1
|
Cichon J, Joseph TT, Lu X, Wasilczuk AZ, Kelz MB, Mennerick SJ, Zorumski CF, Nagele P. Nitrous oxide activates layer 5 prefrontal neurons via SK2 channel inhibition for antidepressant effect. Nat Commun 2025; 16:2999. [PMID: 40180931 PMCID: PMC11968965 DOI: 10.1038/s41467-025-57951-y] [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: 09/23/2024] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
Nitrous oxide (N2O) induces rapid and durable antidepressant effects. The cellular and circuit mechanisms mediating this process are not known. Here we find that a single dose of inhaled N2O induces rapid and specific activation of layer V (L5) pyramidal neurons in the cingulate cortex of rodents exposed to chronic stress conditions. N2O-induced L5 activation rescues a stress-associated hypoactivity state, persists following exposure, and is necessary for its antidepressant-like activity. Although NMDA-receptor antagonism is believed to be a primary mechanism of action for N2O, L5 neurons activate even when NMDA-receptor function is attenuated through both pharmacological and genetic approaches. By examining different molecular and circuit targets, we identify N2O-induced inhibition of calcium-sensitive potassium (SK2) channels as a key molecular interaction responsible for driving specific L5 activity along with ensuing antidepressant-like effects. These results suggest that N2O-induced L5 activation is crucial for its fast antidepressant action and this effect involves novel and specific molecular actions in distinct cortical cell types.
Collapse
Affiliation(s)
- Joseph Cichon
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Thomas T Joseph
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xinguo Lu
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrzej Z Wasilczuk
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Max B Kelz
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven J Mennerick
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Charles F Zorumski
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Peter Nagele
- Department of Anesthesia and Critical Care, University of Chicago, Chicago, IL, USA
| |
Collapse
|
2
|
Kayyal H, Cruciani F, Chandran SK, Edry E, Schif-Zuck S, Koren T, Yiannakas A, Rolls A, Ariel A, Rosenblum K. Retrieval of conditioned immune response in male mice is mediated by an anterior-posterior insula circuit. Nat Neurosci 2025; 28:589-601. [PMID: 39870921 DOI: 10.1038/s41593-024-01864-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 12/05/2024] [Indexed: 01/29/2025]
Abstract
To protect the body from infections, the brain has evolved the ability to coordinate behavioral and immunological responses. The conditioned immune response (CIR) is a form of Pavlovian conditioning wherein a sensory (for example, taste) stimulus, when paired with an immunomodulatory agent, evokes aversive behavior and an anticipatory immune response after re-experiencing the taste. Although taste and its valence are represented in the anterior insular cortex and immune response in the posterior insula and although the insula is pivotal for CIRs, the precise circuitry underlying CIRs remains unknown. Here, we demonstrated that a bidirectional circuit connecting the anterior and posterior (aIC-pIC) insula mediates the CIR in male mice. Retrieving the behavioral dimension of the association requires activity of aIC-to-pIC neurons, whereas modulating the anticipatory immunological dimension requires bidirectional projections. These results illuminate a mechanism by which experience shapes interactions between sensory internal representations and the immune system. Moreover, this newly described intrainsular circuit contributes to the preservation of brain-dependent immune homeostasis.
Collapse
Affiliation(s)
- Haneen Kayyal
- Sagol Department of Neuroscience, The Integrated Brain and Behavior Center, University of Haifa, Haifa, Israel.
| | - Federica Cruciani
- Sagol Department of Neuroscience, The Integrated Brain and Behavior Center, University of Haifa, Haifa, Israel.
| | | | - Efrat Edry
- Sagol Department of Neuroscience, The Integrated Brain and Behavior Center, University of Haifa, Haifa, Israel
- Center for Gene Manipulation in the Brain, University of Haifa, Haifa, Israel
| | - Sagie Schif-Zuck
- Department of Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
- Departments of Human Biology, University of Haifa, Haifa, Israel
| | - Tamar Koren
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Adonis Yiannakas
- Sagol Department of Neuroscience, The Integrated Brain and Behavior Center, University of Haifa, Haifa, Israel
- European University of Cyprus Medical School, Frankfurt am Main, Germany
| | - Asya Rolls
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Amiram Ariel
- Department of Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
- Departments of Human Biology, University of Haifa, Haifa, Israel
| | - Kobi Rosenblum
- Sagol Department of Neuroscience, The Integrated Brain and Behavior Center, University of Haifa, Haifa, Israel.
- Center for Gene Manipulation in the Brain, University of Haifa, Haifa, Israel.
| |
Collapse
|
3
|
Qiu J, Voliotis M, Bosch MA, Li XF, Zweifel LS, Tsaneva-Atanasova K, O'Byrne KT, Rønnekleiv OK, Kelly MJ. Estradiol elicits distinct firing patterns in arcuate nucleus kisspeptin neurons of females through altering ion channel conductances. eLife 2024; 13:RP96691. [PMID: 39671233 PMCID: PMC11643640 DOI: 10.7554/elife.96691] [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] [Indexed: 12/14/2024] Open
Abstract
Hypothalamic kisspeptin (Kiss1) neurons are vital for pubertal development and reproduction. Arcuate nucleus Kiss1 (Kiss1ARH) neurons are responsible for the pulsatile release of gonadotropin-releasing hormone (GnRH). In females, the behavior of Kiss1ARH neurons, expressing Kiss1, neurokinin B (NKB), and dynorphin (Dyn), varies throughout the ovarian cycle. Studies indicate that 17β-estradiol (E2) reduces peptide expression but increases Slc17a6 (Vglut2) mRNA and glutamate neurotransmission in these neurons, suggesting a shift from peptidergic to glutamatergic signaling. To investigate this shift, we combined transcriptomics, electrophysiology, and mathematical modeling. Our results demonstrate that E2 treatment upregulates the mRNA expression of voltage-activated calcium channels, elevating the whole-cell calcium current that contributes to high-frequency burst firing. Additionally, E2 treatment decreased the mRNA levels of canonical transient receptor potential (TPRC) 5 and G protein-coupled K+ (GIRK) channels. When Trpc5 channels in Kiss1ARH neurons were deleted using CRISPR/SaCas9, the slow excitatory postsynaptic potential was eliminated. Our data enabled us to formulate a biophysically realistic mathematical model of Kiss1ARH neurons, suggesting that E2 modifies ionic conductances in these neurons, enabling the transition from high-frequency synchronous firing through NKB-driven activation of TRPC5 channels to a short bursting mode facilitating glutamate release. In a low E2 milieu, synchronous firing of Kiss1ARH neurons drives pulsatile release of GnRH, while the transition to burst firing with high, preovulatory levels of E2 would facilitate the GnRH surge through its glutamatergic synaptic connection to preoptic Kiss1 neurons.
Collapse
Affiliation(s)
- Jian Qiu
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science UniversityPortlandUnited States
| | - Margaritis Voliotis
- Department of Mathematics and Statistics, University of ExeterExeterUnited Kingdom
- Living Systems Institute, University of ExeterExeterUnited Kingdom
| | - Martha A Bosch
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science UniversityPortlandUnited States
| | - Xiao Feng Li
- Department of Women and Children’s Health, School of Life Course and Population Sciences, King’s College LondonLondonUnited Kingdom
| | - Larry S Zweifel
- Department of Psychiatry and Behavioral Sciences, University of WashingtonSeattleUnited States
- Depatment of Pharmacology, University of WashingtonSeattleUnited States
| | - Krasimira Tsaneva-Atanasova
- Department of Mathematics and Statistics, University of ExeterExeterUnited Kingdom
- Living Systems Institute, University of ExeterExeterUnited Kingdom
| | - Kevin T O'Byrne
- Department of Women and Children’s Health, School of Life Course and Population Sciences, King’s College LondonLondonUnited Kingdom
| | - Oline K Rønnekleiv
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science UniversityPortlandUnited States
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| | - Martin J Kelly
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science UniversityPortlandUnited States
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| |
Collapse
|
4
|
Pakalapati N, Chiang CC, Durand DM. Low-frequency stimulation of corpus callosum suppresses epileptiform activity in the cortex through γ-aminobutyric acid type B receptor and slow afterhyperpolarization-mediated reduction in tissue excitability. Epilepsia 2024; 65:3689-3702. [PMID: 39425912 DOI: 10.1111/epi.18135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 10/21/2024]
Abstract
OBJECTIVE Deep brain stimulation, particularly low-frequency stimulation (LFS) targeting fiber tracts, has emerged as a potential therapy for drug-resistant epilepsy (DRE) and for generalized epilepsy, both of which pose significant treatment challenges. LFS diffusely suppresses seizures in the cortex when applied to fiber tracts like the corpus callosum (CC). Nevertheless, the specific processes responsible for suppressing epileptic activity in the cortex induced by LFS remain unclear. This study investigates the mechanisms underlying the antiepileptic effect in the cortex of LFS of the CC in coronal rodent brain slices. METHODS An in vitro 4-aminopyridine (4-AP) seizure model of cortical seizures was generated. LFS stimulation parameters were optimized to provide the largest antiepileptic effect in the cortex when applied to the CC. Changes to tissue excitability and percent time spent seizing were measured due to LFS in artificial cerebrospinal fluid, 4-AP, and in the presence of various specific and nonspecific γ-aminobutyric acid type B (GABAB) and slow afterhyperpolarization (sAHP) antagonists. RESULTS LFS significantly suppressed seizure activity in the cortex, with an optimal frequency of 5 Hz (76.5%). Tissue excitability during LFS reduces across a wide range of interstimulus intervals, with a maximum reduction at 200 ms. Notably, the tissue excitability remains depressed at 1000 ms. LFS, in the presence of GABAB antagonists, had diminished seizure reduction (<15%) and failed to reduce tissue excitability in the 50-400-ms range. Tissue excitability measured with paired pulses in the 600-1000-ms range was depressed in the presence of GABAB antagonists, suggesting a different antiepileptic mechanism was active. Upon administering sAHP antagonists, seizure reduction was once again diminished (<15%). Upon administration of both sAHP and GABAB antagonists, LFS failed to provide any meaningful seizure reduction (<5%). SIGNIFICANCE LFS of the CC provides an antiepileptic effect in the cortex with well-understood mechanisms and could be an alternative to surgical intervention for patients suffering from DRE.
Collapse
Affiliation(s)
- Nrupen Pakalapati
- Neural Engineering Center, Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Chia-Chu Chiang
- Neural Engineering Center, Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Dominique M Durand
- Neural Engineering Center, Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
5
|
Cichon J, Joseph TT, Lu X, Wasilczuk AZ, Kelz MB, Mennerick SJ, Zorumski CF, Nagele P. Nitrous Oxide activates layer 5 prefrontal neurons via SK2 channel inhibition for antidepressant effect. RESEARCH SQUARE 2024:rs.3.rs-5141491. [PMID: 39606485 PMCID: PMC11601843 DOI: 10.21203/rs.3.rs-5141491/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Nitrous oxide (N2O) induces rapid and durable antidepressant effects. The cellular and circuit mechanisms mediating this process are not known. Here we find that a single dose of inhaled N2O induces rapid and specific activation of layer V (L5) pyramidal neurons in the cingulate cortex of rodents exposed to chronic stress conditions. N2O-induced L5 activation rescues a stress-associated hypoactivity state, persists following exposure, and is necessary for its antidepressant-like activity. Although NMDA-receptor antagonism is believed to be a primary mechanism of action for N2O, L5 neurons activate even when NMDA-receptor function is attenuated through both pharmacological and genetic approaches. By examining different molecular and circuit targets, we identify N2O-induced inhibition of calcium-sensitive potassium (SK2) channels as a key molecular interaction responsible for driving specific L5 activity along with ensuing antidepressant-like effects. These results suggest that N2O-induced L5 activation is crucial for its fast antidepressant action and this effect involves novel and specific molecular actions in distinct cortical cell types.
Collapse
Affiliation(s)
- Joseph Cichon
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas T. Joseph
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xinguo Lu
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine
| | - Andrzej Z. Wasilczuk
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Max B. Kelz
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven J. Mennerick
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine
| | - Charles F. Zorumski
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine
| | - Peter Nagele
- Department of Anesthesia and Critical Care, University of Chicago, Chicago, Illinois
| |
Collapse
|
6
|
Golinski SR, Soriano K, Briegel AC, Burke MC, Yu TW, Nakayama T, Hu R, Smith RS. Gene therapy for targeting a prenatally enriched potassium channel associated with severe childhood epilepsy and premature death. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.620125. [PMID: 39484453 PMCID: PMC11527169 DOI: 10.1101/2024.10.24.620125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Dysfunction of the sodium-activated potassium channel KNa1.1 (encoded by KCNT1) is associated with a severe condition characterized by frequent seizures (up to hundreds per day) and is often fatal by age three years. We defined the early developmental onset of KNa1.1 channels in prenatal and neonatal brain tissue, establishing a timeline for pathophysiology and a window for therapeutic intervention. Using patch-clamp electrophysiology, we observed age-dependent increases in KNa1.1 K+ conductance. In neurons derived from a child with a gain-of-function KCNT1 pathogenic variant (p.R474H), we detected abnormal excitability and action potential afterhyperpolarization kinetics. In a clinical trial, two individuals with the p.R474H variant showed dramatic reductions in seizure occurrence and severity with a first-in-human antisense oligonucleotide (ASO) RNA therapy. ASO-treated p.R474H neurons in vitro exhibited normalized spiking and burst properties. Finally, we demonstrated the feasibility of ASO knockdown of KNa1.1 in mid-gestation human neurons, suggesting potential for early therapeutic intervention before the onset of epileptic encephalopathy.
Collapse
Affiliation(s)
- Sean R Golinski
- Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611
| | - Karla Soriano
- Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611
| | - Alex C Briegel
- Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611
| | - Madeline C Burke
- Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611
| | - Timothy W Yu
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, 02115
| | - Tojo Nakayama
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, 02115
| | - Ruilong Hu
- Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611
| | - Richard S Smith
- Northwestern University, Feinberg School of Medicine, Chicago, IL, 60611
| |
Collapse
|
7
|
Qiu J, Voliotis M, Bosch MA, Li XF, Zweifel LS, Tsaneva-Atanasova K, O’Byrne KT, Rønnekleiv OK, Kelly MJ. Estradiol elicits distinct firing patterns in arcuate nucleus kisspeptin neurons of females through altering ion channel conductances. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.20.581121. [PMID: 38915596 PMCID: PMC11195100 DOI: 10.1101/2024.02.20.581121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Hypothalamic kisspeptin (Kiss1) neurons are vital for pubertal development and reproduction. Arcuate nucleus Kiss1 (Kiss1ARH) neurons are responsible for the pulsatile release of Gonadotropin-releasing Hormone (GnRH). In females, the behavior of Kiss1ARH neurons, expressing Kiss1, Neurokinin B (NKB), and Dynorphin (Dyn), varies throughout the ovarian cycle. Studies indicate that 17β-estradiol (E2) reduces peptide expression but increases Vglut2 mRNA and glutamate neurotransmission in these neurons, suggesting a shift from peptidergic to glutamatergic signaling. To investigate this shift, we combined transcriptomics, electrophysiology, and mathematical modeling. Our results demonstrate that E2 treatment upregulates the mRNA expression of voltage-activated calcium channels, elevating the whole-cell calcium current and that contribute to high-frequency burst firing. Additionally, E2 treatment decreased the mRNA levels of Canonical Transient Receptor Potential (TPRC) 5 and G protein-coupled K+ (GIRK) channels. When TRPC5 channels in Kiss1ARH neurons were deleted using CRISPR, the slow excitatory postsynaptic potential (sEPSP) was eliminated. Our data enabled us to formulate a biophysically realistic mathematical model of the Kiss1ARH neuron, suggesting that E2 modifies ionic conductances in Kiss1ARH neurons, enabling the transition from high frequency synchronous firing through NKB-driven activation of TRPC5 channels to a short bursting mode facilitating glutamate release. In a low E2 milieu, synchronous firing of Kiss1ARH neurons drives pulsatile release of GnRH, while the transition to burst firing with high, preovulatory levels of E2 would facilitate the GnRH surge through its glutamatergic synaptic connection to preoptic Kiss1 neurons.
Collapse
Affiliation(s)
- Jian Qiu
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science U., Portland, OR 97239, USA
| | - Margaritis Voliotis
- Department of Mathematics and Statistics, University of Exeter, Stocker Rd, Exeter, EX4 4PY, UK
- Living Systems Institute, University of Exeter, Stocker Rd, Exeter, EX4 4PY, UK
| | - Martha A. Bosch
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science U., Portland, OR 97239, USA
| | - Xiao Feng Li
- Department of Women and Children’s Health, School of Life Course and Population Sciences, King’s College London, Guy’s Campus, London SE1 1UL, UK
| | - Larry S. Zweifel
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA
- Depatment of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Krasimira Tsaneva-Atanasova
- Department of Mathematics and Statistics, University of Exeter, Stocker Rd, Exeter, EX4 4PY, UK
- Living Systems Institute, University of Exeter, Stocker Rd, Exeter, EX4 4PY, UK
| | - Kevin T. O’Byrne
- Department of Women and Children’s Health, School of Life Course and Population Sciences, King’s College London, Guy’s Campus, London SE1 1UL, UK
| | - Oline K. Rønnekleiv
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science U., Portland, OR 97239, USA
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| | - Martin J. Kelly
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science U., Portland, OR 97239, USA
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| |
Collapse
|
8
|
Kirchner MK, Althammer F, Campos-Lira E, Montanez J, Stern JE. Endoplasmic Reticulum and Mitochondrial Calcium Handling Dynamically Shape Slow Afterhyperpolarizations in Vasopressin Magnocellular Neurons. J Neurosci 2024; 44:e0003242024. [PMID: 38937101 PMCID: PMC11270521 DOI: 10.1523/jneurosci.0003-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 05/15/2024] [Accepted: 06/10/2024] [Indexed: 06/29/2024] Open
Abstract
Many neurons including vasopressin (VP) magnocellular neurosecretory cells (MNCs) of the hypothalamic supraoptic nucleus (SON) generate afterhyperpolarizations (AHPs) during spiking to slow firing, a phenomenon known as spike frequency adaptation. The AHP is underlain by Ca2+-activated K+ currents, and while slow component (sAHP) features are well described, its mechanism remains poorly understood. Previous work demonstrated that Ca2+ influx through N-type Ca2+ channels is a primary source of sAHP activation in SON oxytocin neurons, but no obvious channel coupling was described for VP neurons. Given this, we tested the possibility of an intracellular source of sAHP activation, namely, the Ca2+-handling organelles endoplasmic reticulum (ER) and mitochondria in male and female Wistar rats. We demonstrate that ER Ca2+ depletion greatly inhibits sAHPs without a corresponding decrease in Ca2+ signal. Caffeine sensitized AHP activation by Ca2+ In contrast to ER, disabling mitochondria with CCCP or blocking mitochondria Ca2+ uniporters (MCUs) enhanced sAHP amplitude and duration, implicating mitochondria as a vital buffer for sAHP-activating Ca2+ Block of mitochondria Na+-dependent Ca2+ release via triphenylphosphonium (TPP+) failed to affect sAHPs, indicating that mitochondria Ca2+ does not contribute to sAHP activation. Together, our results suggests that ER Ca2+-induced Ca2+ release activates sAHPs and mitochondria shape the spatiotemporal trajectory of the sAHP via Ca2+ buffering in VP neurons. Overall, this implicates organelle Ca2+, and specifically ER-mitochondria-associated membrane contacts, as an important site of Ca2+ microdomain activity that regulates sAHP signaling pathways. Thus, this site plays a major role in influencing VP firing activity and systemic hormonal release.
Collapse
Affiliation(s)
- Matthew K Kirchner
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, Georgia 30303
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303
| | - Ferdinand Althammer
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, Georgia 30303
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303
| | - Elba Campos-Lira
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, Georgia 30303
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303
| | - Juliana Montanez
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, Georgia 30303
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303
| | - Javier E Stern
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, Georgia 30303
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303
| |
Collapse
|
9
|
Yang X, La Camera G. Co-existence of synaptic plasticity and metastable dynamics in a spiking model of cortical circuits. PLoS Comput Biol 2024; 20:e1012220. [PMID: 38950068 PMCID: PMC11244818 DOI: 10.1371/journal.pcbi.1012220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 07/12/2024] [Accepted: 06/01/2024] [Indexed: 07/03/2024] Open
Abstract
Evidence for metastable dynamics and its role in brain function is emerging at a fast pace and is changing our understanding of neural coding by putting an emphasis on hidden states of transient activity. Clustered networks of spiking neurons have enhanced synaptic connections among groups of neurons forming structures called cell assemblies; such networks are capable of producing metastable dynamics that is in agreement with many experimental results. However, it is unclear how a clustered network structure producing metastable dynamics may emerge from a fully local plasticity rule, i.e., a plasticity rule where each synapse has only access to the activity of the neurons it connects (as opposed to the activity of other neurons or other synapses). Here, we propose a local plasticity rule producing ongoing metastable dynamics in a deterministic, recurrent network of spiking neurons. The metastable dynamics co-exists with ongoing plasticity and is the consequence of a self-tuning mechanism that keeps the synaptic weights close to the instability line where memories are spontaneously reactivated. In turn, the synaptic structure is stable to ongoing dynamics and random perturbations, yet it remains sufficiently plastic to remap sensory representations to encode new sets of stimuli. Both the plasticity rule and the metastable dynamics scale well with network size, with synaptic stability increasing with the number of neurons. Overall, our results show that it is possible to generate metastable dynamics over meaningful hidden states using a simple but biologically plausible plasticity rule which co-exists with ongoing neural dynamics.
Collapse
Affiliation(s)
- Xiaoyu Yang
- Graduate Program in Physics and Astronomy, Stony Brook University, Stony Brook, New York, United States of America
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York, United States of America
- Center for Neural Circuit Dynamics, Stony Brook University, Stony Brook, New York, United States of America
| | - Giancarlo La Camera
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York, United States of America
- Center for Neural Circuit Dynamics, Stony Brook University, Stony Brook, New York, United States of America
| |
Collapse
|
10
|
Yang X, La Camera G. Co-existence of synaptic plasticity and metastable dynamics in a spiking model of cortical circuits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.07.570692. [PMID: 38106233 PMCID: PMC10723399 DOI: 10.1101/2023.12.07.570692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Evidence for metastable dynamics and its role in brain function is emerging at a fast pace and is changing our understanding of neural coding by putting an emphasis on hidden states of transient activity. Clustered networks of spiking neurons have enhanced synaptic connections among groups of neurons forming structures called cell assemblies; such networks are capable of producing metastable dynamics that is in agreement with many experimental results. However, it is unclear how a clustered network structure producing metastable dynamics may emerge from a fully local plasticity rule, i.e., a plasticity rule where each synapse has only access to the activity of the neurons it connects (as opposed to the activity of other neurons or other synapses). Here, we propose a local plasticity rule producing ongoing metastable dynamics in a deterministic, recurrent network of spiking neurons. The metastable dynamics co-exists with ongoing plasticity and is the consequence of a self-tuning mechanism that keeps the synaptic weights close to the instability line where memories are spontaneously reactivated. In turn, the synaptic structure is stable to ongoing dynamics and random perturbations, yet it remains sufficiently plastic to remap sensory representations to encode new sets of stimuli. Both the plasticity rule and the metastable dynamics scale well with network size, with synaptic stability increasing with the number of neurons. Overall, our results show that it is possible to generate metastable dynamics over meaningful hidden states using a simple but biologically plausible plasticity rule which co-exists with ongoing neural dynamics.
Collapse
Affiliation(s)
- Xiaoyu Yang
- Graduate Program in Physics and Astronomy, Stony Brook University
- Department of Neurobiology & Behavior, Stony Brook University
- Center for Neural Circuit Dynamics, Stony Brook University
| | - Giancarlo La Camera
- Department of Neurobiology & Behavior, Stony Brook University
- Center for Neural Circuit Dynamics, Stony Brook University
| |
Collapse
|
11
|
Abstract
Dystonia is a clinically and genetically highly heterogeneous neurological disorder characterized by abnormal movements and postures caused by involuntary sustained or intermittent muscle contractions. A number of groundbreaking genetic and molecular insights have recently been gained. While they enable genetic testing and counseling, their translation into new therapies is still limited. However, we are beginning to understand shared pathophysiological pathways and molecular mechanisms. It has become clear that dystonia results from a dysfunctional network involving the basal ganglia, cerebellum, thalamus, and cortex. On the molecular level, more than a handful of, often intertwined, pathways have been linked to pathogenic variants in dystonia genes, including gene transcription during neurodevelopment (e.g., KMT2B, THAP1), calcium homeostasis (e.g., ANO3, HPCA), striatal dopamine signaling (e.g., GNAL), endoplasmic reticulum stress response (e.g., EIF2AK2, PRKRA, TOR1A), autophagy (e.g., VPS16), and others. Thus, different forms of dystonia can be molecularly grouped, which may facilitate treatment development in the future.
Collapse
Affiliation(s)
- Mirja Thomsen
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany;
| | - Lara M Lange
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany;
| | - Michael Zech
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Katja Lohmann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany;
| |
Collapse
|
12
|
Skiteva O, Yao N, Mantas I, Zhang X, Perlmann T, Svenningsson P, Chergui K. Aberrant somatic calcium channel function in cNurr1 and LRRK2-G2019S mice. NPJ Parkinsons Dis 2023; 9:56. [PMID: 37029193 PMCID: PMC10082048 DOI: 10.1038/s41531-023-00500-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 03/23/2023] [Indexed: 04/09/2023] Open
Abstract
In Parkinson's disease (PD), axons of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNc) degenerate before their cell bodies. Calcium influx during pacemaker firing might contribute to neuronal loss, but it is not known if dysfunctions of voltage-gated calcium channels (VGCCs) occur in DA neurons somata and axon terminals. We investigated T-type and L-type VGCCs in SNc-DA neurons of two mouse models of PD: mice with a deletion of the Nurr1 gene in DA neurons from an adult age (cNurr1 mice), and mice bearing the G2019S mutation in the gene coding for LRRK2 (G2019S mice). Adult cNurr1 mice displayed motor and DA deficits, while middle-aged G2019S mice did not. The number and morphology of SNc-DA neurons, most of their intrinsic membrane properties and pacemaker firing were unaltered in cNurr1 and G2019S mice compared to their control and wild-type littermates. L-type VGCCs contributed to the pacemaker firing of SNc-DA neurons in G2019S mice, but not in control, wild-type, and cNurr1 mice. In cNurr1 mice, but not G2019S mice, the contribution of T-type VGCCs to the pacemaker firing of SNc-DA neurons was reduced, and somatic dopamine-D2 autoreceptors desensitized more. Altered contribution of L-type and T-type VGCCs to the pacemaker firing was not observed in the presence of a LRRK2 kinase inhibitor in G2019S mice, and in the presence of a flavonoid with antioxidant activity in G2019S and cNurr1 mice. The role of L-type and T-type VGCCs in controlling dopamine release from axon terminals in the striatum was unaltered in cNurr1 and G2019S mice. Our findings uncover opposite changes, linked to oxidative stress, in the function of two VGCCs in DA neurons somata, but not axon terminals, in two different experimental PD models.
Collapse
Affiliation(s)
- Olga Skiteva
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ning Yao
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ioannis Mantas
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Xiaoqun Zhang
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Perlmann
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Karima Chergui
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
13
|
Lobule-Related Action Potential Shape- and History-Dependent Current Integration in Purkinje Cells of Adult and Developing Mice. Cells 2023; 12:cells12040623. [PMID: 36831290 PMCID: PMC9953991 DOI: 10.3390/cells12040623] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/03/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Purkinje cells (PCs) are the principal cells of the cerebellar cortex and form a central element in the modular organization of the cerebellum. Differentiation of PCs based on gene expression profiles revealed two subpopulations with distinct connectivity, action potential firing and learning-induced activity changes. However, which basal cell physiological features underlie the differences between these subpopulations and to what extent they integrate input differentially remains largely unclear. Here, we investigate the cellular electrophysiological properties of PC subpopulation in adult and juvenile mice. We found that multiple fundamental cell physiological properties, including membrane resistance and various aspects of the action potential shape, differ between PCs from anterior and nodular lobules. Moreover, the two PC subpopulations also differed in the integration of negative and positive current steps as well as in size of the hyperpolarization-activated current. A comparative analysis in juvenile mice confirmed that most of these lobule-specific differences are already present at pre-weaning ages. Finally, we found that current integration in PCs is input history-dependent for both positive and negative currents, but this is not a distinctive feature between anterior and nodular PCs. Our results support the concept of a fundamental differentiation of PCs subpopulations in terms of cell physiological properties and current integration, yet reveals that history-dependent input processing is consistent across PC subtypes.
Collapse
|
14
|
Kolatt Chandran S, Yiannakas A, Kayyal H, Salalha R, Cruciani F, Mizrahi L, Khamaisy M, Stern S, Rosenblum K. Intrinsic Excitability in Layer IV-VI Anterior Insula to Basolateral Amygdala Projection Neurons Correlates with the Confidence of Taste Valence Encoding. eNeuro 2023; 10:ENEURO.0302-22.2022. [PMID: 36635250 PMCID: PMC9850927 DOI: 10.1523/eneuro.0302-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 09/01/2022] [Accepted: 09/11/2022] [Indexed: 12/14/2022] Open
Abstract
Avoiding potentially harmful, and consuming safe food is crucial for the survival of living organisms. However, the perceived valence of sensory information can change following conflicting experiences. Pleasurability and aversiveness are two crucial parameters defining the perceived valence of a taste and can be impacted by novelty. Importantly, the ability of a given taste to serve as the conditioned stimulus (CS) in conditioned taste aversion (CTA) is dependent on its valence. Activity in anterior insula (aIC) Layer IV-VI pyramidal neurons projecting to the basolateral amygdala (BLA) is correlated with and necessary for CTA learning and retrieval, as well as the expression of neophobia toward novel tastants, but not learning taste familiarity. Yet, the cellular mechanisms underlying the updating of taste valence representation in this specific pathway are poorly understood. Here, using retrograde viral tracing and whole-cell patch-clamp electrophysiology in trained mice, we demonstrate that the intrinsic properties of deep-lying Layer IV-VI, but not superficial Layer I-III aIC-BLA neurons, are differentially modulated by both novelty and valence, reflecting the subjective predictability of taste valence arising from prior experience. These correlative changes in the profile of intrinsic properties of LIV-VI aIC-BLA neurons were detectable following both simple taste experiences, as well as following memory retrieval, extinction learning, and reinstatement.
Collapse
Affiliation(s)
| | - Adonis Yiannakas
- Sagol Department of Neurobiology, University of Haifa, Abba Khoushy Ave 199, Haifa, 3498838, Israel
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland
| | - Haneen Kayyal
- Sagol Department of Neurobiology, University of Haifa, Abba Khoushy Ave 199, Haifa, 3498838, Israel
| | - Randa Salalha
- Sagol Department of Neurobiology, University of Haifa, Abba Khoushy Ave 199, Haifa, 3498838, Israel
| | - Federica Cruciani
- Sagol Department of Neurobiology, University of Haifa, Abba Khoushy Ave 199, Haifa, 3498838, Israel
| | - Liron Mizrahi
- Sagol Department of Neurobiology, University of Haifa, Abba Khoushy Ave 199, Haifa, 3498838, Israel
| | - Mohammad Khamaisy
- Sagol Department of Neurobiology, University of Haifa, Abba Khoushy Ave 199, Haifa, 3498838, Israel
| | - Shani Stern
- Sagol Department of Neurobiology, University of Haifa, Abba Khoushy Ave 199, Haifa, 3498838, Israel
| | - Kobi Rosenblum
- Sagol Department of Neurobiology, University of Haifa, Abba Khoushy Ave 199, Haifa, 3498838, Israel
- Center for Gene Manipulation in the Brain, University of Haifa, Haifa, Israel
| |
Collapse
|
15
|
Awasthi R, Chandra N, Barkai E. Olfactory rule learning-induced enhancement in intrinsic neuronal excitability is maintained by shutdown of the cholinergic M-current. Front Cell Neurosci 2022; 16:934838. [PMID: 36246520 PMCID: PMC9556983 DOI: 10.3389/fncel.2022.934838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Training rats in a particularly difficult olfactory discrimination task initiates a period of accelerated learning, manifested as a dramatic increase in the rats' capacity to discriminate between pairs of odors once they have learned the discrimination task, implying that rule learning has taken place. At the cellular biophysical level, rule learning is maintained by reduction in the conductance of the slow current (sIAHP) simultaneously in most piriform cortex layer II pyramidal neurons. Such sIAHP reduction is expressed in attenuation of the post-burst afterhyperpolarization (AHP) potential and thus in enhanced repetitive action potential firing. Previous studies have shown that a causal relationship exists between long-lasting post-burst AHP reduction and rule learning. A specific channel through which the sIAHP flows has not been identified. The sIAHP in pyramidal cells is critically dependent on membrane phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P(2)]. PtdIns(4,5)P(2) regulates the calcium sensitivity of the sIAHP by acting downstream from the rise in intracellular calcium. These findings led to the interesting hypothesis that PtdIns(4,5)P(2) activates a variety of potassium channels. Thus, the sIAHP would not represent a unitary ionic current but the embodiment of a generalized potassium channel gating mechanism. We thus hypothesized that the learning-induced increase in intrinsic excitability is mediated by reduced conductance of one or more of the currents that contribute to the sIAHP. Here we first show, using current-clamp recordings, that the post-burst AHP in piriform cortex pyramidal neurons is also mediated by the Ih, and the contribution of this current to the post-burst AHP is also affected by learning. We also show, using whole-cell patch-clamp recordings, that the sIAHP in neurons from trained rats is not sensitive to blocking membrane phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P(2)], and to the blocking of the current mediated by the cholinergic muscarinic acetylcholine receptor (M-current). Further current-clamp recordings also show that blocking PtdIns(4,5)P(2) synthesis and application of a specific IKCa blocker have no effect on the post-burst AHP in neurons from trained as well as control rats. Taken together with results from our previous studies, these data suggest that rule learning-induced long-lasting enhancement in intrinsic neuronal excitability results from reduced conductance of the M-current and thus the slow potassium currents, which control repetitive spike firing.
Collapse
Affiliation(s)
| | | | - Edi Barkai
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
16
|
Groten CJ, MacVicar BA. Mitochondrial Ca 2+ uptake by the MCU facilitates pyramidal neuron excitability and metabolism during action potential firing. Commun Biol 2022; 5:900. [PMID: 36056095 PMCID: PMC9440007 DOI: 10.1038/s42003-022-03848-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 08/16/2022] [Indexed: 12/12/2022] Open
Abstract
Neuronal activation is fundamental to information processing by the brain and requires mitochondrial energy metabolism. Mitochondrial Ca2+ uptake by the mitochondrial Ca2+ uniporter (MCU) has long been implicated in the control of energy metabolism and intracellular Ca2+ signalling, but its importance to neuronal function in the brain remains unclear. Here, we used in situ electrophysiology and two-photon imaging of mitochondrial Ca2+, cytosolic Ca2+, and NAD(P)H to test the relevance of MCU activation to pyramidal neuron Ca2+ signalling and energy metabolism during action potential firing. We demonstrate that mitochondrial Ca2+ uptake by the MCU is tuned to enhanced firing rate and the strength of this relationship varied between neurons of discrete brain regions. MCU activation promoted electron transport chain activity and chemical reduction of NAD+ to NADH. Moreover, Ca2+ buffering by mitochondria attenuated cytosolic Ca2+ signals and thereby reduced the coupling between activity and the slow afterhyperpolarization, a ubiquitous regulator of excitability. Collectively, we demonstrate that the MCU is engaged by accelerated spike frequency to facilitate neuronal activity through simultaneous control of energy metabolism and excitability. As such, the MCU is situated to promote brain functions associated with high frequency signalling and may represent a target for controlling excessive neuronal activity.
Collapse
Affiliation(s)
- Christopher J Groten
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6T 1Z3, Canada.
| | - Brian A MacVicar
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6T 1Z3, Canada.
| |
Collapse
|
17
|
Smith LA, Goodman AM, McMahon LL. Dentate Granule Cells Are Hyperexcitable in the TgF344-AD Rat Model of Alzheimer's Disease. Front Synaptic Neurosci 2022; 14:826601. [PMID: 35685246 PMCID: PMC9171068 DOI: 10.3389/fnsyn.2022.826601] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
The dentate gyrus is both a critical gatekeeper for hippocampal signal processing and one of the first brain regions to become dysfunctional in Alzheimer's disease (AD). Accordingly, the appropriate balance of excitation and inhibition through the dentate is a compelling target for mechanistic investigation and therapeutic intervention in early AD. Previously, we reported an increased long-term potentiation (LTP) magnitude at medial perforant path-dentate granule cell (MPP-DGC) synapses in slices from both male and acutely ovariectomized female TgF344-AD rats compared with wild type (Wt) as early as 6 months of age that is accompanied by an increase in steady-state postsynaptic depolarization during the high-frequency stimulation used to induce plasticity. Subsequently, we found that heightened function of β-adrenergic receptors (β-ARs) drives the increase in the LTP magnitude, but the increase in steady-state depolarization was only partially due to β-AR activation. As we previously reported no detectable difference in spine density or presynaptic release probability, we entertained the possibility that DGCs themselves might have modified passive or active membrane properties, which may contribute to the significant increase in charge transfer during high-frequency stimulation. Using brain slice electrophysiology from 6-month-old female rats acutely ovariectomized to eliminate variability due to fluctuating plasma estradiol, we found significant changes in passive membrane properties and active membrane properties leading to increased DGC excitability in TgF344-AD rats. Specifically, TgF344-AD DGCs have an increased input resistance and decreased rheobase, decreased sag, and increased action potential (AP) spike accommodation. Importantly, we found that for the same amount of depolarizing current injection, DGCs from TgF344-AD compared with Wt rats have a larger magnitude voltage response, which was accompanied by a decreased delay to fire the first action potential, indicating TgF344-AD DGCs membranes are more excitable. Taken together, DGCs in TgF344-AD rats are more excitable, which likely contributes to the heightened depolarization during high-frequency synaptic activation.
Collapse
|
18
|
Afterhyperpolarization Promotes the Firing of Mitral Cells through a Voltage-Dependent Modification of Action Potential Threshold. eNeuro 2022; 9:ENEURO.0401-21.2021. [PMID: 35277450 PMCID: PMC8982644 DOI: 10.1523/eneuro.0401-21.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/22/2021] [Accepted: 12/13/2021] [Indexed: 11/21/2022] Open
Abstract
In the olfactory bulb, mitral cells (MCs) display a spontaneous firing that is characterized by bursts of action potentials (APs) intermixed with silent periods. Intraburst firing frequency and duration are heterogeneous among MCs and increase with membrane depolarization. By using patch-clamp recording on rat slices, we dissected out the intrinsic properties responsible for this bursting activity. We showed that the threshold of AP generation dynamically changes as a function of the preceding trajectory of the membrane potential. In fact, the AP threshold became more negative when the membrane was hyperpolarized and had a recovery rate inversely proportional to the membrane repolarization rate. Such variations appeared to be produced by changes in the inactivation state of voltage-dependent Na+ channels. Thus, AP initiation was favored by hyperpolarizing events, such as negative membrane oscillations or inhibitory synaptic input. After the first AP, the following fast afterhyperpolarization (AHP) brought the threshold to more negative values and then promoted the emission of the following AP. This phenomenon was repeated for each AP of the burst making the fast AHP a regenerative mechanism that sustained the firing, AHP with larger amplitudes and faster repolarizations being associated with larger and higher-frequency bursts. Burst termination was found to be because of the development of a slow repolarization component of the AHP (slow AHP). Overall, the AHP characteristics appeared as a major determinant of the bursting properties.
Collapse
|
19
|
Sahu G, Turner RW. The Molecular Basis for the Calcium-Dependent Slow Afterhyperpolarization in CA1 Hippocampal Pyramidal Neurons. Front Physiol 2022; 12:759707. [PMID: 35002757 PMCID: PMC8730529 DOI: 10.3389/fphys.2021.759707] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/01/2021] [Indexed: 12/02/2022] Open
Abstract
Neuronal signal transmission depends on the frequency, pattern, and timing of spike output, each of which are shaped by spike afterhyperpolarizations (AHPs). There are classically three post-spike AHPs of increasing duration categorized as fast, medium and slow AHPs that hyperpolarize a cell over a range of 10 ms to 30 s. Intensive early work on CA1 hippocampal pyramidal cells revealed that all three AHPs incorporate activation of calcium-gated potassium channels. The ionic basis for a fAHP was rapidly attributed to the actions of big conductance (BK) and the mAHP to small conductance (SK) or Kv7 potassium channels. In stark contrast, the ionic basis for a prominent slow AHP of up to 30 s duration remained an enigma for over 30 years. Recent advances in pharmacological, molecular, and imaging tools have uncovered the expression of a calcium-gated intermediate conductance potassium channel (IK, KCa3.1) in central neurons that proves to contribute to the slow AHP in CA1 hippocampal pyramidal cells. Together the data show that the sAHP arises in part from a core tripartite complex between Cav1.3 (L-type) calcium channels, ryanodine receptors, and IK channels at endoplasmic reticulum-plasma membrane junctions. Work on the sAHP in CA1 pyramidal neurons has again quickened pace, with identified contributions by both IK channels and the Na-K pump providing answers to several mysteries in the pharmacological properties of the sAHP.
Collapse
Affiliation(s)
- Giriraj Sahu
- National Institute of Pharmaceutical Education and Research Ahmedabad, Ahmedabad, India
| | - Ray W Turner
- Department Cell Biology & Anatomy, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
20
|
Amakhin DV, Soboleva EB, Chizhov AV, Zaitsev AV. Insertion of Calcium-Permeable AMPA Receptors during Epileptiform Activity In Vitro Modulates Excitability of Principal Neurons in the Rat Entorhinal Cortex. Int J Mol Sci 2021; 22:12174. [PMID: 34830051 PMCID: PMC8621524 DOI: 10.3390/ijms222212174] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/05/2021] [Accepted: 11/07/2021] [Indexed: 12/19/2022] Open
Abstract
Epileptic activity leads to rapid insertion of calcium-permeable α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (CP-AMPARs) into the synapses of cortical and hippocampal glutamatergic neurons, which generally do not express them. The physiological significance of this process is not yet fully understood; however, it is usually assumed to be a pathological process that augments epileptic activity. Using whole-cell patch-clamp recordings in rat entorhinal cortex slices, we demonstrate that the timing of epileptiform discharges, induced by 4-aminopyridine and gabazine, is determined by the shunting effect of Ca2+-dependent slow conductance, mediated predominantly by K+-channels. The blockade of CP-AMPARs by IEM-1460 eliminates this extra conductance and consequently increases the rate of discharge generation. The blockade of NMDARs reduced the additional conductance to a lesser extent than the blockade of CP-AMPARs, indicating that CP-AMPARs are a more significant source of intracellular Ca2+. The study's main findings were implemented in a mathematical model, which reproduces the shunting effect of activity-dependent conductance on the generation of discharges. The obtained results suggest that the expression of CP-AMPARs in principal neurons reduces the discharge generation rate and may be considered as a protective mechanism.
Collapse
Affiliation(s)
- Dmitry V. Amakhin
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Toreza Prospekt 44, 194223 Saint Petersburg, Russia; (D.V.A.); (E.B.S.); (A.V.C.)
| | - Elena B. Soboleva
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Toreza Prospekt 44, 194223 Saint Petersburg, Russia; (D.V.A.); (E.B.S.); (A.V.C.)
| | - Anton V. Chizhov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Toreza Prospekt 44, 194223 Saint Petersburg, Russia; (D.V.A.); (E.B.S.); (A.V.C.)
- Ioffe Institute, Russian Academy of Sciences, Polytekhnicheskaya 26, 194021 Saint Petersburg, Russia
| | - Aleksey V. Zaitsev
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Toreza Prospekt 44, 194223 Saint Petersburg, Russia; (D.V.A.); (E.B.S.); (A.V.C.)
| |
Collapse
|
21
|
Ferdous ZI, Yu A, Zeng Y, Guo X, Yan Z, Berdichevsky Y. Efficient and Accurate Computational Model of Neuron with Spike Frequency Adaptation. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2021; 2021:6496-6499. [PMID: 34892598 DOI: 10.1109/embc46164.2021.9629799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Simplified models of neurons are widely used in computational investigations of large networks. One of the most important performance metrics of simplified models is their accuracy in reproducing action potential (spike) timing. In this article, we developed a simple, computationally efficient neuron model by modifying the adaptive exponential integrate and fire (AdEx) model [1] with sigmoid afterhyperpolarization current (Sigmoid AHP). Our model can precisely match the spike times and spike frequency adaptation of cortical pyramidal neurons. The accuracy was similar to a more complex two compartment biophysically realistic model of the same neurons. This work provides a simplified neuronal model with improved spike timing accuracy for use in modeling of large neural networks.Clinical Relevance- Accurate and computationally efficient single neuron model will enable large network modeling of brain regions involved in neurological and psychiatric disorders and may lead to a better understanding of the disorder mechanisms.
Collapse
|
22
|
Piggott CA, Jin Y. Junctophilins: Key Membrane Tethers in Muscles and Neurons. Front Mol Neurosci 2021; 14:709390. [PMID: 34305529 PMCID: PMC8295595 DOI: 10.3389/fnmol.2021.709390] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 06/15/2021] [Indexed: 12/26/2022] Open
Abstract
Contacts between the endoplasmic reticulum (ER) and plasma membrane (PM) contain specialized tethering proteins that bind both ER and PM membranes. In excitable cells, ER–PM contacts play an important role in calcium signaling and transferring lipids. Junctophilins are a conserved family of ER–PM tethering proteins. They are predominantly expressed in muscles and neurons and known to simultaneously bind both ER- and PM-localized ion channels. Since their discovery two decades ago, functional studies using junctophilin-deficient animals have provided a deep understanding of their roles in muscles and neurons, including excitation-contraction coupling, store-operated calcium entry (SOCE), and afterhyperpolarization (AHP). In this review, we highlight key findings from mouse, fly, and worm that support evolutionary conservation of junctophilins.
Collapse
Affiliation(s)
- Christopher A Piggott
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, San Diego, CA, United States
| | - Yishi Jin
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
23
|
Laker D, Tolle F, Stegen M, Heerdegen M, Köhling R, Kirschstein T, Wolfart J. K v7 and K ir6 Channels Shape the Slow AHP in Mouse Dentate Gyrus Granule Cells and Control Burst-like Firing Behavior. Neuroscience 2021; 467:56-72. [PMID: 34048798 DOI: 10.1016/j.neuroscience.2021.05.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 11/29/2022]
Abstract
The slow afterhyperpolarizing potential (sAHP) can silence a neuron for hundreds of milliseconds. Thereby, the sAHP determines the discharge behavior of many types of neurons. In dentate granule cells (DGCs), serving as a filter into the hippocampal network, mostly tonic or adapting discharge properties have been described. As under standard whole-cell recording conditions the sAHP is inhibited, we reevaluated the intrinsic functional phenotype of DGCs and the conductances underlying the sAHP, using gramicidine-perforated patch-clamp technique. We found that in 97/113 (86%) of the DGCs, a burst of action potentials (APs) to excitation ended by a large sAHP, despite continued depolarization. This result suggests that burst-like firing is the default functional phenotype of DGCs and that sAHPs are important for it. Indeed, burst-like firing DGCs showed a significantly higher sAHP-current (IsAHP) amplitude compared to spike-frequency adapting cells (16/113 = 14%). The IsAHP was mediated by Kv7 and Kir6 channels by pharmacological inhibition using XE991 and tolbutamide, although heterogeneously among DGCs. The percent inhibition of IsAHP by these compounds also correlated with the AP number and AP burst length. Application of 100 µM nickel after XE991 and tolbutamide detected a third conductance contributing to burst-like firing and the sAHP, most likely mediated by T-type calcium channels. Lastly, medial perforant path-dentate gyrus long-term potentiation was amplified by XE991 and tolbutamide. In conclusion, the sAHP shapes intrinsic burst-like firing which, under physiological circumstances, could be controlled via cholinergic afferents and ATP metabolism.
Collapse
Affiliation(s)
- Debora Laker
- Oscar Langendorff Institute of Physiology, University Medicine Rostock, Rostock, Germany
| | - Frederik Tolle
- Oscar Langendorff Institute of Physiology, University Medicine Rostock, Rostock, Germany
| | - Michael Stegen
- Department of Neurosurgery, University of Freiburg, Germany
| | - Marco Heerdegen
- Oscar Langendorff Institute of Physiology, University Medicine Rostock, Rostock, Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, University Medicine Rostock, Rostock, Germany
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology, University Medicine Rostock, Rostock, Germany.
| | - Jakob Wolfart
- Oscar Langendorff Institute of Physiology, University Medicine Rostock, Rostock, Germany
| |
Collapse
|
24
|
Angstadt JD, Rebel MI, Connolly MK. Effects of calcium-activated potassium channel modulators on afterhyperpolarizing potentials in identified motor and mechanosensory neurons of the medicinal leech. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2021; 207:69-85. [PMID: 33483833 DOI: 10.1007/s00359-021-01462-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/30/2020] [Accepted: 01/02/2021] [Indexed: 11/26/2022]
Abstract
Calcium-activated potassium (KCa) channels contribute to multiple neuronal properties including spike frequency and afterhyperpolarizing potentials (AHPs). KCa channels are classified as KCa1.1, KCa2, or KCa3.1 based on single-channel conductance and pharmacology. Ca2+-dependent AHPs in vertebrates are categorized as fast, medium, or slow. Fast and medium AHPs are generated by KCa1.1 and KCa2 channels, respectively. The KCa subtype responsible for slow AHPs is unclear. Prolonged, Ca2+-dependent AHPs have been described in several leech neurons. Unfortunately, apamin and other KCa blockers often prove ineffective in the leech. An alternative approach is to utilize KCa modulators, which alter channel sensitivity to Ca2+. Vertebrate KCa2 channels are targeted selectively by the positive modulator CyPPA and the negative modulator NS8593. Here we show that AHPs in identified motor and mechanosensory leech neurons are enhanced by CyPPA and suppressed by NS8593. Our results indicate that KCa2 channels underlie prolonged AHPs in these neurons and suggest that KCa2 modulators may serve as effective tools to explore the role of KCa channels in leech physiology.
Collapse
Affiliation(s)
| | - Matthew I Rebel
- Siena College, Loudonville, NY, USA
- College of Medicine, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Megan K Connolly
- Siena College, Loudonville, NY, USA
- Physician Assistant Studies Department, Marist College, Poughkeepsie, NY, USA
| |
Collapse
|
25
|
The aging mouse brain: cognition, connectivity and calcium. Cell Calcium 2021; 94:102358. [PMID: 33517250 DOI: 10.1016/j.ceca.2021.102358] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/16/2021] [Accepted: 01/18/2021] [Indexed: 02/08/2023]
Abstract
Aging is a complex process that differentially impacts multiple cognitive, sensory, neuronal and molecular processes. Technological innovations now allow for parallel investigation of neuronal circuit function, structure and molecular composition in the brain of awake behaving adult mice. Thus, mice have become a critical tool to better understand how aging impacts the brain. However, a more granular systems-based approach, which considers the impact of age on key features relating to neural processing, is required. Here, we review evidence probing the impact of age on the mouse brain. We focus on a range of processes relating to neuronal function, including cognitive abilities, sensory systems, synaptic plasticity and calcium regulation. Across many systems, we find evidence for prominent age-related dysregulation even before 12 months of age, suggesting that emerging age-related alterations can manifest by late adulthood. However, we also find reports suggesting that some processes are remarkably resilient to aging. The evidence suggests that aging does not drive a parallel, linear dysregulation of all systems, but instead impacts some processes earlier, and more severely, than others. We propose that capturing the more fine-scale emerging features of age-related vulnerability and resilience may provide better opportunities for the rejuvenation of the aged brain.
Collapse
|
26
|
Bacon TJ, Pickering AE, Mellor JR. Noradrenaline Release from Locus Coeruleus Terminals in the Hippocampus Enhances Excitation-Spike Coupling in CA1 Pyramidal Neurons Via β-Adrenoceptors. Cereb Cortex 2020; 30:6135-6151. [PMID: 32607551 PMCID: PMC7609922 DOI: 10.1093/cercor/bhaa159] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/13/2020] [Accepted: 05/13/2020] [Indexed: 12/29/2022] Open
Abstract
Release of the neuromodulator noradrenaline signals salience during wakefulness, flagging novel or important experiences to reconfigure information processing and memory representations in the hippocampus. Noradrenaline is therefore expected to enhance hippocampal responses to synaptic input; however, noradrenergic agonists have been found to have mixed and sometimes contradictory effects on Schaffer collateral synapses and the resulting CA1 output. Here, we examine the effects of endogenous, optogenetically driven noradrenaline release on synaptic transmission and spike output in mouse hippocampal CA1 pyramidal neurons. We show that endogenous noradrenaline release enhances the probability of CA1 pyramidal neuron spiking without altering feedforward excitatory or inhibitory synaptic inputs in the Schaffer collateral pathway. β-adrenoceptors mediate this enhancement of excitation-spike coupling by reducing the charge required to initiate action potentials, consistent with noradrenergic modulation of voltage-gated potassium channels. Furthermore, we find the likely effective concentration of endogenously released noradrenaline is sub-micromolar. Surprisingly, although comparable concentrations of exogenous noradrenaline cause robust depression of slow afterhyperpolarization currents, endogenous release of noradrenaline does not, indicating that endogenous noradrenaline release is targeted to specific cellular locations. These findings provide a mechanism by which targeted endogenous release of noradrenaline can enhance information transfer in the hippocampus in response to salient events.
Collapse
Affiliation(s)
- Travis J Bacon
- Centre for Synaptic Plasticity, University of Bristol, Bristol, UK
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| | - Anthony E Pickering
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol BS8 1TD, UK
- Bristol Anaesthesia, Pain & Critical Care Sciences, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol BS2 8HW, UK
| | - Jack R Mellor
- Centre for Synaptic Plasticity, University of Bristol, Bristol, UK
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
27
|
Severin D, Gallagher M, Kirkwood A. Afterhyperpolarization amplitude in CA1 pyramidal cells of aged Long-Evans rats characterized for individual differences. Neurobiol Aging 2020; 96:43-48. [PMID: 32932137 DOI: 10.1016/j.neurobiolaging.2020.07.022] [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: 04/09/2020] [Revised: 07/06/2020] [Accepted: 07/25/2020] [Indexed: 11/18/2022]
Abstract
Altered neural excitability is considered a prominent contributing factor to cognitive decline during aging. A clear example is the excess neural activity observed in several temporal lobe structures of cognitively impaired older individuals in rodents and humans. At a cellular level, aging-related changes in mechanisms regulating intrinsic excitability have been well examined in pyramidal cells of the CA1 hippocampal subfield. Studies in the inbred Fisher 344 rat strain document an age-related increase in the slow afterhyperpolarization (AHP) that normally occurs after a burst of action potentials, and serves to reduce subsequent firing. We evaluated the status of the AHP in the outbred Long-Evans rat, a well-established model for studying individual differences in neurocognitive aging. In contrast to the findings reported in the Fisher 344 rats, in the Long-Evan rats we detected a selective reduction in AHP in cognitively impaired aged individuals. We discuss plausible scenarios to account for these differences and also discuss possible implications of these differences.
Collapse
Affiliation(s)
- Daniel Severin
- Department of Neurosciences, Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Michela Gallagher
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Alfredo Kirkwood
- Department of Neurosciences, Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
28
|
Ca 2+-activated KCa3.1 potassium channels contribute to the slow afterhyperpolarization in L5 neocortical pyramidal neurons. Sci Rep 2020; 10:14484. [PMID: 32879404 PMCID: PMC7468258 DOI: 10.1038/s41598-020-71415-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/07/2020] [Indexed: 01/15/2023] Open
Abstract
Layer 5 neocortical pyramidal neurons are known to display slow Ca2+-dependent afterhyperpolarization (sAHP) after bursts of spikes, which is similar to the sAHP in CA1 hippocampal cells. However, the mechanisms of sAHP in the neocortex remain poorly understood. Here, we identified the Ca2+-gated potassium KCa3.1 channels as contributors to sAHP in ER81-positive neocortical pyramidal neurons. Moreover, our experiments strongly suggest that the relationship between sAHP and KCa3.1 channels in a feedback mechanism underlies the adaptation of the spiking frequency of layer 5 pyramidal neurons. We demonstrated the relationship between KCa3.1 channels and sAHP using several parallel methods: electrophysiology, pharmacology, immunohistochemistry, and photoactivatable probes. Our experiments demonstrated that ER81 immunofluorescence in layer 5 co-localized with KCa3.1 immunofluorescence in the soma. Targeted Ca2+ uncaging confirmed two major features of KCa3.1 channels: preferential somatodendritic localization and Ca2+-driven gating. In addition, both the sAHP and the slow Ca2+-induced hyperpolarizing current were sensitive to TRAM-34, a selective blocker of KCa3.1 channels.
Collapse
|
29
|
Forelimb movements evoked by optogenetic stimulation of the macaque motor cortex. Nat Commun 2020; 11:3253. [PMID: 32591505 PMCID: PMC7319997 DOI: 10.1038/s41467-020-16883-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 05/28/2020] [Indexed: 12/15/2022] Open
Abstract
Optogenetics has become an indispensable tool for investigating brain functions. Although non-human primates are particularly useful models for understanding the functions and dysfunctions of the human brain, application of optogenetics to non-human primates is still limited. In the present study, we generate an effective adeno-associated viral vector serotype DJ to express channelrhodopsin-2 (ChR2) under the control of a strong ubiquitous CAG promoter and inject into the somatotopically identified forelimb region of the primary motor cortex in macaque monkeys. ChR2 is strongly expressed around the injection sites, and optogenetic intracortical microstimulation (oICMS) through a homemade optrode induces prominent cortical activity: Even single-pulse, short-duration oICMS evokes long-lasting repetitive firings of cortical neurons. In addition, oICMS elicits distinct forelimb movements and muscle activity, which are comparable to those elicited by conventional electrical ICMS. The present study removes obstacles to optogenetic manipulation of neuronal activity and behaviors in non-human primates. Non-human primates are useful models for understanding the human brain but application of optogenetics to non-human primates is challenging. The authors used optogenetic intracortical microstimulation in the primary motor cortex of macaques to elicit distinct forelimb movements and muscle activity.
Collapse
|
30
|
Kopach O, Esteras N, Wray S, Rusakov DA, Abramov AY. Maturation and phenotype of pathophysiological neuronal excitability of human cells in tau-related dementia. J Cell Sci 2020; 133:jcs241687. [PMID: 32299835 PMCID: PMC7272359 DOI: 10.1242/jcs.241687] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 04/01/2020] [Indexed: 01/23/2023] Open
Abstract
Frontotemporal dementia and parkinsonism (FTDP-17) caused by the 10+16 splice-site mutation in the gene encoding microtubule-associated protein tau (MAPT) provides an established platform to model tau-related dementia in vitro Neurons derived from human induced pluripotent stem cells (iPSCs) have been shown to recapitulate the neurodevelopmental profile of tau pathology during in vitro corticogenesis, as in the adult human brain. However, the neurophysiological phenotype of these cells has remained unknown, leaving unanswered questions regarding the functional relevance and the gnostic power of this disease model. In this study, we used electrophysiology to explore the membrane properties and intrinsic excitability of the generated neurons and found that human cells mature by ∼150 days of neurogenesis to become compatible with matured cortical neurons. In earlier FTDP-17, however, neurons exhibited a depolarized resting membrane potential associated with increased resistance and reduced voltage-gated Na+- and K+-channel-mediated conductance. Expression of the Nav1.6 protein was reduced in FTDP-17. These effects led to reduced cell capability of induced firing and changed the action potential waveform in FTDP-17. The revealed neuropathology might thus contribute to the clinicopathological profile of the disease. This sheds new light on the significance of human in vitro models of dementia.
Collapse
Affiliation(s)
- Olga Kopach
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Noemí Esteras
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Selina Wray
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WC1N 1PJ, UK
| | - Dmitri A Rusakov
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Andrey Y Abramov
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| |
Collapse
|
31
|
Tubert C, Murer MG. What’s wrong with the striatal cholinergic interneurons in Parkinson’s disease? Focus on intrinsic excitability. Eur J Neurosci 2020; 53:2100-2116. [DOI: 10.1111/ejn.14742] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/04/2020] [Accepted: 04/05/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Cecilia Tubert
- Instituto de Fisiología y Biofísica “Bernardo Houssay”, (IFIBIO‐Houssay) Grupo de Neurociencia de Sistemas Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires Argentina
| | - Mario Gustavo Murer
- Instituto de Fisiología y Biofísica “Bernardo Houssay”, (IFIBIO‐Houssay) Grupo de Neurociencia de Sistemas Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires Argentina
| |
Collapse
|
32
|
Zhang Y, Garcia E, Sack AS, Snutch TP. L-type calcium channel contributions to intrinsic excitability and synaptic activity during basolateral amygdala postnatal development. J Neurophysiol 2020; 123:1216-1235. [PMID: 31967931 DOI: 10.1152/jn.00606.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The amygdala contributes toward emotional processes such as fear, anxiety, and social cognition. Furthermore, evidence suggests that increased excitability of basolateral amygdala (BLA) principal neurons underlie certain neuropsychiatric disorders. Gain-of-function mutations in neuronal L-type calcium channels (LTCCs) are linked to neurodevelopmental diseases, including autism spectrum disorders (ASDs). While LTCCs are expressed throughout the BLA, direct evidence for increased LTCC activity affecting BLA excitability and potentially contributing to disease pathophysiology is lacking. In this study, we utilized a pharmacological approach to examine the contributions of LTCCs to BLA principal cell excitability and synaptic activity at immature (postnatal day 7, P7) and juvenile (P21) developmental stages. Acute upregulation of LTCC activity in brain slices by application of the agonist (S)-Bay K 8644 resulted in increased intrinsic excitability properties including firing frequency response, plateau potential, and spike-frequency adaptation selectively in P7 neurons. Contrastingly, for P21 neurons, the main effect of (S)-Bay K 8644 was to enhance burst firing. (S)-Bay K 8644 increased spontaneous inhibitory synaptic currents at both P7 and P21 but did not affect evoked synaptic currents at either stage. (S)-Bay K 8644 did not alter P7 spontaneous excitatory synaptic currents, although it increased current amplitude in P21 neurons. Overall, the results provide support for the notion that alteration of LTCC activity at specific periods of early brain development may lead to functional alterations to neuronal network activity and subsequently contribute to underlying mechanisms of amygdala-related neurological disorders.NEW & NOTEWORTHY The role of L-type calcium channels (LTCCs) in regulating neuronal electrophysiological properties during development remains unclear. We show that in basolateral amygdala principal neurons, an increase of LTCC activity alters both neuronal excitability and synaptic activity. The results also provide evidence for the distinct contributions of LTCCs at different stages of neurodevelopment and shed insight into our understanding of LTCC dysfunction in amygdala-related neurological disorders.
Collapse
Affiliation(s)
- Yiming Zhang
- Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Esperanza Garcia
- Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Anne-Sophie Sack
- Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Terrance P Snutch
- Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
33
|
Perturbed Ca2+-dependent signaling of DYT2 hippocalcin mutant as mechanism of autosomal recessive dystonia. Neurobiol Dis 2019; 132:104529. [DOI: 10.1016/j.nbd.2019.104529] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/22/2019] [Accepted: 07/09/2019] [Indexed: 11/23/2022] Open
|
34
|
Calcium Signaling in Neurons and Glial Cells: Role of Cav1 channels. Neuroscience 2019; 421:95-111. [DOI: 10.1016/j.neuroscience.2019.09.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 11/18/2022]
|
35
|
Kuznik BI, Davydov SO, Popravka ES, Lin’kova NS, Kozina LS, Khavinson VK. Epigenetic Mechanisms of Peptide-Driven Regulation and Neuroprotective Protein FKBP1b. Mol Biol 2019. [DOI: 10.1134/s0026893319020092] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
36
|
Chen KF, Lowe S, Lamaze A, Krätschmer P, Jepson J. Neurocalcin regulates nighttime sleep and arousal in Drosophila. eLife 2019; 8:e38114. [PMID: 30865587 PMCID: PMC6415939 DOI: 10.7554/elife.38114] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 01/29/2019] [Indexed: 01/28/2023] Open
Abstract
Sleep-like states in diverse organisms can be separated into distinct stages, each with a characteristic arousal threshold. However, the molecular pathways underlying different sleep stages remain unclear. The fruit fly, Drosophila melanogaster, exhibits consolidated sleep during both day and night, with night sleep associated with higher arousal thresholds compared to day sleep. Here we identify a role for the neuronal calcium sensor protein Neurocalcin (NCA) in promoting sleep during the night but not the day by suppressing nocturnal arousal and hyperactivity. We show that both circadian and light-sensing pathways define the temporal window in which NCA promotes sleep. Furthermore, we find that NCA promotes sleep by suppressing synaptic release from a dispersed wake-promoting neural network and demonstrate that the mushroom bodies, a sleep-regulatory center, are a module within this network. Our results advance the understanding of how sleep stages are genetically defined.
Collapse
Affiliation(s)
- Ko-Fan Chen
- Department of Clinical and Experimental EpilepsyUCL Institute of NeurologyLondonUnited Kingdom
| | - Simon Lowe
- Department of Clinical and Experimental EpilepsyUCL Institute of NeurologyLondonUnited Kingdom
| | - Angélique Lamaze
- Department of Clinical and Experimental EpilepsyUCL Institute of NeurologyLondonUnited Kingdom
| | - Patrick Krätschmer
- Department of Clinical and Experimental EpilepsyUCL Institute of NeurologyLondonUnited Kingdom
| | - James Jepson
- Department of Clinical and Experimental EpilepsyUCL Institute of NeurologyLondonUnited Kingdom
| |
Collapse
|
37
|
Bajorat R, Porath K, Kuhn J, Goßla E, Goerss D, Sellmann T, Köhling R, Kirschstein T. Oral administration of the casein kinase 2 inhibitor TBB leads to persistent K Ca2.2 channel up-regulation in the epileptic CA1 area and cortex, but lacks anti-seizure efficacy in the pilocarpine epilepsy model. Epilepsy Res 2018; 147:42-50. [PMID: 30219695 DOI: 10.1016/j.eplepsyres.2018.08.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/31/2018] [Accepted: 08/29/2018] [Indexed: 02/04/2023]
Abstract
Temporal lobe epilepsy (TLE) is the most common epileptic syndrome in adults and often presents with seizures that prove intractable with currently available anticonvulsants. Thus, there is still a need for new anti-seizure drugs in this condition. Recently, we found that the casein kinase 2 inhibitor 4,5,6,7-tetrabromotriazole (TBB) prevented the emergence of spontaneous epileptic discharges in an acute in vitro epilepsy model. This prompted us to study the anti-seizure effects of TBB in the pilocarpine model of chronic epilepsy in vivo. To this end, we performed long-term video-EEG monitoring lasting 78-167 days of nine chronically epileptic rats and obtained a baseline seizure rate of 3.3 ± 1.3 per day (baseline of 27-80 days). We found a significant age effect with more pronounced seizure rates in older animals as compared to younger ones. However, the seizure rate increased to 6.3 ± 2.2 per day during the oral TBB administration (treatment period of 21-50 days), and following discontinuation of TBB, this rate remained stable with 5.2 ± 1.4 seizures per day (follow-up of 30-55 days). After completing the video-EEG during the follow-up the hippocampal tissue was prepared and studied for the expression of the Ca2+-activated K+ channel KCa2.2. We found a significant up-regulation of KCa2.2 in the epileptic CA1 region and in the neocortex, but in no other hippocampal subfield. Hence, our findings indicate that oral administration of TBB leads to persistent up-regulation of KCa2.2 in the epileptic CA1 subfield and in the neocortex, but lacks anti-seizure efficacy in the pilocarpine epilepsy model.
Collapse
Affiliation(s)
- Rika Bajorat
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany; Department of Anesthesiology and Intensive Care Medicine, Rostock University Medical Centre, Rostock, Germany.
| | - Katrin Porath
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany.
| | - Johannes Kuhn
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany.
| | - Elke Goßla
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany.
| | - Doreen Goerss
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany; Department of Psychosomatic and Psychotherapeutic Medicine, Rostock University Medical Centre, Rostock, Germany.
| | - Tina Sellmann
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany.
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany.
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology, Rostock University Medical Centre, Rostock, Germany.
| |
Collapse
|
38
|
Kirchner MK, Foehring RC, Callaway J, Armstrong WE. Specificity in the interaction of high-voltage-activated Ca 2+ channel types with Ca 2+-dependent afterhyperpolarizations in magnocellular supraoptic neurons. J Neurophysiol 2018; 120:1728-1739. [PMID: 30020842 DOI: 10.1152/jn.00285.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Magnocellular oxytocin (OT) and vasopressin (VP) neurons express an afterhyperpolarization (AHP) following spike trains that attenuates firing rate and contributes to burst patterning. This AHP includes contributions from an apamin-sensitive, medium-duration AHP (mAHP) and from an apamin-insensitive, slow-duration AHP (sAHP). These AHPs are Ca2+ dependent and activated by Ca2+ influx through voltage-gated Ca2+ channels. Across central nervous system neurons that generate Ca2+-dependent AHPs, the Ca2+ channels that couple to the mAHP and sAHP differ greatly, but for magnocellular neurosecretory cells this relationship is unknown. Using simultaneous whole cell recording and Ca2+ imaging, we evaluated the effect of specific high-voltage-activated (HVA) Ca2+ channel blockers on the mAHP and sAHP. Block of all HVA channels via 400 μM Cd2+ inhibited almost the entire AHP. We tested nifedipine, conotoxin GVIA, agatoxin IVA, and SNX-482, specific blockers of L-, N-, P/Q-, and R-type channels, respectively. The N-type channel blocker conotoxin GVIA (1 μM) was the only toxin that inhibited the mAHP in either OT or VP neurons although the effect on VP neurons was weaker by comparison. The sAHP was significantly inhibited by N-type block in OT neurons and by R-type block in VP neurons although neither accounted for the entirety of the sAHP. Thus the mAHP appears to be elicited by Ca2+ from mostly N-type channels in both OT and VP neurons, but the contributions of specific Ca2+ channel types to the sAHP in each cell type are different. Alternative sources to HVA channels may contribute Ca2+ for the sAHP. NEW & NOTEWORTHY Despite the importance of afterhyperpolarization (AHP) mechanisms for regulating firing behavior of oxytocin (OT) and vasopressin (VP) neurons of supraoptic nucleus, which types of high-voltage-activated Ca2+ channels elicit AHPs in these cells was unknown. We found that N-type channels couple to the medium AHP in both cell types. For the slow AHP, N-type channels contribute in OT neurons, whereas R-type contribute in VP neurons. No single Ca2+ channel blocker abolished the entire AHP, suggesting that additional Ca2+ sources are involved.
Collapse
Affiliation(s)
- Matthew K Kirchner
- University of Tennessee Health Science Center, Department of Anatomy & Neurobiology
| | - Robert C Foehring
- University of Tennessee Health Science Center, Department of Anatomy & Neurobiology
| | - Joseph Callaway
- University of Tennessee Health Science Center, Department of Anatomy & Neurobiology
| | - William E Armstrong
- University of Tennessee Health Science Center, Department of Anatomy & Neurobiology
| |
Collapse
|
39
|
Fardet T, Ballandras M, Bottani S, Métens S, Monceau P. Understanding the Generation of Network Bursts by Adaptive Oscillatory Neurons. Front Neurosci 2018; 12:41. [PMID: 29467607 PMCID: PMC5808224 DOI: 10.3389/fnins.2018.00041] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 01/17/2018] [Indexed: 12/26/2022] Open
Abstract
Experimental and numerical studies have revealed that isolated populations of oscillatory neurons can spontaneously synchronize and generate periodic bursts involving the whole network. Such a behavior has notably been observed for cultured neurons in rodent's cortex or hippocampus. We show here that a sufficient condition for this network bursting is the presence of an excitatory population of oscillatory neurons which displays spike-driven adaptation. We provide an analytic model to analyze network bursts generated by coupled adaptive exponential integrate-and-fire neurons. We show that, for strong synaptic coupling, intrinsically tonic spiking neurons evolve to reach a synchronized intermittent bursting state. The presence of inhibitory neurons or plastic synapses can then modulate this dynamics in many ways but is not necessary for its appearance. Thanks to a simple self-consistent equation, our model gives an intuitive and semi-quantitative tool to understand the bursting behavior. Furthermore, it suggests that after-hyperpolarization currents are sufficient to explain bursting termination. Through a thorough mapping between the theoretical parameters and ion-channel properties, we discuss the biological mechanisms that could be involved and the relevance of the explored parameter-space. Such an insight enables us to propose experimentally-testable predictions regarding how blocking fast, medium or slow after-hyperpolarization channels would affect the firing rate and burst duration, as well as the interburst interval.
Collapse
Affiliation(s)
- Tanguy Fardet
- Laboratoire Matière et Systèmes Complexes, UMR 7057, Université Paris Diderot, USPC, Paris, France
| | - Mathieu Ballandras
- Laboratoire Matière et Systèmes Complexes, UMR 7057, Université Paris Diderot, USPC, Paris, France
| | - Samuel Bottani
- Laboratoire Matière et Systèmes Complexes, UMR 7057, Université Paris Diderot, USPC, Paris, France
| | - Stéphane Métens
- Laboratoire Matière et Systèmes Complexes, UMR 7057, Université Paris Diderot, USPC, Paris, France
| | - Pascal Monceau
- Laboratoire Matière et Systèmes Complexes, UMR 7057, Université Paris Diderot, USPC, Paris, France.,Department of Physics, Université d'Evry-Val d'Essonne, Évry, France
| |
Collapse
|
40
|
Beining M, Mongiat LA, Schwarzacher SW, Cuntz H, Jedlicka P. T2N as a new tool for robust electrophysiological modeling demonstrated for mature and adult-born dentate granule cells. eLife 2017; 6:e26517. [PMID: 29165247 PMCID: PMC5737656 DOI: 10.7554/elife.26517] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 11/21/2017] [Indexed: 12/18/2022] Open
Abstract
Compartmental models are the theoretical tool of choice for understanding single neuron computations. However, many models are incomplete, built ad hoc and require tuning for each novel condition rendering them of limited usability. Here, we present T2N, a powerful interface to control NEURON with Matlab and TREES toolbox, which supports generating models stable over a broad range of reconstructed and synthetic morphologies. We illustrate this for a novel, highly detailed active model of dentate granule cells (GCs) replicating a wide palette of experiments from various labs. By implementing known differences in ion channel composition and morphology, our model reproduces data from mouse or rat, mature or adult-born GCs as well as pharmacological interventions and epileptic conditions. This work sets a new benchmark for detailed compartmental modeling. T2N is suitable for creating robust models useful for large-scale networks that could lead to novel predictions. We discuss possible T2N application in degeneracy studies.
Collapse
Affiliation(s)
- Marcel Beining
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck SocietyFrankfurtGermany
- Frankfurt Institute for Advanced StudiesFrankfurtGermany
- Institute of Clinical Neuroanatomy, Neuroscience CenterGoethe UniversityFrankfurtGermany
- Faculty of BiosciencesGoethe UniversityFrankfurtGermany
| | - Lucas Alberto Mongiat
- Instituto de Investigación en Biodiversidad y MedioambienteUniversidad Nacional del Comahue-CONICETSan Carlos de BarilocheArgentina
| | | | - Hermann Cuntz
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck SocietyFrankfurtGermany
- Frankfurt Institute for Advanced StudiesFrankfurtGermany
| | - Peter Jedlicka
- Institute of Clinical Neuroanatomy, Neuroscience CenterGoethe UniversityFrankfurtGermany
| |
Collapse
|
41
|
Brown DA. Regulation of neural ion channels by muscarinic receptors. Neuropharmacology 2017; 136:383-400. [PMID: 29154951 DOI: 10.1016/j.neuropharm.2017.11.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 10/26/2017] [Accepted: 11/13/2017] [Indexed: 12/20/2022]
Abstract
The excitable behaviour of neurons is determined by the activity of their endogenous membrane ion channels. Since muscarinic receptors are not themselves ion channels, the acute effects of muscarinic receptor stimulation on neuronal function are governed by the effects of the receptors on these endogenous neuronal ion channels. This review considers some principles and factors determining the interaction between subtypes and classes of muscarinic receptors with neuronal ion channels, and summarizes the effects of muscarinic receptor stimulation on a number of different channels, the mechanisms of receptor - channel transduction and their direct consequences for neuronal activity. Ion channels considered include potassium channels (voltage-gated, inward rectifier and calcium activated), voltage-gated calcium channels, cation channels and chloride channels. This article is part of the Special Issue entitled 'Neuropharmacology on Muscarinic Receptors'.
Collapse
Affiliation(s)
- David A Brown
- Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
42
|
Activity-Dependent Facilitation of Ca V1.3 Calcium Channels Promotes KCa3.1 Activation in Hippocampal Neurons. J Neurosci 2017; 37:11255-11270. [PMID: 29038242 DOI: 10.1523/jneurosci.0967-17.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 10/02/2017] [Accepted: 10/07/2017] [Indexed: 11/21/2022] Open
Abstract
CaV1 L-type calcium channels are key to regulating neuronal excitability, with the range of functional roles enhanced by interactions with calmodulin, accessory proteins, or CaMKII that modulate channel activity. In hippocampal pyramidal cells, a prominent elevation of CaV1 activity is apparent in late channel openings that can last for seconds following a depolarizing stimulus train. The current study tested the hypothesis that a reported interaction among CaV1.3 channels, the scaffolding protein densin, and CaMKII could generate a facilitation of channel activity that outlasts a depolarizing stimulus. We found that CaV1.3 but not CaV1.2 channels exhibit a long-duration calcium-dependent facilitation (L-CDF) that lasts up to 8 s following a brief 50 Hz stimulus train, but only when coexpressed with densin and CaMKII. To test the physiological role for CaV1.3 L-CDF, we coexpressed the intermediate-conductance KCa3.1 potassium channel, revealing a strong functional coupling to CaV1.3 channel activity that was accentuated by densin and CaMKII. Moreover, the CaV1.3-densin-CaMKII interaction gave rise to an outward tail current of up to 8 s duration following a depolarizing stimulus in both tsA-201 cells and male rat CA1 pyramidal cells. A slow afterhyperpolarization in pyramidal cells was reduced by a selective block of CaV1 channels by isradipine, a CaMKII blocker, and siRNA knockdown of densin, and spike frequency increased upon selective block of CaV1 channel conductance. The results are important in revealing a CaV1.3-densin-CaMKII interaction that extends the contribution of CaV1.3 calcium influx to a time frame well beyond a brief input train.SIGNIFICANCE STATEMENT CaV1 L-type calcium channels play a key role in regulating the output of central neurons by providing calcium influx during repetitive inputs. This study identifies a long-duration calcium-dependent facilitation (L-CDF) of CaV1.3 channels that depends on the scaffolding protein densin and CaMKII and that outlasts a depolarizing stimulus by seconds. We further show a tight functional coupling between CaV1.3 calcium influx and the intermediate-conductance KCa3.1 potassium channel that promotes an outward tail current of up to 8 s following a depolarizing stimulus. Tests in CA1 hippocampal pyramidal cells reveal that a slow AHP is reduced by blocking different components of the CaV1.3-densin-CaMKII interaction, identifying an important role for CaV1.3 L-CDF in regulating neuronal excitability.
Collapse
|
43
|
McKiernan EC, Marrone DF. CA1 pyramidal cells have diverse biophysical properties, affected by development, experience, and aging. PeerJ 2017; 5:e3836. [PMID: 28948109 PMCID: PMC5609525 DOI: 10.7717/peerj.3836] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 08/31/2017] [Indexed: 12/04/2022] Open
Abstract
Neuron types (e.g., pyramidal cells) within one area of the brain are often considered homogeneous, despite variability in their biophysical properties. Here we review literature demonstrating variability in the electrical activity of CA1 hippocampal pyramidal cells (PCs), including responses to somatic current injection, synaptic stimulation, and spontaneous network-related activity. In addition, we describe how responses of CA1 PCs vary with development, experience, and aging, and some of the underlying ionic currents responsible. Finally, we suggest directions that may be the most impactful in expanding this knowledge, including the use of text and data mining to systematically study cellular heterogeneity in more depth; dynamical systems theory to understand and potentially classify neuron firing patterns; and mathematical modeling to study the interaction between cellular properties and network output. Our goals are to provide a synthesis of the literature for experimentalists studying CA1 PCs, to give theorists an idea of the rich diversity of behaviors models may need to reproduce to accurately represent these cells, and to provide suggestions for future research.
Collapse
Affiliation(s)
- Erin C McKiernan
- Departamento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Diano F Marrone
- Department of Psychology, Wilfrid Laurier University, Waterloo, Ontario, Canada.,McKnight Brain Institute, University of Arizona, Tucson, AZ, United States of America
| |
Collapse
|
44
|
Kirchner MK, Foehring RC, Wang L, Chandaka GK, Callaway JC, Armstrong WE. Phosphatidylinositol 4,5-bisphosphate (PIP 2 ) modulates afterhyperpolarizations in oxytocin neurons of the supraoptic nucleus. J Physiol 2017; 595:4927-4946. [PMID: 28383826 DOI: 10.1113/jp274219] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 04/04/2017] [Indexed: 01/19/2023] Open
Abstract
KEY POINTS Afterhyperpolarizations (AHPs) generated by repetitive action potentials in supraoptic magnocellular neurons regulate repetitive firing and spike frequency adaptation but relatively little is known about PIP2 's control of these AHPs. We examined how changes in PIP2 levels affected AHPs, somatic [Ca2+ ]i , and whole cell Ca2+ currents. Manipulations of PIP2 levels affected both medium and slow AHP currents in oxytocin (OT) neurons of the supraoptic nucleus. Manipulations of PIP2 levels did not modulate AHPs by influencing Ca2+ release from IP3 -triggered Ca2+ stores, suggesting more direct modulation of channels by PIP2 . PIP2 depletion reduced spike-evoked Ca2+ entry and voltage-gated Ca2+ currents. PIP2 appears to influence AHPs in OT neurons by reducing Ca2+ influx during spiking. ABSTRACT Oxytocin (OT)- and vasopressin (VP)-secreting magnocellular neurons of the supraoptic nucleus (SON) display calcium-dependent afterhyperpolarizations (AHPs) following a train of action potentials that are critical to shaping the firing patterns of these cells. Previous work demonstrated that the lipid phosphatidylinositol 4,5-bisphosphate (PIP2 ) enabled the slow AHP component (sAHP) in cortical pyramidal neurons. We investigated whether this phenomenon occurred in OT and VP neurons of the SON. Using whole cell recordings in coronal hypothalamic slices from adult female rats, we demonstrated that inhibition of PIP2 synthesis with wortmannin robustly blocked both the medium and slow AHP currents (ImAHP and IsAHP ) of OT, but not VP neurons with high affinity. We further tested this by introducing a water-soluble PIP2 analogue (diC8 -PIP2 ) into neurons, which in OT neurons not only prevented wortmannin's inhibitory effect, but slowed rundown of the ImAHP and IsAHP . Inhibition of phospholipase C (PLC) with U73122 did not inhibit either ImAHP or IsAHP in OT neurons, consistent with wortmannin's effects not being due to reducing diacylglycerol (DAG) or IP3 availability, i.e. PIP2 modulation of AHPs is not likely to involve downstream Ca2+ release from inositol 1,4,5-trisphosphate (IP3 )-triggered Ca2+ -store release, or channel modulation via DAG and protein kinase C (PKC). We found that wortmannin reduced [Ca2+ ]i increase induced by spike trains in OT neurons, but had no effect on AHPs evoked by uncaging intracellular Ca2+ . Finally, wortmannin selectively reduced whole cell Ca2+ currents in OT neurons while leaving VP neurons unaffected. The results indicate that PIP2 modulates both the ImAHP and IsAHP in OT neurons, most likely by controlling Ca2+ entry through voltage-gated Ca2+ channels opened during spike trains.
Collapse
Affiliation(s)
- Matthew K Kirchner
- Department of Anatomy and Neurobiology and Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Robert C Foehring
- Department of Anatomy and Neurobiology and Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Lie Wang
- Department of Anatomy and Neurobiology and Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Giri Kumar Chandaka
- Department of Anatomy and Neurobiology and Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Joseph C Callaway
- Department of Anatomy and Neurobiology and Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| | - William E Armstrong
- Department of Anatomy and Neurobiology and Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
45
|
Angstadt JD, Giordano JR, Goncalves AJ. 9-Phenanthrol modulates postinhibitory rebound and afterhyperpolarizing potentials in an excitatory motor neuron of the medicinal leech. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2017; 203:613-633. [DOI: 10.1007/s00359-017-1178-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 05/01/2017] [Accepted: 05/02/2017] [Indexed: 12/19/2022]
|
46
|
Kim KS, Duignan KM, Hawryluk JM, Soh H, Tzingounis AV. The Voltage Activation of Cortical KCNQ Channels Depends on Global PIP2 Levels. Biophys J 2016; 110:1089-98. [PMID: 26958886 DOI: 10.1016/j.bpj.2016.01.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 01/12/2016] [Accepted: 01/13/2016] [Indexed: 01/22/2023] Open
Abstract
The slow afterhyperpolarization (sAHP) is a calcium-activated potassium conductance with critical roles in multiple physiological processes. Pharmacological and genetic data suggest that KCNQ channels partly mediate the sAHP. However, these channels are not typically open within the observed voltage range of the sAHP. Recent work has shown that the sAHP is gated by increased PIP2 levels, which are generated downstream of calcium binding by neuronal calcium sensors such as hippocalcin. Here, we examined whether changes in PIP2 levels could shift the voltage-activation range of KCNQ channels. In HEK293T cells, expression of the PIP5 kinase PIPKIγ90, which increases global PIP2 levels, shifted the KCNQ voltage activation to within the operating range of the sAHP. Further, the sensitivity of this effect on KCNQ3 channels appeared to be higher than that on KCNQ2. Therefore, we predict that KCNQ3 plays an essential role in maintaining the sAHP under low PIP2 conditions. In support of this notion, we find that sAHP inhibition by muscarinic receptors that increase phosphoinositide turnover in neurons is enhanced in Kcnq3-knockout mice. Likewise, the presence of KCNQ3 is essential for maintaining the sAHP when hippocalcin is ablated, a condition that likely impairs PIP2 generation. Together, our results establish the relationship between PIP2 and the voltage dependence of cortical KCNQ channels (KCNQ2/3, KCNQ3/5, and KCNQ5), and suggest a possible mechanism for the involvement of KCNQ channels in the sAHP.
Collapse
Affiliation(s)
- Kwang S Kim
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut
| | - Kevin M Duignan
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut
| | - Joanna M Hawryluk
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut
| | - Heun Soh
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut
| | | |
Collapse
|
47
|
Hypocretin/Orexin Peptides Alter Spike Encoding by Serotonergic Dorsal Raphe Neurons through Two Distinct Mechanisms That Increase the Late Afterhyperpolarization. J Neurosci 2016; 36:10097-115. [PMID: 27683906 DOI: 10.1523/jneurosci.0635-16.2016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 08/11/2016] [Indexed: 01/02/2023] Open
Abstract
UNLABELLED Orexins (hypocretins) are neuropeptides that regulate multiple homeostatic processes, including reward and arousal, in part by exciting serotonergic dorsal raphe neurons, the major source of forebrain serotonin. Here, using mouse brain slices, we found that, instead of simply depolarizing these neurons, orexin-A altered the spike encoding process by increasing the postspike afterhyperpolarization (AHP) via two distinct mechanisms. This orexin-enhanced AHP (oeAHP) was mediated by both OX1 and OX2 receptors, required Ca(2+) influx, reversed near EK, and decayed with two components, the faster of which resulted from enhanced SK channel activation, whereas the slower component decayed like a slow AHP (sAHP), but was not blocked by UCL2077, an antagonist of sAHPs in some neurons. Intracellular phospholipase C inhibition (U73122) blocked the entire oeAHP, but neither component was sensitive to PKC inhibition or altered PKA signaling, unlike classical sAHPs. The enhanced SK current did not depend on IP3-mediated Ca(2+) release but resulted from A-current inhibition and the resultant spike broadening, which increased Ca(2+) influx and Ca(2+)-induced-Ca(2+) release, whereas the slower component was insensitive to these factors. Functionally, the oeAHP slowed and stabilized orexin-induced firing compared with firing produced by a virtual orexin conductance lacking the oeAHP. The oeAHP also reduced steady-state firing rate and firing fidelity in response to stimulation, without affecting the initial rate or fidelity. Collectively, these findings reveal a new orexin action in serotonergic raphe neurons and suggest that, when orexin is released during arousal and reward, it enhances the spike encoding of phasic over tonic inputs, such as those related to sensory, motor, and reward events. SIGNIFICANCE STATEMENT Orexin peptides are known to excite neurons via slow postsynaptic depolarizations. Here we elucidate a significant new orexin action that increases and prolongs the postspike afterhyperpolarization (AHP) in 5-HT dorsal raphe neurons and other arousal-system neurons. Our mechanistic studies establish involvement of two distinct Ca(2+)-dependent AHP currents dependent on phospholipase C signaling but independent of IP3 or PKC. Our functional studies establish that this action preserves responsiveness to phasic inputs while attenuating responsiveness to tonic inputs. Thus, our findings bring new insight into the actions of an important neuropeptide and indicate that, in addition to producing excitation, orexins can tune postsynaptic excitability to better encode the phasic sensory, motor, and reward signals expected during aroused states.
Collapse
|
48
|
Greene DL, Hoshi N. Modulation of Kv7 channels and excitability in the brain. Cell Mol Life Sci 2016; 74:495-508. [PMID: 27645822 DOI: 10.1007/s00018-016-2359-y] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 08/25/2016] [Accepted: 09/06/2016] [Indexed: 11/26/2022]
Abstract
Neuronal Kv7 channels underlie a voltage-gated non-inactivating potassium current known as the M-current. Due to its particular characteristics, Kv7 channels show pronounced control over the excitability of neurons. We will discuss various factors that have been shown to drastically alter the activity of this channel such as protein and phospholipid interactions, phosphorylation, calcium, and numerous neurotransmitters. Kv7 channels locate to key areas for the control of action potential initiation and propagation. Moreover, we will explore the dynamic surface expression of the channel modulated by neurotransmitters and neural activity. We will also focus on known principle functions of neural Kv7 channels: control of resting membrane potential and spiking threshold, setting the firing frequency, afterhyperpolarization after burst firing, theta resonance, and transient hyperexcitability from neurotransmitter-induced suppression of the M-current. Finally, we will discuss the contribution of altered Kv7 activity to pathologies such as epilepsy and cognitive deficits.
Collapse
Affiliation(s)
- Derek L Greene
- Department of Pharmacology, University of California, 360 Med Surge II, Irvine, CA, 92697, USA
| | - Naoto Hoshi
- Department of Pharmacology, University of California, 360 Med Surge II, Irvine, CA, 92697, USA.
- Department of Physiology and Biophysics, University of California, Irvine, USA.
| |
Collapse
|
49
|
Decrease of a Current Mediated by K v 1.3 Channels Causes Striatal Cholinergic Interneuron Hyperexcitability in Experimental Parkinsonism. Cell Rep 2016; 16:2749-2762. [DOI: 10.1016/j.celrep.2016.08.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 06/16/2016] [Accepted: 08/05/2016] [Indexed: 02/05/2023] Open
|
50
|
Turner RW, Asmara H, Engbers JDT, Miclat J, Rizwan AP, Sahu G, Zamponi GW. Assessing the role of IKCa channels in generating the sAHP of CA1 hippocampal pyramidal cells. Channels (Austin) 2016; 10:313-9. [PMID: 26950800 PMCID: PMC4954577 DOI: 10.1080/19336950.2016.1161988] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Our previous work reported that KCa3.1 (IKCa) channels are expressed in CA1 hippocampal pyramidal cells and contribute to the slow afterhyperpolarization that regulates spike accommodation in these cells. The current report presents data from single cell RT-PCR that further reveals mRNA in CA1 cells that corresponds to the sequence of an IKCa channel from transmembrane segments 5 through 6 including the pore region, revealing the established binding sites for 4 different IKCa channel blockers. A comparison of methods to internally apply the IKCa channel blocker TRAM-34 shows that including the drug in an electrode from the onset of an experiment is unviable given the speed of drug action upon gaining access for whole-cell recordings. Together the data firmly establish IKCa channel expression in CA1 neurons and clarify methodological requirements to obtain a block of IKCa channel activity through internal application of TRAM-34.
Collapse
Affiliation(s)
- Ray W Turner
- a Hotchkiss Brain Institute, University of Calgary , Calgary , AB , Canada
| | - Hadhimulya Asmara
- a Hotchkiss Brain Institute, University of Calgary , Calgary , AB , Canada
| | - Jordan D T Engbers
- a Hotchkiss Brain Institute, University of Calgary , Calgary , AB , Canada
| | - Jason Miclat
- a Hotchkiss Brain Institute, University of Calgary , Calgary , AB , Canada
| | - Arsalan P Rizwan
- a Hotchkiss Brain Institute, University of Calgary , Calgary , AB , Canada
| | - Giriraj Sahu
- a Hotchkiss Brain Institute, University of Calgary , Calgary , AB , Canada
| | - Gerald W Zamponi
- a Hotchkiss Brain Institute, University of Calgary , Calgary , AB , Canada
| |
Collapse
|