1
|
Krebs A, Kautzmann S, Klämbt C. Many faces of neuronal activity manipulation in Drosophila. Neural Regen Res 2025; 20:2574-2576. [PMID: 39503419 PMCID: PMC11801296 DOI: 10.4103/nrr.nrr-d-24-00524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/29/2024] [Accepted: 07/15/2024] [Indexed: 02/08/2025] Open
Affiliation(s)
- Amber Krebs
- Institut für Neuro- und Verhaltensbiologie, Münster, Germany
| | | | | |
Collapse
|
2
|
Richartz N, Pietka W, Yadav A, Bostad M, Bhagwat S, Naderi S, Naderi EH, Stokke T, Ruud E, Blomhoff HK. N-acetyl cysteine turns EPAC activators into potent killers of acute lymphoblastic leukemia cells. J Biol Chem 2024; 300:105509. [PMID: 38042493 PMCID: PMC10772734 DOI: 10.1016/j.jbc.2023.105509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 12/04/2023] Open
Abstract
Today, the majority of patients with pediatric B cell precursor acute lymphoblastic leukemia (BCP-ALL, hereafter ALL) survive their disease, but many of the survivors suffer from life-limiting late effects of the treatment. ALL develops in the bone marrow, where the cells are exposed to cAMP-generating prostaglandin E2. We have previously identified the cAMP signaling pathway as a putative target for improved efficacy of ALL treatment, based on the ability of cAMP signaling to reduce apoptosis induced by DNA damaging agents. In the present study, we have identified the antioxidant N-acetyl cysteine (NAC) as a powerful modifier of critical events downstream of the cell-permeable cAMP analog 8-(4-chlorophenylthio) adenosine-3', 5'- cyclic monophosphate (8-CPT). Accordingly, we found NAC to turn 8-CPT into a potent killer of ALL cells in vitro both in the presence and absence of DNA damaging treatment. Furthermore, we revealed that NAC in combination with 8-CPT is able to delay the progression of ALL in a xenograft model in NOD-scid IL2Rγnull mice. NAC was shown to rely on the ability of 8-CPT to activate the guanine-nucleotide exchange factor EPAC, and we demonstrated that the ALL cells are killed by apoptosis involving sustained elevated levels of calcium imposed by the combination of the two drugs. Taken together, we propose that 8-CPT in the presence of NAC might be utilized as a novel strategy for treating pediatric ALL patients, and that this powerful combination might be exploited to enhance the therapeutic index of current ALL targeting therapies.
Collapse
Affiliation(s)
- Nina Richartz
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Wojciech Pietka
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Ajay Yadav
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Monica Bostad
- Department of Core Facilities, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Sampada Bhagwat
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Soheil Naderi
- Division of Laboratory Medicine, Department of Pharmacology, Oslo University Hospital, Oslo, Norway
| | - Elin Hallan Naderi
- Section of Head and Neck Oncology, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Trond Stokke
- Department of Core Facilities, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Ellen Ruud
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Heidi Kiil Blomhoff
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
3
|
Piatkevich KD, Boyden ES. Optogenetic control of neural activity: The biophysics of microbial rhodopsins in neuroscience. Q Rev Biophys 2023; 57:e1. [PMID: 37831008 DOI: 10.1017/s0033583523000033] [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] [Indexed: 10/14/2023]
Abstract
Optogenetics, the use of microbial rhodopsins to make the electrical activity of targeted neurons controllable by light, has swept through neuroscience, enabling thousands of scientists to study how specific neuron types contribute to behaviors and pathologies, and how they might serve as novel therapeutic targets. By activating a set of neurons, one can probe what functions they can initiate or sustain, and by silencing a set of neurons, one can probe the functions they are necessary for. We here review the biophysics of these molecules, asking why they became so useful in neuroscience for the study of brain circuitry. We review the history of the field, including early thinking, early experiments, applications of optogenetics, pre-optogenetics targeted neural control tools, and the history of discovering and characterizing microbial rhodopsins. We then review the biophysical attributes of rhodopsins that make them so useful to neuroscience - their classes and structure, their photocycles, their photocurrent magnitudes and kinetics, their action spectra, and their ion selectivity. Our hope is to convey to the reader how specific biophysical properties of these molecules made them especially useful to neuroscientists for a difficult problem - the control of high-speed electrical activity, with great precision and ease, in the brain.
Collapse
Affiliation(s)
- Kiryl D Piatkevich
- School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Edward S Boyden
- McGovern Institute and Koch Institute, Departments of Brain and Cognitive Sciences, Media Arts and Sciences, and Biological Engineering, K. Lisa Yang Center for Bionics and Center for Neurobiological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Cambridge, MA, USA
| |
Collapse
|
4
|
Single-Cell Labeling Strategies to Dissect Neuronal Structures and Local Functions. BIOLOGY 2023; 12:biology12020321. [PMID: 36829594 PMCID: PMC9953318 DOI: 10.3390/biology12020321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
The brain network consists of ten billion neurons and is the most complex structure in the universe. Understanding the structure of complex brain networks and neuronal functions is one of the main goals of modern neuroscience. Since the seminal invention of Golgi staining, single-cell labeling methods have been among the most potent approaches for dissecting neuronal structures and neural circuits. Furthermore, the development of sparse single-cell transgenic methods has enabled single-cell gene knockout studies to examine the local functions of various genes in neural circuits and synapses. Here, we review non-transgenic single-cell labeling methods and recent advances in transgenic strategies for sparse single neuronal labeling. These methods and strategies will fundamentally contribute to the understanding of brain structure and function.
Collapse
|
5
|
Bustamante HA, Ehrich MF, Klein BG. Intracellular potassium depletion enhances apoptosis induced by staurosporine in cultured trigeminal satellite glial cells. Somatosens Mot Res 2021; 38:194-201. [PMID: 34187291 DOI: 10.1080/08990220.2021.1941843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE Satellite glial cells (SGC) surrounding neurons in sensory ganglia can buffer extracellular potassium, regulating the excitability of injured neurons and possibly influencing a shift from acute to neuropathic pain. SGC apoptosis may be a key component in this process. This work evaluated induction or enhancement of apoptosis in cultured trigeminal SGC following changes in intracellular potassium [K]ic. MATERIALS AND METHODS We developed SGC primary cultures from rat trigeminal ganglia (TG). Purity of our cultures was confirmed using immunofluorescence and western blot analysis for the presence of the specific marker of SGC, glutamine synthetase (GS). SGC [K]ic was depleted using hypo-osmotic shock and 4 mM bumetanide plus 10 mM ouabain. [K]ic was measured using the K+ fluorescent indicator potassium benzofuran isophthalate (PBFI-AM). RESULTS SGC tested positive for GS and hypo-osmotic shock induced a significant decrease in [K]ic at every evaluated time. Cells were then incubated for 5 h with either 2 mM staurosporine (STS) or 20 ng/ml of TNF-α and evaluated for early apoptosis and late apoptosis/necrosis by flow cytometry using annexin V and propidium iodide. A significant increase in early apoptosis, from 16 to 38%, was detected in SGC with depleted [K]ic after incubation with STS. In contrast, TNF-α did not increase early apoptosis in normal or [K]ic depleted SGC. CONCLUSION Hypo-osmotic shock induced a decrease in intracellular potassium in cultured trigeminal SGC and this enhanced apoptosis induced by STS that is associated with the mitochondrial pathway. These results suggest that K+ dysregulation may underlie apoptosis in trigeminal SGC.
Collapse
Affiliation(s)
- Hedie A Bustamante
- Faculty of Veterinary Sciences, Veterinary Clinical Sciences Institute, Universidad Austral de Chile, Valdivia, Chile
| | - Marion F Ehrich
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Bradley G Klein
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| |
Collapse
|
6
|
Yin S, Liu J, Kang Y, Lin Y, Li D, Shao L. Interactions of nanomaterials with ion channels and related mechanisms. Br J Pharmacol 2019; 176:3754-3774. [PMID: 31290152 DOI: 10.1111/bph.14792] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 06/10/2019] [Accepted: 07/01/2019] [Indexed: 12/15/2022] Open
Abstract
The pharmacological potential of nanotechnology, especially in drug delivery and bioengineering, has developed rapidly in recent decades. Ion channels, which are easily targeted by external agents, such as nanomaterials (NMs) and synthetic drugs, due to their unique structures, have attracted increasing attention in the fields of nanotechnology and pharmacology for the treatment of ion channel-related diseases. NMs have significant effects on ion channels, and these effects are manifested in many ways, including changes in ion currents, kinetic characteristics and channel distribution. Subsequently, intracellular ion homeostasis, signalling pathways, and intracellular ion stores are affected, leading to the initiation of a range of biological processes. However, the effect of the interactions of NMs with ion channels is an interesting topic that remains obscure. In this review, we have summarized the recent research progress on the direct and indirect interactions between NMs and ion channels and discussed the related molecular mechanisms, which are crucial to the further development of ion channel-related nanotechnological applications.
Collapse
Affiliation(s)
- Suhan Yin
- Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Jia Liu
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yiyuan Kang
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuqing Lin
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dongjian Li
- Liwan District Stomatology Hospital, Guangzhou, China
| | - Longquan Shao
- Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
7
|
Chen KF, Lowe S, Lamaze A, Krätschmer P, Jepson J. Neurocalcin regulates nighttime sleep and arousal in Drosophila. eLife 2019; 8:e38114. [PMID: 30865587 PMCID: PMC6415939 DOI: 10.7554/elife.38114] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 01/29/2019] [Indexed: 01/28/2023] Open
Abstract
Sleep-like states in diverse organisms can be separated into distinct stages, each with a characteristic arousal threshold. However, the molecular pathways underlying different sleep stages remain unclear. The fruit fly, Drosophila melanogaster, exhibits consolidated sleep during both day and night, with night sleep associated with higher arousal thresholds compared to day sleep. Here we identify a role for the neuronal calcium sensor protein Neurocalcin (NCA) in promoting sleep during the night but not the day by suppressing nocturnal arousal and hyperactivity. We show that both circadian and light-sensing pathways define the temporal window in which NCA promotes sleep. Furthermore, we find that NCA promotes sleep by suppressing synaptic release from a dispersed wake-promoting neural network and demonstrate that the mushroom bodies, a sleep-regulatory center, are a module within this network. Our results advance the understanding of how sleep stages are genetically defined.
Collapse
Affiliation(s)
- Ko-Fan Chen
- Department of Clinical and Experimental EpilepsyUCL Institute of NeurologyLondonUnited Kingdom
| | - Simon Lowe
- Department of Clinical and Experimental EpilepsyUCL Institute of NeurologyLondonUnited Kingdom
| | - Angélique Lamaze
- Department of Clinical and Experimental EpilepsyUCL Institute of NeurologyLondonUnited Kingdom
| | - Patrick Krätschmer
- Department of Clinical and Experimental EpilepsyUCL Institute of NeurologyLondonUnited Kingdom
| | - James Jepson
- Department of Clinical and Experimental EpilepsyUCL Institute of NeurologyLondonUnited Kingdom
| |
Collapse
|
8
|
Aldrin-Kirk P, Björklund T. Practical Considerations for the Use of DREADD and Other Chemogenetic Receptors to Regulate Neuronal Activity in the Mammalian Brain. Methods Mol Biol 2019; 1937:59-87. [PMID: 30706390 DOI: 10.1007/978-1-4939-9065-8_4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Chemogenetics is the process of genetically expressing a macromolecule receptor capable of modulating the activity of the cell in response to selective chemical ligand. This chapter will cover the chemogenetic technologies that are available to date, focusing on the commonly available engineered or otherwise modified ligand-gated ion channels and G-protein-coupled receptors in the context of neuromodulation. First, we will give a brief overview of each chemogenetic approach as well as in vitro/in vivo applications, then we will list their strengths and weaknesses. Finally, we will provide tips for ligand application in each case.Each technology has specific limitations that make them more or less suitable for different applications in neuroscience although we will focus mainly on the most commonly used and versatile family named designer receptors exclusively activated by designer drugs or DREADDs. We here describe the most common cases where these can be implemented and provide tips on how and where these technologies can be applied in the field of neuroscience.
Collapse
Affiliation(s)
- Patrick Aldrin-Kirk
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Tomas Björklund
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Lund University, Lund, Sweden.
| |
Collapse
|
9
|
Oxygen-glucose deprivation enhancement of cell death/apoptosis in PC12 cells and hippocampal neurons correlates with changes in neuronal excitatory amino acid neurotransmitter signaling and potassium currents. Neuroreport 2018; 27:617-26. [PMID: 27082843 DOI: 10.1097/wnr.0000000000000588] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Neuronal death is a pathophysiological process that is often caused by hypoxia/ischemia. However, the causes of hypoxia/ischemia-induced neuronal death are debated, and additional experimental data are needed to resolve this debate. In the present study, we applied oxygen-glucose deprivation (OGD) to PC12 cells and hippocampal neurons to establish a hypoxia/ischemia model. We evaluated the effects of OGD on cell death/apoptosis and on the levels of two excitatory amino acid neurotransmitters, aspartic acid and glutamic acid, in both hippocampal neurons and the medium used to culture the hippocampal neurons. We also evaluated GluR2 expression in hippocampal neurons as well as the effects of OGD on whole-cell potassium currents in PC12 cells and hippocampal neurons. Our experimental results showed that OGD significantly decreased cell viability and markedly enhanced apoptosis in PC12 cells and hippocampal neurons. OGD treatment for 3 h increased the levels of Asp and Glu in the medium used to culture hippocampal neurons, but decreased both the levels of Asp and Glu and GluR2 expression in hippocampal neurons. Furthermore, OGD altered the electrophysiological properties of voltage-dependent potassium channels in PC12 cells and hippocampal neurons in different ways; OGD decreased the voltage-dependent potassium current in PC12 cells, but increased this current in hippocampal neurons. On the basis of these results, we concluded that OGD enhanced neuronal cell death/apoptosis in addition to altering neuronal excitatory amino acid neurotransmitter signaling and whole-cell voltage-dependent potassium currents.
Collapse
|
10
|
Ma XL, Zhang F, Wang YX, He CC, Tian K, Wang HG, An D, Heng B, Liu YQ. Genistein inhibition of OGD-induced brain neuron death correlates with its modulation of apoptosis, voltage-gated potassium and sodium currents and glutamate signal pathway. Chem Biol Interact 2016; 254:73-82. [PMID: 27238724 DOI: 10.1016/j.cbi.2016.05.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 05/06/2016] [Accepted: 05/25/2016] [Indexed: 01/25/2023]
Abstract
In the present study, we established an in vitro model of hypoxic-ischemia via exposing primary neurons of newborn rats to oxygen-glucose deprivation (OGD) and observing the effects of genistein, a soybean isoflavone, on hypoxic-ischemic neuron viability, apoptosis, voltage-activated potassium (Kv) and sodium (Nav) currents, and glutamate receptor subunits. The results indicated that OGD exposure reduced the viability and increased the apoptosis of brain neurons. Meanwhile, OGD exposure caused changes in the current-voltage curves and current amplitude values of voltage-activated potassium and sodium currents; OGD exposure also decreased GluR2 expression and increased NR2 expression. However, genistein at least partially reversed the effects caused by OGD. The results suggest that hypoxic-ischemia-caused neuronal apoptosis/death is related to an increase in K(+) efflux, a decrease in Na(+) influx, a down-regulation of GluR2, and an up-regulation of NR2. Genistein may exert some neuroprotective effects via the modulation of Kv and Nav currents and the glutamate signal pathway, mediated by GluR2 and NR2.
Collapse
Affiliation(s)
- Xue-Ling Ma
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Feng Zhang
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yu-Xiang Wang
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Cong-Cong He
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Kun Tian
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Hong-Gang Wang
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Di An
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Bin Heng
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yan-Qiang Liu
- College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
11
|
Sesti F. Oxidation of K(+) Channels in Aging and Neurodegeneration. Aging Dis 2016; 7:130-5. [PMID: 27114846 PMCID: PMC4809605 DOI: 10.14336/ad.2015.0901] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 09/01/2015] [Indexed: 01/26/2023] Open
Abstract
Reversible regulation of proteins by reactive oxygen species (ROS) is an important mechanism of neuronal plasticity. In particular, ROS have been shown to act as modulatory molecules of ion channels-which are key to neuronal excitability-in several physiological processes. However ROS are also fundamental contributors to aging vulnerability. When the level of excess ROS increases in the cell during aging, DNA is damaged, proteins are oxidized, lipids are degraded and more ROS are produced, all culminating in significant cell injury. From this arose the idea that oxidation of ion channels by ROS is one of the culprits for neuronal aging. Aging-dependent oxidative modification of voltage-gated potassium (K(+)) channels was initially demonstrated in the nematode Caenorhabditis elegans and more recently in the mammalian brain. Specifically, oxidation of the delayed rectifier KCNB1 (Kv2.1) and of Ca(2+)- and voltage sensitive K(+) channels have been established suggesting that their redox sensitivity contributes to altered excitability, progression of healthy aging and of neurodegenerative disease. Here I discuss the implications that oxidation of K(+) channels by ROS may have for normal aging, as well as for neurodegenerative disease.
Collapse
Affiliation(s)
- Federico Sesti
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
12
|
Calstabin 2: An important regulator for learning and memory in mice. Sci Rep 2016; 6:21087. [PMID: 26888649 PMCID: PMC4758079 DOI: 10.1038/srep21087] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 01/14/2016] [Indexed: 12/20/2022] Open
Abstract
Calstabin2, also named FK506 binding protein 12.6 (FKBP12.6), is a subunit of ryanodine receptor subtype 2 (RyR2) macromolecular complex, which is an intracellular calcium channel and abundant in the brain. Previous studies identified a role of leaky neuronal RyR2 in posttraumatic stress disorder (PTSD). However, the functional role of Calstabin2 in the cognitive function remains unclear. Herein, we used a mouse model of genetic deletion of Calstabin2 to investigate the function of Calstabin2 in cognitive dysfunction. We found that Calstabin2 knockout (KO) mice showed significantly reduced performance in Morris Water Maze (MWM), long-term memory (LTM) contextual fear testing, and rotarod test when compared to wild type (WT) littermates. Indeed, genetic deletion of Calstabin2 reduced long-term potentiation (LTP) at the hippocampal CA3-CA1 connection, increased membrane excitability, and induced RyR2 leak. Finally, we demonstrated that the increase in cytoplasmic calcium activated Ca(2+) dependent potassium currents and led to neuronal apoptosis in KO hippocampal neurons. Thus, these results suggest that neuronal RyR2 Ca(2+) leak due to Calstabin2 deletion contributes to learning deficiency and memory impairment.
Collapse
|
13
|
Increase in the titer of lentiviral vectors expressing potassium channels by current blockade during viral vector production. BMC Neurosci 2015; 16:30. [PMID: 25940378 PMCID: PMC4425897 DOI: 10.1186/s12868-015-0159-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 04/01/2015] [Indexed: 12/02/2022] Open
Abstract
Background High titers of lentiviral vectors are required for the efficient transduction of a gene of interest. During preparation of lentiviral the vectors, the protein of interest is inevitably expressed in the viral vector-producing cells. This expression may affect the production of the lentiviral vector. Methods We prepared lentiviral vectors expressing inwardly rectifying potassium channel (Lv-Kir2.1), its dominant-negative form (Lv-Kir-DN), and other K+ channels, using the ubiquitously active β-actin and neuron-specific synapsin I promoters. Results The titer of Lv-Kir-DN was higher than that of Lv-Kir2.1, suggesting a negative effect of induced K+ currents on viral titer. We then blocked Kir2.1 currents with the selective blocker Ba2+ during Lv-Kir2.1 production, and obtained about a 5-fold increase in the titer. Higher extracellular K+ concentrations increased the titer of Lv-Kir2.1 about 9-fold. With a synapsin I promoter Ba2+ increased the titer because of the moderate expression of Kir2.1 channel. Channel blockade also increased the titers of the lentivirus expressing Kv1.4 and TREK channels, but not HERG. The increase in titer correlated with the K+ currents generated by the channels expressed. Conclusion In the production of lentivirus expressing K+ channels, titers are increased by blocking K+ currents in the virus-producing cells. This identifies a crucial issue in the production of viruses expressing membrane channels, and should facilitate basic and gene therapeutic research on channelopathies.
Collapse
|
14
|
Kondratskyi A, Kondratska K, Skryma R, Prevarskaya N. Ion channels in the regulation of apoptosis. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:2532-46. [PMID: 25450339 DOI: 10.1016/j.bbamem.2014.10.030] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 10/08/2014] [Accepted: 10/20/2014] [Indexed: 02/07/2023]
Abstract
Apoptosis, a type of genetically controlled cell death, is a fundamental cellular mechanism utilized by multicellular organisms for disposal of cells that are no longer needed or potentially detrimental. Given the crucial role of apoptosis in physiology, deregulation of apoptotic machinery is associated with various diseases as well as abnormalities in development. Acquired resistance to apoptosis represents the common feature of most and perhaps all types of cancer. Therefore, repairing and reactivating apoptosis represents a promising strategy to fight cancer. Accumulated evidence identifies ion channels as essential regulators of apoptosis. However, the contribution of specific ion channels to apoptosis varies greatly depending on cell type, ion channel type and intracellular localization, pathology as well as intracellular signaling pathways involved. Here we discuss the involvement of major types of ion channels in apoptosis regulation. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
Affiliation(s)
- Artem Kondratskyi
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale Contre le Cancer, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université Lille 1, Villeneuve d'Ascq, France
| | - Kateryna Kondratska
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale Contre le Cancer, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université Lille 1, Villeneuve d'Ascq, France
| | - Roman Skryma
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale Contre le Cancer, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université Lille 1, Villeneuve d'Ascq, France
| | - Natalia Prevarskaya
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale Contre le Cancer, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université Lille 1, Villeneuve d'Ascq, France.
| |
Collapse
|
15
|
Lee CH, Liou HH. Pregabalin activates ROMK1 channels via cAMP-dependent protein kinase and protein kinase C. Eur J Pharmacol 2014; 740:35-44. [DOI: 10.1016/j.ejphar.2014.06.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 05/21/2014] [Accepted: 06/16/2014] [Indexed: 02/01/2023]
|
16
|
Abstract
Elucidating the roles of neuronal cell types for physiology and behavior is essential for understanding brain functions. Perturbation of neuron electrical activity can be used to probe the causal relationship between neuronal cell types and behavior. New genetically encoded neuron perturbation tools have been developed for remotely controlling neuron function using small molecules that activate engineered receptors that can be targeted to cell types using genetic methods. Here we describe recent progress for approaches using genetically engineered receptors that selectively interact with small molecules. Called "chemogenetics," receptors with diverse cellular functions have been developed that facilitate the selective pharmacological control over a diverse range of cell-signaling processes, including electrical activity, for molecularly defined cell types. These tools have revealed remarkably specific behavioral physiological influences for molecularly defined cell types that are often intermingled with populations having different or even opposite functions.
Collapse
Affiliation(s)
- Scott M Sternson
- Janelia Farm Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia 20147;
| | | |
Collapse
|
17
|
Kang JY, Kawaguchi D, Coin I, Xiang Z, O'Leary DDM, Slesinger PA, Wang L. In vivo expression of a light-activatable potassium channel using unnatural amino acids. Neuron 2014; 80:358-70. [PMID: 24139041 DOI: 10.1016/j.neuron.2013.08.016] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2013] [Indexed: 01/28/2023]
Abstract
Optical control of protein function provides excellent spatial-temporal resolution for studying proteins in situ. Although light-sensitive exogenous proteins and ligands have been used to manipulate neuronal activity, a method for optical control of neuronal proteins using unnatural amino acids (Uaa) in vivo is lacking. Here, we describe the genetic incorporation of a photoreactive Uaa into the pore of an inwardly rectifying potassium channel Kir2.1. The Uaa occluded the pore, rendering the channel nonconducting, and, on brief light illumination, was released to permit outward K(+) current. Expression of this photoinducible inwardly rectifying potassium (PIRK) channel in rat hippocampal neurons created a light-activatable PIRK switch for suppressing neuronal firing. We also expanded the genetic code of mammals to express PIRK channels in embryonic mouse neocortex in vivo and demonstrated a light-activated PIRK current in cortical neurons. These principles could be generally expanded to other proteins expressed in the brain to enable optical regulation.
Collapse
Affiliation(s)
- Ji-Yong Kang
- The Jack H. Skirball Center for Chemical Biology and Proteomics, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Shah NH, Aizenman E. Voltage-gated potassium channels at the crossroads of neuronal function, ischemic tolerance, and neurodegeneration. Transl Stroke Res 2013; 5:38-58. [PMID: 24323720 DOI: 10.1007/s12975-013-0297-7] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 09/14/2013] [Accepted: 10/14/2013] [Indexed: 11/29/2022]
Abstract
Voltage-gated potassium (Kv) channels are widely expressed in the central and peripheral nervous system and are crucial mediators of neuronal excitability. Importantly, these channels also actively participate in cellular and molecular signaling pathways that regulate the life and death of neurons. Injury-mediated increased K(+) efflux through Kv2.1 channels promotes neuronal apoptosis, contributing to widespread neuronal loss in neurodegenerative disorders such as Alzheimer's disease and stroke. In contrast, some forms of neuronal activity can dramatically alter Kv2.1 channel phosphorylation levels and influence their localization. These changes are normally accompanied by modifications in channel voltage dependence, which may be neuroprotective within the context of ischemic injury. Kv1 and Kv7 channel dysfunction leads to neuronal hyperexcitability that critically contributes to the pathophysiology of human clinical disorders such as episodic ataxia and epilepsy. This review summarizes the neurotoxic, neuroprotective, and neuroregulatory roles of Kv channels and highlights the consequences of Kv channel dysfunction on neuronal physiology. The studies described in this review thus underscore the importance of normal Kv channel function in neurons and emphasize the therapeutic potential of targeting Kv channels in the treatment of a wide range of neurological diseases.
Collapse
Affiliation(s)
- Niyathi Hegde Shah
- Department of Neurobiology, University of Pittsburgh School of Medicine, 3500 Terrace Street, E1456 BST, Pittsburgh, PA, 15261, USA,
| | | |
Collapse
|
19
|
Chen M, Sun HY, Hu P, Wang CF, Li BX, Li SJ, Li JJ, Tan HY, Gao TM. Activation of BKCa Channels Mediates Hippocampal Neuronal Death After Reoxygenation and Reperfusion. Mol Neurobiol 2013; 48:794-807. [DOI: 10.1007/s12035-013-8467-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Accepted: 04/26/2013] [Indexed: 11/29/2022]
|
20
|
Canonico B, Luchetti F, Ambrogini P, Arcangeletti M, Betti M, Cesarini E, Lattanzi D, Ciuffoli S, Palma F, Cuppini R, Papa S. Pharmacological doses of melatonin induce alterations in mitochondrial mass and potential, bcl-2 levels and K+currents in UVB-exposed U937 cells. Cell Biol Int 2013; 37:213-26. [DOI: 10.1002/cbin.10030] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 12/06/2012] [Indexed: 12/19/2022]
Affiliation(s)
- Barbara Canonico
- DiSTeVA, Department of Earth, Life and Environmental Sciences; University of Urbino ‘Carlo Bo’; Urbino; Italy
| | - Francesca Luchetti
- DiSTeVA, Department of Earth, Life and Environmental Sciences; University of Urbino ‘Carlo Bo’; Urbino; Italy
| | - Patrizia Ambrogini
- DiSTeVA, Department of Earth, Life and Environmental Sciences; University of Urbino ‘Carlo Bo’; Urbino; Italy
| | - Marcella Arcangeletti
- DiSTeVA, Department of Earth, Life and Environmental Sciences; University of Urbino ‘Carlo Bo’; Urbino; Italy
| | - Michele Betti
- DiSTeVA, Department of Earth, Life and Environmental Sciences; University of Urbino ‘Carlo Bo’; Urbino; Italy
| | - Erica Cesarini
- DiSTeVA, Department of Earth, Life and Environmental Sciences; University of Urbino ‘Carlo Bo’; Urbino; Italy
| | - Davide Lattanzi
- DiSTeVA, Department of Earth, Life and Environmental Sciences; University of Urbino ‘Carlo Bo’; Urbino; Italy
| | - Stefano Ciuffoli
- DiSTeVA, Department of Earth, Life and Environmental Sciences; University of Urbino ‘Carlo Bo’; Urbino; Italy
| | - Fulvio Palma
- DiSTeVA, Department of Earth, Life and Environmental Sciences; University of Urbino ‘Carlo Bo’; Urbino; Italy
| | - Riccardo Cuppini
- DiSTeVA, Department of Earth, Life and Environmental Sciences; University of Urbino ‘Carlo Bo’; Urbino; Italy
| | - Stefano Papa
- DiSTeVA, Department of Earth, Life and Environmental Sciences; University of Urbino ‘Carlo Bo’; Urbino; Italy
| |
Collapse
|
21
|
Zschüntzsch J, Schütze S, Hülsmann S, Dibaj P, Neusch C. Heterologous expression of a glial Kir channel (KCNJ10) in a neuroblastoma spinal cord (NSC-34) cell line. Physiol Res 2012; 62:95-105. [PMID: 23173681 DOI: 10.33549/physiolres.932264] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Heterologous expression of Kir channels offers a tool to modulate excitability of neurons which provide insight into Kir channel functions in general. Inwardly-rectifying K+ channels (Kir channels) are potential candidate proteins to hyperpolarize neuronal cell membranes. However, heterologous expression of inwardly-rectifying K+ channels has previously proven to be difficult. This was mainly due to a high toxicity of the respective Kir channel expression. We investigated the putative role of a predominantly glial-expressed, weakly rectifying Kir channel (Kir4.1 channel subunit; KCNJ10) in modulating electrophysiological properties of a motoneuron-like cell culture (NSC-34). Transfection procedures using an EGFP-tagged Kir4.1 protein in this study proved to have no toxic effects on NSC-34 cells. Using whole cell-voltage clamp, a substantial increase of inward rectifying K+ currents as well as hyperpolarization of the cell membrane was observed in Kir4.1-transfected cells. Na+ inward currents, observed in NSC-34 controls, were absent in Kir4.1/EGFP motoneuronal cells. The Kir4.1-transfection did not influence the NaV1.6 sodium channel expression. This study demonstrates the general feasibility of a heterologous expression of a weakly inward-rectifying K+ channel (Kir4.1 subunit) and shows that in vitro overexpression of Kir4.1 shifts electrophysiological properties of neuronal cells to a more glial-like phenotype and may therefore be a candidate tool to dampen excitability of neurons in experimental paradigms.
Collapse
Affiliation(s)
- J Zschüntzsch
- Department of Neurology, Georg-August-University, Göttingen, Germany
| | | | | | | | | |
Collapse
|
22
|
Ozcan M, Ayar A. Modulation of action potential and calcium signaling by levetiracetam in rat sensory neurons. J Recept Signal Transduct Res 2012; 32:156-62. [DOI: 10.3109/10799893.2012.672993] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
23
|
Depetris-Chauvin A, Berni J, Aranovich EJ, Muraro NI, Beckwith EJ, Ceriani MF. Adult-specific electrical silencing of pacemaker neurons uncouples molecular clock from circadian outputs. Curr Biol 2011; 21:1783-93. [PMID: 22018542 DOI: 10.1016/j.cub.2011.09.027] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 09/09/2011] [Accepted: 09/13/2011] [Indexed: 01/24/2023]
Abstract
BACKGROUND Circadian rhythms regulate physiology and behavior through transcriptional feedback loops of clock genes running within specific pacemaker cells. In Drosophila, molecular oscillations in the small ventral lateral neurons (sLNvs) command rhythmic behavior under free-running conditions releasing the neuropeptide PIGMENT DISPERSING FACTOR (PDF) in a circadian fashion. Electrical activity in the sLNvs is also required for behavioral rhythmicity. Yet, how temporal information is transduced into behavior remains unclear. RESULTS Here we developed a new tool for temporal control of gene expression to obtain adult-restricted electrical silencing of the PDF circuit, which led to reversible behavioral arrhythmicity. Remarkably, PERIOD (PER) oscillations during the silenced phase remained unaltered, indicating that arrhythmicity is a direct consequence of the silenced activity. Accordingly, circadian axonal remodeling and PDF accumulation were severely affected during the silenced phase. CONCLUSIONS Although electrical activity of the sLNvs is not a clock component, it coordinates circuit outputs leading to rhythmic behavior.
Collapse
Affiliation(s)
- Ana Depetris-Chauvin
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir and Instituto de Investigaciones Bioquímicas-Buenos Aires, Av. Patricias Argentinas 435, 1405-BWE Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
24
|
Bhave G, Lonergan D, Chauder BA, Denton JS. Small-molecule modulators of inward rectifier K+ channels: recent advances and future possibilities. Future Med Chem 2010; 2:757-74. [PMID: 20543968 PMCID: PMC2883187 DOI: 10.4155/fmc.10.179] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Inward rectifier potassium (Kir) channels have been postulated as therapeutic targets for several common disorders including hypertension, cardiac arrhythmias and pain. With few exceptions, however, the small-molecule pharmacology of this family is limited to nonselective cardiovascular and neurologic drugs with off-target activity toward inward rectifiers. Consequently, the actual therapeutic potential and 'drugability' of most Kir channels has not yet been determined experimentally. The purpose of this review is to provide a comprehensive summary of publicly disclosed Kir channel small-molecule modulators and highlight recent targeted drug-discovery efforts toward Kir1.1 and Kir2.1. The review concludes with a brief speculation on how the field of Kir channel pharmacology will develop over the coming years and a discussion of the increasingly important role academic laboratories will play in this progress.
Collapse
Affiliation(s)
- Gautam Bhave
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Daniel Lonergan
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Brian A Chauder
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jerod S Denton
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Digestive Disease Research Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
25
|
Hu D, Liu J, Keblesh J, Xiong H. Involvement of the 4-aminopyridine-sensitive transient A-type K+ current in macrophage-induced neuronal injury. Eur J Neurosci 2010; 31:214-22. [PMID: 20074219 DOI: 10.1111/j.1460-9568.2009.07063.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Through their capacity to secrete, upon activation, a variety of bioactive molecules, brain macrophages (and resident microglia) play an important role in brain immune and inflammatory responses. To test our hypothesis that activated macrophages induce neuronal injury by enhancing neuronal outward K(+) current, we studied the effects of lipopolysaccharide (LPS)-stimulated human monocyte-derived macrophage (MDM) on neuronal transient A-type K(+) current (I(A)) and resultant neuronal injury in primary rat hippocampal neuronal cultures. Bath application of LPS-stimulated MDM-conditioned media (MCM+) enhanced neuronal I(A) in a concentration-dependent manner. Non-stimulated MCM (MCM-) failed to alter I(A). The enhancement of neuronal I(A) was recapitulated in neurons co-cultured with macrophages. The link of MCM(+)-induced enhancement of I(A) to MCM(+)-associated neuronal injury, as detected by propidium iodide and 4'',6-diamidino-2-phenylindol staining (DAPI) and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, was demonstrated by experimental results showing that addition of I(A) blocker 4-aminopyridine to the cultures protected hippocampal neurons from MCM(+)-induced neuronal injury. Further investigation revealed that glutamate was involved in MCM(+)-induced enhancement of neuronal I(A). These results suggest that during brain inflammation macrophages (and microglia) might mediate neuronal injury via enhancement of neuronal I(A), and that neuronal K(v) channel might be a potential target for the development of therapeutic strategies for some neurodegenerative disorders by which immune and inflammatory responses are believed to be involved in the pathogenesis.
Collapse
Affiliation(s)
- Dehui Hu
- Center for Neurovirology and Neurodegenerative Disorders, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | | | | | | |
Collapse
|
26
|
Oxidation of potassium channels by ROS: a general mechanism of aging and neurodegeneration? Trends Cell Biol 2010; 20:45-51. [DOI: 10.1016/j.tcb.2009.09.008] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Revised: 09/23/2009] [Accepted: 09/24/2009] [Indexed: 12/13/2022]
|
27
|
Lewis LM, Bhave G, Chauder BA, Banerjee S, Lornsen KA, Redha R, Fallen K, Lindsley CW, Weaver CD, Denton JS. High-throughput screening reveals a small-molecule inhibitor of the renal outer medullary potassium channel and Kir7.1. Mol Pharmacol 2009; 76:1094-103. [PMID: 19706730 PMCID: PMC2774996 DOI: 10.1124/mol.109.059840] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Accepted: 08/25/2009] [Indexed: 11/22/2022] Open
Abstract
The renal outer medullary potassium channel (ROMK) is expressed in the kidney tubule and critically regulates sodium and potassium balance. The physiological functions of other inward rectifying K(+) (Kir) channels expressed in the nephron, such as Kir7.1, are less well understood in part due to the lack of selective pharmacological probes targeting inward rectifiers. In an effort to identify Kir channel probes, we performed a fluorescence-based, high-throughput screen (HTS) of 126,009 small molecules for modulators of ROMK function. Several antagonists were identified in the screen. One compound, termed VU590, inhibits ROMK with submicromolar affinity, but has no effect on Kir2.1 or Kir4.1. Low micromolar concentrations inhibit Kir7.1, making VU590 the first small-molecule inhibitor of Kir7.1. Structure-activity relationships of VU590 were defined using small-scale parallel synthesis. Electrophysiological analysis indicates that VU590 is an intracellular pore blocker. VU590 and other compounds identified by HTS will be instrumental in defining Kir channel structure, physiology, and therapeutic potential.
Collapse
Affiliation(s)
- L Michelle Lewis
- Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Hodge JJL. Ion channels to inactivate neurons in Drosophila. Front Mol Neurosci 2009; 2:13. [PMID: 19750193 PMCID: PMC2741205 DOI: 10.3389/neuro.02.013.2009] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2009] [Accepted: 08/11/2009] [Indexed: 02/05/2023] Open
Abstract
Ion channels are the determinants of excitability; therefore, manipulation of their levels and properties provides an opportunity for the investigator to modulate neuronal and circuit function. There are a number of ways to suppress electrical activity in Drosophila neurons, for instance, over-expression of potassium channels (i.e. Shaker Kv1, Shaw Kv3, Kir2.1 and DORK) that are open at resting membrane potential. This will result in increased potassium efflux and membrane hyperpolarisation setting resting membrane potential below the threshold required to fire action potentials. Alternatively over-expression of other channels, pumps or co-transporters that result in a hyperpolarised membrane potential will also prevent firing. Lastly, neurons can be inactivated by, disrupting or reducing the level of functional voltage-gated sodium (Nav1 paralytic) or calcium (Cav2 cacophony) channels that mediate the depolarisation phase of action potentials. Similarly, strategies involving the opposite channel manipulation should allow net depolarisation and hyperexcitation in a given neuron. These changes in ion channel expression can be brought about by the versatile transgenic (i.e. Gal4/UAS based) systems available in Drosophila allowing fine temporal and spatial control of (channel) transgene expression. These systems are making it possible to electrically inactivate (or hyperexcite) any neuron or neural circuit in the fly brain, and much like an exquisite lesion experiment, potentially elucidate whatever interesting behaviour or phenotype each network mediates. These techniques are now being used in Drosophila to reprogram electrical activity of well-defined circuits and bring about robust and easily quantifiable changes in behaviour, allowing different models and hypotheses to be rapidly tested.
Collapse
Affiliation(s)
- James J L Hodge
- Physiology and Pharmacology Department, University of Bristol Bristol, UK
| |
Collapse
|
29
|
Koeberle PD, Wang Y, Schlichter LC. Kv1.1 and Kv1.3 channels contribute to the degeneration of retinal ganglion cells after optic nerve transection in vivo. Cell Death Differ 2009; 4:337-46. [DOI: 10.1038/cdd.2009.113] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
30
|
Zhang M, Fei XW, He YL, Yang G, Mei YA. Bradykinin inhibits the transient outward K+ current in mouse Schwann cells via the cAMP/PKA pathway. Am J Physiol Cell Physiol 2009; 296:C1364-72. [PMID: 19339513 DOI: 10.1152/ajpcell.00014.2009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Bradykinin (BK) is an endogenous peptide with diverse biological actions and is considered to be an important mediator of the inflammatory response in both the peripheral and the central nervous systems. BK has attracted recent interest as a potential mediator of K(+) conductance, Cl(-) channels, and Ca(2+)-activated K(+) channels. However, few reports have associated BK with the voltage-gated K(+) current. In this study, we demonstrated that BK suppressed the transient outward potassium current (I(A)) in mouse Schwann cells using whole cell recording techniques. At a concentration of 0.1 muM to 5 muM, BK reversibly inhibited I(A) in a dose-dependent manner with the modulation of steady-state activation and inactivation properties. The effect of BK on I(A) current was abolished after preincubation with a B(2) receptor antagonist but could not be eliminated by B(1) receptor antagonist. Intracellular application of GTP-gammaS induced an irreversible decrease in I(A), and the inhibition of G(s) using NF449 provoked a gradual augmentation in I(A) and eliminated the BK-induced effect on I(A,) while the G(i)/(o) antagonist NF023 did not. The application of forskolin or dibutyryl-cAMP mimicked the inhibitory effect of BK on I(A) and abolished the BK-induced effect on I(A). H-89, an inhibitor of PKA, augmented I(A) amplitude and completely eliminated the BK-induced inhibitory effect on I(A). In contrast, activation of PKC by PMA augmented I(A) amplitude. A cAMP assay revealed that BK significantly increased intracellular cAMP level. It is therefore concluded that BK inhibits the I(A) current in Schwann cells by cAMP/PKA-dependent pathways via activation of the B(2) receptor.
Collapse
Affiliation(s)
- Man Zhang
- Institute of Brain Science, School of Life Sciences and State Key Lab of Medical Neurobiology, Fudan University, Shanghai 200433, P.R. China
| | | | | | | | | |
Collapse
|
31
|
Fallen K, Banerjee S, Sheehan J, Addison D, Lewis LM, Meiler J, Denton JS. The Kir channel immunoglobulin domain is essential for Kir1.1 (ROMK) thermodynamic stability, trafficking and gating. Channels (Austin) 2009; 3:57-68. [PMID: 19221509 DOI: 10.4161/chan.3.1.7817] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The renal inward rectifying potassium channel Kir1.1 plays key roles in regulating electrolyte homeostasis and blood pressure. Loss-of-function mutations in the channel cause a life-threatening salt and water balance disorder in infants called antenatal Bartter syndrome (ABS). Of more than 30 ABS mutations identified, approximately half are located in the intracellular domain of the channel. The mechanisms underlying channel dysfunction for most of these mutations are unknown. By mapping intracellular mutations onto an atomic model of Kir1.1, we found that several of these are localized to a phylogenetically ancient immunoglobulin (Ig)-like domain (IgLD) that has not been characterized previously, prompting us to examine this structure in detail. The IgLD is assembled from two beta-pleated sheets packed face-to-face, creating a beta-sheet interface or core, populated by highly conserved side chains. Thermodynamic calculations on computationally mutated channels suggest that IgLD core residues are among the most important residues for determining cytoplasmic domain stability. Consistent with this notion, we show that two ABS mutations (A198T and Y314C) located within the IgLD core impair channel biosynthesis and trafficking in mammalian cells. A fraction of core mutant channels reach the cell surface, but are electrically silent due to closure of the helix-bundle gate. Compensatory mutation-induced rescue of channel function revealed that IgLD core mutants fail to rectify. Our study sheds new light on the pathogenesis of ABS and establishes the IgLD as an essential structure within the Kir channel family.
Collapse
Affiliation(s)
- Katherine Fallen
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Sättler MB, Williams SK, Neusch C, Otto M, Pehlke JR, Bähr M, Diem R. Flupirtine as neuroprotective add-on therapy in autoimmune optic neuritis. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 173:1496-507. [PMID: 18832577 DOI: 10.2353/ajpath.2008.080491] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) is a common inflammatory disease of the central nervous system that results in persistent impairment in young adults. During chronic progressive disease stages, there is a strong correlation between neurodegeneration and disability. Current therapies fail to prevent progression of neurological impairment during these disease stages. Flupirtine, a drug approved for oral use in patients suffering from chronic pain, was used in a rat model of autoimmune optic neuritis and significantly increased the survival of retinal ganglion cells, the neurons that form the axons of the optic nerve. When flupirtine was combined with interferon-beta, an established immunomodulatory therapy for MS, visual functions of the animals were improved during the acute phase of optic neuritis. Furthermore, flupirtine protected retinal ganglion cells from degeneration in a noninflammatory animal model of optic nerve transection. Although flupirtine was shown previously to increase neuronal survival by Bcl-2 up-regulation, this mechanism does not appear to play a role in flupirtine-mediated protection of retinal ganglion cells either in vitro or in vivo. Instead, we showed through patch-clamp investigations that the activation of inwardly rectifying potassium channels is involved in flupirtine-mediated neuroprotection. Considering the few side effects reported in patients who receive long-term flupirtine treatment for chronic pain, our results indicate that this drug is an interesting candidate for further evaluation of its neuroprotective potential in MS.
Collapse
Affiliation(s)
- Muriel B Sättler
- Department of Neurology, University of Göttingen, Göttingen, Germany.
| | | | | | | | | | | | | |
Collapse
|
33
|
Okada M, Matsuda H. Chronic lentiviral expression of inwardly rectifying K+ channels (Kir2.1) reduces neuronal activity and downregulates voltage-gated potassium currents in hippocampus. Neuroscience 2008; 156:289-97. [PMID: 18713648 DOI: 10.1016/j.neuroscience.2008.07.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Revised: 06/19/2008] [Accepted: 07/17/2008] [Indexed: 10/21/2022]
|
34
|
Hu CL, Zeng XM, Zhou MH, Shi YT, Cao H, Mei YA. Kv 1.1 is associated with neuronal apoptosis and modulated by protein kinase C in the rat cerebellar granule cell. J Neurochem 2008; 106:1125-1137. [PMID: 18466331 DOI: 10.1111/j.1471-4159.2008.05449.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2025]
Abstract
Previously, we reported that apoptosis of cerebellar granular neurons induced by low-K+ and serum-free (LK-S) was associated with an increase in the A-type K+ channel current (I(A)), and an elevated expression of main alpha-subunit of the I(A) channel, which is known as Kv4.2 and Kv4.3. Here, we show, as assessed by quantitative RT-PCR and whole-cell recording, that besides Kv4.2 and Kv4.3, Kv1.1 is very important for I(A) channel. The expression of Kv1.1 was elevated in the apoptotic neurons, while silencing Kv1.1 expression by siRNA reduced the I(A) amplitude of the apoptotic neuron, and increased neuron viability. Inhibiting Kv1.1 current by dendrotoxin-K evoked a similar effect of reduction of I(A) amplitude and protection of neurons. Applying a protein kinase C (PKC) activator, phorbol ester acetate A (PMA) mimicked the LK-S-induced neuronal apoptotic effect, enhanced the I(A) amplitude and reduced the granule cell viability. The PKC inhibitor, bisindolylmaleimide I and Gö6976 protected the cell against apoptosis induced by LK-S. After silencing the Kv1.1 gene, the effect of PMA on the residual K+ current was reduced significantly. Quantitative RT-PCR and Western immunoblot techniques revealed that LK-S treatment and PMA increased the level of the expression of Kv1.1, in contrast, bisindolylmaleimide I inhibited Kv1.1 expression. In addition, the activation of the PKC isoform was identified in apoptotic neurons. We thus conclude that in the rat cerebellar granule cell, the I(A) channel associated with apoptotic neurons is encoded mainly by the Kv1.1 gene, and that the PKC pathway promotes neuronal apoptosis by a brief modulation of the I(A) amplitude and a permanent increase in the levels of expression of the Kv1.1 alpha-subunit.
Collapse
Affiliation(s)
- Chang-Long Hu
- The Institute of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
35
|
PKA-mediated phosphorylation is a novel mechanism for levetiracetam, an antiepileptic drug, activating ROMK1 channels. Biochem Pharmacol 2008; 76:225-35. [DOI: 10.1016/j.bcp.2008.04.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2008] [Revised: 04/22/2008] [Accepted: 04/22/2008] [Indexed: 11/22/2022]
|
36
|
d-Amphetamine inhibits inwardly rectifying potassium channels in Xenopus oocytes expression system. Neurotoxicology 2008; 29:638-46. [DOI: 10.1016/j.neuro.2008.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2007] [Revised: 05/01/2008] [Accepted: 05/05/2008] [Indexed: 12/13/2022]
|
37
|
Jessica Chen M, Sepramaniam S, Armugam A, Shyan Choy M, Manikandan J, Melendez AJ, Jeyaseelan K, Sang Cheung N. Water and ion channels: crucial in the initiation and progression of apoptosis in central nervous system? Curr Neuropharmacol 2008; 6:102-16. [PMID: 19305791 PMCID: PMC2647147 DOI: 10.2174/157015908784533879] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2007] [Revised: 09/09/2007] [Accepted: 10/01/2007] [Indexed: 12/14/2022] Open
Abstract
Programmed cell death (PCD), is a highly regulated and sophisticated cellular mechanism that commits cell to isolated death fate. PCD has been implicated in the pathogenesis of numerous neurodegenerative disorders. Countless molecular events underlie this phenomenon, with each playing a crucial role in death commitment. A precedent event, apoptotic volume decrease (AVD), is ubiquitously observed in various forms of PCD induced by different cellular insults. Under physiological conditions, cells when subjected to osmotic fluctuations will undergo regulatory volume increase/decrease (RVI/RVD) to achieve homeostatic balance with neurons in the brain being additionally protected by the blood-brain-barrier. However, during AVD following apoptotic trigger, cell undergoes anistonic shrinkage that involves the loss of water and ions, particularly monovalent ions e.g. K(+), Na(+) and Cl(-). It is worthwhile to concentrate on the molecular implications underlying the loss of these cellular components which posed to be significant and crucial in the successful propagation of the apoptotic signals. Microarray and real-time PCR analyses demonstrated several ion and water channel genes are regulated upon the onset of lactacystin (a proteosomal inhibitor)-mediated apoptosis. A time course study revealed that gene expressions of water and ion channels are being modulated just prior to apoptosis, some of which are aquaporin 4 and 9, potassium channels and chloride channels. In this review, we shall looked into the molecular protein machineries involved in the execution of AVD in the central nervous system (CNS), and focus on the significance of movements of each cellular component in affecting PCD commitment, thus provide some pharmacological advantages in the global apoptotic cell death.
Collapse
Affiliation(s)
- Minghui Jessica Chen
- Departments of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Sugunavathi Sepramaniam
- Departments of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Arunmozhiarasi Armugam
- Departments of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Meng Shyan Choy
- Departments of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Jayapal Manikandan
- Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Alirio J Melendez
- Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Kandiah Jeyaseelan
- Departments of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Nam Sang Cheung
- Departments of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
38
|
Sjulson L, Miesenböck G. Rational optimization and imaging in vivo of a genetically encoded optical voltage reporter. J Neurosci 2008; 28:5582-93. [PMID: 18495892 PMCID: PMC2714581 DOI: 10.1523/jneurosci.0055-08.2008] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Revised: 04/10/2008] [Accepted: 04/14/2008] [Indexed: 11/21/2022] Open
Abstract
The hybrid voltage sensor (hVOS) combines membrane-targeted green fluorescent protein and the hydrophobic anion dipicrylamine (DPA) to provide a promising tool for optical recording of electrical activity from genetically defined populations of neurons. However, large fluorescence signals are obtained only at high DPA concentrations (>3 mum) that increase membrane capacitance to a level that suppresses neural activity. Here, we develop a quantitative model of the sensor to guide its optimization and achieved an approximate threefold increase in fractional fluorescence change at a lower DPA concentration of 2 mum. Using this optimized voltage reporter, we perform optical recordings of evoked activity in the Drosophila antennal lobe with millisecond temporal resolution but fail to detect action potentials, presumably because spike initiation and/or propagation are inhibited by the capacitive load added even at reduced DPA membrane densities. We evaluate strategies for potential further improvement of hVOS quantitatively and derive theoretical performance limits for optical voltage reporters in general.
Collapse
Affiliation(s)
- Lucas Sjulson
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06520, and
| | - Gero Miesenböck
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06520, and
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom
| |
Collapse
|
39
|
Over-expression of the potassium channel Kir2.3 using the dopamine-1 receptor promoter selectively inhibits striatal neurons. Neuroscience 2008; 155:114-27. [PMID: 18571331 DOI: 10.1016/j.neuroscience.2008.04.075] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2007] [Revised: 04/03/2008] [Accepted: 04/16/2008] [Indexed: 10/22/2022]
Abstract
Dysfunction of basal ganglia circuits underlies a variety of movement disorders and neuropsychiatric conditions. Selective control of the electrical activity of striatal outflow pathways by manipulation of ion channel function presents a novel therapeutic approach. Toward this end, we have constructed and studied in vitro an adenoviral gene transfer vector that employs the promoter region of the dopamine-1 receptor to drive expression of the inward rectifier K(+) channel Kir2.3. The use of this neuronal promoter confers cell-type specificity and a physiological level of trans-gene expression in rat primary striatal cultures. The electrophysiological properties were confirmed in transfected human embryonic kidney cells, in which an inwardly-rectifying, Cs(+)-sensitive current was measured by voltage clamp. Current clamp studies of transduced striatal neurons demonstrated an increase in the firing threshold, latency to first action potential and decrease in neuronal excitability. Neurotoxin-induced activation of c-Fos, a marker of neuronal activity, was blocked in transduced neurons indicating that the decrease in electrical excitability was physiologically significant. When used in vivo, this strategy may have the potential to positively impact movement disorders by selectively changing activity of neurons belonging to the direct striatal pathway, characterized by the expression of dopamine-1 receptors.
Collapse
|
40
|
Sjulson L, Miesenböck G. Photocontrol of neural activity: biophysical mechanisms and performance in vivo. Chem Rev 2008; 108:1588-602. [PMID: 18447399 DOI: 10.1021/cr078221b] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Lucas Sjulson
- Department of Cell Biology, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520, USA
| | | |
Collapse
|
41
|
Lee CH, Tsai TS, Liou HH. Gabapentin activates ROMK1 channels by a protein kinase A (PKA)-dependent mechanism. Br J Pharmacol 2008; 154:216-25. [PMID: 18311184 DOI: 10.1038/bjp.2008.73] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Gabapentin is an effective anticonvulsant. The major physiological function of renal outer medullary potassium (ROMK1) channels is to maintain the resting membrane potential (RMP). We investigated the effect of gabapentin on ROMK1 channels and the mechanism involved. EXPERIMENTAL APPROACH Xenopus oocytes were injected with mRNA coding for wild-type or mutant ROMK1 channels and giant inside-out patch-clamp recordings were performed. KEY RESULTS Gabapentin increased the activity of ROMK1 channels, concentration-dependently and enhanced the activity of wild-type and an intracellular pH (pH(i))-gating residue mutant (K80M) channels over a range of pH(i). Gabapentin also increased activity of channels mutated at phosphatidylinositol 4,5-bisphosphate (PIP(2))-binding sites (R188Q, R217A and K218A). However, gabapentin failed to enhance channel activity in the presence of protein kinase A (PKA) inhibitors and did not activate phosphorylation site mutants (S44A, S219A or S313A), mutants that mimicked the negative charge carried by a phosphate group bound to a serine (S44D, S219D or S313D), or a mutated channel with a positive charge (S219R). These findings show that gabapentin activates ROMK1 channels independently of the pH(i) and not via a PIP(2)-dependent pathway. The effects of gabapentin on ROMK1 channels may be due to a PKA-mediated phosphorylation-induced conformational change, but not to charge-charge interactions. CONCLUSIONS AND IMPLICATIONS ROMK1 channels are the main channels responsible for maintaining the RMP during cellular excitation. Gabapentin increased the activity of ROMK1 channels by a PKA-dependent mechanism, reducing neuronal excitability, and this may play an important role in its antiepileptic effect.
Collapse
Affiliation(s)
- C-H Lee
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | |
Collapse
|
42
|
Lerchner W, Xiao C, Nashmi R, Slimko EM, van Trigt L, Lester HA, Anderson DJ. Reversible silencing of neuronal excitability in behaving mice by a genetically targeted, ivermectin-gated Cl- channel. Neuron 2007; 54:35-49. [PMID: 17408576 DOI: 10.1016/j.neuron.2007.02.030] [Citation(s) in RCA: 138] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2006] [Revised: 02/09/2007] [Accepted: 02/22/2007] [Indexed: 02/03/2023]
Abstract
Several genetic strategies for inhibiting neuronal function in mice have been described, but no system that directly suppresses membrane excitability and is triggered by a systemically administered drug, has been validated in awake behaving animals. We expressed unilaterally in mouse striatum a modified heteromeric ivermectin (IVM)-gated chloride channel from C. elegans (GluClalphabeta), systemically administered IVM, and then assessed amphetamine-induced rotational behavior. Rotation was observed as early as 4 hr after a single intraperitoneal IVM injection (10 mg/kg), reached maximal levels by 12 hr, and was almost fully reversed by 4 days. Multiple cycles of silencing and recovery could be performed in a single animal. In striatal slice preparations from GluClalphabeta-expressing animals, IVM rapidly suppressed spiking. The two-subunit GluCl/IVM system permits "intersectional" strategies designed to increase the cellular specificity of silencing in transgenic animals.
Collapse
Affiliation(s)
- Walter Lerchner
- Division of Biology 216-76, California Institute of Technology, Pasadena, CA 91125, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Luca T, Givogri MI, Perani L, Galbiati F, Follenzi A, Naldini L, Bongarzone ER. Axons mediate the distribution of arylsulfatase A within the mouse hippocampus upon gene delivery. Mol Ther 2006; 12:669-79. [PMID: 16087406 DOI: 10.1016/j.ymthe.2005.06.438] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2005] [Revised: 06/21/2005] [Accepted: 06/23/2005] [Indexed: 11/22/2022] Open
Abstract
Axonal transport of the lysosomal enzyme arylsulfatase A (ARSA) may be an additional mechanism of enzyme distribution after in vivo brain gene transfer in an animal model of metachromatic leukodystrophy (MLD). Direct molecular demonstration of the movement of this lysosomal enzyme within axonal networks was missing. We generated lentiviral vectors carrying the ARSA cDNA tagged with hemagglutinin or the green fluorescent protein and examined the subcellular localization and anatomical distribution of the tagged enzymes within the MLD hippocampus after in vivo lentiviral gene transfer. The use of tagged ARSA allowed direct real-time observation and tracking of axon-dendritic transport of the enzyme after lentiviral gene therapy. Tagged ARSA was expressed in transduced pyramidal, granule, and hilar neurons within the lentiviral-injected side and was robustly contained in vesicles within ipsilateral axon-dendritic processes as well as in vesicles associated with contralateral axons and commissural axons of the ventral hippocampal commissure. Axonal transport of tagged ARSA led to the correction of hippocampal defects in long-term treated MLD mice, which was accompanied by enzyme uptake in nontransduced contralateral neurons, enzyme accumulation within the lysosomal compartment, and clearance of sulfatide storage deposits in this region of the MLD brain. These results contribute to the understanding of the mechanisms of distribution of lysosomal enzymes within the mammalian brain after direct gene therapy, demonstrating the use of neural processes for enzyme transport.
Collapse
Affiliation(s)
- Tonia Luca
- Telethon Institute for Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
44
|
Tan EM, Yamaguchi Y, Horwitz GD, Gosgnach S, Lein ES, Goulding M, Albright TD, Callaway EM. Selective and quickly reversible inactivation of mammalian neurons in vivo using the Drosophila allatostatin receptor. Neuron 2006; 51:157-70. [PMID: 16846851 DOI: 10.1016/j.neuron.2006.06.018] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2006] [Revised: 06/05/2006] [Accepted: 06/19/2006] [Indexed: 01/24/2023]
Abstract
Genetic strategies for perturbing activity of selected neurons hold great promise for understanding circuitry and behavior. Several such strategies exist, but there has been no direct demonstration of reversible inactivation of mammalian neurons in vivo. We previously reported quickly reversible inactivation of neurons in vitro using expression of the Drosophila allatostatin receptor (AlstR). Here, adeno-associated viral vectors are used to express AlstR in vivo in cortical and thalamic neurons of rats, ferrets, and monkeys. Application of the receptor's ligand, allatostatin (AL), leads to a dramatic reduction in neural activity, including responses of visual neurons to optimized visual stimuli. Additionally, AL eliminates activity in spinal cords of transgenic mice conditionally expressing AlstR. This reduction occurs selectively in AlstR-expressing neurons. Inactivation can be reversed within minutes upon washout of the ligand and is repeatable, demonstrating that the AlstR/AL system is effective for selective, quick, and reversible silencing of mammalian neurons in vivo.
Collapse
Affiliation(s)
- Elaine M Tan
- Systems Neurobiology, The Salk Institute for Biological Studies, and Graduate Program in Neurosciences, University of California, San Diego, La Jolla, California 92093, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Nothwang HG, Koehl A, Friauf E. Comparative gene expression analysis reveals a characteristic molecular profile of the superior olivary complex. ACTA ACUST UNITED AC 2006; 288:409-23. [PMID: 16550588 DOI: 10.1002/ar.a.20301] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The superior olivary complex (SOC) is a very conspicuous structure in the mammalian auditory brainstem. It represents the first binaural processing center and is important for sound localization in the azimuth and in feedback regulation of cochlear function. In order to define molecular determinants of the SOC, which are of potential functional relevance, we have performed a comprehensive analysis of its transcriptome by serial analysis of gene expression in adult rats. Here, we performed a detailed analysis of the SOC's gene expression profile compared to that of two other neural tissues, the striatum and the hippocampus, and with extraocular muscle tissue. This tested the hypothesis that SOC-specific or significantly upregulated transcripts provide candidates for the specific function of auditory neurons. Thirty-three genes were significantly upregulated in the SOC when compared to the two other neural tissues. Thirteen encoded proteins involved in neurotransmission, including action potential propagation, exocytosis, and myelination; five genes are important for the energy metabolism, and five transcripts are unknown or poorly characterized and have yet to be described in the nervous system. The comparison of functional gene classes indicates that the SOC has the highest energy demand of the three neural tissues, yet protein turnover is apparently not increased. This suggests a high energy demand for fueling auditory neurotransmission. Such a demand may have implications on auditory-specific tasks and relate to central auditory processing disorders. Ultimately, these data provide new avenues to foster investigations of auditory function and to advance molecular physiology in the central auditory system.
Collapse
Affiliation(s)
- Hans Gerd Nothwang
- Abteilung Tierphysiologie, Technische Universität Kaiserslautern, Kaiserslautern, Germany.
| | | | | |
Collapse
|
46
|
Karpova AY, Tervo DGR, Gray NW, Svoboda K. Rapid and reversible chemical inactivation of synaptic transmission in genetically targeted neurons. Neuron 2006; 48:727-35. [PMID: 16337911 DOI: 10.1016/j.neuron.2005.11.015] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2004] [Revised: 05/05/2005] [Accepted: 11/08/2005] [Indexed: 10/25/2022]
Abstract
Inducible and reversible silencing of selected neurons in vivo is critical to understanding the structure and dynamics of brain circuits. We have developed Molecules for Inactivation of Synaptic Transmission (MISTs) that can be genetically targeted to allow the reversible inactivation of neurotransmitter release. MISTs consist of modified presynaptic proteins that interfere with the synaptic vesicle cycle when crosslinked by small molecule "dimerizers." MISTs based on the vesicle proteins VAMP2/Synaptobrevin and Synaptophysin induced rapid ( approximately 10 min) and reversible block of synaptic transmission in cultured neurons and brain slices. In transgenic mice expressing MISTs selectively in Purkinje neurons, administration of dimerizer reduced learning and performance of the rotarod behavior. MISTs allow for specific, inducible, and reversible lesions in neuronal circuits and may provide treatment of disorders associated with neuronal hyperactivity.
Collapse
|
47
|
Falk T, Xiang S, Erbe EL, Sherman SJ. Neurochemical and electrophysiological characteristics of rat striatal neurons in primary culture. J Comp Neurol 2006; 494:275-89. [PMID: 16320238 PMCID: PMC2923039 DOI: 10.1002/cne.20819] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Neurons maintained in dispersed primary culture offer a number of advantages as a model system and are particularly well-suited for studies of the intrinsic electrical properties of neurons by patch clamp. We have characterized the immunocytochemical and electrophysiological properties of cultured rat striatal neurons as they develop in vitro in order to compare this model system with the known properties found in vivo. We found a high abundance of cells in vitro corresponding to the principal striatal output neuron, the medium spiny neuron. Immunocytochemical studies indicate that these cells have both dopamine-1 and dopamine-2 receptors and that there is overlap in their expression within the population of neurons. Semiquantitative analysis revealed bimodal distributions of dopamine receptor expression among the population of neurons. The principal peptide neurotransmitters substance P and enkephalin were present but at reduced levels compared with adult preparations. Other striatal markers such as calbindin, calretinin, and the cannabinoid-1 receptor were abundant. An immunocytochemical survey of voltage-gated K(+) channel subunits characteristic of adult tissue demonstrated the presence in vitro of Kv1.1, Kv1.4, Kv4.2, Kv4.3, and Kvbeta1.1, which have been associated with the rapidly inactivating currents. Electrophysiological studies employing voltage clamp revealed that outward currents had a large inactivating (A-type) component characteristic of mature basal ganglia. Current clamp studies reveal complex spontaneous firing patterns in a subset of neurons, including bursting behaviors superimposed on a slow depolarization. The inward rectifying channels Kir2.1 and Kir2.3, which are specific to particular compartments in adult striatum, were present in culture.
Collapse
Affiliation(s)
- Torsten Falk
- The University of Arizona, Depts. of Neurology and Physiology
| | - ShiLing Xiang
- The University of Arizona, Depts. of Neurology and Physiology
| | - Emilie L. Erbe
- The University of Arizona, Depts. of Neurology and Physiology
| | | |
Collapse
|
48
|
Grishin A, Ford H, Wang J, Li H, Salvador-Recatala V, Levitan ES, Zaks-Makhina E. Attenuation of apoptosis in enterocytes by blockade of potassium channels. Am J Physiol Gastrointest Liver Physiol 2005; 289:G815-21. [PMID: 16020659 DOI: 10.1152/ajpgi.00001.2005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Apoptosis plays an important role in maintaining the balance between proliferation and cell loss in the intestinal epithelium. Apoptosis rates may increase in intestinal pathologies such as inflammatory bowel disease and necrotizing enterocolitis, suggesting pharmacological prevention of apoptosis as a therapy for these conditions. Here, we explore the feasibility of this approach using the rat epithelial cell line IEC-6 as a model. On the basis of the known role of K+ efflux in apoptosis in various cell types, we hypothesized that K+ efflux is essential for apoptosis in enterocytes and that pharmacological blockade of this efflux would inhibit apoptosis. By probing intracellular [K+] with the K+-sensitive fluorescent dye and measuring the efflux of 86Rb+, we found that apoptosis-inducing treatment with the proteasome inhibitor MG-132 leads to a twofold increase in K+ efflux from IEC-6 cells. Blockade of K+ efflux with tetraethylammonium, 4-aminopyridine, stromatoxin, chromanol 293B, and the recently described K+ channel inhibitor 48F10 prevents DNA fragmentation, caspase activation, release of cytochrome c from mitochondria, and loss of mitochondrial membrane potential. Thus K+ efflux occurs early in the apoptotic program and is required for the execution of later events. Apoptotic K+ efflux critically depends on activation of p38 MAPK. These results demonstrate for the first time the requirement of K+ channel-mediated K+ efflux for progression of apoptosis in enterocytes and suggest the use of K+ channel blockers to prevent apoptotic cell loss occurring in intestinal pathologies.
Collapse
Affiliation(s)
- Anatoly Grishin
- Division of Pediatric Surgery, Children's Hospital of Los Angeles, Los Angeles, California, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Chen X, Chi S, Liu M, Yang W, Wei T, Qi Z, Yang F. Inhibitory effect of ganglioside GD1b on K+ current in hippocampal neurons and its involvement in apoptosis suppression. J Lipid Res 2005; 46:2580-5. [PMID: 16199892 DOI: 10.1194/jlr.m500252-jlr200] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Gangliosides are endogenous membrane components enriched in neuronal cells. They have been shown to play regulatory roles in many cellular processes. Here, we show for the first time that ganglioside GD1b plays an antiapoptotic role in cultured hippocampal neurons. GD1b inhibited the voltage-dependent outward delayed rectifier current (I(K)) but not the transient outward A-type current in a dose-dependent manner, with an IC50 value of 15.2 microM. This effect appears to be somehow specific, because GD1b, but not GM1, GM2, GM3, GD1a, GD3, or GT1b, was effective in inhibiting I(K). Intracellular application of staurosporine (STS; 0.1 microM) resulted in rapid activation of I(K), which was partially reversed upon addition of the K+ channel blocker tetraethylammonium (TEA; 5 mM) and GD1b (10 microM). Furthermore, GD1b (10 microM) attenuated STS-induced neuronal apoptosis by nearly the same amount as 5 mM TEA. In addition, GD1b suppressed the apoptosis-associated caspase 3 activation that was activated by STS. Collectively, these findings suggest that GD1b plays an antiapoptotic role in cultured hippocampal neurons through its inhibitory effect on the I(K) and caspase activity.
Collapse
Affiliation(s)
- Xuesong Chen
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
50
|
Pannaccione A, Secondo A, Scorziello A, Calì G, Taglialatela M, Annunziato L. Nuclear factor-κB activation by reactive oxygen species mediates voltage-gated K+ current enhancement by neurotoxic β-amyloid peptides in nerve growth factor-differentiated PC-12 cells and hippocampal neurones. J Neurochem 2005; 94:572-86. [PMID: 15969743 DOI: 10.1111/j.1471-4159.2005.03075.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Increased activity of plasma membrane K+ channels, leading to decreased cytoplasmic K+ concentrations, occurs during neuronal cell death. In the present study, we showed that the neurotoxic beta-amyloid peptide Abeta(25-35) caused a dose-dependent (0.1-10 microm) and time-dependent (> 12 h) enhancement of both inactivating and non-inactivating components of voltage-dependent K+ (VGK) currents in nerve growth factor-differentiated rat phaeochromocytoma (PC-12) cells and primary rat hippocampal neurones. Similar effects were exerted by Abeta(1-42), but not by the non-neurotoxic Abeta(35-25) peptide. Abeta(25-35) and Abeta(1-42) caused an early (15-20 min) increase in intracellular Ca(2+) concentration. This led to an increased production of reactive oxygen species (ROS), which peaked at 3 h and lasted for 24 h; ROS production seemed to trigger the VGK current increase as vitamin E (50 microm) blocked both the Abeta(25-35)- and Abeta(1-42)-induced ROS increase and VGK current enhancement. Inhibition of protein synthesis (cycloheximide, 1 microg/mL) and transcription (actinomycin D, 50 ng/mL) blocked Abeta(25-35)-induced VGK current enhancement, suggesting that this potentiation is mediated by transcriptional activation induced by ROS. Interestingly, the specific nuclear factor-kappaB inhibitor SN-50 (5 microm), but not its inactive analogue SN-50M (5 microm), fully counteracted Abeta(1-42)- or Abeta(25-35)-induced enhancement of VGK currents, providing evidence for a role of this family of transcription factors in regulating neuronal K+ channel function during exposure to Abeta.
Collapse
Affiliation(s)
- Anna Pannaccione
- Division of Pharmacology, Department of Neuroscience, School of Medicine, Federico II University of Naples, Naples, Italy
| | | | | | | | | | | |
Collapse
|