1
|
Mayadali ÜS, Chertes CAM, Sinicina I, Shaikh AG, Horn AKE. Ion channel profiles of extraocular motoneurons and internuclear neurons in human abducens and trochlear nuclei. Front Neuroanat 2024; 18:1411154. [PMID: 38957435 PMCID: PMC11217180 DOI: 10.3389/fnana.2024.1411154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 05/27/2024] [Indexed: 07/04/2024] Open
Abstract
Introduction Extraocular muscles are innervated by two anatomically and histochemically distinct motoneuron populations: motoneurons of multiply-innervated fibers (MIF), and of singly-innervated fibers (SIF). Recently, it has been established by our research group that these motoneuron types of monkey abducens and trochlear nuclei express distinct ion channel profiles: SIF motoneurons, as well as abducens internuclear neurons (INT), express strong Kv1.1 and Kv3.1b immunoreactivity, indicating their fast-firing capacity, whereas MIF motoneurons do not. Moreover, low voltage activated cation channels, such as Cav3.1 and HCN1 showed differences between MIF and SIF motoneurons, indicating distinct post-inhibitory rebound characteristics. However, the ion channel profiles of MIF and SIF motoneurons have not been established in human brainstem tissue. Methods Therefore, we used immunohistochemical methods with antibodies against Kv, Cav3 and HCN channels to (1) examine the human trochlear nucleus in terms of anatomical organization of MIF and SIF motoneurons, (2) examine immunolabeling patterns of ion channel proteins in the distinct motoneurons populations in the trochlear and abducens nuclei. Results In the examination of the trochlear nucleus, a third motoneuron subgroup was consistently encountered with weak perineuronal nets (PN). The neurons of this subgroup had -on average- larger diameters than MIF motoneurons, and smaller diameters than SIF motoneurons, and PN expression strength correlated with neuronal size. Immunolabeling of various ion channels revealed that, in general, human MIF and SIF motoneurons did not differ consistently, as opposed to the findings in monkey trochlear and abducens nuclei. Kv1.1, Kv3.1b and HCN channels were found on both MIF and SIF motoneurons and the immunolabeling density varied for multiple ion channels. On the other hand, significant differences between SIF motoneurons and INTs were found in terms of HCN1 immunoreactivity. Discussion These results indicated that motoneurons may be more variable in human in terms of histochemical and biophysiological characteristics, than previously thought. This study therefore establishes grounds for any histochemical examination of motor nuclei controlling extraocular muscles in eye movement related pathologies in the human brainstem.
Collapse
Affiliation(s)
- Ümit S. Mayadali
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, LMU Munich, Munich, Germany
| | - Christina A. M. Chertes
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, LMU Munich, Munich, Germany
| | - Inga Sinicina
- Faculty of Medicine, Institute of Legal Medicine, LMU Munich, Munich, Germany
| | - Aasef G. Shaikh
- Department of Neurology, University Hospitals, Cleveland VA Medical Center, Case Western Reserve University, Cleveland, OH, United States
| | - Anja K. E. Horn
- Chair of Vegetative Anatomy, Faculty of Medicine, Institute of Anatomy, LMU Munich, Munich, Germany
| |
Collapse
|
2
|
Yu B, Lu Q, Li J, Cheng X, Hu H, Li Y, Che T, Hua Y, Jiang H, Zhang Y, Xian C, Yang T, Fu Y, Chen Y, Nan W, McCormick PJ, Xiong B, Duan J, Zeng B, Li Y, Fu Y, Zhang J. Cryo-EM structure of human HCN3 channel and its regulation by cAMP. J Biol Chem 2024; 300:107288. [PMID: 38636662 PMCID: PMC11126801 DOI: 10.1016/j.jbc.2024.107288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 03/30/2024] [Accepted: 04/08/2024] [Indexed: 04/20/2024] Open
Abstract
HCN channels are important for regulating heart rhythm and nerve activity and have been studied as potential drug targets for treating depression, arrhythmia, nerve pain, and epilepsy. Despite possessing unique pharmacological properties, HCN channels share common characteristics in that they are activated by hyperpolarization and modulated by cAMP and other membrane lipids. However, the mechanisms of how these ligands bind and modulate HCN channels are unclear. In this study, we solved structures of full-length human HCN3 using cryo-EM and captured two different states, including a state without any ligand bound and a state with cAMP bound. Our structures reveal the novel binding sites for cholesteryl hemisuccinate in apo state and show how cholesteryl hemisuccinate and cAMP binding cause conformational changes in different states. These findings explain how these small modulators are sensed in mammals at the molecular level. The results of our study could help to design more potent and specific compounds to influence HCN channel activity and offer new therapeutic possibilities for diseases that lack effective treatment.
Collapse
Affiliation(s)
- Bo Yu
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China; The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qiuyuan Lu
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jian Li
- College of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Xinyu Cheng
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China; The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Han Hu
- Shenzhen Crystalo Biopharmaceutical Co, Ltd, Shenzhen, Guangdong, China
| | - Yuanshuo Li
- Shenzhen Crystalo Biopharmaceutical Co, Ltd, Shenzhen, Guangdong, China
| | - Tong Che
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China; The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yaoguang Hua
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China; The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Haihai Jiang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China; The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yuting Zhang
- Shenzhen Crystalo Biopharmaceutical Co, Ltd, Shenzhen, Guangdong, China
| | - Cuiling Xian
- Shenzhen Crystalo Biopharmaceutical Co, Ltd, Shenzhen, Guangdong, China
| | - Tingting Yang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China; The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Ying Fu
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China; The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yixiang Chen
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China; The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Weiwei Nan
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China; The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Peter J McCormick
- William Harvey Research Institute, Bart's and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Bing Xiong
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jingjing Duan
- Human Aging Research Institute (HARI), School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Bo Zeng
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Yanyan Li
- Department of Chemical Biology, School of Life Southern University of Science and Technology, Southern University of Science and Technology, Shenzhen, Guangdong, China; Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Yang Fu
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Jin Zhang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China; The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
3
|
Flerlage WJ, Simmons SC, Thomas EH, Gouty S, Tsuda MC, Wu TJ, Armstrong RC, Cox BM, Nugent FS. Effects of Repetitive Mild Traumatic Brain Injury on Corticotropin-Releasing Factor Modulation of Lateral Habenula Excitability and Motivated Behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.16.589760. [PMID: 38798343 PMCID: PMC11118357 DOI: 10.1101/2024.04.16.589760] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Mild traumatic brain injury (mTBI) is a significant health burden due to mTBI-related chronic debilitating cognitive and psychiatric morbidities. Recent evidence from our laboratory suggests a possible dysregulation within reward/motivational circuit function at the level of a subcortical structure, the lateral habenula (LHb), where we demonstrated a causal role for hyperactive LHb in mTBI-induced motivational deficits in self-care grooming behavior in young adult male mice when exposed to mTBI injury during late adolescence (at ~8 weeks old). Here we extended this observation by further characterizing neurobehavioral effects of this repetitive closed head injury model of mTBI in both young adult male and female mice on LHb excitability, corticotropin releasing factor (CRF) modulation of LHb activity, and behavioral responses of motivation to self-care behavior, and approach versus avoidance behavior in the presence of a social- or threat-related stimulus. We show that mTBI increases LHb spontaneous tonic activity in female mice similar to what we previously observed in male mice as well as promoting LHb neuronal hyperexcitability and hyperpolarization-induced LHb bursting in both male and female mice. Interestingly, mTBI only increases LHb intrinsic excitability in male mice coincident with higher levels of the hyperpolarization-activated cation currents (HCN/Ih) and reduces levels of the M-type potassium currents while potentiating M-currents without altering intrinsic excitability in LHb neurons of female mice. Since persistent dysregulation of brain CRF systems is suggested to contribute to chronic psychiatric morbidities and that LHb neurons are highly responsive to CRF, we then tested whether LHb CRF subsystem becomes engaged following mTBI. We found that in vitro inhibition of CRF receptor type 1 (CRFR1) within the LHb normalizes mTBI-induced enhancement of LHb tonic activity and hyperexcitability in both sexes, suggesting that an augmented intra-LHb CRF-CRFR1-mediated signaling contributes to the overall LHb hyperactivity following mTBI. Behaviorally, mTBI diminishes motivation for self-care grooming in female mice as in male mice. mTBI also alters defensive behaviors in the looming shadow task by shifting the innate defensive behaviors towards more passive action-locking rather than escape behaviors in response to an aerial threat in both male and female mice as well as prolonging the latency to escape responses in female mice. While, this model of mTBI reduces social preference in male mice, it induces higher social novelty seeking during the novel social encounters in both male and female mice. Overall, our study provides further translational validity for the use of this preclinical model of mTBI for investigation of mTBI-related reward circuit dysfunction and mood/motivation-related behavioral deficits in both sexes while uncovering a few sexually dimorphic neurobehavioral effects of this model that may differentially affect young males and females when exposed to this type of mTBI injury during late adolescence.
Collapse
Affiliation(s)
- William J. Flerlage
- Uniformed Services University of the Health Sciences, Department of Pharmacology and Molecular Therapeutics, Bethesda, Maryland 20814, USA
| | - Sarah C. Simmons
- Uniformed Services University of the Health Sciences, Department of Pharmacology and Molecular Therapeutics, Bethesda, Maryland 20814, USA
| | - Emily H. Thomas
- Uniformed Services University of the Health Sciences, Department of Pharmacology and Molecular Therapeutics, Bethesda, Maryland 20814, USA
| | - Shawn Gouty
- Uniformed Services University of the Health Sciences, Department of Pharmacology and Molecular Therapeutics, Bethesda, Maryland 20814, USA
| | - Mumeko C. Tsuda
- Preclinical Behavior and Modeling Core, Uniformed Services University of the Health Sciences, Bethesda, MD
| | - T. John Wu
- Uniformed Services University of the Health Sciences, Department of Gynecologic Surgery and Obstetrics, Bethesda, MD 20814
| | - Regina C. Armstrong
- Uniformed Services University of the Health Sciences, Department of Anatomy, Physiology and Genetics, Bethesda, Maryland 20814, USA
| | - Brian M. Cox
- Uniformed Services University of the Health Sciences, Department of Pharmacology and Molecular Therapeutics, Bethesda, Maryland 20814, USA
| | - Fereshteh S. Nugent
- Uniformed Services University of the Health Sciences, Department of Pharmacology and Molecular Therapeutics, Bethesda, Maryland 20814, USA
| |
Collapse
|
4
|
Burrack N, Yitzhaky A, Mizrahi L, Wang M, Stern S, Hertzberg L. Altered Expression of PDE4 Genes in Schizophrenia: Insights from a Brain and Blood Sample Meta-Analysis and iPSC-Derived Neurons. Genes (Basel) 2024; 15:609. [PMID: 38790238 PMCID: PMC11121586 DOI: 10.3390/genes15050609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 05/03/2024] [Accepted: 05/05/2024] [Indexed: 05/26/2024] Open
Abstract
Schizophrenia symptomatology includes negative symptoms and cognitive impairment. Several studies have linked schizophrenia with the PDE4 family of enzymes due to their genetic association and function in cognitive processes such as long-term potentiation. We conducted a systematic gene expression meta-analysis of four PDE4 genes (PDE4A-D) in 10 brain sample datasets (437 samples) and three blood sample datasets (300 samples). Subsequently, we measured mRNA levels in iPSC-derived hippocampal dentate gyrus neurons generated from fibroblasts of three groups: healthy controls, healthy monozygotic twins (MZ), and their MZ siblings with schizophrenia. We found downregulation of PDE4B in brain tissues, further validated by independent data of the CommonMind consortium (515 samples). Interestingly, the downregulation signal was present in a subgroup of the patients, while the others showed no differential expression or even upregulation. Notably, PDE4A, PDE4B, and PDE4D exhibited upregulation in iPSC-derived neurons compared to healthy controls, whereas in blood samples, PDE4B was found to be upregulated while PDE4A was downregulated. While the precise mechanism and direction of altered PDE4 expression necessitate further investigation, the observed multilevel differential expression across the brain, blood, and iPSC-derived neurons compellingly suggests the involvement of PDE4 genes in the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Nitzan Burrack
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel;
- Clinical Research Center, Soroka University Medical Center, Beer-Sheva 84101, Israel
| | - Assif Yitzhaky
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Liron Mizrahi
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3103301, Israel
| | - Meiyan Wang
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3103301, Israel
| | - Libi Hertzberg
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot 76100, Israel
- Shalvata Mental Health Center, Affiliated with the Faculty of Medicine, Tel-Aviv University, 13 Aliat Hanoar St., Hod Hasharon 45100, Israel
| |
Collapse
|
5
|
Abstract
Repetitive transcranial magnetic stimulation (rTMS) has become an increasingly popular tool to modulate neural excitability and induce neural plasticity in clinical and preclinical models; however, the physiological mechanisms in which it exerts these effects remain largely unknown. To date, studies have primarily focused on characterizing rTMS-induced changes occurring at the synapse, with little attention given to changes in intrinsic membrane properties. However, accumulating evidence suggests that rTMS may induce its effects, in part, via intrinsic plasticity mechanisms, suggesting a new and potentially complementary understanding of how rTMS alters neural excitability and neural plasticity. In this review, we provide an overview of several intrinsic plasticity mechanisms before reviewing the evidence for rTMS-induced intrinsic plasticity. In addition, we discuss a select number of neurological conditions where rTMS-induced intrinsic plasticity has therapeutic potential before speculating on the temporal relationship between rTMS-induced intrinsic and synaptic plasticity.
Collapse
Affiliation(s)
- Emily S King
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Perth, Australia
- Perron Institute for Neurological and Translational Science, Perth, Australia
| | - Alexander D Tang
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Perth, Australia
- Perron Institute for Neurological and Translational Science, Perth, Australia
| |
Collapse
|
6
|
Kim D, Roh H, Lee HM, Kim SJ, Im M. Localization of hyperpolarization-activated cyclic nucleotide-gated channels in the vertebrate retinas across species and their physiological roles. Front Neuroanat 2024; 18:1385932. [PMID: 38562955 PMCID: PMC10982330 DOI: 10.3389/fnana.2024.1385932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
Transmembrane proteins known as hyperpolarization-activated cyclic nucleotide-gated (HCN) channels control the movement of Na+ and K+ ions across cellular membranes. HCN channels are known to be involved in crucial physiological functions in regulating neuronal excitability and rhythmicity, and pacemaker activity in the heart. Although HCN channels have been relatively well investigated in the brain, their distribution and function in the retina have received less attention, remaining their physiological roles to be comprehensively understood. Also, because recent studies reported HCN channels have been somewhat linked with the dysfunction of photoreceptors which are affected by retinal diseases, investigating HCN channels in the retina may offer valuable insights into disease mechanisms and potentially contribute to identifying novel therapeutic targets for retinal degenerative disorders. This paper endeavors to summarize the existing literature on the distribution and function of HCN channels reported in the vertebrate retinas of various species and discuss the potential implications for the treatment of retinal diseases. Then, we recapitulate current knowledge regarding the function and regulation of HCN channels, as well as their relevance to various neurological disorders.
Collapse
Affiliation(s)
- Daniel Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University (SNU), Seoul, Republic of Korea
| | - Hyeonhee Roh
- School of Electrical Engineering, College of Engineering, Korea University, Seoul, Republic of Korea
| | - Hyung-Min Lee
- School of Electrical Engineering, College of Engineering, Korea University, Seoul, Republic of Korea
| | - Sang Jeong Kim
- Department of Biomedical Sciences, College of Medicine, Seoul National University (SNU), Seoul, Republic of Korea
| | - Maesoon Im
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, University of Science & Technology (UST), Seoul, Republic of Korea
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
7
|
Nguyen LH, Xu Y, Nair M, Bordey A. The mTOR pathway genes MTOR, Rheb, Depdc5, Pten, and Tsc1 have convergent and divergent impacts on cortical neuron development and function. eLife 2024; 12:RP91010. [PMID: 38411613 PMCID: PMC10942629 DOI: 10.7554/elife.91010] [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: 02/28/2024] Open
Abstract
Brain somatic mutations in various components of the mTOR complex 1 (mTORC1) pathway have emerged as major causes of focal malformations of cortical development and intractable epilepsy. While these distinct gene mutations converge on excessive mTORC1 signaling and lead to common clinical manifestations, it remains unclear whether they cause similar cellular and synaptic disruptions underlying cortical network hyperexcitability. Here, we show that in utero activation of the mTORC1 activator genes, Rheb or MTOR, or biallelic inactivation of the mTORC1 repressor genes, Depdc5, Tsc1, or Pten in the mouse medial prefrontal cortex leads to shared alterations in pyramidal neuron morphology, positioning, and membrane excitability but different changes in excitatory synaptic transmission. Our findings suggest that, despite converging on mTORC1 signaling, mutations in different mTORC1 pathway genes differentially impact cortical excitatory synaptic activity, which may confer gene-specific mechanisms of hyperexcitability and responses to therapeutic intervention.
Collapse
Affiliation(s)
- Lena H Nguyen
- Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at DallasRichardsonUnited States
- Departments of Neurosurgery and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of MedicineNew HavenUnited States
| | - Youfen Xu
- Departments of Neurosurgery and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of MedicineNew HavenUnited States
| | - Maanasi Nair
- Departments of Neurosurgery and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of MedicineNew HavenUnited States
| | - Angelique Bordey
- Departments of Neurosurgery and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of MedicineNew HavenUnited States
| |
Collapse
|
8
|
Nguyen LH, Xu Y, Nair M, Bordey A. The mTOR pathway genes mTOR, Rheb, Depdc5, Pten, and Tsc1 have convergent and divergent impacts on cortical neuron development and function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.11.553034. [PMID: 37609221 PMCID: PMC10441381 DOI: 10.1101/2023.08.11.553034] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Brain somatic mutations in various components of the mTOR complex 1 (mTORC1) pathway have emerged as major causes of focal malformations of cortical development and intractable epilepsy. While these distinct gene mutations converge on excessive mTORC1 signaling and lead to common clinical manifestations, it remains unclear whether they cause similar cellular and synaptic disruptions underlying cortical network hyperexcitability. Here, we show that in utero activation of the mTORC1 activators, Rheb or mTOR, or biallelic inactivation of the mTORC1 repressors, Depdc5, Tsc1, or Pten in mouse medial prefrontal cortex leads to shared alterations in pyramidal neuron morphology, positioning, and membrane excitability but different changes in excitatory synaptic transmission. Our findings suggest that, despite converging on mTORC1 signaling, mutations in different mTORC1 pathway genes differentially impact cortical excitatory synaptic activity, which may confer gene-specific mechanisms of hyperexcitability and responses to therapeutic intervention.
Collapse
Affiliation(s)
- Lena H. Nguyen
- Department Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
- Departments of Neurosurgery and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Youfen Xu
- Departments of Neurosurgery and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Maanasi Nair
- Departments of Neurosurgery and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Angelique Bordey
- Departments of Neurosurgery and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
9
|
Su T, Lu Y, Fu C, Geng Y, Chen Y. GluN2A mediates ketamine-induced rapid antidepressant-like responses. Nat Neurosci 2023; 26:1751-1761. [PMID: 37709995 DOI: 10.1038/s41593-023-01436-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 08/16/2023] [Indexed: 09/16/2023]
Abstract
Ketamine was thought to induce rapid antidepressant responses by inhibiting GluN2B-containing N-methyl-D-aspartic acid (NMDA) receptors (NMDARs), which presents a promising opportunity to develop better antidepressants. However, adverse side effects limit the broader application of ketamine and GluN2B inhibitors are yet to be approved for clinical use. It is unclear whether ketamine acts solely through GluN2B-dependent mechanisms. The present study reports that the loss of another major NMDAR subunit, GluN2A, in adult mouse brains elicits robust antidepressant-like responses with limited impact on the behaviors that mimic the psychomimetic effects of ketamine. The antidepressant-like behavioral effects of broad NMDAR channel blockers, such as ketamine and MK-801 (dizocilpine), were mediated by the suppression of GluN2A, but not by the inhibition of GluN2B. Moreover, treatment with ketamine or MK-801 rapidly increased the intrinsic excitability of hippocampal principal neurons through GluN2A, but not GluN2B. Together, these findings indicate that GluN2A mediates ketamine-triggered rapid antidepressant-like responses.
Collapse
Affiliation(s)
- Tonghui Su
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Synphatec (Shanghai) Biopharmaceutical Technology Co., Ltd, Shanghai, China
| | - Yi Lu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chaoying Fu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Yang Geng
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
- Synphatec (Shanghai) Biopharmaceutical Technology Co., Ltd, Shanghai, China.
| | - Yelin Chen
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
10
|
Pruunsild P, Bengtson CP, Loss I, Lohrer B, Bading H. Expression of the primate-specific LINC00473 RNA in mouse neurons promotes excitability and CREB-regulated transcription. J Biol Chem 2023; 299:104671. [PMID: 37019214 DOI: 10.1016/j.jbc.2023.104671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
The LINC00473 (Lnc473) gene has previously been shown to be associated with cancer and psychiatric disorders. Its expression is elevated in several types of tumors and decreased in the brains of patients diagnosed with schizophrenia or major depression. In neurons, Lnc473 transcription is strongly responsive to synaptic activity, suggesting a role in adaptive, plasticity-related mechanisms. However, the function of Lnc473 is largely unknown. Here, using a recombinant adeno-associated viral vector, we introduced a primate-specific human Lnc473 RNA into mouse primary neurons. We show that this resulted in a transcriptomic shift comprising downregulation of epilepsy-associated genes and a rise in cAMP response element binding protein (CREB) activity, which was driven by augmented CREB-regulated transcription coactivator 1 (CRTC1) nuclear localization. Moreover, we demonstrate that ectopic Lnc473 expression increased neuronal excitability as well as network excitability. These findings suggest that primates may possess a lineage-specific activity-dependent modulator of CREB-regulated neuronal excitability.
Collapse
|
11
|
Involvement of Potassium Channel Signalling in Migraine Pathophysiology. Pharmaceuticals (Basel) 2023; 16:ph16030438. [PMID: 36986537 PMCID: PMC10057509 DOI: 10.3390/ph16030438] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/28/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Abstract
Migraine is a primary headache disorder ranked as the leading cause of years lived with disability among individuals younger than 50 years. The aetiology of migraine is complex and might involve several molecules of different signalling pathways. Emerging evidence implicates potassium channels, predominantly ATP-sensitive potassium (KATP) channels and large (big) calcium-sensitive potassium (BKCa) channels in migraine attack initiation. Basic neuroscience revealed that stimulation of potassium channels activated and sensitized trigeminovascular neurons. Clinical trials showed that administration of potassium channel openers caused headache and migraine attack associated with dilation of cephalic arteries. The present review highlights the molecular structure and physiological function of KATP and BKCa channels, presents recent insights into the role of potassium channels in migraine pathophysiology, and discusses possible complementary effects and interdependence of potassium channels in migraine attack initiation.
Collapse
|
12
|
Depolarization and Hyperexcitability of Cortical Motor Neurons after Spinal Cord Injury Associates with Reduced HCN Channel Activity. Int J Mol Sci 2023; 24:ijms24054715. [PMID: 36902146 PMCID: PMC10003573 DOI: 10.3390/ijms24054715] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/05/2023] Open
Abstract
A spinal cord injury (SCI) damages the axonal projections of neurons residing in the neocortex. This axotomy changes cortical excitability and results in dysfunctional activity and output of infragranular cortical layers. Thus, addressing cortical pathophysiology after SCI will be instrumental in promoting recovery. However, the cellular and molecular mechanisms of cortical dysfunction after SCI are poorly resolved. In this study, we determined that the principal neurons of the primary motor cortex layer V (M1LV), those suffering from axotomy upon SCI, become hyperexcitable following injury. Therefore, we questioned the role of hyperpolarization cyclic nucleotide gated channels (HCN channels) in this context. Patch clamp experiments on axotomized M1LV neurons and acute pharmacological manipulation of HCN channels allowed us to resolve a dysfunctional mechanism controlling intrinsic neuronal excitability one week after SCI. Some axotomized M1LV neurons became excessively depolarized. In those cells, the HCN channels were less active and less relevant to control neuronal excitability because the membrane potential exceeded the window of HCN channel activation. Care should be taken when manipulating HCN channels pharmacologically after SCI. Even though the dysfunction of HCN channels partakes in the pathophysiology of axotomized M1LV neurons, their dysfunctional contribution varies remarkably between neurons and combines with other pathophysiological mechanisms.
Collapse
|
13
|
Szegedi V, Bakos E, Furdan S, Kovács BH, Varga D, Erdélyi M, Barzó P, Szücs A, Tamás G, Lamsa K. HCN channels at the cell soma ensure the rapid electrical reactivity of fast-spiking interneurons in human neocortex. PLoS Biol 2023; 21:e3002001. [PMID: 36745683 PMCID: PMC9934405 DOI: 10.1371/journal.pbio.3002001] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 02/16/2023] [Accepted: 01/17/2023] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence indicates that there are substantial species differences in the properties of mammalian neurons, yet theories on circuit activity and information processing in the human brain are based heavily on results obtained from rodents and other experimental animals. This knowledge gap may be particularly important for understanding the neocortex, the brain area responsible for the most complex neuronal operations and showing the greatest evolutionary divergence. Here, we examined differences in the electrophysiological properties of human and mouse fast-spiking GABAergic basket cells, among the most abundant inhibitory interneurons in cortex. Analyses of membrane potential responses to current input, pharmacologically isolated somatic leak currents, isolated soma outside-out patch recordings, and immunohistochemical staining revealed that human neocortical basket cells abundantly express hyperpolarization-activated cyclic nucleotide-gated cation (HCN) channel isoforms HCN1 and HCN2 at the cell soma membrane, whereas these channels are sparse at the rodent basket cell soma membrane. Antagonist experiments showed that HCN channels in human neurons contribute to the resting membrane potential and cell excitability at the cell soma, accelerate somatic membrane potential kinetics, and shorten the lag between excitatory postsynaptic potentials and action potential generation. These effects are important because the soma of human fast-spiking neurons without HCN channels exhibit low persistent ion leak and slow membrane potential kinetics, compared with mouse fast-spiking neurons. HCN channels speed up human cell membrane potential kinetics and help attain an input-output rate close to that of rodent cells. Computational modeling demonstrated that HCN channel activity at the human fast-spiking cell soma membrane is sufficient to accelerate the input-output function as observed in cell recordings. Thus, human and mouse fast-spiking neurons exhibit functionally significant differences in ion channel composition at the cell soma membrane to set the speed and fidelity of their input-output function. These HCN channels ensure fast electrical reactivity of fast-spiking cells in human neocortex.
Collapse
Affiliation(s)
- Viktor Szegedi
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
- Hungarian Centre of Excellence for Molecular Medicine Research Group for Human neuron physiology and therapy, Szeged, Hungary
| | - Emőke Bakos
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
- Hungarian Centre of Excellence for Molecular Medicine Research Group for Human neuron physiology and therapy, Szeged, Hungary
| | - Szabina Furdan
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
- Hungarian Centre of Excellence for Molecular Medicine Research Group for Human neuron physiology and therapy, Szeged, Hungary
| | - Bálint H. Kovács
- Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - Dániel Varga
- Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - Miklós Erdélyi
- Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - Pál Barzó
- Department of Neurosurgery, University of Szeged, Szeged, Hungary
| | - Attila Szücs
- Hungarian Centre of Excellence for Molecular Medicine Research Group for Human neuron physiology and therapy, Szeged, Hungary
- Neuronal Cell Biology Research Group, Eötvös Loránd University, Budapest, Budapest, Hungary
| | - Gábor Tamás
- MTA-SZTE Research Group for Cortical Microcircuits, Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
| | - Karri Lamsa
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
- Hungarian Centre of Excellence for Molecular Medicine Research Group for Human neuron physiology and therapy, Szeged, Hungary
- * E-mail: ,
| |
Collapse
|
14
|
Miura Y, Shanley MR, Urbaez A, Friedman AK. Electrophysiologically distinct bed nucleus of the stria terminalis projections to the ventral tegmental area in mice. Front Neural Circuits 2023; 16:1081099. [PMID: 36698552 PMCID: PMC9870318 DOI: 10.3389/fncir.2022.1081099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/13/2022] [Indexed: 01/11/2023] Open
Abstract
The bed nucleus of the stria terminalis (BNST) is a highly heterogeneous limbic forebrain structure that serves as a relay connecting autonomic, neuroendocrine and behavioral function. It can be divided into over 16 individual subregions with distinct neuronal subpopulations based on receptors, transmitters, and neuropeptides. Specifically, the BNST projection to the ventral tegmental area (VTA), the dopamine hub of the brain, has been shown to have a crucial role in the stress response. However, in mice there is a lack of unbiased data on the functional diversity of this sub-population which serves as an upstream input to the VTA. The dopaminergic neurons in the VTA modify their ion channel activity and intrinsic membrane properties to adapt to stress in part from inputs from BNST projections. Therefore, we aimed to perform a multi-component characterization of the functional diversity of the BNST-VTA pathway. We studied the passive and active electrophysiological properties of virally identified population of BNST neurons that project to the VTA. We used a comprehensive series of in vitro recordings of electrophysiological variables and performed hierarchical clustering to determine the functional diversity of the projection neurons in the BNST-VTA pathway. Our study revealed four subpopulations in the BNST-VTA pathway, all of which differ in their activation profiles and likely have distinct inputs and function in the VTA. Our results will help resolve the discord in interpretation of the various roles of this electrophysiologically diverse projection and builds a foundation for understanding how the different neuronal types integrate signals.
Collapse
Affiliation(s)
- Yuka Miura
- Department of Biological Sciences, Hunter College of the City University of New York, New York, NY, United States
- Program in Biology, Graduate Center of the City University of New York, New York, NY, United States
| | - Mary Regis Shanley
- Department of Biological Sciences, Hunter College of the City University of New York, New York, NY, United States
- Program in Biology, Graduate Center of the City University of New York, New York, NY, United States
| | - Ashley Urbaez
- Department of Biological Sciences, Hunter College of the City University of New York, New York, NY, United States
| | - Allyson K. Friedman
- Department of Biological Sciences, Hunter College of the City University of New York, New York, NY, United States
- Program in Biology, Graduate Center of the City University of New York, New York, NY, United States
| |
Collapse
|
15
|
Al‐Karagholi MA, Hakbilen CC, Ashina M. The role of high-conductance calcium-activated potassium channel in headache and migraine pathophysiology. Basic Clin Pharmacol Toxicol 2022; 131:347-354. [PMID: 36028922 PMCID: PMC9826089 DOI: 10.1111/bcpt.13787] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/17/2022] [Accepted: 08/25/2022] [Indexed: 01/11/2023]
Abstract
Migraine is a common, neurovascular headache disorder with a complex molecular interplay. The involvement of ion channels in the pathogenesis of migraine gathered considerable attention with the findings that different ion channels subfamilies are expressed in trigeminovascular system, the physiological substrate of migraine pain, and several ion channel openers investigated in clinical trials with diverse primary endpoints caused headache as a frequent side effect. High-conductance (big) calcium-activated potassium (BKCa ) channel is expressed in the cranial arteries and the trigeminal pain pathway. Recent clinical research revealed that infusion of BKCa channel opener MaxiPost caused vasodilation, headache and migraine attack. Thus, BKCa channel is involved in pathophysiological mechanisms underlying headache and migraine, and targeting BKCa channel presents a new potential strategy for migraine treatment.
Collapse
Affiliation(s)
- Mohammad Al‐Mahdi Al‐Karagholi
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Cemile Ceren Hakbilen
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Messoud Ashina
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
16
|
Mäki-Marttunen T, Mäki-Marttunen V. Excitatory and inhibitory effects of HCN channel modulation on excitability of layer V pyramidal cells. PLoS Comput Biol 2022; 18:e1010506. [PMID: 36099307 PMCID: PMC9506642 DOI: 10.1371/journal.pcbi.1010506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 09/23/2022] [Accepted: 08/19/2022] [Indexed: 11/19/2022] Open
Abstract
Dendrites of cortical pyramidal cells are densely populated by hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, a.k.a. Ih channels. Ih channels are targeted by multiple neuromodulatory pathways, and thus are one of the key ion-channel populations regulating the pyramidal cell activity. Previous observations and theories attribute opposing effects of the Ih channels on neuronal excitability due to their mildly hyperpolarized reversal potential. These effects are difficult to measure experimentally due to the fine spatiotemporal landscape of the Ih activity in the dendrites, but computational models provide an efficient tool for studying this question in a reduced but generalizable setting. In this work, we build upon existing biophysically detailed models of thick-tufted layer V pyramidal cells and model the effects of over- and under-expression of Ih channels as well as their neuromodulation. We show that Ih channels facilitate the action potentials of layer V pyramidal cells in response to proximal dendritic stimulus while they hinder the action potentials in response to distal dendritic stimulus at the apical dendrite. We also show that the inhibitory action of the Ih channels in layer V pyramidal cells is due to the interactions between Ih channels and a hot zone of low voltage-activated Ca2+ channels at the apical dendrite. Our simulations suggest that a combination of Ih-enhancing neuromodulation at the proximal part of the apical dendrite and Ih-inhibiting modulation at the distal part of the apical dendrite can increase the layer V pyramidal excitability more than either of the two alone. Our analyses uncover the effects of Ih-channel neuromodulation of layer V pyramidal cells at a single-cell level and shed light on how these neurons integrate information and enable higher-order functions of the brain.
Collapse
Affiliation(s)
- Tuomo Mäki-Marttunen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Biosciences, University of Oslo, Oslo, Norway
- Simula Research Laboratory, Oslo, Norway
- * E-mail:
| | - Verónica Mäki-Marttunen
- Cognitive Psychology Unit, Faculty of Social Sciences, University of Leiden, Leiden, Netherlands
| |
Collapse
|
17
|
Ozturk H, Basoglu H, Yorulmaz N, Aydin-Abidin S, Abidin I. Fisetin decreases the duration of ictal-like discharges in mouse hippocampal slices. J Biol Phys 2022; 48:355-368. [PMID: 35948819 PMCID: PMC9411310 DOI: 10.1007/s10867-022-09612-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/27/2022] [Indexed: 11/24/2022] Open
Abstract
There is an increasing interest in the biological and therapeutic effects of fisetin, a natural phenolic compound. Fisetin has affinity on some neuronal targets and may have the potential to modulate neuronal activity. In this study the effects of acute application of fisetin on synchronized events were evaluated electro-physiologically. Besides, interaction of fisetin with closely related channels were investigated in silico. Acute horizontal hippocampal slices were obtained from 32- to 36-day-old C57BL/6 mice. Extracellular field potentials were recorded from CA3 region of the hippocampus. Bath application of 4 aminopyridine (4AP, 100 µM) initiated ictal- and interictal-like synchronized epileptiform discharges in the brain slices. Fifty micromolar fisetin was applied to the recording chamber during the epileptiform activity. The duration and frequencies of both ictal-like and interictal-like activities were calculated from the electrophysiological records. Molecular docking was performed to reveal interaction of fisetin on GABA-A, NMDA, AMPA receptors, and HCN2 channel, which are neuronal structures directly involved in recorded activity. Although fisetin does not affect basal neuronal activity in brain slice, it reduced the duration of ictal-like discharges significantly. Molecular docking results indicated that fisetin has no effect on GABA-A, NMDA, and AMPA receptors. However, fisetin binds to the (5JON) HCN2 channel strongly with the binding energy of -7.66 kcal/mol. Reduction on the duration of 4AP-induced ictal-like discharges can be explained as HCN channels can cause an inhibitory effect via enhancing M-type K + channels which increase K outward currents.
Collapse
Affiliation(s)
- Hilal Ozturk
- Department of Biophysics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
- Department of Biophysics, Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Harun Basoglu
- Department of Biophysics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey.
| | - Nuri Yorulmaz
- Department of Physics, Faculty of Science, Harran University, Sanliurfa, Turkey
| | - Selcen Aydin-Abidin
- Department of Biophysics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Ismail Abidin
- Department of Biophysics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| |
Collapse
|
18
|
ATP-Sensitive Potassium Channels in Migraine: Translational Findings and Therapeutic Potential. Cells 2022; 11:cells11152406. [PMID: 35954249 PMCID: PMC9367966 DOI: 10.3390/cells11152406] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 12/10/2022] Open
Abstract
Globally, migraine is a leading cause of disability with a huge impact on both the work and private life of affected persons. To overcome the societal migraine burden, better treatment options are needed. Increasing evidence suggests that ATP-sensitive potassium (KATP) channels are involved in migraine pathophysiology. These channels are essential both in blood glucose regulation and cardiovascular homeostasis. Experimental infusion of the KATP channel opener levcromakalim to healthy volunteers and migraine patients induced headache and migraine attacks in 82-100% of participants. Thus, this is the most potent trigger of headache and migraine identified to date. Levcromakalim likely induces migraine via dilation of cranial arteries. However, other neuronal mechanisms are also proposed. Here, basic KATP channel distribution, physiology, and pharmacology are reviewed followed by thorough review of clinical and preclinical research on KATP channel involvement in migraine. KATP channel opening and blocking have been studied in a range of preclinical migraine models and, within recent years, strong evidence on the importance of their opening in migraine has been provided from human studies. Despite major advances, translational difficulties exist regarding the possible anti-migraine efficacy of KATP channel blockage. These are due to significant species differences in the potency and specificity of pharmacological tools targeting the various KATP channel subtypes.
Collapse
|
19
|
Saccadic premotor burst neurons and histochemical correlates of their firing patterns in rhesus monkey. J Neurol Sci 2022; 439:120328. [DOI: 10.1016/j.jns.2022.120328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 11/20/2022]
|
20
|
Nguyen LH, Xu Y, Mahadeo T, Zhang L, Lin TV, Born HA, Anderson AE, Bordey A. Expression of 4E-BP1 in juvenile mice alleviates mTOR-induced neuronal dysfunction and epilepsy. Brain 2022; 145:1310-1325. [PMID: 34849602 PMCID: PMC9128821 DOI: 10.1093/brain/awab390] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/01/2021] [Accepted: 09/22/2021] [Indexed: 11/13/2022] Open
Abstract
Hyperactivation of the mTOR pathway during foetal neurodevelopment alters neuron structure and function, leading to focal malformation of cortical development and intractable epilepsy. Recent evidence suggests a role for dysregulated cap-dependent translation downstream of mTOR signalling in the formation of focal malformation of cortical development and seizures. However, it is unknown whether modifying translation once the developmental pathologies are established can reverse neuronal abnormalities and seizures. Addressing these issues is crucial with regards to therapeutics because these neurodevelopmental disorders are predominantly diagnosed during childhood, when patients present with symptoms. Here, we report increased phosphorylation of the mTOR effector and translational repressor, 4E-BP1, in patient focal malformation of cortical development tissue and in a mouse model of focal malformation of cortical development. Using temporally regulated conditional gene expression systems, we found that expression of a constitutively active form of 4E-BP1 that resists phosphorylation by focal malformation of cortical development in juvenile mice reduced neuronal cytomegaly and corrected several neuronal electrophysiological alterations, including depolarized resting membrane potential, irregular firing pattern and aberrant expression of HCN4 ion channels. Further, 4E-BP1 expression in juvenile focal malformation of cortical development mice after epilepsy onset resulted in improved cortical spectral activity and decreased spontaneous seizure frequency in adults. Overall, our study uncovered a remarkable plasticity of the juvenile brain that facilitates novel therapeutic opportunities to treat focal malformation of cortical development-related epilepsy during childhood with potentially long-lasting effects in adults.
Collapse
Affiliation(s)
- Lena H Nguyen
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Youfen Xu
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Travorn Mahadeo
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Longbo Zhang
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Tiffany V Lin
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Heather A Born
- Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Anne E Anderson
- Cain Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Angélique Bordey
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
21
|
Kessi M, Peng J, Duan H, He H, Chen B, Xiong J, Wang Y, Yang L, Wang G, Kiprotich K, Bamgbade OA, He F, Yin F. The Contribution of HCN Channelopathies in Different Epileptic Syndromes, Mechanisms, Modulators, and Potential Treatment Targets: A Systematic Review. Front Mol Neurosci 2022; 15:807202. [PMID: 35663267 PMCID: PMC9161305 DOI: 10.3389/fnmol.2022.807202] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 04/06/2022] [Indexed: 12/04/2022] Open
Abstract
Background Hyperpolarization-activated cyclic nucleotide-gated (HCN) current reduces dendritic summation, suppresses dendritic calcium spikes, and enables inhibitory GABA-mediated postsynaptic potentials, thereby suppressing epilepsy. However, it is unclear whether increased HCN current can produce epilepsy. We hypothesized that gain-of-function (GOF) and loss-of-function (LOF) variants of HCN channel genes may cause epilepsy. Objectives This systematic review aims to summarize the role of HCN channelopathies in epilepsy, update genetic findings in patients, create genotype–phenotype correlations, and discuss animal models, GOF and LOF mechanisms, and potential treatment targets. Methods The review was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement, for all years until August 2021. Results We identified pathogenic variants of HCN1 (n = 24), HCN2 (n = 8), HCN3 (n = 2), and HCN4 (n = 6) that were associated with epilepsy in 74 cases (43 HCN1, 20 HCN2, 2 HCN3, and 9 HCN4). Epilepsy was associated with GOF and LOF variants, and the mechanisms were indeterminate. Less than half of the cases became seizure-free and some developed drug-resistant epilepsy. Of the 74 cases, 12 (16.2%) died, comprising HCN1 (n = 4), HCN2 (n = 2), HCN3 (n = 2), and HCN4 (n = 4). Of the deceased cases, 10 (83%) had a sudden unexpected death in epilepsy (SUDEP) and 2 (16.7%) due to cardiopulmonary failure. SUDEP affected more adults (n = 10) than children (n = 2). HCN1 variants p.M234R, p.C329S, p.V414M, p.M153I, and p.M305L, as well as HCN2 variants p.S632W and delPPP (p.719–721), were associated with different phenotypes. HCN1 p.L157V and HCN4 p.R550C were associated with genetic generalized epilepsy. There are several HCN animal models, pharmacological targets, and modulators, but precise drugs have not been developed. Currently, there are no HCN channel openers. Conclusion We recommend clinicians to include HCN genes in epilepsy gene panels. Researchers should explore the possible underlying mechanisms for GOF and LOF variants by identifying the specific neuronal subtypes and neuroanatomical locations of each identified pathogenic variant. Researchers should identify specific HCN channel openers and blockers with high binding affinity. Such information will give clarity to the involvement of HCN channelopathies in epilepsy and provide the opportunity to develop targeted treatments.
Collapse
Affiliation(s)
- Miriam Kessi
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
- Department of Pediatrics, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Jing Peng
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Haolin Duan
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Hailan He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Baiyu Chen
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Juan Xiong
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Ying Wang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Lifen Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Guoli Wang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Karlmax Kiprotich
- Department of Epidemiology and Medical Statistics, School of Public Health, Moi University, Eldoret, Kenya
| | - Olumuyiwa A. Bamgbade
- Department of Anesthesiology and Pharmacology, University of British Columbia, Vancouver, BC, Canada
| | - Fang He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Fei Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
- *Correspondence: Fei Yin
| |
Collapse
|
22
|
Oberländer K, Witte V, Mallien AS, Gass P, Bengtson CP, Bading H. Dysregulation of Npas4 and Inhba expression and an altered excitation-inhibition balance are associated with cognitive deficits in DBA/2 mice. Learn Mem 2022; 29:55-70. [PMID: 35042829 PMCID: PMC8774195 DOI: 10.1101/lm.053527.121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/28/2021] [Indexed: 02/03/2023]
Abstract
Differences in the learning associated transcriptional profiles between mouse strains with distinct learning abilities could provide insight into the molecular basis of learning and memory. The inbred mouse strain DBA/2 shows deficits in hippocampus-dependent memory, yet the transcriptional responses to learning and the underlying mechanisms of the impairments are unknown. Comparing DBA/2J mice with the reference standard C57BL/6N mouse strain we verify an enhanced susceptibility to kainic acid induced seizures, confirm impairments in hippocampus-dependent spatial memory tasks and uncover additional behavioral abnormalities including deficits in hippocampus-independent learning. Surprisingly, we found no broad dysfunction of the DBA/2J strain in immediate early gene (IEG) activation but instead report brain region-specific and gene-specific alterations. The learning-associated IEGs Arc, c-Fos, and Nr4a1 showed no DBA/2J deficits in basal or synaptic activity induced gene expression in hippocampal or cortical primary neuronal cultures or in the CA1, CA3, or retrosplenial cortex following spatial object recognition (SOR) training in vivo. However, the parietal cortex showed reduced and the dentate gyrus showed enhanced SOR-evoked induction of most IEGs. All DBA/2J hippocampal regions exhibited elevated basal expression of inhibin β A (Inhba) and a learning-associated superinduction of the transcription factor neuronal Per-Arnt-Sim domain protein 4 (Npas4) known to regulate the synaptic excitation-inhibition balance. In line with this, CA1 pyramidal neurons of DBA/2J mice showed fewer inhibitory and more excitatory miniature postsynaptic currents but no alteration in most other electrophysiological properties or gross dendritic morphology. The dysregulation of Npas4 and Inhba expression and synaptic connectivity may underlie the cognitive deficits and increased susceptibility to seizures of DBA/2J mice.
Collapse
Affiliation(s)
- Kristin Oberländer
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Victoria Witte
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Anne Stephanie Mallien
- Department of Psychiatry and Psychotherapy, Research Group Animal Models in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, 68159 Mannheim, Germany
| | - Peter Gass
- Department of Psychiatry and Psychotherapy, Research Group Animal Models in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, 68159 Mannheim, Germany
| | - C. Peter Bengtson
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
23
|
Chalif JI, de Lourdes Martínez-Silva M, Pagiazitis JG, Murray AJ, Mentis GZ. Control of mammalian locomotion by ventral spinocerebellar tract neurons. Cell 2022; 185:328-344.e26. [PMID: 35063074 PMCID: PMC8852337 DOI: 10.1016/j.cell.2021.12.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 11/09/2021] [Accepted: 12/13/2021] [Indexed: 01/22/2023]
Abstract
Locomotion is a complex behavior required for animal survival. Vertebrate locomotion depends on spinal interneurons termed the central pattern generator (CPG), which generates activity responsible for the alternation of flexor and extensor muscles and the left and right side of the body. It is unknown whether multiple or a single neuronal type is responsible for the control of mammalian locomotion. Here, we show that ventral spinocerebellar tract neurons (VSCTs) drive generation and maintenance of locomotor behavior in neonatal and adult mice. Using mouse genetics, physiological, anatomical, and behavioral assays, we demonstrate that VSCTs exhibit rhythmogenic properties and neuronal circuit connectivity consistent with their essential role in the locomotor CPG. Importantly, optogenetic activation and chemogenetic silencing reveals that VSCTs are necessary and sufficient for locomotion. These findings identify VSCTs as critical components for mammalian locomotion and provide a paradigm shift in our understanding of neural control of complex behaviors.
Collapse
Affiliation(s)
- Joshua I. Chalif
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA,Dept. of Neurology, Columbia University, New York, NY 10032, USA
| | - María de Lourdes Martínez-Silva
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA,Dept. of Neurology, Columbia University, New York, NY 10032, USA
| | - John G. Pagiazitis
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA,Dept. of Neurology, Columbia University, New York, NY 10032, USA
| | - Andrew J. Murray
- Sainsbury Wellcome Centre, University College London, 25 Howland Street, London W1T 4JG, UK
| | - George Z. Mentis
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA,Dept. of Neurology, Columbia University, New York, NY 10032, USA,Dept. of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA,Corresponding author & Lead contact: Tel: +1-212-305-9846,
| |
Collapse
|
24
|
Bosque-Cordero KY, Vazquez-Torres R, Calo-Guadalupe C, Consuegra-Garcia D, Fois GR, Georges F, Jimenez-Rivera CA. I h blockade reduces cocaine-induced firing patterns of putative dopaminergic neurons of the ventral tegmental area in the anesthetized rat. Prog Neuropsychopharmacol Biol Psychiatry 2022; 112:110431. [PMID: 34454991 PMCID: PMC8489561 DOI: 10.1016/j.pnpbp.2021.110431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/23/2021] [Accepted: 08/23/2021] [Indexed: 01/12/2023]
Abstract
The hyperpolarization-activated cation current (Ih) is a determinant of intrinsic excitability in various cells, including dopaminergic neurons (DA) of the ventral tegmental area (VTA). In contrast to other cellular conductances, Ih is activated by hyperpolarization negative to -55 mV and activating Ih produces a time-dependent depolarizing current. Our laboratory demonstrated that cocaine sensitization, a chronic cocaine behavioral model, significantly reduces Ih amplitude in VTA DA neurons. Despite this reduction in Ih, the spontaneous firing of VTA DA cells after cocaine sensitization remained similar to control groups. Although the role of Ih in controlling VTA DA excitability is still poorly understood, our hypothesis is that Ih reduction could play a role of a homeostatic controller compensating for cocaine-induced change in excitability. Using in vivo single-unit extracellular electrophysiology in isoflurane anesthetized rats, we explored the contribution of Ih on spontaneous firing patterns of VTA DA neurons. A key feature of spontaneous excitability is bursting activity; bursting is defined as trains of two or more spikes occurring within a short interval and followed by a prolonged period of inactivity. Burst activity increases the reliability of information transfer. To elucidate the contribution of Ih to spontaneous firing patterns of VTA DA neurons, we locally infused an Ih blocker (ZD 7288, 8.3 μM) and evaluated its effect. Ih blockade significantly reduced firing rate, bursting frequency, and percent of spikes within a burst. In addition, Ih blockade significantly reduced acute cocaine-induced spontaneous firing rate, bursting frequency, and percent of spikes within a burst. Using whole-cell patch-clamp, we determine the progressive reduction of Ih after acute and chronic cocaine administration (15 mg/k.g intraperitoneally). Our data show a significant reduction (~25%) in Ih amplitude after 24 but not 2 h of acute cocaine administration. These results suggest that a progressive reduction of Ih could serve as a homeostatic regulator of cocaine-induced spontaneous firing patterns related to VTA DA excitability.
Collapse
Affiliation(s)
| | | | | | | | - Giulia R Fois
- University of Bordeaux, Neurodegeneratives Diseases Institute, IMN-UMR-CNRS 5293, 146 rue Léo Saignat, 33076 Bordeaux, France; CNRS, Neurodegeneratives Diseases Institute, IMN-UMR-CNRS 5293, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - François Georges
- University of Bordeaux, Neurodegeneratives Diseases Institute, IMN-UMR-CNRS 5293, 146 rue Léo Saignat, 33076 Bordeaux, France; CNRS, Neurodegeneratives Diseases Institute, IMN-UMR-CNRS 5293, 146 rue Léo Saignat, 33076 Bordeaux, France
| | | |
Collapse
|
25
|
Eren-Koçak E, Dalkara T. Ion Channel Dysfunction and Neuroinflammation in Migraine and Depression. Front Pharmacol 2021; 12:777607. [PMID: 34858192 PMCID: PMC8631474 DOI: 10.3389/fphar.2021.777607] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/22/2021] [Indexed: 01/15/2023] Open
Abstract
Migraine and major depression are debilitating disorders with high lifetime prevalence rates. Interestingly these disorders are highly comorbid and show significant heritability, suggesting shared pathophysiological mechanisms. Non-homeostatic function of ion channels and neuroinflammation may be common mechanisms underlying both disorders: The excitation-inhibition balance of microcircuits and their modulation by monoaminergic systems, which depend on the expression and function of membrane located K+, Na+, and Ca+2 channels, have been reported to be disturbed in both depression and migraine. Ion channels and energy supply to synapses not only change excitability of neurons but can also mediate the induction and maintenance of inflammatory signaling implicated in the pathophysiology of both disorders. In this respect, Pannexin-1 and P2X7 large-pore ion channel receptors can induce inflammasome formation that triggers release of pro-inflammatory mediators from the cell. Here, the role of ion channels involved in the regulation of excitation-inhibition balance, synaptic energy homeostasis as well as inflammatory signaling in migraine and depression will be reviewed.
Collapse
Affiliation(s)
- Emine Eren-Koçak
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey.,Department of Psychiatry, Medical Faculty, Hacettepe University, Ankara, Turkey
| | - Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| |
Collapse
|
26
|
Vaiciuleviciute R, Bironaite D, Uzieliene I, Mobasheri A, Bernotiene E. Cardiovascular Drugs and Osteoarthritis: Effects of Targeting Ion Channels. Cells 2021; 10:cells10102572. [PMID: 34685552 PMCID: PMC8534048 DOI: 10.3390/cells10102572] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 12/24/2022] Open
Abstract
Osteoarthritis (OA) and cardiovascular diseases (CVD) share many similar features, including similar risk factors and molecular mechanisms. A great number of cardiovascular drugs act via different ion channels and change ion balance, thus modulating cell metabolism, osmotic responses, turnover of cartilage extracellular matrix and inflammation. These drugs are consumed by patients with CVD for many years; however, information about their effects on the joint tissues has not been fully clarified. Nevertheless, it is becoming increasingly likely that different cardiovascular drugs may have an impact on articular tissues in OA. Here, we discuss the potential effects of direct and indirect ion channel modulating drugs, including inhibitors of voltage gated calcium and sodium channels, hyperpolarization-activated cyclic nucleotide-gated channels, β-adrenoreceptor inhibitors and angiotensin-aldosterone system affecting drugs. The aim of this review was to summarize the information about activities of cardiovascular drugs on cartilage and subchondral bone and to discuss their possible consequences on the progression of OA, focusing on the modulation of ion channels in chondrocytes and other joint cells, pain control and regulation of inflammation. The implication of cardiovascular drug consumption in aetiopathogenesis of OA should be considered when prescribing ion channel modulators, particularly in long-term therapy protocols.
Collapse
Affiliation(s)
- Raminta Vaiciuleviciute
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (R.V.); (D.B.); (I.U.); (A.M.)
| | - Daiva Bironaite
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (R.V.); (D.B.); (I.U.); (A.M.)
| | - Ilona Uzieliene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (R.V.); (D.B.); (I.U.); (A.M.)
| | - Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (R.V.); (D.B.); (I.U.); (A.M.)
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, FI-90014 Oulu, Finland
- Departments of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, 508 GA Utrecht, The Netherlands
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania; (R.V.); (D.B.); (I.U.); (A.M.)
- Correspondence:
| |
Collapse
|
27
|
Yang YS, Choi JH, Rah JC. Hypoxia with inflammation and reperfusion alters membrane resistance by dynamically regulating voltage-gated potassium channels in hippocampal CA1 neurons. Mol Brain 2021; 14:147. [PMID: 34556177 PMCID: PMC8461870 DOI: 10.1186/s13041-021-00857-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/07/2021] [Indexed: 02/01/2023] Open
Abstract
Hypoxia typically accompanies acute inflammatory responses in patients and animal models. However, a limited number of studies have examined the effect of hypoxia in combination with inflammation (Hypo-Inf) on neural function. We previously reported that neuronal excitability in hippocampal CA1 neurons decreased during hypoxia and greatly rebounded upon reoxygenation. We attributed this altered excitability mainly to the dynamic regulation of hyperpolarization-activated cyclic nucleotide-gated cation (HCN) channels and input resistance. However, the molecular mechanisms underlying input resistance changes by Hypo-Inf and reperfusion remained unclear. In the present study, we found that a change in the density of the delayed rectifier potassium current (IDR) can explain the input resistance variability. Furthermore, voltage-dependent inactivation of A-type potassium (IA) channels shifted in the depolarizing direction during Hypo-Inf and reverted to normal upon reperfusion without a significant alteration in the maximum current density. Our results indicate that changes in the input resistance, and consequently excitability, caused by Hypo-Inf and reperfusion are at least partially regulated by the availability and voltage dependence of KV channels. Moreover, these results suggest that selective KV channel modulators can be used as potential neuroprotective drugs to minimize hypoxia- and reperfusion-induced neuronal damage.
Collapse
Affiliation(s)
- Yoon-Sil Yang
- Korea Brain Research Institute, 61 Cheomdan-ro, Dong-gu, Daegu, 41062 South Korea
| | - Joon Ho Choi
- Korea Brain Research Institute, 61 Cheomdan-ro, Dong-gu, Daegu, 41062 South Korea
| | - Jong-Cheol Rah
- Korea Brain Research Institute, 61 Cheomdan-ro, Dong-gu, Daegu, 41062 South Korea
- Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, 333 Techno Jungang-daero, Dalseong-gun, Daegu, 42988 South Korea
| |
Collapse
|
28
|
Zhang XQ, Xu L, Yang SY, Hu LB, Dong FY, Sun BG, Shen HW. Reduced Synaptic Transmission and Intrinsic Excitability of a Subtype of Pyramidal Neurons in the Medial Prefrontal Cortex in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2021; 84:129-140. [PMID: 34487044 DOI: 10.3233/jad-210585] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Abnormal morphology and function of neurons in the prefrontal cortex (PFC) are associated with cognitive deficits in rodent models of Alzheimer's disease (AD), particularly in cortical layer-5 pyramidal neurons that integrate inputs from different sources and project outputs to cortical or subcortical structures. Pyramidal neurons in layer-5 of the PFC can be classified as two subtypes depending on the inducibility of prominent hyperpolarization-activated cation currents (h-current). However, the differences in the neurophysiological alterations between these two subtypes in rodent models of AD remain poorly understood. OBJECTIVE To investigate the neurophysiological alterations between two subtypes of pyramidal neurons in hAPP-J20 mice, a transgenic model for early onset AD. METHODS The synaptic transmission and intrinsic excitability of pyramidal neurons were investigated using whole-cell patch recordings. The morphological complexity of pyramidal neurons was detected by biocytin labelling and subsequent Sholl analysis. RESULTS We found reduced synaptic transmission and intrinsic excitability of the prominent h-current (PH) cells but not the non-PH cells in hAPP-J20 mice. Furthermore, the function of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels which mediated h-current was disrupted in the PH cells of hAPP-J20 mice. Sholl analysis revealed that PH cells had less dendritic intersections in hAPP-J20 mice comparing to control mice, implying that a lower morphological complexity might contribute to the reduced neuronal activity. CONCLUSION These results suggest that the PH cells in the medial PFC may be more vulnerable to degeneration in hAPP-J20 mice and play a sustainable role in frontal dysfunction in AD.
Collapse
Affiliation(s)
- Xiao-Qin Zhang
- Department of Pharmacology, School of Medicine, Zhejiang Key Laboratory of Pathophysiology, Ningbo University, Ningbo, Zhejiang, China.,Key Laboratory of Addiction Research of Zhejiang Province, Ningbo Kangning Hospital, Ningbo, Zhejiang, China
| | - Le Xu
- Department of Pharmacology, School of Medicine, Zhejiang Key Laboratory of Pathophysiology, Ningbo University, Ningbo, Zhejiang, China
| | - Si-Yu Yang
- Department of Pharmacology, School of Medicine, Zhejiang Key Laboratory of Pathophysiology, Ningbo University, Ningbo, Zhejiang, China
| | - Lin-Bo Hu
- Department of Pharmacology, School of Medicine, Zhejiang Key Laboratory of Pathophysiology, Ningbo University, Ningbo, Zhejiang, China
| | - Fei-Yuan Dong
- Department of Pharmacology, School of Medicine, Zhejiang Key Laboratory of Pathophysiology, Ningbo University, Ningbo, Zhejiang, China
| | - Bing-Gui Sun
- Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Hao-Wei Shen
- Department of Pharmacology, School of Medicine, Zhejiang Key Laboratory of Pathophysiology, Ningbo University, Ningbo, Zhejiang, China.,Key Laboratory of Addiction Research of Zhejiang Province, Ningbo Kangning Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
29
|
Binini N, Talpo F, Spaiardi P, Maniezzi C, Pedrazzoli M, Raffin F, Mattiello N, Castagno AN, Masetto S, Yanagawa Y, Dickson CT, Ramat S, Toselli M, Biella GR. Membrane Resonance in Pyramidal and GABAergic Neurons of the Mouse Perirhinal Cortex. Front Cell Neurosci 2021; 15:703407. [PMID: 34366789 PMCID: PMC8339929 DOI: 10.3389/fncel.2021.703407] [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: 04/30/2021] [Accepted: 06/16/2021] [Indexed: 11/13/2022] Open
Abstract
The perirhinal cortex (PRC) is a polymodal associative region of the temporal lobe that works as a gateway between cortical areas and hippocampus. In recent years, an increasing interest arose in the role played by the PRC in learning and memory processes, such as object recognition memory, in contrast with certain forms of hippocampus-dependent spatial and episodic memory. The integrative properties of the PRC should provide all necessary resources to select and enhance the information to be propagated to and from the hippocampus. Among these properties, we explore in this paper the ability of the PRC neurons to amplify the output voltage to current input at selected frequencies, known as membrane resonance. Within cerebral circuits the resonance of a neuron operates as a filter toward inputs signals at certain frequencies to coordinate network activity in the brain by affecting the rate of neuronal firing and the precision of spike timing. Furthermore, the ability of the PRC neurons to resonate could have a fundamental role in generating subthreshold oscillations and in the selection of cortical inputs directed to the hippocampus. Here, performing whole-cell patch-clamp recordings from perirhinal pyramidal neurons and GABAergic interneurons of GAD67-GFP+ mice, we found, for the first time, that the majority of PRC neurons are resonant at their resting potential, with a resonance frequency of 0.5–1.5 Hz at 23°C and of 1.5–2.8 Hz at 36°C. In the presence of ZD7288 (blocker of HCN channels) resonance was abolished in both pyramidal neurons and interneurons, suggesting that Ih current is critically involved in resonance generation. Otherwise, application of TTx (voltage-dependent Na+ channel blocker) attenuates the resonance in pyramidal neurons but not in interneurons, suggesting that only in pyramidal neurons the persistent sodium current has an amplifying effect. These experimental results have also been confirmed by a computational model. From a functional point of view, the resonance in the PRC would affect the reverberating activity between neocortex and hippocampus, especially during slow wave sleep, and could be involved in the redistribution and strengthening of memory representation in cortical regions.
Collapse
Affiliation(s)
- Noemi Binini
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Francesca Talpo
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Paolo Spaiardi
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Claudia Maniezzi
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Matteo Pedrazzoli
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Francesca Raffin
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Niccolò Mattiello
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Antonio N Castagno
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Sergio Masetto
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Gunma University, Maebashi, Japan
| | - Clayton T Dickson
- Department of Psychology, University of Alberta, Edmonton, AB, Canada
| | - Stefano Ramat
- Department of Industrial and Information Engineering, University of Pavia, Pavia, Italy
| | - Mauro Toselli
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Gerardo Rosario Biella
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| |
Collapse
|
30
|
Loss of HCN2 in Dorsal Hippocampus of Young Adult Mice Induces Specific Apoptosis of the CA1 Pyramidal Neuron Layer. Int J Mol Sci 2021; 22:ijms22136699. [PMID: 34206649 PMCID: PMC8269412 DOI: 10.3390/ijms22136699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/08/2021] [Accepted: 06/18/2021] [Indexed: 11/20/2022] Open
Abstract
Neurons inevitably rely on a proper repertoire and distribution of membrane-bound ion-conducting channels. Among these proteins, the family of hyperpolarization-activated and cyclic nucleotide-gated (HCN) channels possesses unique properties giving rise to the corresponding Ih-current that contributes to various aspects of neural signaling. In mammals, four genes (hcn1-4) encode subunits of HCN channels. These subunits can assemble as hetero- or homotetrameric ion-conducting channels. In order to elaborate on the specific role of the HCN2 subunit in shaping electrical properties of neurons, we applied an Adeno-associated virus (AAV)-mediated, RNAi-based knock-down strategy of hcn2 gene expression both in vitro and in vivo. Electrophysiological measurements showed that HCN2 subunit knock-down resulted in specific yet anticipated changes in Ih-current properties in primary hippocampal neurons and, in addition, corroborated that the HCN2 subunit participates in postsynaptic signal integration. To further address the role of the HCN2 subunit in vivo, we injected recombinant (r)AAVs into the dorsal hippocampus of young adult male mice. Behavioral and biochemical analyses were conducted to assess the contribution of HCN2-containing channels in shaping hippocampal network properties. Surprisingly, knock-down of hcn2 expression resulted in a severe degeneration of the CA1 pyramidal cell layer, which did not occur in mice injected with control rAAV constructs. This finding might pinpoint to a vital and yet unknown contribution of HCN2 channels in establishing or maintaining the proper function of CA1 pyramidal neurons of the dorsal hippocampus.
Collapse
|
31
|
Roles of the Functional Interaction between Brain Cholinergic and Dopaminergic Systems in the Pathogenesis and Treatment of Schizophrenia and Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22094299. [PMID: 33919025 PMCID: PMC8122651 DOI: 10.3390/ijms22094299] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/14/2022] Open
Abstract
Most physiologic processes in the brain and related diseases involve more than one neurotransmitter system. Thus, elucidation of the interaction between different neurotransmitter systems could allow for better therapeutic approaches to the treatments of related diseases. Dopaminergic (DAergic) and cholinergic neurotransmitter system regulate various brain functions that include cognition, movement, emotion, etc. This review focuses on the interaction between the brain DAergic and cholinergic systems with respect to the pathogenesis and treatment of schizophrenia and Parkinson’s disease (PD). We first discussed the selection of motor plans at the level of basal ganglia, the major DAergic and cholinergic pathways in the brain, and the receptor subtypes involved in the interaction between the two signaling systems. Next, the roles of each signaling system were discussed in the context of the negative symptoms of schizophrenia, with a focus on the α7 nicotinic cholinergic receptor and the dopamine D1 receptor in the prefrontal cortex. In addition, the roles of the nicotinic and dopamine receptors were discussed in the context of regulation of striatal cholinergic interneurons, which play crucial roles in the degeneration of nigrostriatal DAergic neurons and the development of L-DOPA-induced dyskinesia in PD patients. Finally, we discussed the general mechanisms of nicotine-induced protection of DAergic neurons.
Collapse
|
32
|
Kelley C, Dura-Bernal S, Neymotin SA, Antic SD, Carnevale NT, Migliore M, Lytton WW. Effects of Ih and TASK-like shunting current on dendritic impedance in layer 5 pyramidal-tract neurons. J Neurophysiol 2021; 125:1501-1516. [PMID: 33689489 PMCID: PMC8282219 DOI: 10.1152/jn.00015.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
Pyramidal neurons in neocortex have complex input-output relationships that depend on their morphologies, ion channel distributions, and the nature of their inputs, but which cannot be replicated by simple integrate-and-fire models. The impedance properties of their dendritic arbors, such as resonance and phase shift, shape neuronal responses to synaptic inputs and provide intraneuronal functional maps reflecting their intrinsic dynamics and excitability. Experimental studies of dendritic impedance have shown that neocortical pyramidal tract neurons exhibit distance-dependent changes in resonance and impedance phase with respect to the soma. We, therefore, investigated how well several biophysically detailed multicompartment models of neocortical layer 5 pyramidal tract neurons reproduce the location-dependent impedance profiles observed experimentally. Each model tested here exhibited location-dependent impedance profiles, but most captured either the observed impedance amplitude or phase, not both. The only model that captured features from both incorporates hyperpolarization-activated cyclic nucleotide-gated (HCN) channels and a shunting current, such as that produced by Twik-related acid-sensitive K+ (TASK) channels. TASK-like channel density in this model was proportional to local HCN channel density. We found that although this shunting current alone is insufficient to produce resonance or realistic phase response, it modulates all features of dendritic impedance, including resonance frequencies, resonance strength, synchronous frequencies, and total inductive phase. We also explored how the interaction of HCN channel current (Ih) and a TASK-like shunting current shape synaptic potentials and produce degeneracy in dendritic impedance profiles, wherein different combinations of Ih and shunting current can produce the same impedance profile.NEW & NOTEWORTHY We simulated chirp current stimulation in the apical dendrites of 5 biophysically detailed multicompartment models of neocortical pyramidal tract neurons and found that a combination of HCN channels and TASK-like channels produced the best fit to experimental measurements of dendritic impedance. We then explored how HCN and TASK-like channels can shape the dendritic impedance as well as the voltage response to synaptic currents.
Collapse
Affiliation(s)
- Craig Kelley
- Program in Biomedical Engineering, SUNY Downstate Health Sciences University and NYU Tandon School of Engineering, Brooklyn, New York
| | - Salvador Dura-Bernal
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, New York
- Center for Biomedical Imaging and Neuromodulation, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York
| | - Samuel A Neymotin
- Center for Biomedical Imaging and Neuromodulation, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York
- Department of Psychiatry, NYU Grossman School of Medicine, New York City, New York
| | - Srdjan D Antic
- Neuroscience Department, Institute of Systems Genomics, University of Connecticut Health, Farmington, Connecticut
| | | | - Michele Migliore
- Institute of Biophysics, National Research Council, Palermo, Italy
| | - William W Lytton
- Program in Biomedical Engineering, SUNY Downstate Health Sciences University and NYU Tandon School of Engineering, Brooklyn, New York
- Department of Physiology and Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, New York
- Department of Neurology, SUNY Downstate Health Sciences University, Brooklyn, New York
- Department of Neurology, Kings County Hospital Center, Brooklyn, New York
- The Robert F. Furchgott Center for Neural and Behavioral Science, Brooklyn, New York
| |
Collapse
|
33
|
Homeostatic plasticity and burst activity are mediated by hyperpolarization-activated cation currents and T-type calcium channels in neuronal cultures. Sci Rep 2021; 11:3236. [PMID: 33547341 PMCID: PMC7864958 DOI: 10.1038/s41598-021-82775-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 01/20/2021] [Indexed: 01/27/2023] Open
Abstract
Homeostatic plasticity stabilizes neuronal networks by adjusting the responsiveness of neurons according to their global activity and the intensity of the synaptic inputs. We investigated the homeostatic regulation of hyperpolarization-activated cyclic nucleotide-gated (HCN) and T-type calcium (CaV3) channels in dissociated and organotypic slice cultures. After 48 h blocking of neuronal activity by tetrodotoxin (TTX), our patch-clamp experiments revealed an increase in the depolarizing voltage sag and post-inhibitory rebound mediated by HCN and CaV3 channels, respectively. All HCN subunits (HCN1 to 4) and T-type Ca-channel subunits (CaV3.1, 3.2 and 3.3) were expressed in both control and activity-deprived hippocampal cultures. Elevated expression levels of CaV3.1 mRNA and a selective increase in the expression of TRIP8b exon 4 isoforms, known to regulate HCN channel localization, were also detected in TTX-treated cultured hippocampal neurons. Immunohistochemical staining in TTX-treated organotypic slices verified a more proximal translocation of HCN1 channels in CA1 pyramidal neurons. Computational modeling also implied that HCN and T-type calcium channels have important role in the regulation of synchronized bursting evoked by previous activity-deprivation. Thus, our findings indicate that HCN and T-type Ca-channels contribute to the homeostatic regulation of excitability and integrative properties of hippocampal neurons.
Collapse
|
34
|
Pelot NA, Catherall DC, Thio BJ, Titus ND, Liang ED, Henriquez CS, Grill WM. Excitation properties of computational models of unmyelinated peripheral axons. J Neurophysiol 2021; 125:86-104. [PMID: 33085556 PMCID: PMC8087387 DOI: 10.1152/jn.00315.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/09/2020] [Accepted: 09/14/2020] [Indexed: 12/19/2022] Open
Abstract
Biophysically based computational models of nerve fibers are important tools for designing electrical stimulation therapies, investigating drugs that affect ion channels, and studying diseases that affect neurons. Although peripheral nerves are primarily composed of unmyelinated axons (i.e., C-fibers), most modeling efforts focused on myelinated axons. We implemented the single-compartment model of vagal afferents from Schild et al. (1994) (Schild JH, Clark JW, Hay M, Mendelowitz D, Andresen MC, Kunze DL. J Neurophysiol 71: 2338-2358, 1994) and extended the model into a multicompartment axon, presenting the first cable model of a C-fiber vagal afferent. We also implemented the updated parameters from the Schild and Kunze (1997) model (Schild JH, Kunze DL. J Neurophysiol 78: 3198-3209, 1997). We compared the responses of these novel models with those of three published models of unmyelinated axons (Rattay F, Aberham M. IEEE Trans Biomed Eng 40: 1201-1209, 1993; Sundt D, Gamper N, Jaffe DB. J Neurophysiol 114: 3140-3153, 2015; Tigerholm J, Petersson ME, Obreja O, Lampert A, Carr R, Schmelz M, Fransén E. J Neurophysiol 111: 1721-1735, 2014) and with experimental data from single-fiber recordings. Comparing the two models by Schild et al. (1994, 1997) revealed that differences in rest potential and action potential shape were driven by changes in maximum conductances rather than changes in sodium channel dynamics. Comparing the five model axons, the conduction speeds and strength-duration responses were largely within expected ranges, but none of the models captured the experimental threshold recovery cycle-including a complete absence of late subnormality in the models-and their action potential shapes varied dramatically. The Tigerholm et al. (2014) model best reproduced the experimental data, but these modeling efforts make clear that additional data are needed to parameterize and validate future models of autonomic C-fibers.NEW & NOTEWORTHY Peripheral nerves are primarily composed of unmyelinated axons, and there is growing interest in electrical stimulation of the autonomic nervous system to treat various diseases. We present the first cable model of an unmyelinated vagal nerve fiber and compare its ion channel isoforms and conduction responses with other published models of unmyelinated axons, establishing important tools for advancing modeling of autonomic nerves.
Collapse
Affiliation(s)
- Nicole A Pelot
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - David C Catherall
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Brandon J Thio
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Nathan D Titus
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Edward D Liang
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Craig S Henriquez
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina
| | - Warren M Grill
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
- Department of Electrical and Computer Engineering, Duke University, Durham, North Carolina
- Department of Neurobiology, Duke University, Durham, North Carolina
- Department of Neurosurgery, Duke University, Durham, North Carolina
| |
Collapse
|
35
|
Rivero-Echeto MC, Perissinotti PP, González-Inchauspe C, Kargieman L, Bisagno V, Urbano FJ. Simultaneous administration of cocaine and caffeine dysregulates HCN and T-type channels. Psychopharmacology (Berl) 2021; 238:787-810. [PMID: 33241481 PMCID: PMC7688300 DOI: 10.1007/s00213-020-05731-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/18/2020] [Indexed: 12/13/2022]
Abstract
RATIONALE The abuse of psychostimulants has adverse consequences on the physiology of the central nervous system. In Argentina, and other South American countries, coca paste or "PACO" (cocaine and caffeine are its major components) is massively consumed with deleterious clinical consequences for the health and well-being of the general population. A scant number of studies have addressed the consequences of stimulant combination of cocaine and caffeine on the physiology of the somatosensory thalamocortical (ThCo) system. OBJECTIVES Our aim was to study ion conductances that have important implications regulating sleep-wake states 24-h after an acute or chronic binge-like administration of a cocaine and caffeine mixture following previously analyzed pasta base samples ("PACO"-like binge") using mice. METHODS We randomly injected (i.p.) male C57BL/6JFcen mice with a binge-like psychostimulants regimen during either 1 day (acute) or 1 day on/1 day off during 13 days for a total of 7 binges (chronic). Single-cell patch-clamp recordings of VB neurons were performed in thalamocortical slices 24 h after the last psychostimulant injection. We also recorded EEG/EMG from mice 24 h after being systemically treated with chronic administration of cocaine + caffeine versus saline, vehicle. RESULTS Our results showed notorious changes in the intrinsic properties of the VB nucleus neurons that persist after 24-h of either acute or chronic binge administrations of combined cocaine and caffeine ("PACO"-like binge). Functional dysregulation of HCN (hyperpolarization-activated cyclic nucleotide-gated) and T-type VGC (voltage-gated calcium) channels was described 24-h after acute/chronic "PACO"-like administrations. Furthermore, intracellular basal [Ca2+] disturbances resulted a key factor that modulated the availability and the activation of T-type channels, altering T-type "window currents." As a result, all these changes ultimately shaped the low-threshold spikes (LTS)-associated Ca2+ transients, regulated the membrane excitability, and altered sleep-wake transitions. CONCLUSION Our results suggest that deleterious consequences of stimulants cocaine and caffeine combination on the thalamocortical physiology as a whole might be related to potential neurotoxic effects of soaring intracellular [Ca2+].
Collapse
Affiliation(s)
- María Celeste Rivero-Echeto
- grid.7345.50000 0001 0056 1981CONICET-Universidad de Buenos Aires, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Ciudad de Buenos Aires, Argentina
| | - Paula P. Perissinotti
- grid.7345.50000 0001 0056 1981CONICET-Universidad de Buenos Aires, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Ciudad de Buenos Aires, Argentina
| | - Carlota González-Inchauspe
- grid.7345.50000 0001 0056 1981CONICET-Universidad de Buenos Aires, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Ciudad de Buenos Aires, Argentina
| | - Lucila Kargieman
- grid.7345.50000 0001 0056 1981CONICET-Universidad de Buenos Aires, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Ciudad de Buenos Aires, Argentina
| | - Verónica Bisagno
- grid.7345.50000 0001 0056 1981CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Farmacológicas (ININFA), Ciudad de Buenos Aires, Argentina
| | - Francisco J. Urbano
- grid.7345.50000 0001 0056 1981CONICET-Universidad de Buenos Aires, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Ciudad de Buenos Aires, Argentina ,grid.7345.50000 0001 0056 1981Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Fisiología, Biología Molecular y Celular “Dr. Héctor Maldonado”, Ciudad de Buenos Aires, Argentina ,grid.7345.50000 0001 0056 1981IFIBYNE (UBA-CONICET), Intendente Güiraldes 2160, Ciudad Universitaria, C1428EGA Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
36
|
Cuddy SR, Schinlever AR, Dochnal S, Seegren PV, Suzich J, Kundu P, Downs TK, Farah M, Desai BN, Boutell C, Cliffe AR. Neuronal hyperexcitability is a DLK-dependent trigger of herpes simplex virus reactivation that can be induced by IL-1. eLife 2020; 9:e58037. [PMID: 33350386 PMCID: PMC7773336 DOI: 10.7554/elife.58037] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Herpes simplex virus-1 (HSV-1) establishes a latent infection in neurons and periodically reactivates to cause disease. The stimuli that trigger HSV-1 reactivation have not been fully elucidated. We demonstrate HSV-1 reactivation from latently infected mouse neurons induced by forskolin requires neuronal excitation. Stimuli that directly induce neurons to become hyperexcitable also induced HSV-1 reactivation. Forskolin-induced reactivation was dependent on the neuronal pathway of DLK/JNK activation and included an initial wave of viral gene expression that was independent of histone demethylase activity and linked to histone phosphorylation. IL-1β is released under conditions of stress, fever and UV exposure of the epidermis; all known triggers of clinical HSV reactivation. We found that IL-1β induced histone phosphorylation and increased the excitation in sympathetic neurons. Importantly, IL-1β triggered HSV-1 reactivation, which was dependent on DLK and neuronal excitability. Thus, HSV-1 co-opts an innate immune pathway resulting from IL-1 stimulation of neurons to induce reactivation.
Collapse
Affiliation(s)
- Sean R Cuddy
- Department of Microbiology, Immunology and Cancer Biology, University of VirginiaCharlottesvilleUnited States
- Neuroscience Graduate Program, University of VirginiaCharlottesvilleUnited States
| | - Austin R Schinlever
- Department of Microbiology, Immunology and Cancer Biology, University of VirginiaCharlottesvilleUnited States
| | - Sara Dochnal
- Department of Microbiology, Immunology and Cancer Biology, University of VirginiaCharlottesvilleUnited States
| | - Philip V Seegren
- Department of Pharmacology, University of VirginiaCharlottesvilleUnited States
| | - Jon Suzich
- Department of Microbiology, Immunology and Cancer Biology, University of VirginiaCharlottesvilleUnited States
| | - Parijat Kundu
- Department of Microbiology, Immunology and Cancer Biology, University of VirginiaCharlottesvilleUnited States
| | - Taylor K Downs
- Department of Pharmacology, University of VirginiaCharlottesvilleUnited States
| | - Mina Farah
- Department of Microbiology, Immunology and Cancer Biology, University of VirginiaCharlottesvilleUnited States
| | - Bimal N Desai
- Department of Pharmacology, University of VirginiaCharlottesvilleUnited States
| | - Chris Boutell
- MRC-University of Glasgow Centre for Virus Research (CVR), Garscube CampusGlasgowUnited Kingdom
| | - Anna R Cliffe
- Department of Microbiology, Immunology and Cancer Biology, University of VirginiaCharlottesvilleUnited States
| |
Collapse
|
37
|
Santoro B, Shah MM. Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels as Drug Targets for Neurological Disorders. Annu Rev Pharmacol Toxicol 2020; 60:109-131. [PMID: 31914897 DOI: 10.1146/annurev-pharmtox-010919-023356] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are voltage-gated ion channels that critically modulate neuronal activity. Four HCN subunits (HCN1-4) have been cloned, each having a unique expression profile and distinctive effects on neuronal excitability within the brain. Consistent with this, the expression and function of these subunits are altered in diverse ways in neurological disorders. Here, we review current knowledge on the structure and distribution of the individual HCN channel isoforms, their effects on neuronal activity under physiological conditions, and how their expression and function are altered in neurological disorders, particularly epilepsy, neuropathic pain, and affective disorders. We discuss the suitability of HCN channels as therapeutic targets and how drugs might be strategically designed to specifically act on particular isoforms. We conclude that medicines that target individual HCN isoforms and/or their auxiliary subunit, TRIP8b, may provide valuable means of treating distinct neurological conditions.
Collapse
Affiliation(s)
- Bina Santoro
- Department of Neuroscience, Columbia University, New York, NY 10027, USA
| | - Mala M Shah
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, United Kingdom;
| |
Collapse
|
38
|
Trompoukis G, Rigas P, Leontiadis LJ, Papatheodoropoulos C. I h, GIRK, and KCNQ/Kv7 channels differently modulate sharp wave - ripples in the dorsal and ventral hippocampus. Mol Cell Neurosci 2020; 107:103531. [PMID: 32711112 DOI: 10.1016/j.mcn.2020.103531] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 07/11/2020] [Accepted: 07/16/2020] [Indexed: 12/14/2022] Open
Abstract
Sharp waves and ripples (SPW-Rs) are endogenous transient patterns of hippocampus local network activity implicated in several functions including memory consolidation, and they are diversified between the dorsal and the ventral hippocampus. Ion channels in the neuronal membrane play important roles in cell and local network function. In this study, using transverse slices and field potential recordings from the CA1 field of rat hippocampus we show that GIRK and KCNQ2/3 potassium channels play a higher role in modulating SPW-Rs in the dorsal hippocampus, while Ih and other KCNQ (presumably KCNQ5) channels, contribute to shaping SPW-R activity more in the ventral than in dorsal hippocampus. Specifically, blockade of Ih channels by ZD 7288 reduced the rate of occurrence of SPW-Rs and increased the generation of SPW-Rs in the form of clusters in both hippocampal segments, while enhanced the amplitude of SPW-Rs only in the ventral hippocampus. Most effects of ZD 7288 appeared to be independent of NMDA receptors' activity. However, the effects of blockade of NMDA receptors depended on the functional state of Ih channels in both hippocampal segments. Blockade of GIRK channels by Tertiapin-Q increased the rate of occurrence of SPW-Rs only in the dorsal hippocampus and the probability of clusters in both segments of the hippocampus. Blockade of KCNQ2/3 channels by XE 991 increased the rate of occurrence of SPW-Rs and the probability of clusters in the dorsal hippocampus, and only reduced the clustered generation of SPW-Rs in the ventral hippocampus. The blocker of KCNQ1/2 channels, that also enhances KCNQ5 channels, UCL 2077, increased the probability of clusters and the power of the ripple oscillation in the ventral hippocampus only. These results suggest that GIRK, KCNQ and Ih channels represent a key mechanism for modulation of SPW-R activity which act differently in the dorsal and ventral hippocampus, fundamentally supporting functional diversification along the dorsal-ventral axis of the hippocampus.
Collapse
Affiliation(s)
- George Trompoukis
- Laboratory of Physiology, Department of Medicine, University of Patras, Rion, Greece
| | - Pavlos Rigas
- Laboratory of Physiology, Department of Medicine, University of Patras, Rion, Greece
| | - Leonidas J Leontiadis
- Laboratory of Physiology, Department of Medicine, University of Patras, Rion, Greece
| | | |
Collapse
|
39
|
Lignani G, Baldelli P, Marra V. Homeostatic Plasticity in Epilepsy. Front Cell Neurosci 2020; 14:197. [PMID: 32676011 PMCID: PMC7333442 DOI: 10.3389/fncel.2020.00197] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/05/2020] [Indexed: 11/26/2022] Open
Abstract
In the healthy brain, neuronal excitability and synaptic strength are homeostatically regulated to keep neuronal network activity within physiological boundaries. Epilepsy is characterized by episodes of highly synchronized firing across in widespread neuronal populations, due to a failure in regulation of network activity. Here we consider epilepsy as a failure of homeostatic plasticity or as a maladaptive response to perturbations in the activity. How homeostatic compensation is involved in epileptogenic processes or in the chronic phase of epilepsy, is still debated. Although several theories have been proposed, there is relatively little experimental evidence to evaluate them. In this perspective, we will discuss recent results that shed light on the potential role of homeostatic plasticity in epilepsy. First, we will present some recent insights on how homeostatic compensations are probably active before and during epileptogenesis and how their actions are temporally regulated and closely dependent on the progression of pathology. Then, we will consider the dual role of transcriptional regulation during epileptogenesis, and finally, we will underline the importance of homeostatic plasticity in the context of therapeutic interventions for epilepsy. While classic pharmacological interventions may be counteracted by the epileptic brain to maintain its potentially dysfunctional set point, novel therapeutic approaches may provide the neuronal network with the tools necessary to restore its physiological balance.
Collapse
Affiliation(s)
- Gabriele Lignani
- Department of Clinical and Experimental Epilepsy, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Pietro Baldelli
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Vincenzo Marra
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
40
|
Rivolta I, Binda A, Masi A, DiFrancesco JC. Cardiac and neuronal HCN channelopathies. Pflugers Arch 2020; 472:931-951. [PMID: 32424620 DOI: 10.1007/s00424-020-02384-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/21/2020] [Accepted: 04/24/2020] [Indexed: 12/31/2022]
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are expressed as four different isoforms (HCN1-4) in the heart and in the central and peripheral nervous systems. In the voltage range of activation, HCN channels carry an inward current mediated by Na+ and K+, termed If in the heart and Ih in neurons. Altered function of HCN channels, mainly HCN4, is associated with sinus node dysfunction and other arrhythmias such as atrial fibrillation, ventricular tachycardia, and atrioventricular block. In recent years, several data have also shown that dysfunctional HCN channels, in particular HCN1, but also HCN2 and HCN4, can play a pathogenic role in epilepsy; these include experimental data from animal models, and data collected over genetic mutations of the channels identified and characterized in epileptic patients. In the central nervous system, alteration of the Ih current could predispose to the development of neurodegenerative diseases such as Parkinson's disease; since HCN channels are widely expressed in the peripheral nervous system, their dysfunctional behavior could also be associated with the pathogenesis of neuropathic pain. Given the fundamental role played by the HCN channels in the regulation of the discharge activity of cardiac and neuronal cells, the modulation of their function for therapeutic purposes is under study since it could be useful in various pathological conditions. Here we review the present knowledge of the HCN-related channelopathies in cardiac and neurological diseases, including clinical, genetic, therapeutic, and physiopathological aspects.
Collapse
Affiliation(s)
- Ilaria Rivolta
- School of Medicine and Surgery, Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Monza, Italy
| | - Anna Binda
- School of Medicine and Surgery, Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Monza, Italy
| | - Alessio Masi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Jacopo C DiFrancesco
- School of Medicine and Surgery, Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Monza, Italy. .,Department of Neurology, ASST San Gerardo Hospital, University of Milano-Bicocca, Via Pergolesi, 33, 20900, Monza, MB, Italy.
| |
Collapse
|
41
|
Wei F, Wang Q, Han J, Goswamee P, Gupta A, McQuiston AR, Liu Q, Zhou L. Photodynamic Modification of Native HCN Channels Expressed in Thalamocortical Neurons. ACS Chem Neurosci 2020; 11:851-863. [PMID: 32078767 DOI: 10.1021/acschemneuro.9b00475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The photodynamic process requires three elements: light, oxygen, and photosensitizer, and involves the formation of singlet oxygen, the molecular oxygen in excited electronic states. Previously, we reported that heterologously expressed hyperpolarization-activated cAMP-gated (HCN) channels in excised membrane patches are sensitive to photodynamic modification (PDM). Here we extend this study to native HCN channels expressed in thalamocortical (TC) neurons in the ventrobasal (VB) complex of the thalamus and dopaminergic neurons (DA) of the ventral tegmental area (VTA). To do this, we introduced the photosensitizer FITC-cAMP into TCs or DAs of rodent brain slices via a whole-cell patch-clamp recording pipette. After illumination with blue light pulses, we observed an increase in the voltage-insensitive, instantaneous Iinst component, accompanied by a long-lasting decrease in the hyperpolarization-dependent Ih component. Both Ih and the increased Iinst after PDM could be blocked by the HCN blockers Cs+ and ZD7288. When FITC and cAMP were dissociated and loaded into neurons as two separate chemicals, light application did not result in any long-lasting changes of the HCN currents. In contrast, light pulses applied to HCN2-/- neurons loaded with FITC-cAMP generated a much greater reduction in the Iinst component compared to that of WT neurons. Next, we investigated the impact of the long-lasting increases in Iinst after PDM on the cellular physiology of VB neurons. Consistent with an upregulation of HCN channel function, PDM elicited a depolarization of the resting membrane potential (RMP). Importantly, Trolox-C, an effective quencher for singlet oxygen, could block the PDM-dependent increase in Iinst and depolarization of the RMP. We propose that PDM of native HCN channels under physiological conditions may provide a photodynamic approach to alleviate HCN channelopathy in certain pathological conditions.
Collapse
Affiliation(s)
- Fusheng Wei
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330031, Jiangxi, China
| | - Qiang Wang
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Jizhong Han
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Priyodarshan Goswamee
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Ankush Gupta
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Adam Rory McQuiston
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Qinglian Liu
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Lei Zhou
- Department of Physiology and Biophysics, Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| |
Collapse
|
42
|
Islas JF, Abbasgholizadeh R, Dacso C, Potaman VN, Navran S, Bond RA, Iyer D, Birla R, Schwartz RJ. β-Adrenergic stimuli and rotating suspension culture enhance conversion of human adipogenic mesenchymal stem cells into highly conductive cardiac progenitors. J Tissue Eng Regen Med 2020; 14:306-318. [PMID: 31821703 DOI: 10.1002/term.2994] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 11/08/2019] [Accepted: 11/19/2019] [Indexed: 12/26/2022]
Abstract
Clinical trials using human adipogenic mesenchymal stem cells (hAdMSCs) for the treatment of cardiac diseases have shown improvement in cardiac function and were proven safe. However, hAdMSCs do not convert efficiently into cardiomyocytes (CMs) or vasculature. Thus, reprogramming hAdMSCs into myocyte progenitors may fare better in future investigations. To reprogramme hAdMSCs into electrically conductive cardiac progenitor cells, we pioneered a three-step reprogramming strategy that uses proven MESP1/ETS2 transcription factors, β-adrenergic and hypoxic signalling induced in three-dimensional (3D) cardiospheres. In Stage 1, ETS2 and MESP1 activated NNKX2.5, TBX5, MEF2C, dHAND, and GATA4 during the conversion of hAdMSCs into cardiac progenitor cells. Next, in Stage 2, β2AR activation repositioned cardiac progenitors into de novo immature conductive cardiac cells, along with the appearance of RYR2, CAV2.1, CAV3.1, NAV1.5, SERCA2, and CX45 gene transcripts and displayed action potentials. In Stage 3, electrical conduction that was fostered by 3D cardiospheres formed in a Synthecon®, Inc. rotating bioreactor induced the appearance of hypoxic genes: HIF-1α/β, PCG 1α/β, and NOS2, which coincided with the robust activation of adult contractile genes including MLC2v, TNNT2, and TNNI3, ion channel genes, and the appearance of hyperpolarization-activated and cyclic nucleotide-gated channels (HCN1-4). Conduction velocities doubled to ~200 mm/s after hypoxia and doubled yet again after dissociation of the 3D cell clusters to ~400 mm/s. By comparison, normal conduction velocities within working ventricular myocytes in the whole heart range from 0.5 to 1 m/s. Epinephrine stimulation of stage 3 cardiac cells in patches resulted in an increase in amplitude of the electrical wave, indicative of conductive cardiac cells. Our efficient protocol that converted hAdMSCs into highly conductive cardiac progenitors demonstrated the potential utilization of stage 3 cells for tissue engineering applications for cardiac repair.
Collapse
Affiliation(s)
- Jose Francisco Islas
- Texas Heart Institute, Texas Medical Center, Houston, TX.,Departamento de Bioquímica y Medicina Molecular, Faculta de Medicina, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | | | - Clifford Dacso
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Texas Medical Center, Houston, TX
| | | | | | - Richard A Bond
- College of Pharmacy, Science and Engineering Research Center, University of Houston, Houston, TX
| | - Dinakar Iyer
- Department of Biology and Biochemistry, University of Houston, Houston, TX
| | - Ravi Birla
- Department of Biomedical Engineering, University of Houston, Houston, TX
| | - Robert J Schwartz
- Texas Heart Institute, Texas Medical Center, Houston, TX.,Department of Biology and Biochemistry, University of Houston, Houston, TX
| |
Collapse
|
43
|
Zhong LY, Fan XR, Shi ZJ, Fan ZC, Luo J, Lin N, Liu YC, Wu L, Zeng XR, Cao JM, Wei Y. Hyperpolarization-Activated Cyclic Nucleotide-Gated Ion (HCN) Channels Regulate PC12 Cell Differentiation Toward Sympathetic Neuron. Front Cell Neurosci 2019; 13:415. [PMID: 31616252 PMCID: PMC6763607 DOI: 10.3389/fncel.2019.00415] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 08/28/2019] [Indexed: 12/15/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated ion channels (HCN channels) are widely expressed in the central and peripheral nervous systems and organs, while their functions are not well elucidated especially in the sympathetic nerve. The present study aimed to investigate the roles of HCN channel isoforms in the differentiation of sympathetic neurons using PC12 cell as a model. PC12 cells derived from rat pheochromocytoma were cultured and induced by nerve growth factor (NGF) (25 ng/ml) to differentiate to sympathetic neuron-like cells. Sympathetic directional differentiation of PC12 cells were evaluated by expressions of growth-associated protein 43 (GAP-43) (a growth cone marker), tyrosine hydroxylase (TH) (a sympathetic neuron marker) and neurite outgrowth. Results show that the HCN channel isoforms (HCN1-4) were all expressed in PC12 cells; blocking HCN channels with ivabradine suppressed NGF-induced GAP-43 expression and neurite outgrowth; silencing the expression of HCN2 and HCN4 using silenced using small interfering RNAs (siRNA), rather than HCN1 and HCN3, restrained GAP-43 expression and neurite outgrowth, while overexpression of HCN2 and HCN4 channels with gene transfer promoted GAP-43 expression and neurite outgrowth. Patch clamp experiments show that PC12 cells exhibited resting potentials (RP) of about −65 to −70 mV, and also presented inward HCN channel currents and outward (K+) currents, but no inward voltage-gated Na+ current was induced; NGF did not significantly affect the RP but promoted the establishment of excitability as indicated by the increased ability to depolarize and repolarize in the evoked suspicious action potentials (AP). We conclude that HCN2 and HCN4 channel isoforms, but not HCN1 and HCN3, promote the differentiation of PC12 cells toward sympathetic neurons. NGF potentiates the establishment of excitability during PC12 cell differentiation.
Collapse
Affiliation(s)
- Li-Ying Zhong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xin-Rong Fan
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zhang-Jing Shi
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Zhong-Cai Fan
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jian Luo
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Na Lin
- Department of Respiratory Medicine, Rongcheng People's Hospital, Rongcheng, China
| | - Ying-Cai Liu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lin Wu
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.,Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Xiao-Rong Zeng
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Ji-Min Cao
- Key Laboratory of Cellular Physiology of Ministry of Education, Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Yan Wei
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
44
|
Song C, Orlandi C, Sutton LP, Martemyanov KA. The signaling proteins GPR158 and RGS7 modulate excitability of L2/3 pyramidal neurons and control A-type potassium channel in the prelimbic cortex. J Biol Chem 2019; 294:13145-13157. [PMID: 31311860 DOI: 10.1074/jbc.ra119.007533] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 07/11/2019] [Indexed: 12/18/2022] Open
Abstract
Stress profoundly affects physiological properties of neurons across brain circuits and thereby increases the risk for depression. However, the molecular and cellular mechanisms mediating these effects are poorly understood. In this study, we report that chronic physical restraint stress in mice decreases excitability specifically in layer 2/3 of pyramidal neurons within the prelimbic subarea of the prefrontal cortex (PFC) accompanied by the induction of depressive-like behavioral states. We found that a complex between G protein-coupled receptor (GPCR) 158 (GPR158) and regulator of G protein signaling 7 (RGS7), a regulatory GPCR signaling node recently discovered to be a key modulator of affective behaviors, plays a key role in controlling stress-induced changes in excitability in this neuronal population. Deletion of GPR158 or RGS7 enhanced excitability of layer 2/3 PFC neurons and prevented the impact of stress. Investigation of the underlying molecular mechanisms revealed that the A-type potassium channel Kv4.2 subunit is a molecular target of the GPR158-RGS7 complex. We further report that GPR158 physically associates with Kv4.2 channel and promotes its function by suppressing inhibitory modulation by cAMP-protein kinase A (PKA)-mediated phosphorylation. Taken together, our observations reveal a critical mechanism that adjusts neuronal excitability in L2/3 pyramidal neurons of the PFC and may thereby modulate the effects of stress on depression.
Collapse
Affiliation(s)
- Chenghui Song
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458
| | - Cesare Orlandi
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458
| | - Laurie P Sutton
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458.
| |
Collapse
|
45
|
Hajisoltani R, Karimi SA, Rahdar M, Davoudi S, Borjkhani M, Hosseinmardi N, Behzadi G, Janahmadi M. Hyperexcitability of hippocampal CA1 pyramidal neurons in male offspring of a rat model of autism spectrum disorder (ASD) induced by prenatal exposure to valproic acid: A possible involvement of Ih channel current. Brain Res 2019; 1708:188-199. [DOI: 10.1016/j.brainres.2018.12.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 11/30/2018] [Accepted: 12/07/2018] [Indexed: 11/25/2022]
|
46
|
da Silva MP, Moraes DJA, Bonagamba LGH, Mecawi ADS, Varanda WA, Machado BH. Hyperexcitability and plasticity induced by sustained hypoxia on rectus abdominis motoneurons. J Physiol 2019; 597:1935-1956. [PMID: 30747446 DOI: 10.1113/jp277030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 02/06/2019] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Acute hypoxia induces active expiration in rectus abdominis (RA) muscles in conscious freely moving rats, although its overall contribution is smaller than in internal oblique (IO) muscles. Tonically active and silent RA motoneurons were identified in in vitro preparations of rat spinal cords. Sustained hypoxia (SH) increased the synaptic strength and induced morphological changes in tonically active RA motoneurons. Expiratory RA motoneurons were recorded in the in situ preparation and SH enhanced both the excitability and the synaptic transmission in those firing during the stage 2 expiration. The present study contributes to a better understanding of the mechanisms involved in SH recruitment of RA motoneurons to induce active expiration in rats. ABSTRACT Rectus abdominis (RA) motoneurons translate the complex respiratory brainstem inputs into effective muscle contractions. Despite their fundamental role in respiration, their functional and morphological properties are not fully understood. In the present study, we investigated for the first time the contribution of RA muscle to active expiration and characterized RA motoneurons regarding their electrical, molecular and morphological profiles in control rats and in rats submitted to sustained hypoxia (SH), which induces chronic recruitment of abdominal muscles. Electromyographic experiments in conscious freely moving control rats and SH rats showed that RA contributes to active expiration induced by acute hypoxia, although its contribution is smaller than in internal oblique muscles. in vitro whole-cell patch clamp recordings from RA motoneurons revealed two populations of cells: tonically active and silent. SH induced hyperexcitability in the tonically active cells by changing their action potential properties, and EPSCs. Three-dimensional morphological reconstructions of these cells showed that SH increased the dendritic complexity, stimulated the appearance of dendrite spines, and increased the somatic area and volume. Physiologically identified RA motoneurons, firing in two distinct phases of expiration, were recorded in the brainstem-spinal cord in situ preparation of rats. SH increased the firing frequency and EPSCs of neurons firing during stage 2 expiration. Taken together, our results show that RA motoneurons reconfigure their biophysical properties, morphology and synaptic strength to produce an appropriate expiratory drive in response to SH in rats.
Collapse
Affiliation(s)
- Melina P da Silva
- School of Medicine of Ribeirão Preto, Department of Physiology, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Davi José A Moraes
- School of Medicine of Ribeirão Preto, Department of Physiology, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Leni G H Bonagamba
- School of Medicine of Ribeirão Preto, Department of Physiology, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - André de Souza Mecawi
- Paulista School of Medicine, Department of Biophysics, Federal University of São Paulo, São Paulo, Brazil
| | - Wamberto A Varanda
- School of Medicine of Ribeirão Preto, Department of Physiology, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Benedito H Machado
- School of Medicine of Ribeirão Preto, Department of Physiology, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
47
|
Abstract
Chronic pain is a frequent condition that affects an estimated 20% of people worldwide, accounting for 15%-20% of doctors' appointments (Treede et al., 2015). It lacks the acute warning function of physiologic nociception, and instead involves the activation of multiple neurophysiologic mechanisms in the somatosensory system, a complex neuronal network under the control of powerful autoregulatory loops and able to undergo rapid neuroplastic alteration (Verdu et al., 2008). There is a growing body of research suggesting that some such pathways are shared by major psychologic disorders such as depression and anxiety, opening new avenues in co-treatment strategies. In particular, besides anticonvulsants, which are today used as analgesics, other psychopharmaceuticals, such as the tricyclic antidepressants, are displaying efficacy in the treatment of neuropathic and nociceptive chronic pain. The state of the art regarding the mechanisms of nociception and the pharmacology of both the neurotransmitters involved and the wide range of psychoactive compounds that may be useful in the treatment of chronic pain are discussed.
Collapse
|
48
|
Urbanska M, Kazmierska-Grebowska P, Kowalczyk T, Caban B, Nader K, Pijet B, Kalita K, Gozdz A, Devijver H, Lechat B, Jaworski T, Grajkowska W, Sadowski K, Jozwiak S, Kotulska K, Konopacki J, Van Leuven F, van Vliet EA, Aronica E, Jaworski J. GSK3β activity alleviates epileptogenesis and limits GluA1 phosphorylation. EBioMedicine 2018; 39:377-387. [PMID: 30502054 PMCID: PMC6355642 DOI: 10.1016/j.ebiom.2018.11.040] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/16/2018] [Accepted: 11/16/2018] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Glycogen synthase kinase-3β (GSK3β) is a key regulator of cellular homeostasis. In neurons, GSK3β contributes to the control of neuronal transmission and plasticity, but its role in epilepsy remains to be defined. METHODS Biochemical and electrophysiological methods were used to assess the role of GSK3β in regulating neuronal transmission and epileptogenesis. GSK3β activity was increased genetically in GSK3β[S9A] mice. Its effects on neuronal transmission and epileptogenesis induced by kainic acid were assessed by field potential recordings in mice brain slices and video electroencephalography in vivo. The ion channel expression was measured in brain samples from mice and followed by analysis in samples from patients with temporal lobe epilepsy or focal cortical dysplasia in correlation to GSK3β phosphorylation. FINDINGS Higher GSK3β activity decreased the progression of kainic acid induced epileptogenesis. At the biochemical level, higher GSK3β activity increased the expression of hyperpolarization-activated cyclic nucleotide-gated (HCN) channel 4 under basal conditions and in the epileptic mouse brain and decreased phosphorylation of the glutamate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluA1 at Serine 831 under basal conditions. Moreover, we found a significant correlation between higher inhibitory GSK3β phosphorylation at Serine 9 and higher activating GluA1 phosphorylation at Serine 845 in brain samples from epileptic patients. INTERPRETATION Our data imply GSK3β activity in the protection of neuronal networks from hyper-activation in response to epileptogenic stimuli and indicate that the anti-epileptogenic function of GSK3β involves modulation of HCN4 level and the synaptic AMPA receptors pool.
Collapse
Affiliation(s)
- Malgorzata Urbanska
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw 02-109, Poland; Department of Neurology and Epileptology, Children's Memorial Health Institute, Warsaw 04-730, Poland.
| | - Paulina Kazmierska-Grebowska
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz 90-236, Poland
| | - Tomasz Kowalczyk
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz 90-236, Poland
| | - Bartosz Caban
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz 90-236, Poland
| | - Karolina Nader
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Science, Warsaw 02-093, Poland
| | - Barbara Pijet
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Science, Warsaw 02-093, Poland
| | - Katarzyna Kalita
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Science, Warsaw 02-093, Poland
| | - Agata Gozdz
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw 02-109, Poland
| | - Herman Devijver
- Experimental Genetics Group - LEGTEGG, Department of Human Genetics, KULeuven, Leuven 3000, Belgium
| | - Benoit Lechat
- Experimental Genetics Group - LEGTEGG, Department of Human Genetics, KULeuven, Leuven 3000, Belgium
| | - Tomasz Jaworski
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Science, Warsaw 02-093, Poland
| | - Wieslawa Grajkowska
- Department of Pathology, Children's Memorial Health Institute, Warsaw 04-730, Poland
| | - Krzysztof Sadowski
- Department of Neurology and Epileptology, Children's Memorial Health Institute, Warsaw 04-730, Poland
| | - Sergiusz Jozwiak
- Department of Neurology and Epileptology, Children's Memorial Health Institute, Warsaw 04-730, Poland; Department of Child Neurology, Medical University of Warsaw, Warsaw 02-091, Poland
| | - Katarzyna Kotulska
- Department of Neurology and Epileptology, Children's Memorial Health Institute, Warsaw 04-730, Poland
| | - Jan Konopacki
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz 90-236, Poland
| | - Fred Van Leuven
- Experimental Genetics Group - LEGTEGG, Department of Human Genetics, KULeuven, Leuven 3000, Belgium
| | - Erwin A van Vliet
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, the Netherlands; Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, 1098 XH, the Netherlands
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, the Netherlands; Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, the Netherlands
| | - Jacek Jaworski
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw 02-109, Poland.
| |
Collapse
|
49
|
Zhou R, Jiang G, Tian X, Wang X. Progress in the molecular mechanisms of genetic epilepsies using patient-induced pluripotent stem cells. Epilepsia Open 2018; 3:331-339. [PMID: 30187003 PMCID: PMC6119748 DOI: 10.1002/epi4.12238] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2018] [Indexed: 12/29/2022] Open
Abstract
Research findings on the molecular mechanisms of epilepsy almost always originate from animal experiments, and the development of induced pluripotent stem cell (iPSC) technology allows the use of human cells with genetic defects for studying the molecular mechanisms of genetic epilepsy (GE) for the first time. With iPSC technology, terminally differentiated cells collected from GE patients with specific genetic etiologies can be differentiated into many relevant cell subtypes that carry all of the GE patient's genetic information. iPSCs have opened up a new research field involving the pathogenesis of GE. Using this approach, studies have found that gene mutations induce GE by altering the balance between neuronal excitation and inhibition, which is associated. among other factors, with neuronal developmental disturbances, ion channel abnormalities, and synaptic dysfunction. Simultaneously, astrocyte activation, mitochondrial dysfunction, and abnormal signaling pathway activity are also important factors in the molecular mechanisms of GE.
Collapse
Affiliation(s)
- Ruijiao Zhou
- Department of Neurology the First Affiliated Hospital of Chongqing Medical University Chongqing Key Laboratory of Neurology Chongqing China
| | - Guohui Jiang
- Department of Neurology Institute of Neurological Diseases Affiliated Hospital of North Sichuan Medical College Nanchong China
| | - Xin Tian
- Department of Neurology the First Affiliated Hospital of Chongqing Medical University Chongqing Key Laboratory of Neurology Chongqing China
| | - Xuefeng Wang
- Department of Neurology the First Affiliated Hospital of Chongqing Medical University Chongqing Key Laboratory of Neurology Chongqing China
| |
Collapse
|
50
|
Balakrishnan S, Mironov SL. Rescue of hyperexcitability in hippocampal CA1 neurons from Mecp2 (-/y) mouse through surface potential neutralization. PLoS One 2018; 13:e0195094. [PMID: 29621262 PMCID: PMC5886422 DOI: 10.1371/journal.pone.0195094] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 03/18/2018] [Indexed: 11/19/2022] Open
Abstract
Hyperventilation is a known feature of Rett syndrome (RTT). However, how hyperventilation is related to other RTT symptoms such as hyperexcitability is unknown. Intense breathing during hyperventilation induces hypocapnia and culminates in respiratory alkalosis. Alkalinization of extracellular milieu can trigger epilepsy in patients who already have neuronal hyperexcitability. By combining patch-clamp electrophysiology and quantitative glutamate imaging, we compared excitability of CA1 neurons of WT and Mecp2 (-/y) mice, and analyzed the biophysical properties of subthreshold membrane channels. The results show that Mecp2 (-/y) CA1 neurons are hyperexcitable in normal pH (7.4) and are increasingly vulnerable to alkaline extracellular pH (8.4), during which their excitability increased further. Under normal pH conditions, an abnormal negative shift in the voltage-dependencies of HCN (hyperpolarization-activated cyclic nucleotide-gated) and calcium channels in the CA1 neurons of Mecp2 (-/y) mice was observed. Alkaline pH also enhanced excitability in wild-type (WT) CA1 neurons through modulation of the voltage dependencies of HCN- and calcium channels. Additionally alkaline pH augmented spontaneous glutamate release and burst firing in WT CA1 neurons. Conversely, acidic pH (6.4) and 8 mM Mg2+ exerted the opposite effect, and diminished hyperexcitability in Mecp2 (-/y) CA1 neurons. We propose that the observed effects of pH and Mg2+ are mediated by changes in the neuronal membrane surface potential, which consecutively modulates the gating of HCN and calcium channels. The results provide insight to pivotal cellular mechanisms that can regulate neuronal excitability and help to devise treatment strategies for hyperexcitability induced symptoms of Rett syndrome.
Collapse
Affiliation(s)
- Saju Balakrishnan
- CNMPB (Centre for Nanoscale Microscopy and Molecular Physiology of the Brain, Cluster of Excellence 171, DFG Research Center 103), Institute of Neuro and Sensory Physiology, Georg-August-University, Göttingen, Germany
| | - Sergej L. Mironov
- CNMPB (Centre for Nanoscale Microscopy and Molecular Physiology of the Brain, Cluster of Excellence 171, DFG Research Center 103), Institute of Neuro and Sensory Physiology, Georg-August-University, Göttingen, Germany
| |
Collapse
|