - Daniel Levenstein
- Montréal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, 3801 Rue University, Montréal, QC H3A 2B4, Canada; MILA, 6666 Rue Saint-Urbain, Montréal, QC H2S 3H1, Canada
| | - Adrian J Duszkiewicz
- Montréal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, 3801 Rue University, Montréal, QC H3A 2B4, Canada; Division of Psychology, Faculty of Natural Sciences, University of Stirling, Stirling FK9 4LA, UK; Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Sofia Skromne Carrasco
- Montréal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, 3801 Rue University, Montréal, QC H3A 2B4, Canada; Integrated Program in Neuroscience, McGill University, 3801 Rue University, Montréal, QC H3A 2B4, Canada
| | - Sam A Booker
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; Patrick Wild Centre for Research into Autism, Fragile X Syndrome & Intellectual Disabilities, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Angelika Kwiatkowska
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Adrien Peyrache
- Montréal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, 3801 Rue University, Montréal, QC H3A 2B4, Canada.
| |
Collapse
|
23
|
Abdallah BM, Elshoeibi AM, ElTantawi N, Arif M, Hourani RF, Akomolafe AF, Hamwi MN, Mahmood FR, Saracoglu KT, Saracoglu A, Chivese T. Comparison of postoperative pain in children after maintenance anaesthesia with propofol or sevoflurane: a systematic review and meta-analysis. Br J Anaesth 2024; 133:93-102. [PMID: 38670899 PMCID: PMC11213989 DOI: 10.1016/j.bja.2024.03.022] [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: 12/27/2023] [Revised: 03/04/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Propofol and sevoflurane are two of the most commonly used anaesthetics for paediatric surgery. Data from some clinical trials suggest that postoperative pain incidence is lower when propofol is used for maintenance of anaesthesia compared with sevoflurane, although this is not clear. METHODS This meta-analysis compared postoperative pain following maintenance of anaesthesia with propofol or sevoflurane in paediatric surgeries. PubMed Medline, Embase, Scopus, Web of Science and Cochrane Library were searched for randomised controlled trials (RCTs) that compared postoperative pain between sevoflurane and propofol anaesthesia in children. After quality assessment, a meta-analysis was carried out using bias-adjusted inverse heterogeneity methods, heterogeneity using I2 and publication bias using Doi plots. RESULTS In total, 13 RCTs with 1174 children were included. The overall synthesis suggested nearly two-fold higher odds of overall postoperative pain in the sevoflurane group compared with the propofol group (odds ratio [OR] 1.88, 95% confidence interval [CI] 1.12-3.15, I2=58.2%). Further, children in the sevoflurane group had higher odds of having higher pain scores (OR 3.18, 95% CI 1.83-5.53, I2=20.9%), and a 60% increase in the odds of requiring postoperative rescue analgesia compared with propofol (OR 1.60, 95% CI 0.89-2.88, I2=58.2%). CONCLUSIONS Children maintained on inhalational sevoflurane had higher odds of postoperative pain compared with those maintained on propofol. The results also suggest that sevoflurane is associated with higher odds of needing postoperative rescue analgesia compared with propofol. REGISTRATION The protocol for this systematic review and meta-analysis was registered on the International Prospective Register of Systematic Reviews (PROSPERO) with registration ID CRD42023445913.
Collapse
Affiliation(s)
| | | | | | - Mariah Arif
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Razan F Hourani
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | - Mahmoud N Hamwi
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | - Kemal T Saracoglu
- College of Medicine, QU Health, Qatar University, Doha, Qatar; Department of Anaesthesiology, ICU, and Perioperative Medicine, Hazm Mebaireek General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Ayten Saracoglu
- College of Medicine, QU Health, Qatar University, Doha, Qatar; Department of Anaesthesiology, ICU, and Perioperative Medicine, Aisha Bint Hamad Al-Attiyah Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Tawanda Chivese
- College of Medicine, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
24
|
Burtscher V, Mount J, Huang J, Cowgill J, Chang Y, Bickel K, Chen J, Yuan P, Chanda B. Structural basis for hyperpolarization-dependent opening of human HCN1 channel. Nat Commun 2024; 15:5216. [PMID: 38890331 PMCID: PMC11189445 DOI: 10.1038/s41467-024-49599-x] [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: 08/14/2023] [Accepted: 06/11/2024] [Indexed: 06/20/2024] Open
Abstract
Hyperpolarization and cyclic nucleotide (HCN) activated ion channels are critical for the automaticity of action potentials in pacemaking and rhythmic electrical circuits in the human body. Unlike most voltage-gated ion channels, the HCN and related plant ion channels activate upon membrane hyperpolarization. Although functional studies have identified residues in the interface between the voltage-sensing and pore domain as crucial for inverted electromechanical coupling, the structural mechanisms for this unusual voltage-dependence remain unclear. Here, we present cryo-electron microscopy structures of human HCN1 corresponding to Closed, Open, and a putative Intermediate state. Our structures reveal that the downward motion of the gating charges past the charge transfer center is accompanied by concomitant unwinding of the inner end of the S4 and S5 helices, disrupting the tight gating interface observed in the Closed state structure. This helix-coil transition at the intracellular gating interface accompanies a concerted iris-like dilation of the pore helices and underlies the reversed voltage dependence of HCN channels.
Collapse
Affiliation(s)
- Verena Burtscher
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jonathan Mount
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jian Huang
- Department of Chemistry, University of Massachusetts, Amherst, MA, USA
| | - John Cowgill
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Applied Physics, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Yongchang Chang
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kathleen Bickel
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jianhan Chen
- Department of Chemistry, University of Massachusetts, Amherst, MA, USA
| | - Peng Yuan
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Baron Chanda
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA.
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Neuroscience, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
25
|
Südkamp N, Shchyglo O, Manahan-Vaughan D. GluN2A or GluN2B subunits of the NMDA receptor contribute to changes in neuronal excitability and impairments in LTP in the hippocampus of aging mice but do not mediate detrimental effects of oligomeric Aβ (1-42). Front Aging Neurosci 2024; 16:1377085. [PMID: 38832073 PMCID: PMC11144909 DOI: 10.3389/fnagi.2024.1377085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/26/2024] [Indexed: 06/05/2024] Open
Abstract
Studies in rodent models have revealed that oligomeric beta-amyloid protein [Aβ (1-42)] plays an important role in the pathogenesis of Alzheimer's disease. Early elevations in hippocampal neuronal excitability caused by Aβ (1-42) have been proposed to be mediated via enhanced activation of GluN2B-containing N-methyl-D-aspartate receptors (NMDAR). To what extent GluN2A or GluN2B-containing NMDAR contribute to Aβ (1-42)-mediated impairments of hippocampal function in advanced rodent age is unclear. Here, we assessed hippocampal long-term potentiation (LTP) and neuronal responses 4-5 weeks after bilateral intracerebral inoculation of 8-15 month old GluN2A+/- or GluN2B+/- transgenic mice with oligomeric Aβ (1-42), or control peptide. Whole-cell patch-clamp recordings in CA1 pyramidal neurons revealed a more positive resting membrane potential and increased total spike time in GluN2A+/-, but not GluN2B+/--hippocampi following treatment with Aβ (1-42) compared to controls. Action potential 20%-width was increased, and the descending slope was reduced, in Aβ-treated GluN2A+/-, but not GluN2B+/- hippocampi. Sag ratio was increased in Aβ-treated GluN2B+/--mice. Firing frequency was unchanged in wt, GluN2A+/-, and GluN2B+/-hippocampi after Aβ-treatment. Effects were not significantly different from responses detected under the same conditions in wt littermates, however. LTP that lasted for over 2 h in wt hippocampal slices was significantly reduced in GluN2A+/- and was impaired for 15 min in GluN2B+/--hippocampi compared to wt littermates. Furthermore, LTP (>2 h) was significantly impaired in Aβ-treated hippocampi of wt littermates compared to wt treated with control peptide. LTP induced in Aβ-treated GluN2A+/- and GluN2B+/--hippocampi was equivalent to LTP in control peptide-treated transgenic and Aβ-treated wt animals. Taken together, our data indicate that knockdown of GluN2A subunits subtly alters membrane properties of hippocampal neurons and reduces the magnitude of LTP. GluN2B knockdown reduces the early phase of LTP but leaves later phases intact. Aβ (1-42)-treatment slightly exacerbates changes in action potential properties in GluN2A+/--mice. However, the vulnerability of the aging hippocampus to Aβ-mediated impairments of LTP is not mediated by GluN2A or GluN2B-containing NMDAR.
Collapse
|
26
|
Zhao K, Li Y, Lai H, Niu R, Li H, He S, Su Z, Gui Y, Ren L, Yang X, Zhou L. Alterations in HCN1 expression and distribution during epileptogenesis in rats. Epilepsy Res 2024; 202:107355. [PMID: 38555654 DOI: 10.1016/j.eplepsyres.2024.107355] [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: 11/30/2023] [Revised: 03/10/2024] [Accepted: 03/21/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND The hyperpolarization-activated cyclic nucleotide-gated cation channel (HCN1) is predominantly located in key regions associated with epilepsy, such as the neocortex and hippocampus. Under normal physiological conditions, HCN1 plays a crucial role in the excitatory and inhibitory regulation of neuronal networks. In temporal lobe epilepsy, the expression of HCN1 is decreased in the hippocampi of both animal models and patients. However, whether HCN1 expression changes during epileptogenesis preceding spontaneous seizures remains unclear. OBJECTIVE The aim of this study was to determine whether the expression of HCN1 is altered during the epileptic prodromal phase, thereby providing evidence for its role in epileptogenesis. METHODS We utilized a cobalt wire-induced rat epilepsy model to observe changes in HCN1 during epileptogenesis and epilepsy. Additionally, we also compared HCN1 alterations in epileptogenic tissues between cobalt wire- and pilocarpine-induced epilepsy rat models. Long-term video EEG recordings were used to confirm seizures development. Transcriptional changes, translation, and distribution of HCN1 were assessed using high-throughput transcriptome sequencing, total protein extraction, membrane and cytoplasmic protein fractionation, western blotting, immunohistochemistry, and immunofluorescence techniques. RESULTS In the cobalt wire-induced rat epilepsy model during the epileptogenesis phase, total HCN1 mRNA and protein levels were downregulated. Specifically, the membrane expression of HCN1 was decreased, whereas cytoplasmic HCN1 expression showed no significant change. The distribution of HCN1 in the distal dendrites of neurons decreased. During the epilepsy period, similar HCN1 alterations were observed in the neocortex of rats with cobalt wire-induced epilepsy and hippocampus of rats with lithium pilocarpine-induced epilepsy, including downregulation of mRNA levels, decreased total protein expression, decreased membrane expression, and decreased distal dendrite expression. CONCLUSIONS Alterations in HCN1 expression and distribution are involved in epileptogenesis beyond their association with seizure occurrence. Similarities in HCN1 alterations observed in epileptogenesis-related tissues from different models suggest a shared pathophysiological pathway in epileptogenesis involving HCN1 dysregulation. Therefore, the upregulation of HCN1 expression in neurons, maintenance of the HCN1 membrane, and distal dendrite distribution in neurons may represent promising disease-modifying strategies in epilepsy.
Collapse
Affiliation(s)
- Ke Zhao
- Department of Neurology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China; Guangzhou National Laboratory, Guangzhou, China; Department of Neurology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Yinchao Li
- Department of Neurology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | | | - Ruili Niu
- Guangzhou National Laboratory, Guangzhou, China
| | - Huifeng Li
- Guangzhou National Laboratory, Guangzhou, China
| | - Shipei He
- Guangzhou National Laboratory, Guangzhou, China
| | - Zhengwei Su
- Department of Neurology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yue Gui
- Guangzhou National Laboratory, Guangzhou, China
| | - Lijie Ren
- Department of Neurology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China.
| | | | - Liemin Zhou
- Department of Neurology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
27
|
Espinosa F, Pop IV, Lai HC. Electrophysiological Properties of Proprioception-Related Neurons in the Intermediate Thoracolumbar Spinal Cord. eNeuro 2024; 11:ENEURO.0331-23.2024. [PMID: 38627062 PMCID: PMC11055654 DOI: 10.1523/eneuro.0331-23.2024] [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: 08/29/2023] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Proprioception, the sense of limb and body position, is required to produce accurate and precise movements. Proprioceptive sensory neurons transmit muscle length and tension information to the spinal cord. The function of excitatory neurons in the intermediate spinal cord, which receive this proprioceptive information, remains poorly understood. Using genetic labeling strategies and patch-clamp techniques in acute spinal cord preparations in mice, we set out to uncover how two sets of spinal neurons, Clarke's column (CC) and Atoh1-lineage neurons, respond to electrical activity and how their inputs are organized. Both sets of neurons are located in close proximity in laminae V-VII of the thoracolumbar spinal cord and have been described to receive proprioceptive signals. We find that a majority of CC neurons have a tonic-firing type and express a distinctive hyperpolarization-activated current (Ih). Atoh1-lineage neurons, which cluster into two spatially distinct populations, are mostly a fading-firing type and display similar electrophysiological properties to each other, possibly due to their common developmental lineage. Finally, we find that CC neurons respond to stimulation of lumbar dorsal roots, consistent with prior knowledge that CC neurons receive hindlimb proprioceptive information. In contrast, using a combination of electrical stimulation, optogenetic stimulation, and transsynaptic rabies virus tracing, we find that Atoh1-lineage neurons receive heterogeneous, predominantly local thoracic inputs that include parvalbumin-lineage sensory afferents and local interneuron presynaptic inputs. Altogether, we find that CC and Atoh1-lineage neurons have distinct membrane properties and sensory input organization, representing different subcircuit modes of proprioceptive information processing.
Collapse
Affiliation(s)
- Felipe Espinosa
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, Texas 75390
| | - Iliodora V Pop
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, Texas 75390
| | - Helen C Lai
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
28
|
Guan L, Gu H, Zhang X. Dynamics of antiphase bursting modulated by the inhibitory synaptic and hyperpolarization-activated cation currents. Front Comput Neurosci 2024; 18:1303925. [PMID: 38404510 PMCID: PMC10884300 DOI: 10.3389/fncom.2024.1303925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/29/2024] [Indexed: 02/27/2024] Open
Abstract
Antiphase bursting related to the rhythmic motor behavior exhibits complex dynamics modulated by the inhibitory synaptic current (Isyn), especially in the presence of the hyperpolarization-activated cation current (Ih). In the present paper, the dynamics of antiphase bursting modulated by the Ih and Isyn is studied in three aspects with a theoretical model. Firstly, the Isyn and the slow Ih with strong strength are the identified to be the necessary conditions for the antiphase bursting. The dependence of the antiphase bursting on the two currents is different for low (escape mode) and high (release mode) threshold voltages (Vth) of the inhibitory synapse. Secondly, more detailed co-regulations of the two currents to induce opposite changes of the bursting period are obtained. For the escape mode, increase of the Ih induces elevated membrane potential of the silence inhibited by a strong Isyn and shortened silence duration to go beyond Vth, resulting in reduced bursting period. For the release mode, increase of the Ih induces elevated tough value of the former part of the burst modulated by a nearly zero Isyn and lengthen burst duration to fall below Vth, resulting in prolonged bursting period. Finally, the fast-slow dynamics of the antiphase bursting are acquired. Using one-and two-parameter bifurcations of the fast subsystem of a single neuron, the burst of the antiphase bursting is related to the stable limit cycle, and the silence modulated by a strong Isyn to the stable equilibrium to a certain extent. The Ih mainly modulates the dynamics within the burst and quiescent state. Furthermore, with the fast subsystem of the coupled neurons, the silence is associated with the unstable equilibrium point. The results present theoretical explanations to the changes in the bursting period and fast-slow dynamics of the antiphase bursting modulated by the Isyn and Ih, which is helpful for understanding the antiphase bursting and modulating rhythmic motor patterns.
Collapse
Affiliation(s)
- Linan Guan
- School of Mathematics and Statistics, North China University of Water Resources and Electric Power, Zhengzhou, China
| | - Huaguang Gu
- School of Aerospace Engineering and Applied Mechanics, Tongji University, Shanghai, China
| | - Xinjing Zhang
- School of Mathematics and Statistics, North China University of Water Resources and Electric Power, Zhengzhou, China
| |
Collapse
|
29
|
Ujfalusi-Pozsonyi K, Bódis E, Nyitrai M, Kengyel A, Telek E, Pécsi I, Fekete Z, Varnyuné Kis-Bicskei N, Mas C, Moussaoui D, Pernot P, Tully MD, Weik M, Schirò G, Kapetanaki SM, Lukács A. ATP-dependent conformational dynamics in a photoactivated adenylate cyclase revealed by fluorescence spectroscopy and small-angle X-ray scattering. Commun Biol 2024; 7:147. [PMID: 38307988 PMCID: PMC10837130 DOI: 10.1038/s42003-024-05842-1] [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: 06/17/2023] [Accepted: 01/22/2024] [Indexed: 02/04/2024] Open
Abstract
Structural insights into the photoactivated adenylate cyclases can be used to develop new ways of controlling cellular cyclic adenosine monophosphate (cAMP) levels for optogenetic and other applications. In this work, we use an integrative approach that combines biophysical and structural biology methods to provide insight on the interaction of adenosine triphosphate (ATP) with the dark-adapted state of the photoactivated adenylate cyclase from the cyanobacterium Oscillatoria acuminata (OaPAC). A moderate affinity of the nucleotide for the enzyme was calculated and the thermodynamic parameters of the interaction have been obtained. Stopped-flow fluorescence spectroscopy and small-angle solution scattering have revealed significant conformational changes in the enzyme, presumably in the adenylate cyclase (AC) domain during the allosteric mechanism of ATP binding to OaPAC with small and large-scale movements observed to the best of our knowledge for the first time in the enzyme in solution upon ATP binding. These results are in line with previously reported drastic conformational changes taking place in several class III AC domains upon nucleotide binding.
Collapse
Affiliation(s)
- K Ujfalusi-Pozsonyi
- Department of Biophysics, Medical School, University of Pécs, 7624, Pécs, Hungary
| | - E Bódis
- Department of Biophysics, Medical School, University of Pécs, 7624, Pécs, Hungary
| | - M Nyitrai
- Department of Biophysics, Medical School, University of Pécs, 7624, Pécs, Hungary
| | - A Kengyel
- Department of Biophysics, Medical School, University of Pécs, 7624, Pécs, Hungary
| | - E Telek
- Department of Biophysics, Medical School, University of Pécs, 7624, Pécs, Hungary
| | - I Pécsi
- Department of Biophysics, Medical School, University of Pécs, 7624, Pécs, Hungary
| | - Z Fekete
- Department of Biophysics, Medical School, University of Pécs, 7624, Pécs, Hungary
| | | | - C Mas
- Univ. Grenoble Alpes, CNRS, CEA, EMBL, ISBG, F-38000, Grenoble, France
| | - D Moussaoui
- European Synchrotron Radiation Facility (ESRF), Grenoble, France
| | - P Pernot
- European Synchrotron Radiation Facility (ESRF), Grenoble, France
| | - M D Tully
- European Synchrotron Radiation Facility (ESRF), Grenoble, France
| | - M Weik
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - G Schirò
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - S M Kapetanaki
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CEA, CNRS, Grenoble, France.
- Department of Biophysics, Medical School, University of Pécs, 7624, Pécs, Hungary.
| | - A Lukács
- Department of Biophysics, Medical School, University of Pécs, 7624, Pécs, Hungary.
| |
Collapse
|
30
|
Kazmierska-Grebowska P, Jankowski MM, MacIver MB. Missing Puzzle Pieces in Dementia Research: HCN Channels and Theta Oscillations. Aging Dis 2024; 15:22-42. [PMID: 37450922 PMCID: PMC10796085 DOI: 10.14336/ad.2023.0607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/07/2023] [Indexed: 07/18/2023] Open
Abstract
Increasing evidence indicates a role of hyperpolarization activated cation (HCN) channels in controlling the resting membrane potential, pacemaker activity, memory formation, sleep, and arousal. Their disfunction may be associated with the development of epilepsy and age-related memory decline. Neuronal hyperexcitability involved in epileptogenesis and EEG desynchronization occur in the course of dementia in human Alzheimer's Disease (AD) and animal models, nevertheless the underlying ionic and cellular mechanisms of these effects are not well understood. Some suggest that theta rhythms involved in memory formation could be used as a marker of memory disturbances in the course of neurogenerative diseases, including AD. This review focusses on the interplay between hyperpolarization HCN channels, theta oscillations, memory formation and their role(s) in dementias, including AD. While individually, each of these factors have been linked to each other with strong supportive evidence, we hope here to expand this linkage to a more inclusive picture. Thus, HCN channels could provide a molecular target for developing new therapeutic agents for preventing and/or treating dementia.
Collapse
Affiliation(s)
| | - Maciej M. Jankowski
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.
- BioTechMed Center, Multimedia Systems Department, Faculty of Electronics, Telecommunications, and Informatics, Gdansk University of Technology, Gdansk, Poland.Telecommunications and Informatics, Gdansk University of Technology, Gdansk, Poland.
| | - M. Bruce MacIver
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of of Medicine, Stanford University, CA, USA.
| |
Collapse
|
31
|
Wang X, Wang Q, Song M, Wang Y, Shen X, Sun Y, Guo C, Geng P, Ma C, Jin X. Chronic but not acute nicotine treatment ameliorates acute inflammation-induced working memory impairment by increasing CRTC1 and HCN2 in adult male mice. CNS Neurosci Ther 2024; 30:e14627. [PMID: 38353058 PMCID: PMC10865150 DOI: 10.1111/cns.14627] [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: 08/05/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Systemic inflammation in which lipopolysaccharide (LPS) is released into circulation can cause cognitive dysfunction and we have previously shown that LPS impaired working memory (WM) which refers to the ability to guide incoming behavior by retrieving recently acquired information. However, the mechanism is not very clear, and currently, there is no approved strategy to improve inflammation-induced WM deficit. Notably, epidemiological studies have demonstrated a lower occurrence rate of inflammatory-related diseases in smoking patients, suggesting that inflammation-induced WM impairment may be improved by nicotine treatment. Here, our object is to investigate the effect and potential mechanisms of acute and chronic nicotine treatment on LPS-produced WM deficiency. METHODS Delayed alternation T-maze task (DAT) was applied for evaluating WM which includes both the short-term information storage and the ability to correct errors in adult male mice. Immunofluorescence staining and immunoblotting were used for assessing the levels and distribution of CREB-regulated transcription coactivator 1 (CRTC1) and hyperpolarization-activated cation channels 2 (HCN2) in the medial prefrontal cortex (mPFC) and hippocampus. Quantitative PCR and ELISA were employed for analyzing the mRNA and protein levels of TNF-α and IL-1β. RESULTS Our results revealed that administration of LPS (i.p.) at a dose of 0.5 mg/kg significantly produced WM impairment in the DAT task accompanied by an increase in IL-1β and TNF-α expression in the mPFC. Moreover, intra-mPFC infusion of IL-1Ra, an IL-1 antagonist, markedly alleviated LPS-induced WM deficiency. More important, chronic (2 weeks) but not acute nicotine (0.2 mg/kg, subcutaneous) treatment significantly alleviated LPS-induced WM deficiency by upregulating CRTC1 and HCN2. Of note, intra-mPFC infusion of HCN blocker ZD7288 produced significant WM deficiency. CONCLUSIONS In summary, in this study, we show that chronic nicotine treatment ameliorates acute inflammation-induced working memory deficiency by increasing CRTC1 and HCN2 in adult male mice.
Collapse
Affiliation(s)
- Xiaona Wang
- School of Life ScienceNanchang UniversityNanchangChina
- Institute of Biomedical Innovation, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Qian Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Beijing Key Laboratory of Cancer Invasion and Metastasis ResearchCapital Medical UniversityBeijingChina
| | - Min Song
- Institute of NeuroscienceThe Second The Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yihui Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Beijing Key Laboratory of Cancer Invasion and Metastasis ResearchCapital Medical UniversityBeijingChina
| | - Xianzhi Shen
- Institute of NeuroscienceThe Second The Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yanyun Sun
- Institute of NeuroscienceThe Second The Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Chun Guo
- School of BiosciencesUniversity of SheffieldSheffieldUK
| | - Panpan Geng
- Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Beijing Key Laboratory of Cancer Invasion and Metastasis ResearchCapital Medical UniversityBeijingChina
| | - Chaolin Ma
- School of Life ScienceNanchang UniversityNanchangChina
- Institute of Biomedical Innovation, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Xinchun Jin
- Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Beijing Key Laboratory of Cancer Invasion and Metastasis ResearchCapital Medical UniversityBeijingChina
- Institute of NeuroscienceThe Second The Second Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
32
|
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
|
33
|
Stengl M, Schneider AC. Contribution of membrane-associated oscillators to biological timing at different timescales. Front Physiol 2024; 14:1243455. [PMID: 38264332 PMCID: PMC10803594 DOI: 10.3389/fphys.2023.1243455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 12/12/2023] [Indexed: 01/25/2024] Open
Abstract
Environmental rhythms such as the daily light-dark cycle selected for endogenous clocks. These clocks predict regular environmental changes and provide the basis for well-timed adaptive homeostasis in physiology and behavior of organisms. Endogenous clocks are oscillators that are based on positive feedforward and negative feedback loops. They generate stable rhythms even under constant conditions. Since even weak interactions between oscillators allow for autonomous synchronization, coupling/synchronization of oscillators provides the basis of self-organized physiological timing. Amongst the most thoroughly researched clocks are the endogenous circadian clock neurons in mammals and insects. They comprise nuclear clockworks of transcriptional/translational feedback loops (TTFL) that generate ∼24 h rhythms in clock gene expression entrained to the environmental day-night cycle. It is generally assumed that this TTFL clockwork drives all circadian oscillations within and between clock cells, being the basis of any circadian rhythm in physiology and behavior of organisms. Instead of the current gene-based hierarchical clock model we provide here a systems view of timing. We suggest that a coupled system of autonomous TTFL and posttranslational feedback loop (PTFL) oscillators/clocks that run at multiple timescales governs adaptive, dynamic homeostasis of physiology and behavior. We focus on mammalian and insect neurons as endogenous oscillators at multiple timescales. We suggest that neuronal plasma membrane-associated signalosomes constitute specific autonomous PTFL clocks that generate localized but interlinked oscillations of membrane potential and intracellular messengers with specific endogenous frequencies. In each clock neuron multiscale interactions of TTFL and PTFL oscillators/clocks form a temporally structured oscillatory network with a common complex frequency-band comprising superimposed multiscale oscillations. Coupling between oscillator/clock neurons provides the next level of complexity of an oscillatory network. This systemic dynamic network of molecular and cellular oscillators/clocks is suggested to form the basis of any physiological homeostasis that cycles through dynamic homeostatic setpoints with a characteristic frequency-band as hallmark. We propose that mechanisms of homeostatic plasticity maintain the stability of these dynamic setpoints, whereas Hebbian plasticity enables switching between setpoints via coupling factors, like biogenic amines and/or neuropeptides. They reprogram the network to a new common frequency, a new dynamic setpoint. Our novel hypothesis is up for experimental challenge.
Collapse
Affiliation(s)
- Monika Stengl
- Department of Biology, Animal Physiology/Neuroethology, University of Kassel, Kassel, Germany
| | | |
Collapse
|
34
|
Zhu M, Lu J, Li X, An Y, Li B, Liu W. Ivabradine Alleviates Experimental Autoimmune Myocarditis-Mediated Myocardial Injury. Int Heart J 2024; 65:109-118. [PMID: 38296563 DOI: 10.1536/ihj.23-330] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Ivabradine (IVA) reduces heart rate by inhibiting hyperpolarization-activated cyclic nucleotide-gated channels (HCNs), which play a role in the promotion of pacemaker activity in cardiac sinoatrial node cells. HCNs are highly expressed in neural and myocardial tissues and are involved in the modulation of inflammatory neuropathic pain. However, whether IVA exerts any effect on myocardial inflammation in the pathogenesis of heart failure is unclear. We employed single-cell RNA sequencing (scRNA-seq) in porcine cardiac myosin-induced experimental autoimmune myocarditis rat model to determine the effects and mechanisms of IVA. Lewis rats (n = 32) were randomly divided into the normal, control, high-dose-IVA, and low-dose-IVA groups. Heart rate and blood pressure were measured on days 0 and 21, respectively. Echocardiography was performed on day 22, and inflammation of the myocardium was evaluated via histopathological examination. Western blot was employed to detect the expression of HCN1-4, MinK-related protein 1 (MiRP1), matrix metalloproteinase 2 (MMP-2), MMP-9, and transforming growth factor-β (TGF-β). Furthermore, enzyme-linked immunosorbent assay was performed to measure serum IL-1, IL-6, and TNF-α. The relative mRNA levels of collagen I, collagen III, and α-smooth muscle actin (α-SMA) were determined via qRT-PCR. We found that IVA reduced the total number of cells infiltrated into the myocardium, particularly in the subset of fibroblasts, endocardia, and monocytes. IVA administration ameliorated cardiac inflammation and reduced collagen production. Results of the echocardiography indicated that left ventricular internal diameter at end-systole LVIDs increased whereas left ventricular ejection fraction and left ventricular fractional shortening decreased in the control group. IVA improved cardiac performance. The expression of HCN4 and MiRP1 protein and the level of serum IL-1, IL-6, and TNF-α were decreased by IVA treatment. In conclusion, HCNs and the helper proteins were increased in the profile of myocardial inflammation. HCNs may be involved in the regulation of myocardial inflammation by inhibiting immune cell infiltration. Our findings can contribute to the development of IVA-based combination therapies for the future treatment of cardiac inflammation and heart failure.
Collapse
Affiliation(s)
- Manlin Zhu
- Department of Cardiology, Fourth Affiliated Hospital, Harbin Medical University
| | - Jingjing Lu
- Department of Cardiology, Fourth Affiliated Hospital, Harbin Medical University
| | - Xiaomin Li
- Department of Cardiology, Fourth Affiliated Hospital, Harbin Medical University
| | - Yongqiang An
- Department of Cardiology, First Affiliated Hospital, Hebei Medical University
| | | | - Wei Liu
- Department of Cardiology, Fourth Affiliated Hospital, Harbin Medical University
| |
Collapse
|
35
|
Mӓnnikkӧ R, Kullmann DM. Structure-function and pharmacologic aspects of ion channels relevant to neurologic channelopathies. HANDBOOK OF CLINICAL NEUROLOGY 2024; 203:1-23. [PMID: 39174242 DOI: 10.1016/b978-0-323-90820-7.00009-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Ion channels are membrane proteins that allow the passage of ions across the membrane. They characteristically contain a pore where the selectivity of certain ion species is determined and gates that open and close the pore are found. The pore is often connected to additional domains or subunits that regulate its function. Channels are grouped into families based on their selectivity for specific ions and the stimuli that control channel opening and closing, such as voltage or ligands. Ion channels are fundamental to the electrical properties of excitable tissues. Dysfunction of channels can lead to abnormal electrical signaling of neurons and muscle cells, accompanied by clinical manifestations, known as channelopathies. Many naturally occurring toxins target ion channels and affect excitable cells where the channels are expressed. Furthermore, ion channels, as membrane proteins and key regulators of a number of physiologic functions, are an important target for drugs in clinical use. In this chapter, we give a general overview of the classification, genetics and structure-function features of the main ion channel families, and address some pharmacologic aspects relevant to neurologic channelopathies.
Collapse
Affiliation(s)
- Roope Mӓnnikkӧ
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.
| | - Dimitri M Kullmann
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.
| |
Collapse
|
36
|
Alaee E, Pachenari N, Khani F, Semnanian S, Shojaei A, Azizi H. Enhancement of neuronal excitability in the medial prefrontal cortex following prenatal morphine exposure. Brain Res Bull 2023; 204:110803. [PMID: 37913849 DOI: 10.1016/j.brainresbull.2023.110803] [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: 09/28/2023] [Revised: 10/26/2023] [Accepted: 10/28/2023] [Indexed: 11/03/2023]
Abstract
The clinical use and abuse of opioids during human pregnancy have been widely reported. Several studies have demonstrated that opioids cross the placenta in rats during late gestation, and prenatal morphine exposure has been shown to have negative outcomes in cognitive function. The medial prefrontal cortex (mPFC) is believed to play a crucial role in cognitive processes, motivation, and emotion, integrating neural information from several brain areas and sending converted information to other structures. Dysfunctions in this area have been observed in numerous psychiatric and neurological disorders, including addiction. This current study aimed to compare the electrophysiological properties of mPFC neurons in rat offspring prenatally exposed to morphine. Pregnant rats were injected with morphine or saline twice a day from gestational days 11-18. Whole-cell patch-clamp recordings were performed in male offspring on postnatal days 14-18. All recordings were obtained in current-clamp configuration from mPFC pyramidal neurons to assess their electrophysiological properties. The results revealed that prenatal exposure to morphine shifted the resting membrane potential (RMP) to less negative voltages and increased input resistance and duration of action potentials. However, the amplitude, rise slope, and afterhyperpolarization (AHP) amplitude of the first elicited action potentials were significantly decreased in rats prenatally exposed to morphine. Moreover, the sag voltage ratio was significantly decreased in the prenatal morphine group. Our results suggest that the changes observed in the electrophysiological properties of mPFC neurons indicate an elevation in neuronal excitability following prenatal exposure to morphine.
Collapse
Affiliation(s)
- Elham Alaee
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Narges Pachenari
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Fatemeh Khani
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeed Semnanian
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Amir Shojaei
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Hossein Azizi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
37
|
Handlin LJ, Dai G. Direct regulation of the voltage sensor of HCN channels by membrane lipid compartmentalization. Nat Commun 2023; 14:6595. [PMID: 37852983 PMCID: PMC10584925 DOI: 10.1038/s41467-023-42363-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/09/2023] [Indexed: 10/20/2023] Open
Abstract
Ion channels function within a membrane environment characterized by dynamic lipid compartmentalization. Limited knowledge exists regarding the response of voltage-gated ion channels to transmembrane potential within distinct membrane compartments. By leveraging fluorescence lifetime imaging microscopy (FLIM) and Förster resonance energy transfer (FRET), we visualized the localization of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels in membrane domains. HCN4 exhibits a greater propensity for incorporation into ordered lipid domains compared to HCN1. To investigate the conformational changes of the S4 helix voltage sensor of HCN channels, we used dual stop-codon suppression to incorporate different noncanonical amino acids, orthogonal click chemistry for site-specific fluorescence labeling, and transition metal FLIM-FRET. Remarkably, altered FRET levels were observed between VSD sites within HCN channels upon disruption of membrane domains. We propose that the voltage-sensor rearrangements, directly influenced by membrane lipid domains, can explain the heightened activity of pacemaker HCN channels when localized in cholesterol-poor, disordered lipid domains, leading to membrane hyperexcitability and diseases.
Collapse
Affiliation(s)
- Lucas J Handlin
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, 1100 South Grand Blvd., St. Louis, MO, 63104, USA
| | - Gucan Dai
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, 1100 South Grand Blvd., St. Louis, MO, 63104, USA.
| |
Collapse
|
38
|
Chartrand T, Dalley R, Close J, Goriounova NA, Lee BR, Mann R, Miller JA, Molnar G, Mukora A, Alfiler L, Baker K, Bakken TE, Berg J, Bertagnolli D, Braun T, Brouner K, Casper T, Csajbok EA, Dee N, Egdorf T, Enstrom R, Galakhova AA, Gary A, Gelfand E, Goldy J, Hadley K, Heistek TS, Hill D, Jorstad N, Kim L, Kocsis AK, Kruse L, Kunst M, Leon G, Long B, Mallory M, McGraw M, McMillen D, Melief EJ, Mihut N, Ng L, Nyhus J, Oláh G, Ozsvár A, Omstead V, Peterfi Z, Pom A, Potekhina L, Rajanbabu R, Rozsa M, Ruiz A, Sandle J, Sunkin SM, Szots I, Tieu M, Toth M, Trinh J, Vargas S, Vumbaco D, Williams G, Wilson J, Yao Z, Barzo P, Cobbs C, Ellenbogen RG, Esposito L, Ferreira M, Gouwens NW, Grannan B, Gwinn RP, Hauptman JS, Jarsky T, Keene CD, Ko AL, Koch C, Ojemann JG, Patel A, Ruzevick J, Silberberg DL, Smith K, Sorensen SA, Tasic B, Ting JT, Waters J, de Kock CP, Mansvelder HD, Tamas G, Zeng H, Kalmbach B, Lein ES. Morphoelectric and transcriptomic divergence of the layer 1 interneuron repertoire in human versus mouse neocortex. Science 2023; 382:eadf0805. [PMID: 37824667 PMCID: PMC11864503 DOI: 10.1126/science.adf0805] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 09/09/2023] [Indexed: 10/14/2023]
Abstract
Neocortical layer 1 (L1) is a site of convergence between pyramidal-neuron dendrites and feedback axons where local inhibitory signaling can profoundly shape cortical processing. Evolutionary expansion of human neocortex is marked by distinctive pyramidal neurons with extensive L1 branching, but whether L1 interneurons are similarly diverse is underexplored. Using Patch-seq recordings from human neurosurgical tissue, we identified four transcriptomic subclasses with mouse L1 homologs, along with distinct subtypes and types unmatched in mouse L1. Subclass and subtype comparisons showed stronger transcriptomic differences in human L1 and were correlated with strong morphoelectric variability along dimensions distinct from mouse L1 variability. Accompanied by greater layer thickness and other cytoarchitecture changes, these findings suggest that L1 has diverged in evolution, reflecting the demands of regulating the expanded human neocortical circuit.
Collapse
Affiliation(s)
| | | | | | - Natalia A. Goriounova
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit; Amsterdam, The Netherlands
| | | | - Rusty Mann
- Allen Institute for Brain Science; Seattle, USA
| | | | - Gabor Molnar
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | | | | | | | | | - Jim Berg
- Allen Institute for Brain Science; Seattle, USA
| | | | | | | | | | - Eva Adrienn Csajbok
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | - Nick Dee
- Allen Institute for Brain Science; Seattle, USA
| | - Tom Egdorf
- Allen Institute for Brain Science; Seattle, USA
| | | | - Anna A. Galakhova
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit; Amsterdam, The Netherlands
| | - Amanda Gary
- Allen Institute for Brain Science; Seattle, USA
| | | | - Jeff Goldy
- Allen Institute for Brain Science; Seattle, USA
| | | | - Tim S. Heistek
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit; Amsterdam, The Netherlands
| | - DiJon Hill
- Allen Institute for Brain Science; Seattle, USA
| | - Nik Jorstad
- Allen Institute for Brain Science; Seattle, USA
| | - Lisa Kim
- Allen Institute for Brain Science; Seattle, USA
| | - Agnes Katalin Kocsis
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | | | | | | | - Brian Long
- Allen Institute for Brain Science; Seattle, USA
| | | | | | | | - Erica J. Melief
- Department of Laboratory Medicine and Pathology, University of Washington; Seattle, USA
| | - Norbert Mihut
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | - Lindsay Ng
- Allen Institute for Brain Science; Seattle, USA
| | - Julie Nyhus
- Allen Institute for Brain Science; Seattle, USA
| | - Gáspár Oláh
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | - Attila Ozsvár
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | | | - Zoltan Peterfi
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | - Alice Pom
- Allen Institute for Brain Science; Seattle, USA
| | | | | | - Marton Rozsa
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | | | - Joanna Sandle
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | | | - Ildiko Szots
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | | | - Martin Toth
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | | | - Sara Vargas
- Allen Institute for Brain Science; Seattle, USA
| | | | | | | | - Zizhen Yao
- Allen Institute for Brain Science; Seattle, USA
| | - Pal Barzo
- Department of Neurosurgery, University of Szeged; Szeged, Hungary
| | | | | | | | - Manuel Ferreira
- Department of Neurological Surgery, University of Washington; Seattle USA
| | | | - Benjamin Grannan
- Department of Neurological Surgery, University of Washington; Seattle USA
| | | | - Jason S. Hauptman
- Department of Neurological Surgery, University of Washington; Seattle USA
| | - Tim Jarsky
- Allen Institute for Brain Science; Seattle, USA
| | - C. Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington; Seattle, USA
| | - Andrew L. Ko
- Department of Neurological Surgery, University of Washington; Seattle USA
| | | | - Jeffrey G. Ojemann
- Department of Neurological Surgery, University of Washington; Seattle USA
| | - Anoop Patel
- Department of Neurological Surgery, University of Washington; Seattle USA
| | - Jacob Ruzevick
- Department of Neurological Surgery, University of Washington; Seattle USA
| | | | | | | | | | - Jonathan T. Ting
- Allen Institute for Brain Science; Seattle, USA
- Department of Physiology and Biophysics, University of Washington; Seattle, USA
- Washington National Primate Research Center, University of Washington; Seattle, USA
| | - Jack Waters
- Allen Institute for Brain Science; Seattle, USA
| | - Christiaan P.J. de Kock
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit; Amsterdam, The Netherlands
| | - Huib D. Mansvelder
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit; Amsterdam, The Netherlands
| | - Gabor Tamas
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | | | - Brian Kalmbach
- Allen Institute for Brain Science; Seattle, USA
- Department of Physiology and Biophysics, University of Washington; Seattle, USA
| | - Ed S. Lein
- Allen Institute for Brain Science; Seattle, USA
- Department of Neurological Surgery, University of Washington; Seattle USA
| |
Collapse
|
39
|
Krumbach JH, Bauer D, Sharifzadeh AS, Saponaro A, Lautenschläger R, Lange K, Rauh O, DiFrancesco D, Moroni A, Thiel G, Hamacher K. Alkali metal cations modulate the geometry of different binding sites in HCN4 selectivity filter for permeation or block. J Gen Physiol 2023; 155:e202313364. [PMID: 37523352 PMCID: PMC10386491 DOI: 10.1085/jgp.202313364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 06/03/2023] [Accepted: 07/13/2023] [Indexed: 08/02/2023] Open
Abstract
Hyperpolarization-activated cyclic-nucleotide gated (HCN) channels are important for timing biological processes like heartbeat and neuronal firing. Their weak cation selectivity is determined by a filter domain with only two binding sites for K+ and one for Na+. The latter acts as a weak blocker, which is released in combination with a dynamic widening of the filter by K+ ions, giving rise to a mixed K+/Na+ current. Here, we apply molecular dynamics simulations to systematically investigate the interactions of five alkali metal cations with the filter of the open HCN4 pore. Simulations recapitulate experimental data like a low Li+ permeability, considerable Rb+ conductance, a block by Cs+ as well as a punch through of Cs+ ions at high negative voltages. Differential binding of the cation species in specific filter sites is associated with structural adaptations of filter residues. This gives rise to ion coordination by a cation-characteristic number of oxygen atoms from the filter backbone and solvent. This ion/protein interplay prevents Li+, but not Na+, from entry into and further passage through the filter. The site equivalent to S3 in K+ channels emerges as a preferential binding and presumably blocking site for Cs+. Collectively, the data suggest that the weak cation selectivity of HCN channels and their block by Cs+ are determined by restrained cation-generated rearrangements of flexible filter residues.
Collapse
Affiliation(s)
- Jan H Krumbach
- Department of Biology, Technical University of Darmstadt, Darmstadt, Germany
| | - Daniel Bauer
- Department of Biology, Technical University of Darmstadt, Darmstadt, Germany
| | | | - Andrea Saponaro
- Department of Biosciences, University of Milan, Milan, Italy
| | - Rene Lautenschläger
- Department of Biology, Technical University of Darmstadt, Darmstadt, Germany
| | - Kristina Lange
- Department of Biology, Technical University of Darmstadt, Darmstadt, Germany
| | - Oliver Rauh
- Department of Biology, Technical University of Darmstadt, Darmstadt, Germany
| | | | - Anna Moroni
- Department of Biosciences, University of Milan, Milan, Italy
| | - Gerhard Thiel
- Department of Biology, Technical University of Darmstadt, Darmstadt, Germany
| | - Kay Hamacher
- Department of Biology, Technical University of Darmstadt, Darmstadt, Germany
- Department of Physics, Technical University of Darmstadt, Darmstadt, Germany
| |
Collapse
|
40
|
Liu YC, So EC, Wu SN. Cannabidiol Modulates M-Type K + and Hyperpolarization-Activated Cation Currents. Biomedicines 2023; 11:2651. [PMID: 37893024 PMCID: PMC10604323 DOI: 10.3390/biomedicines11102651] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 10/29/2023] Open
Abstract
Cannabidiol (CBD) is a naturally occurring compound found in the Cannabis plant that is known for its potential therapeutic effects. However, its impact on membrane ionic currents remains a topic of debate. This study aimed to investigate how CBD modifies various types of ionic currents in pituitary GH3 cells. Results showed that exposure to CBD led to a concentration-dependent decrease in M-type K+ currents (IK(M)), with an IC50 of 3.6 μM, and caused the quasi-steady-state activation curve of the current to shift to a more depolarized potential with no changes in the curve's steepness. The CBD-mediated block of IK(M) was not reversed by naloxone, suggesting that it was not mediated by opioid receptors. The IK(M) elicited by pulse-train stimulation was also decreased upon exposure to CBD. The magnitude of erg-mediated K+ currents was slightly reduced by adding CBD (10 μM), while the density of voltage-gated Na+ currents elicited by a short depolarizing pulse was not affected by it. Additionally, CBD decreased the magnitude of hyperpolarization-activated cation currents (Ih) with an IC50 of 3.3 μM, and the decrease was reversed by oxaliplatin. The quasi-steady-state activation curve of Ih was shifted in the leftward direction with no changes in the slope factor of the curve. CBD also diminished the strength of voltage-dependent hysteresis on Ih elicited by upright isosceles-triangular ramp voltage. Collectively, these findings suggest that CBD's modification of ionic currents presented herein is independent of cannabinoid or opioid receptors and may exert a significant impact on the functional activities of excitable cells occurring in vitro or in vivo.
Collapse
Affiliation(s)
- Yen-Chin Liu
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan;
- Department of Anesthesiology, School of Post-Baccalaureate, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
- Department of Anesthesiology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan
| | - Edmund Cheung So
- Department of Anesthesia, An-Nan Hospital, China Medical University, Tainan 70965, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan 70101, Taiwan
- School of Medicine, National Sun-Yat Sen University College of Medicine, Kaohsiung 80424, Taiwan
- Department of Research and Education, An-Nan Hospital, China Medical University, Tainan 70965, Taiwan
| |
Collapse
|
41
|
Zheng Y, Shao S, Zhang Y, Yuan S, Xing Y, Wang J, Qi X, Cui K, Tong J, Liu F, Cui S, Wan Y, Yi M. HCN2 Channels in the Ventral Hippocampal CA1 Regulate Nociceptive Hypersensitivity in Mice. Int J Mol Sci 2023; 24:13823. [PMID: 37762124 PMCID: PMC10531460 DOI: 10.3390/ijms241813823] [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: 07/12/2023] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 09/29/2023] Open
Abstract
Chronic pain is a significant health problem worldwide. Recent evidence has suggested that the ventral hippocampus is dysfunctional in humans and rodents, with decreased neuronal excitability and connectivity with other brain regions, parallel pain chronicity, and persistent nociceptive hypersensitivity. But the molecular mechanisms underlying hippocampal modulation of pain remain poorly elucidated. In this study, we used ex vivo whole-cell patch-clamp recording, immunofluorescence staining, and behavioral tests to examine whether hyperpolarization-activated cyclic nucleotide-gated channels 2 (HCN2) in the ventral hippocampal CA1 (vCA1) were involved in regulating nociceptive perception and CFA-induced inflammatory pain in mice. Reduced sag potential and firing rate of action potentials were observed in vCA1 pyramidal neurons from CFA-injected mice. Moreover, the expression of HCN2, but not HCN1, in vCA1 decreased in mice injected with CFA. HCN2 knockdown in vCA1 pyramidal neurons induced thermal hypersensitivity, whereas overexpression of HCN2 alleviated thermal hyperalgesia induced by intraplantar injection of CFA in mice. Our findings suggest that HCN2 in the vCA1 plays an active role in pain modulation and could be a promising target for the treatment of chronic pain.
Collapse
Affiliation(s)
- Yawen Zheng
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Z.); (S.S.); (S.Y.); (Y.X.); (J.W.); (X.Q.); (K.C.); (J.T.); (F.L.); (S.C.); (Y.W.)
| | - Shan Shao
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Z.); (S.S.); (S.Y.); (Y.X.); (J.W.); (X.Q.); (K.C.); (J.T.); (F.L.); (S.C.); (Y.W.)
| | - Yu Zhang
- National Health Commission Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS) & Peking Union Medical College (PUMC), Beijing 100101, China;
| | - Shulu Yuan
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Z.); (S.S.); (S.Y.); (Y.X.); (J.W.); (X.Q.); (K.C.); (J.T.); (F.L.); (S.C.); (Y.W.)
| | - Yuanwei Xing
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Z.); (S.S.); (S.Y.); (Y.X.); (J.W.); (X.Q.); (K.C.); (J.T.); (F.L.); (S.C.); (Y.W.)
| | - Jiaxin Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Z.); (S.S.); (S.Y.); (Y.X.); (J.W.); (X.Q.); (K.C.); (J.T.); (F.L.); (S.C.); (Y.W.)
| | - Xuetao Qi
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Z.); (S.S.); (S.Y.); (Y.X.); (J.W.); (X.Q.); (K.C.); (J.T.); (F.L.); (S.C.); (Y.W.)
| | - Kun Cui
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Z.); (S.S.); (S.Y.); (Y.X.); (J.W.); (X.Q.); (K.C.); (J.T.); (F.L.); (S.C.); (Y.W.)
| | - Jifu Tong
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Z.); (S.S.); (S.Y.); (Y.X.); (J.W.); (X.Q.); (K.C.); (J.T.); (F.L.); (S.C.); (Y.W.)
| | - Fengyu Liu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Z.); (S.S.); (S.Y.); (Y.X.); (J.W.); (X.Q.); (K.C.); (J.T.); (F.L.); (S.C.); (Y.W.)
| | - Shuang Cui
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Z.); (S.S.); (S.Y.); (Y.X.); (J.W.); (X.Q.); (K.C.); (J.T.); (F.L.); (S.C.); (Y.W.)
| | - You Wan
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Z.); (S.S.); (S.Y.); (Y.X.); (J.W.); (X.Q.); (K.C.); (J.T.); (F.L.); (S.C.); (Y.W.)
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing 100101, China
| | - Ming Yi
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; (Y.Z.); (S.S.); (S.Y.); (Y.X.); (J.W.); (X.Q.); (K.C.); (J.T.); (F.L.); (S.C.); (Y.W.)
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing 100101, China
| |
Collapse
|
42
|
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
|
43
|
van de Poll Y, Cras Y, Ellender TJ. The neurophysiological basis of stress and anxiety - comparing neuronal diversity in the bed nucleus of the stria terminalis (BNST) across species. Front Cell Neurosci 2023; 17:1225758. [PMID: 37711509 PMCID: PMC10499361 DOI: 10.3389/fncel.2023.1225758] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/03/2023] [Indexed: 09/16/2023] Open
Abstract
The bed nucleus of the stria terminalis (BNST), as part of the extended amygdala, has become a region of increasing interest regarding its role in numerous human stress-related psychiatric diseases, including post-traumatic stress disorder and generalized anxiety disorder amongst others. The BNST is a sexually dimorphic and highly complex structure as already evident by its anatomy consisting of 11 to 18 distinct sub-nuclei in rodents. Located in the ventral forebrain, the BNST is anatomically and functionally connected to many other limbic structures, including the amygdala, hypothalamic nuclei, basal ganglia, and hippocampus. Given this extensive connectivity, the BNST is thought to play a central and critical role in the integration of information on hedonic-valence, mood, arousal states, processing emotional information, and in general shape motivated and stress/anxiety-related behavior. Regarding its role in regulating stress and anxiety behavior the anterolateral group of the BNST (BNSTALG) has been extensively studied and contains a wide variety of neurons that differ in their electrophysiological properties, morphology, spatial organization, neuropeptidergic content and input and output synaptic organization which shape their activity and function. In addition to this great diversity, further species-specific differences are evident on multiple levels. For example, classic studies performed in adult rat brain identified three distinct neuron types (Type I-III) based on their electrophysiological properties and ion channel expression. Whilst similar neurons have been identified in other animal species, such as mice and non-human primates such as macaques, cross-species comparisons have revealed intriguing differences such as their comparative prevalence in the BNSTALG as well as their electrophysiological and morphological properties, amongst other differences. Given this tremendous complexity on multiple levels, the comprehensive elucidation of the BNSTALG circuitry and its role in regulating stress/anxiety-related behavior is a major challenge. In the present Review we bring together and highlight the key differences in BNSTALG structure, functional connectivity, the electrophysiological and morphological properties, and neuropeptidergic profiles of BNSTALG neurons between species with the aim to facilitate future studies of this important nucleus in relation to human disease.
Collapse
Affiliation(s)
- Yana van de Poll
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Yasmin Cras
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Tommas J. Ellender
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
44
|
Burtscher V, Mount J, Cowgill J, Chang Y, Bickel K, Yuan P, Chanda B. Structural Basis for Hyperpolarization-dependent Opening of the Human HCN1 Channel. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.17.553623. [PMID: 37645882 PMCID: PMC10462129 DOI: 10.1101/2023.08.17.553623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Hyperpolarization and cyclic-nucleotide (HCN) activated ion channels play a critical role in generating self-propagating action potentials in pacemaking and rhythmic electrical circuits in the human body. Unlike most voltage-gated ion channels, the HCN channels activate upon membrane hyperpolarization, but the structural mechanisms underlying this gating behavior remain unclear. Here, we present cryo-electron microscopy structures of human HCN1 in Closed, Intermediate, and Open states. Our structures reveal that the inward motion of two gating charges past the charge transfer center (CTC) and concomitant tilting of the S5 helix drives the opening of the central pore. In the intermediate state structure, a single gating charge is positioned below the CTC and the pore appears closed, whereas in the open state structure, both charges move past CTC and the pore is fully open. Remarkably, the downward motion of the voltage sensor is accompanied by progressive unwinding of the inner end of S4 and S5 helices disrupting the tight gating interface that stabilizes the Closed state structure. This "melting" transition at the intracellular gating interface leads to a concerted iris-like displacement of S5 and S6 helices, resulting in pore opening. These findings reveal key structural features that are likely to underlie reversed voltage-dependence of HCN channels.
Collapse
Affiliation(s)
- Verena Burtscher
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jonathan Mount
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John Cowgill
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Applied Physics, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Yongchang Chang
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kathleen Bickel
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
| | - Peng Yuan
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Baron Chanda
- Department of Anesthesiology, Washington University School of Medicine, Saint Louis, MO, USA
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Neuroscience, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
45
|
Catacuzzeno L, Conti F, Franciolini F. Fifty years of gating currents and channel gating. J Gen Physiol 2023; 155:e202313380. [PMID: 37410612 PMCID: PMC10324510 DOI: 10.1085/jgp.202313380] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/12/2023] [Accepted: 06/02/2023] [Indexed: 07/08/2023] Open
Abstract
We celebrate this year the 50th anniversary of the first electrophysiological recordings of the gating currents from voltage-dependent ion channels done in 1973. This retrospective tries to illustrate the context knowledge on channel gating and the impact gating-current recording had then, and how it continued to clarify concepts, elaborate new ideas, and steer the scientific debate in these 50 years. The notion of gating particles and gating currents was first put forward by Hodgkin and Huxley in 1952 as a necessary assumption for interpreting the voltage dependence of the Na and K conductances of the action potential. 20 years later, gating currents were actually recorded, and over the following decades have represented the most direct means of tracing the movement of the gating charges and gaining insights into the mechanisms of channel gating. Most work in the early years was focused on the gating currents from the Na and K channels as found in the squid giant axon. With channel cloning and expression on heterologous systems, other channels as well as voltage-dependent enzymes were investigated. Other approaches were also introduced (cysteine mutagenesis and labeling, site-directed fluorometry, cryo-EM crystallography, and molecular dynamics [MD] modeling) to provide an integrated and coherent view of voltage-dependent gating in biological macromolecules. The layout of this retrospective reflects the past 50 years of investigations on gating currents, first addressing studies done on Na and K channels and then on other voltage-gated channels and non-channel structures. The review closes with a brief overview of how the gating-charge/voltage-sensor movements are translated into pore opening and the pathologies associated with mutations targeting the structures involved with the gating currents.
Collapse
Affiliation(s)
- Luigi Catacuzzeno
- Department of Chemistry Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Franco Conti
- Department of Physics, University of Genova, Genova, Italy
| | - Fabio Franciolini
- Department of Chemistry Biology and Biotechnology, University of Perugia, Perugia, Italy
| |
Collapse
|
46
|
Levenstein D, Okun M. Logarithmically scaled, gamma distributed neuronal spiking. J Physiol 2023; 601:3055-3069. [PMID: 36086892 PMCID: PMC10952267 DOI: 10.1113/jp282758] [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: 05/09/2022] [Accepted: 07/28/2022] [Indexed: 11/08/2022] Open
Abstract
Naturally log-scaled quantities abound in the nervous system. Distributions of these quantities have non-intuitive properties, which have implications for data analysis and the understanding of neural circuits. Here, we review the log-scaled statistics of neuronal spiking and the relevant analytical probability distributions. Recent work using log-scaling revealed that interspike intervals of forebrain neurons segregate into discrete modes reflecting spiking at different timescales and are each well-approximated by a gamma distribution. Each neuron spends most of the time in an irregular spiking 'ground state' with the longest intervals, which determines the mean firing rate of the neuron. Across the entire neuronal population, firing rates are log-scaled and well approximated by the gamma distribution, with a small number of highly active neurons and an overabundance of low rate neurons (the 'dark matter'). These results are intricately linked to a heterogeneous balanced operating regime, which confers upon neuronal circuits multiple computational advantages and has evolutionarily ancient origins.
Collapse
Affiliation(s)
- Daniel Levenstein
- Department of Neurology and NeurosurgeryMcGill UniversityMontrealQCCanada
- MilaMontréalQCCanada
| | - Michael Okun
- Department of Psychology and Neuroscience InstituteUniversity of SheffieldSheffieldUK
| |
Collapse
|
47
|
Häfele M, Kreitz S, Ludwig A, Hess A, Wank I. The impact of HCN4 channels on CNS brain networks as a new target in pain development. FRONTIERS IN NETWORK PHYSIOLOGY 2023; 3:1090502. [PMID: 37496803 PMCID: PMC10368246 DOI: 10.3389/fnetp.2023.1090502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 06/22/2023] [Indexed: 07/28/2023]
Abstract
While it is well established that the isoform 2 of the hyperpolarization-activated cyclic nucleotide-gated cation channel (HCN2) plays an important role in the development and maintenance of pain, the role of the closely related HCN4 isoform in the processing of nociceptive signals is not known. HCN4 channels are highly expressed in the thalamus, a region important for stimulus transmission and information processing. We used a brain-specific HCN4-knockout mouse line (HCN4-KO) to explore the role of HCN4 channels in acute nociceptive processing using several behavioral tests as well as a multimodal magnetic resonance imaging (MRI) approach. Functional MRI (fMRI) brain responses were measured during acute peripheral thermal stimulation complemented by resting state (RS) before and after stimulation. The data were analyzed by conventional and graph-theoretical approaches. Finally, high-resolution anatomical brain data were acquired. HCN4-KO animals showed a central thermal, but not a mechanical hypersensitivity in behavioral experiments. The open field analysis showed no significant differences in motor readouts between HCN4-KO and controls but uncovered increased anxiety in the HCN4-KO mice. Thermal stimulus-driven fMRI (s-fMRI) data revealed increased response volumes and response amplitudes for HCN4-KO, most pronounced at lower stimulation temperatures in the subcortical input, the amygdala as well as in limbic/hippocampal regions, and in the cerebellum. These findings could be cross-validated by graph-theoretical analyses. Assessment of short-term RS before and after thermal stimulation revealed that stimulation-related modulations of the functional connectivity only occurred in control animals. This was consistent with the finding that the hippocampus was found to be smaller in HCN4-KO. In summary, the deletion of HCN4 channels impacts on processing of acute nociception, which is remarkably manifested as a thermal hypersensitive phenotype. This was mediated by the key regions hypothalamus, somatosensory cortex, cerebellum and the amygdala. As consequence, HCN4-KO mice were more anxious, and their brain-wide RS functional connectivity could not be modulated by thermal nociceptive stimulation.
Collapse
Affiliation(s)
- Maximilian Häfele
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Silke Kreitz
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Ludwig
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Hess
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Institute of Neuroradiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- FAU NeW—Research Center for New Bioactive Compounds, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Isabel Wank
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
48
|
Kassab NED, Mehlfeld V, Kass J, Biel M, Schneider G, Rammes G. Xenon's Sedative Effect Is Mediated by Interaction with the Cyclic Nucleotide-Binding Domain (CNBD) of HCN2 Channels Expressed by Thalamocortical Neurons of the Ventrobasal Nucleus in Mice. Int J Mol Sci 2023; 24:ijms24108613. [PMID: 37239964 DOI: 10.3390/ijms24108613] [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: 04/18/2023] [Revised: 04/29/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
Previous studies have shown that xenon reduces hyperpolarization-activated cyclic nucleotide-gated channels type-2 (HCN2) channel-mediated current (Ih) amplitude and shifts the half-maximal activation voltage (V1/2) in thalamocortical circuits of acute brain slices to more hyperpolarized potentials. HCN2 channels are dually gated by the membrane voltage and via cyclic nucleotides binding to the cyclic nucleotide-binding domain (CNBD) on the channel. In this study, we hypothesize that xenon interferes with the HCN2 CNBD to mediate its effect. Using the transgenic mice model HCN2EA, in which the binding of cAMP to HCN2 was abolished by two amino acid mutations (R591E, T592A), we performed ex-vivo patch-clamp recordings and in-vivo open-field test to prove this hypothesis. Our data showed that xenon (1.9 mM) application to brain slices shifts the V1/2 of Ih to more hyperpolarized potentials in wild-type thalamocortical neurons (TC) (V1/2: -97.09 [-99.56--95.04] mV compared to control -85.67 [-94.47--82.10] mV; p = 0.0005). These effects were abolished in HCN2EA neurons (TC), whereby the V1/2 reached only -92.56 [-93.16- -89.68] mV with xenon compared to -90.03 [-98.99--84.59] mV in the control (p = 0.84). After application of a xenon mixture (70% xenon, 30% O2), wild-type mice activity in the open-field test decreased to 5 [2-10] while in HCN2EA mice it remained at 30 [15-42]%, (p = 0.0006). In conclusion, we show that xenon impairs HCN2 channel function by interfering with the HCN2 CNBD site and provide in-vivo evidence that this mechanism contributes to xenon-mediated hypnotic properties.
Collapse
Affiliation(s)
- Nour El Dine Kassab
- Department of Anesthesiology and Intensive Care Medicine, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Verena Mehlfeld
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-Universitñt Mnchen, 81377 Munich, Germany
| | - Jennifer Kass
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-Universitñt Mnchen, 81377 Munich, Germany
| | - Martin Biel
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-Universitñt Mnchen, 81377 Munich, Germany
| | - Gerhard Schneider
- Department of Anesthesiology and Intensive Care Medicine, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Gerhard Rammes
- Department of Anesthesiology and Intensive Care Medicine, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| |
Collapse
|
49
|
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
|
50
|
Pan Y, Pohjolainen E, Schmidpeter PAM, Vaiana AC, Nimigean CM, Grubmüller H, Scheuring S. Discrimination between cyclic nucleotides in a cyclic nucleotide-gated ion channel. Nat Struct Mol Biol 2023; 30:512-520. [PMID: 36973509 DOI: 10.1038/s41594-023-00955-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 02/24/2023] [Indexed: 03/29/2023]
Abstract
Cyclic nucleotide-gated ion channels are crucial in many physiological processes such as vision and pacemaking in the heart. SthK is a prokaryotic homolog with high sequence and structure similarities to hyperpolarization-activated and cyclic nucleotide-modulated and cyclic nucleotide-gated channels, especially at the level of the cyclic nucleotide binding domains (CNBDs). Functional measurements showed that cyclic adenosine monophosphate (cAMP) is a channel activator while cyclic guanosine monophosphate (cGMP) barely leads to pore opening. Here, using atomic force microscopy single-molecule force spectroscopy and force probe molecular dynamics simulations, we unravel quantitatively and at the atomic level how CNBDs discriminate between cyclic nucleotides. We find that cAMP binds to the SthK CNBD slightly stronger than cGMP and accesses a deep-bound state that a cGMP-bound CNBD cannot reach. We propose that the deep binding of cAMP is the discriminatory state that is essential for cAMP-dependent channel activation.
Collapse
Affiliation(s)
- Yangang Pan
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Emmi Pohjolainen
- Max Planck Institute for Multidisciplinary Sciences, Theoretical and Computational Biophysics Department, Goettingen, Germany
| | | | - Andrea C Vaiana
- Max Planck Institute for Multidisciplinary Sciences, Theoretical and Computational Biophysics Department, Goettingen, Germany
- Institute of Biophysics, National Research Council, Palermo, Italy
| | - Crina M Nimigean
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Helmut Grubmüller
- Max Planck Institute for Multidisciplinary Sciences, Theoretical and Computational Biophysics Department, Goettingen, Germany
| | - Simon Scheuring
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA.
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|