1
|
MacDonald E, Johannes L, Wunder C. Acidification on the plasma membrane. Curr Opin Cell Biol 2025; 95:102531. [PMID: 40378645 DOI: 10.1016/j.ceb.2025.102531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 05/19/2025]
Abstract
The pH balance between extracellular and intracellular space is crucial for a multitude of cellular processes. Real-time observation of pH fluctuations in the range 4-9 in live cells and tissues in a sensitive, non-invasive manner has become feasible with advances in pH quantification by organic dyes, genetically encoded fluorescent proteins, and DNA-based probes. We discuss mechanisms through which pH affects cell cycle, transcription, senescence, neurotransmission, glycolipid-lectin driven endocytosis, tissue remodelling, immune responses, and GPCR signalling. Growth factor-stimulated acidification of the extracellular space notably triggers enzymatic reactions like desialylation at the plasma membrane that control processes involving cell migration and bone resorption. Research into the role of pH in cellular physiology continues to be a fertile ground for discovery that underscores its fundamental importance.
Collapse
Affiliation(s)
- Ewan MacDonald
- Montpellier Cell Biology Research Center, CRBM, Université 40 Montpellier, CNRS, Montpellier, France.
| | - Ludger Johannes
- Institut Curie, PSL Research University, U1339 INSERM, UMR3666 CNRS, Chemical Biology of Cancer Unit, Paris, France; Inria Center at University of Rennes, SAIRPICO Team, U1339 INSERM, Institut Curie, Chemical Biology of Cancer Unit, Paris, France.
| | - Christian Wunder
- Institut Curie, PSL Research University, U1339 INSERM, UMR3666 CNRS, Chemical Biology of Cancer Unit, Paris, France; Inria Center at University of Rennes, SAIRPICO Team, U1339 INSERM, Institut Curie, Chemical Biology of Cancer Unit, Paris, France.
| |
Collapse
|
2
|
Mohan M, Mannan A, Singh S, Singh TG. Unlocking the cellular mystery: how proton pump inhibitors may alter the dementia landscape. Brain Res 2025; 1861:149702. [PMID: 40368227 DOI: 10.1016/j.brainres.2025.149702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 04/30/2025] [Accepted: 05/10/2025] [Indexed: 05/16/2025]
Abstract
Proton pump inhibitors (PPIs) have become virtually the sole class of histamine-2 receptor antagonists due to their greater effectiveness and general availability. However, concern has been increasing about long-term use and some possible neurological adverse effects, including a link with dementia. Several studies indicate that long-term use of PPIs can raise the risk for both Alzheimer's disease (AD) and non-Alzheimer's dementia, though there is opposing evidence. Neurological side effects of PPIs are cognitive impairment, neuropathies, depression, anxiety, and hallucinations. The mechanisms are unknown but could be due to PPIs crossing the BBB and interfering with neuronal function or causing systemic deficiencies, e.g., vitamin B12 deficiency. Vitamin B12 is essential for cognitive function, and its deficiency has been linked to dementia. PPIs also cause B12 deficiency by inhibiting gastric acid secretion, which is required for B12 absorption. B12 deficiency causes hyperhomocysteinemia, which facilitates tau hyperphosphorylation and amyloid-β (Aβ) deposition, major pathological hallmarks of AD. PPIs have also been found to disrupt amyloid precursor protein processing, mitochondrial function, and neuroinflammation, further enhancing neurodegenerative processes. Experimental evidence indicates that PPIs affect brain homeostasis through inhibition of vacuolar ATPases, modulation of microglial Aβ phagocytosis, and induction of synaptic dysfunction. While the specific molecular mechanisms are unknown, findings suggest that long-term PPI exposure could contribute to neurodegeneration, especially in elderly patients. With increasing dementia prevalence, additional clinical research is needed to ascertain whether PPIs are a causative agent or a contributing factor to cognitive impairment.
Collapse
Affiliation(s)
- Maneesh Mohan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
3
|
Vaglio-Garro A, Kozlov AV, Smirnova YD, Weidinger A. Pathological Interplay between Inflammation and Mitochondria Aggravates Glutamate Toxicity. Int J Mol Sci 2024; 25:2276. [PMID: 38396952 PMCID: PMC10889519 DOI: 10.3390/ijms25042276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Mitochondrial dysfunction and glutamate toxicity are associated with neural disorders, including brain trauma. A review of the literature suggests that toxic and transmission actions of neuronal glutamate are spatially and functionally separated. The transmission pathway utilizes synaptic GluN2A receptors, rapidly released pool of glutamate, evoked release of glutamate mediated by Synaptotagmin 1 and the amount of extracellular glutamate regulated by astrocytes. The toxic pathway utilizes extrasynaptic GluN2B receptors and a cytoplasmic pool of glutamate, which results from the spontaneous release of glutamate mediated by Synaptotagmin 7 and the neuronal 2-oxoglutarate dehydrogenase complex (OGDHC), a tricarboxylic acid (TCA) cycle enzyme. Additionally, the inhibition of OGDHC observed upon neuro-inflammation is due to an excessive release of reactive oxygen/nitrogen species by immune cells. The loss of OGDHC inhibits uptake of glutamate by mitochondria, thus facilitating its extracellular accumulation and stimulating toxic glutamate pathway without affecting transmission. High levels of extracellular glutamate lead to dysregulation of intracellular redox homeostasis and cause ferroptosis, excitotoxicity, and mitochondrial dysfunction. The latter affects the transmission pathway demanding high-energy supply and leading to cell death. Mitochondria aggravate glutamate toxicity due to impairments in the TCA cycle and become a victim of glutamate toxicity, which disrupts oxidative phosphorylation. Thus, therapies targeting the TCA cycle in neurological disorders may be more efficient than attempting to preserve mitochondrial oxidative phosphorylation.
Collapse
Affiliation(s)
- Annette Vaglio-Garro
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; (A.V.-G.); (Y.D.S.); (A.W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Andrey V. Kozlov
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; (A.V.-G.); (Y.D.S.); (A.W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Yuliya D. Smirnova
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; (A.V.-G.); (Y.D.S.); (A.W.)
- Laboratory of Metagenomics and Food Biotechnology, Voronezh State University of Engineering Technologies, 394036 Voronezh, Russia
| | - Adelheid Weidinger
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria; (A.V.-G.); (Y.D.S.); (A.W.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
4
|
Durbin R, Renden R. Basal lamina: A novel pH regulator at the neuromuscular junction. Sci Prog 2024; 107:368504231225066. [PMID: 38196184 PMCID: PMC10777786 DOI: 10.1177/00368504231225066] [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: 01/11/2024]
Abstract
Proton concentration can change within the cleft during synaptic activity due to vesicular release and Ca2+ extrusion from cellular compartments. These changes within the synaptic cleft can impact neural activity by proton-dependent modulation of ion channel function. The pH transient differs in magnitude and direction between synapses, requiring different synapse types to be measured to generate a complete understanding of this mechanism and its impacts on physiology. With a focus on the mouse neuromuscular junction (NMJ), the recently published "Postsynaptic Calcium Extrusion at the Mouse Neuromuscular Junction Alkalinizes the Synaptic Cleft" measured synaptic cleft pH at a cholinergic synapse and found a biphasic pH transient. The study demonstrated that the changes in proton concentration found were due to postsynaptic signaling when measuring pH at the muscle membrane, despite the expectation of a presynaptic contribution. This result suggests a diffusional barrier within the NMJ isolates pH transients to presynaptic versus postsynaptic compartments. Generating a Donnan equilibrium that impacts protons, evidence suggests the basal lamina may be a key regulator of pH at the NMJ. Exploring synaptic pH, proton regulating factors, and downstream pH transient effects at presynaptic versus postsynaptic membranes may lead to new insight for a variety of diseases.
Collapse
Affiliation(s)
- Ryan Durbin
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, USA
| | - Robert Renden
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, USA
| |
Collapse
|
5
|
Andreasen M, Nedergaard S. Effects of carbonic anhydrase activity on the excitability of hippocampal axons during high-frequency firing. Brain Res 2023; 1821:148604. [PMID: 37748571 DOI: 10.1016/j.brainres.2023.148604] [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: 07/12/2023] [Revised: 08/24/2023] [Accepted: 09/22/2023] [Indexed: 09/27/2023]
Abstract
Epileptic activity is known to cause a lowering of intraneuronal pH, which has been suggested to serve as a feedback signal to terminate seizures. The mechanism of such signaling is unclear, but likely involves an altered function of several types of ligand- and voltage-gated channels in postsynaptic membranes caused by increasing cytosolic and extracellular [H+]. In addition, axonal conduction properties may be altered by endogenous pH signals, but this has not been investigated. In the present study, we have recorded the axonal compound action potential (fiber volley) in hippocampal slices in the presence of glutamatergic and GABAergic antagonists. During high-frequency stimulation (HFS) of the Schaffer collaterals, the fiber volley was depressed and its latency from stimulus to peak increased. In the CA1 stratum radiatum these changes were enhanced when the carbonic anhydrase inhibitor acetazolamide (1 mM) was co-perfused. The enhancing effect of acetazolamide was absent after lowering of [Ca2+] in the perfusion medium. Acetazolamide had no detectable effect on HFS-evoked fiber volleys recorded from a more proximal site along the Schaffer collaterals (at the CA2-CA3 border) or from axons in the alveus of CA1. Intracellular acidification imposed by washout of NH4Cl (5 mM) had qualitatively similar effects on fiber volleys evoked at low frequency as those observed with acetazolamide during HFS in CA1 stratum radiatum. The results suggest that carbonic anhydrase-dependent pH regulation counteracts activity-induced reduction of the excitability of Schaffer collateral axons in CA1. A possible influence from local synaptic terminals on this effect is discussed.
Collapse
Affiliation(s)
- Mogens Andreasen
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Steen Nedergaard
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark.
| |
Collapse
|
6
|
Arkhipov AY, Fedorov NS, Nurullin LF, Khabibrakhmanov AN, Mukhamedyarov MA, Samigullin DV, Malomouzh AI. Activation of TRPV1 Channels Inhibits the Release of Acetylcholine and Improves Muscle Contractility in Mice. Cell Mol Neurobiol 2023; 43:4157-4172. [PMID: 37689594 PMCID: PMC11407716 DOI: 10.1007/s10571-023-01403-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/22/2023] [Indexed: 09/11/2023]
Abstract
TRPV1 represents a non-selective transient receptor potential cation channel found not only in sensory neurons, but also in motor nerve endings and in skeletal muscle fibers. However, the role of TRPV1 in the functioning of the neuromuscular junction has not yet been fully established. In this study, the Levator Auris Longus muscle preparations were used to assess the effect of pharmacological activation of TRPV1 channels on neuromuscular transmission. The presence of TRPV1 channels in the nerve terminal and in the muscle fiber was confirmed by immunohistochemistry. It was verified by electrophysiology that the TRPV1 channel agonist capsaicin inhibits the acetylcholine release, and this effect was completely absent after preliminary application of the TRPV1 channel blocker SB 366791. Nerve stimulation revealed an increase of amplitude of isometric tetanic contractions upon application of capsaicin which was also eliminated after preliminary application of SB 366791. Similar data were obtained during direct muscle stimulation. Thus, pharmacological activation of TRPV1 channels affects the functioning of both the pre- and postsynaptic compartment of the neuromuscular junction. A moderate decrease in the amount of acetylcholine released from the motor nerve allows to maintain a reserve pool of the mediator to ensure a longer signal transmission process, and an increase in the force of muscle contraction, in its turn, also implies more effective physiological muscle activity in response to prolonged stimulation. This assumption is supported by the fact that when muscle was indirect stimulated with a fatigue protocol, muscle fatigue was attenuated in the presence of capsaicin.
Collapse
Affiliation(s)
- Arsenii Y Arkhipov
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center, Kazan Scientific Center of Russian Academy of Sciences, 2/31 Lobachevsky Street, Box 261, Kazan, Russia, 420111
| | - Nikita S Fedorov
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center, Kazan Scientific Center of Russian Academy of Sciences, 2/31 Lobachevsky Street, Box 261, Kazan, Russia, 420111
- Kazan Federal University, 18 Kremlyovskaya Street, Kazan, Russia, 420008
| | - Leniz F Nurullin
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center, Kazan Scientific Center of Russian Academy of Sciences, 2/31 Lobachevsky Street, Box 261, Kazan, Russia, 420111
- Kazan State Medical University, 49 Butlerova Street, Kazan, Russia, 420012
| | | | | | - Dmitry V Samigullin
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center, Kazan Scientific Center of Russian Academy of Sciences, 2/31 Lobachevsky Street, Box 261, Kazan, Russia, 420111
- A.N. Tupolev Kazan National Research Technical University, 10, K. Marx Street, Kazan, Russia, 420111
| | - Artem I Malomouzh
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center, Kazan Scientific Center of Russian Academy of Sciences, 2/31 Lobachevsky Street, Box 261, Kazan, Russia, 420111.
- A.N. Tupolev Kazan National Research Technical University, 10, K. Marx Street, Kazan, Russia, 420111.
| |
Collapse
|
7
|
Durbin RJ, Heredia DJ, Gould TW, Renden RB. Postsynaptic Calcium Extrusion at the Mouse Neuromuscular Junction Alkalinizes the Synaptic Cleft. J Neurosci 2023; 43:5741-5752. [PMID: 37474311 PMCID: PMC10423045 DOI: 10.1523/jneurosci.0815-23.2023] [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] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/07/2023] [Accepted: 07/14/2023] [Indexed: 07/22/2023] Open
Abstract
Neurotransmission is shaped by extracellular pH. Alkalization enhances pH-sensitive transmitter release and receptor activation, whereas acidification inhibits these processes and can activate acid-sensitive conductances in the synaptic cleft. Previous work has shown that the synaptic cleft can either acidify because of synaptic vesicular release and/or alkalize because of Ca2+ extrusion by the plasma membrane ATPase (PMCA). The direction of change differs across synapse types. At the mammalian neuromuscular junction (NMJ), the direction and magnitude of pH transients in the synaptic cleft during transmission remain ambiguous. We set out to elucidate the extracellular pH transients that occur at this cholinergic synapse under near-physiological conditions and identify their sources. We monitored pH-dependent changes in the synaptic cleft of the mouse levator auris longus using viral expression of the pseudoratiometric probe pHusion-Ex in the muscle. Using mice from both sexes, a significant and prolonged alkalization occurred when stimulating the connected nerve for 5 s at 50 Hz, which was dependent on postsynaptic intracellular Ca2+ release. Sustained stimulation for a longer duration (20 s at 50 Hz) caused additional prolonged net acidification at the cleft. To investigate the mechanism underlying cleft alkalization, we used muscle-expressed GCaMP3 to monitor the contribution of postsynaptic Ca2+ Activity-induced liberation of intracellular Ca2+ in muscle positively correlated with alkalization of the synaptic cleft, whereas inhibiting PMCA significantly decreased the extent of cleft alkalization. Thus, cholinergic synapses of the mouse NMJ typically alkalize because of cytosolic Ca2+ liberated in muscle during activity, unless under highly strenuous conditions where acidification predominates.SIGNIFICANCE STATEMENT Changes in synaptic cleft pH alter neurotransmission, acting on receptors and channels on both sides of the synapse. Synaptic acidification has been associated with a myriad of diseases in the central and peripheral nervous system. Here, we report that in near-physiological recording conditions the cholinergic neuromuscular junction shows use-dependent bidirectional changes in synaptic cleft pH-immediate alkalinization and a long-lasting acidification under prolonged stimulation. These results provide further insight into physiologically relevant changes at cholinergic synapses that have not been defined previously. Understanding and identifying synaptic pH transients during and after neuronal activity provides insight into short-term synaptic plasticity synapses and may identify therapeutic targets for diseases.
Collapse
Affiliation(s)
- Ryan J Durbin
- Integrative Neuroscience Graduate Program, University of Nevada, Reno, Reno, Nevada 89557
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, Nevada 89557
| | - Dante J Heredia
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, Nevada 89557
| | - Thomas W Gould
- Integrative Neuroscience Graduate Program, University of Nevada, Reno, Reno, Nevada 89557
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, Nevada 89557
| | - Robert B Renden
- Integrative Neuroscience Graduate Program, University of Nevada, Reno, Reno, Nevada 89557
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, Nevada 89557
| |
Collapse
|
8
|
Zheng Y, Li Y. Past, present, and future of tools for dopamine detection. Neuroscience 2023:S0306-4522(23)00295-6. [PMID: 37419404 DOI: 10.1016/j.neuroscience.2023.06.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/09/2023]
Abstract
Dopamine (DA) is a critical neuromodulator involved in various brain functions. To understand how DA regulates neural circuits and behaviors in the physiological and pathological conditions, it is essential to have tools that enable the direct detection of DA dynamics in vivo. Recently, genetically encoded DA sensors based on G protein-coupled receptors revolutionized this field, as it allows us to track in vivo DA dynamic with unprecedented spatial-temporal resolution, high molecular specificity, and sub-second kinetics. In this review, we first summarize traditional DA detection methods. Then we focus on the development of genetically encoded DA sensors and feature its significance to understanding dopaminergic neuromodulation across diverse behaviors and species. Finally, we present our perspectives about the future direction of the next-generation DA sensors and extend their potential applications. Overall, this review offers a comprehensive perspective on the past, present, and future of DA detection tools, with important implications for the study of DA functions in health and disease.
Collapse
Affiliation(s)
- Yu Zheng
- Peking-Tsinghua Center for Life Sciences, 100871 Beijing, China
| | - Yulong Li
- Peking-Tsinghua Center for Life Sciences, 100871 Beijing, China; State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, 100871 Beijing, China; PKU-IDG/McGovern Institute for Brain Research, 100871 Beijing, China; National Biomedical Imaging Center, Peking University, 100871 Beijing, China.
| |
Collapse
|
9
|
Wu Z, Cui Y, Wang H, Wu H, Wan Y, Li B, Wang L, Pan S, Peng W, Dong A, Yuan Z, Jing M, Xu M, Luo M, Li Y. Neuronal activity-induced, equilibrative nucleoside transporter-dependent, somatodendritic adenosine release revealed by a GRAB sensor. Proc Natl Acad Sci U S A 2023; 120:e2212387120. [PMID: 36996110 PMCID: PMC10083574 DOI: 10.1073/pnas.2212387120] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 02/28/2023] [Indexed: 03/31/2023] Open
Abstract
The purinergic signaling molecule adenosine (Ado) modulates many physiological and pathological functions in the brain. However, the exact source of extracellular Ado remains controversial. Here, utilizing a newly optimized genetically encoded GPCR-Activation-Based Ado fluorescent sensor (GRABAdo), we discovered that the neuronal activity-induced extracellular Ado elevation is due to direct Ado release from somatodendritic compartments of neurons, rather than from the axonal terminals, in the hippocampus. Pharmacological and genetic manipulations reveal that the Ado release depends on equilibrative nucleoside transporters but not the conventional vesicular release mechanisms. Compared with the fast-vesicular glutamate release, the Ado release is slow (~40 s) and requires calcium influx through L-type calcium channels. Thus, this study reveals an activity-dependent second-to-minute local Ado release from the somatodendritic compartments of neurons, potentially serving modulatory functions as a retrograde signal.
Collapse
Affiliation(s)
- Zhaofa Wu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing100871, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing100871, China
| | - Yuting Cui
- National Institute of Biological Sciences, Beijing102206, China
- Chinese Institute for Brain Research, Beijing102206, China
| | - Huan Wang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing100871, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing100871, China
| | - Hao Wu
- School of Life Sciences, Tsinghua University, Beijing100084, China
| | - Yi Wan
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing100871, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing100871, China
| | - Bohan Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing100871, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing100871, China
| | - Lei Wang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing100871, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing100871, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Peking University, Beijing100871, China
| | - Sunlei Pan
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing100871, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing100871, China
| | - Wanling Peng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai200031, China
| | - Ao Dong
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing100871, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing100871, China
| | - Zhengwei Yuan
- National Institute of Biological Sciences, Beijing102206, China
- School of Life Sciences, Tsinghua University, Beijing100084, China
| | - Miao Jing
- Chinese Institute for Brain Research, Beijing102206, China
| | - Min Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai200031, China
| | - Minmin Luo
- National Institute of Biological Sciences, Beijing102206, China
- Chinese Institute for Brain Research, Beijing102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing102206, China
- Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing100005, China
- New Cornerstone Science Institute at Chinese Institute for Brain Research, Beijing102206, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing100871, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing100871, China
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong518055, China
- National Biomedical Imaging Center, Peking University, Beijing100871, China
- New Cornerstone Science Institute at Peking University, Beijing100871, China
| |
Collapse
|
10
|
Müller M, Heckmann M. Linking Protons to Homeostatic Plasticity. Neuroscience 2021; 467:185-187. [PMID: 34029647 DOI: 10.1016/j.neuroscience.2021.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 05/13/2021] [Indexed: 10/21/2022]
Abstract
Editorial on Extracellular protons mediate presynaptic homeostatic potentiation at the mouse neuromuscular junction.
Collapse
Affiliation(s)
- Martin Müller
- Department of Molecular Life Sciences, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Manfred Heckmann
- Institute for Physiology, Department of Neurophysiology, Julius-Maximilians-University Würzburg, D-97070 Würzburg, Germany.
| |
Collapse
|
11
|
Arkhipov AY, Zhilyakov NV, Malomouzh AI, Samigullin DV. Interaction between the Mechanisms
of Suppression of Acetylcholine Quantal Secretion upon Activation
of Vanilloid (TRPV1) and Purine Receptors in the Mouse Neuromuscular Synapse. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021030182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
12
|
Beppu K, Kubo N, Matsui K. Glial amplification of synaptic signals. J Physiol 2021; 599:2085-2102. [PMID: 33527421 DOI: 10.1113/jp280857] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/27/2021] [Indexed: 12/20/2022] Open
Abstract
KEY POINTS Recent studies have repeatedly demonstrated the cross-talk of heterogeneous signals between neuronal and glial circuits. Here, we investigated the mechanism and the influence of physiological interactions between neurons and glia in the cerebellum. We found that the cerebellar astrocytes, Bergmann glial cells, react to exogenously applied glutamate, glutamate transporter substrate (d-aspartate) and synaptically released glutamate. In response, the Bergmann glial cells release glutamate through volume-regulated anion channels. It is generally assumed that all of the postsynaptic current is mediated by presynaptically released glutamate. However, we showed that a part of the postsynaptic current is mediated by glutamate released from Bergmann glial cells. Optogenetic manipulation of Bergmann glial state with archaerhodpsin-T or channelrhodopsin-2 reduced or augmented the amount of glial glutamate release, respectively. Our data indicate that glutamate-induced glutamate release in Bergmann glia serves as an effective amplifier of excitatory information processing in the brain. ABSTRACT Transmitter released from presynaptic neurons has been considered to be the sole generator of postsynaptic excitatory signals. However, astrocytes of the glial cell population have also been shown to release transmitter that can react on postsynaptic receptors. Therefore, we investigated whether astrocytes take part in generation of at least a part of the synaptic current. In this study, mice cerebellar acute slices were prepared and whole cell patch clamp recordings were performed. We found that Bergmann glial cells (BGs), a type of astrocyte in the cerebellum, reacts to a glutamate transporter substrate, d-aspartate (d-Asp) and an anion conductance is generated and glutamate is released from the BGs. Glutamate release was attenuated or augmented by modulating the state of BGs with activation of light-sensitive proteins, archaerhodopsin-T (ArchT) or channelrhodopsin-2 (ChR2) expressed on BGs, respectively. Glutamate release appears to be mediated by anion channels that can be blocked by a volume-regulated anion channel-specific blocker. Synaptic response to a train of parallel fibre stimulation was recorded from Purkinje cells. The latter part of the response was also attenuated or augmented by glial modulation with ArchT or ChR2, respectively. Thus, BGs effectively function as an excitatory signal amplifier, and a part of the 'synaptic' current is actually mediated by glutamate released from BGs. These data show that the state of BGs have potential for having direct and fundamental consequences on the functioning of information processing in the brain.
Collapse
Affiliation(s)
- Kaoru Beppu
- Division of Interdisciplinary Medical Science, Center for Neuroscience, Graduate School of Medicine, Tohoku University, Sendai, 980-8575, Japan
| | - Naoko Kubo
- Division of Interdisciplinary Medical Science, Center for Neuroscience, Graduate School of Medicine, Tohoku University, Sendai, 980-8575, Japan
| | - Ko Matsui
- Division of Interdisciplinary Medical Science, Center for Neuroscience, Graduate School of Medicine, Tohoku University, Sendai, 980-8575, Japan.,Super-network Brain Physiology, Graduate School of Life Sciences, Tohoku University, Sendai, 980-8577, Japan
| |
Collapse
|
13
|
Bocker HT, Heinrich T, Liebmann L, Hennings JC, Seemann E, Gerth M, Jakovčevski I, Preobraschenski J, Kessels MM, Westermann M, Isbrandt D, Jahn R, Qualmann B, Hübner CA. The Na+/H+ Exchanger Nhe1 Modulates Network Excitability via GABA Release. Cereb Cortex 2020; 29:4263-4276. [PMID: 30541023 DOI: 10.1093/cercor/bhy308] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 11/12/2018] [Accepted: 11/13/2018] [Indexed: 01/06/2023] Open
Abstract
Brain functions are extremely sensitive to pH changes because of the pH-dependence of proteins involved in neuronal excitability and synaptic transmission. Here, we show that the Na+/H+ exchanger Nhe1, which uses the Na+ gradient to extrude H+, is expressed at both inhibitory and excitatory presynapses. We disrupted Nhe1 specifically in mice either in Emx1-positive glutamatergic neurons or in parvalbumin-positive cells, mainly GABAergic interneurons. While Nhe1 disruption in excitatory neurons had no effect on overall network excitability, mice with disruption of Nhe1 in parvalbumin-positive neurons displayed epileptic activity. From our electrophysiological analyses in the CA1 of the hippocampus, we conclude that the disruption in parvalbumin-positive neurons impairs the release of GABA-loaded vesicles, but increases the size of GABA quanta. The latter is most likely an indirect pH-dependent effect, as Nhe1 was not expressed in purified synaptic vesicles itself. Conclusively, our data provide first evidence that Nhe1 affects network excitability via modulation of inhibitory interneurons.
Collapse
Affiliation(s)
- Hartmut T Bocker
- Institute of Human Genetics, Jena University Hospital, 07747 Jena, Germany
| | - Theresa Heinrich
- Department GMP Cell and Gene Therapy, Fraunhofer Institute for Cell Therapy and Immunology IZI, 04103 Leipzig, Germany
| | - Lutz Liebmann
- Institute of Human Genetics, Jena University Hospital, 07747 Jena, Germany
| | | | - Eric Seemann
- Institute of Biochemistry I, Jena University Hospital, 07743 Jena, Germany
| | - Melanie Gerth
- Institute of Human Genetics, Jena University Hospital, 07747 Jena, Germany
| | - Igor Jakovčevski
- Institute for Molecular and Behavioral Neuroscience, University of Cologne, 50937 Cologne, Germany, and German Center for Neurodegenerative Diseases (DZNE), 53175 Bonn, Germany
| | - Julia Preobraschenski
- Department of Neurobiology, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | - Michael M Kessels
- Institute of Biochemistry I, Jena University Hospital, 07743 Jena, Germany
| | - Martin Westermann
- Electron Microscopy Center, Jena University Hospital, 07747 Jena, Germany
| | - Dirk Isbrandt
- Institute for Molecular and Behavioral Neuroscience, University of Cologne, 50937 Cologne, Germany, and German Center for Neurodegenerative Diseases (DZNE), 53175 Bonn, Germany
| | - Reinhard Jahn
- Department of Neurobiology, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | - Britta Qualmann
- Institute of Biochemistry I, Jena University Hospital, 07743 Jena, Germany
| | - Christian A Hübner
- Institute of Human Genetics, Jena University Hospital, 07747 Jena, Germany
| |
Collapse
|
14
|
Quantitative Synaptic Biology: A Perspective on Techniques, Numbers and Expectations. Int J Mol Sci 2020; 21:ijms21197298. [PMID: 33023247 PMCID: PMC7582872 DOI: 10.3390/ijms21197298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 12/31/2022] Open
Abstract
Synapses play a central role for the processing of information in the brain and have been analyzed in countless biochemical, electrophysiological, imaging, and computational studies. The functionality and plasticity of synapses are nevertheless still difficult to predict, and conflicting hypotheses have been proposed for many synaptic processes. In this review, we argue that the cause of these problems is a lack of understanding of the spatiotemporal dynamics of key synaptic components. Fortunately, a number of emerging imaging approaches, going beyond super-resolution, should be able to provide required protein positions in space at different points in time. Mathematical models can then integrate the resulting information to allow the prediction of the spatiotemporal dynamics. We argue that these models, to deal with the complexity of synaptic processes, need to be designed in a sufficiently abstract way. Taken together, we suggest that a well-designed combination of imaging and modelling approaches will result in a far more complete understanding of synaptic function than currently possible.
Collapse
|
15
|
Rasmussen R, O'Donnell J, Ding F, Nedergaard M. Interstitial ions: A key regulator of state-dependent neural activity? Prog Neurobiol 2020; 193:101802. [PMID: 32413398 PMCID: PMC7331944 DOI: 10.1016/j.pneurobio.2020.101802] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 02/08/2023]
Abstract
Throughout the nervous system, ion gradients drive fundamental processes. Yet, the roles of interstitial ions in brain functioning is largely forgotten. Emerging literature is now revitalizing this area of neuroscience by showing that interstitial cations (K+, Ca2+ and Mg2+) are not static quantities but change dynamically across states such as sleep and locomotion. In turn, these state-dependent changes are capable of sculpting neuronal activity; for example, changing the local interstitial ion composition in the cortex is sufficient for modulating the prevalence of slow-frequency neuronal oscillations, or potentiating the gain of visually evoked responses. Disturbances in interstitial ionic homeostasis may also play a central role in the pathogenesis of central nervous system diseases. For example, impairments in K+ buffering occur in a number of neurodegenerative diseases, and abnormalities in neuronal activity in disease models disappear when interstitial K+ is normalized. Here we provide an overview of the roles of interstitial ions in physiology and pathology. We propose the brain uses interstitial ion signaling as a global mechanism to coordinate its complex activity patterns, and ion homeostasis failure contributes to central nervous system diseases affecting cognitive functions and behavior.
Collapse
Affiliation(s)
- Rune Rasmussen
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
| | - John O'Donnell
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, United States
| | - Fengfei Ding
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, United States
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark; Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, United States.
| |
Collapse
|
16
|
Zefirov AL, Mukhametzyanov RD, Zakharov AV, Mukhutdinova KA, Odnoshivkina UG, Petrov AM. Intracellular Acidification Suppresses Synaptic Vesicle Mobilization in the Motor Nerve Terminals. Acta Naturae 2020; 12:105-113. [PMID: 33456982 PMCID: PMC7800596 DOI: 10.32607/actanaturae.11054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/07/2020] [Indexed: 01/27/2023] Open
Abstract
Intracellular protons play a special role in the regulation of presynaptic processes, since the functioning of synaptic vesicles and endosomes depends on their acidification by the H+-pump. Furthermore, transient acidification of the intraterminal space occurs during synaptic activity. Using microelectrode recording of postsynaptic responses (an indicator of neurotransmitter release) and exo-endocytic marker FM1-43, we studied the effects of intracellular acidification with propionate on the presynaptic events underlying neurotransmitter release. Cytoplasmic acidification led to a marked decrease in neurotransmitter release during the first minute of a 20-Hz stimulation in the neuromuscular junctions of mouse diaphragm and frog cutaneous pectoris muscle. This was accompanied by a reduction in the FM1-43 loss during synaptic vesicle exocytosis in response to the stimulation. Estimation of the endocytic uptake of FM1-43 showed no disruption in synaptic vesicle endocytosis. Acidification completely prevented the action of the cell-membrane permeable compound 24-hydroxycholesterol, which can enhance synaptic vesicle mobilization. Thus, the obtained results suggest that an increase in [H+]in negatively regulates neurotransmission due to the suppression of synaptic vesicle delivery to the sites of exocytosis at high activity. This mechanism can be a part of the negative feedback loop in regulating neurotransmitter release.
Collapse
Affiliation(s)
- A. L. Zefirov
- Kazan State Medical University, Department of Normal Physiology, Kazan, 420012 Russia
- Institute of Neuroscience, Kazan State Medical University, Kazan, 420012 Russia
| | - R. D. Mukhametzyanov
- Kazan State Medical University, Department of Normal Physiology, Kazan, 420012 Russia
| | - A. V. Zakharov
- Kazan State Medical University, Department of Normal Physiology, Kazan, 420012 Russia
- Kazan Federal University, Kazan, 420008 Russia
| | - K. A. Mukhutdinova
- Institute of Neuroscience, Kazan State Medical University, Kazan, 420012 Russia
| | - U. G. Odnoshivkina
- Kazan State Medical University, Department of Normal Physiology, Kazan, 420012 Russia
| | - A. M. Petrov
- Kazan State Medical University, Department of Normal Physiology, Kazan, 420012 Russia
- Institute of Neuroscience, Kazan State Medical University, Kazan, 420012 Russia
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center “Kazan Scientific Center of RAS”, Kazan, 420111 Russia
| |
Collapse
|
17
|
Peng W, Wu Z, Song K, Zhang S, Li Y, Xu M. Regulation of sleep homeostasis mediator adenosine by basal forebrain glutamatergic neurons. Science 2020; 369:369/6508/eabb0556. [DOI: 10.1126/science.abb0556] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/19/2020] [Accepted: 07/03/2020] [Indexed: 12/15/2022]
Abstract
Sleep and wakefulness are homeostatically regulated by a variety of factors, including adenosine. However, how neural activity underlying the sleep-wake cycle controls adenosine release in the brain remains unclear. Using a newly developed genetically encoded adenosine sensor, we found an activity-dependent rapid increase in the concentration of extracellular adenosine in mouse basal forebrain (BF), a critical region controlling sleep and wakefulness. Although the activity of both BF cholinergic and glutamatergic neurons correlated with changes in the concentration of adenosine, optogenetic activation of these neurons at physiological firing frequencies showed that glutamatergic neurons contributed much more to the adenosine increase. Mice with selective ablation of BF glutamatergic neurons exhibited a reduced adenosine increase and impaired sleep homeostasis regulation. Thus, cell type–specific neural activity in the BF dynamically controls sleep homeostasis.
Collapse
Affiliation(s)
- Wanling Peng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhaofa Wu
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
- PKU-IDG–McGovern Institute for Brain Research, Beijing 100871, China
| | - Kun Song
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Siyu Zhang
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China
- PKU-IDG–McGovern Institute for Brain Research, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Min Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China
| |
Collapse
|
18
|
Activity and Cytosolic Na + Regulate Synaptic Vesicle Endocytosis. J Neurosci 2020; 40:6112-6120. [PMID: 32605936 DOI: 10.1523/jneurosci.0119-20.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 06/18/2020] [Accepted: 06/24/2020] [Indexed: 11/21/2022] Open
Abstract
Retrieval of synaptic vesicles via endocytosis is essential for maintaining sustained synaptic transmission, especially for neurons that fire action potentials at high frequencies. However, how neuronal activity regulates synaptic vesicle recycling is largely unknown. Here we report that Na+ substantially accumulated in the mouse calyx of Held terminals of either sex during repetitive high-frequency spiking. Elevated presynaptic Na+ accelerated both slow and rapid forms of endocytosis and facilitated endocytosis overshoot, but did not affect the readily releasable pool size, Ca2+ influx, or exocytosis. To examine whether this facilitation of endocytosis is related to the Na+-dependent vesicular content change, we dialyzed glutamate into the presynaptic cytosol or blocked the vesicular glutamate uptake with bafilomycin and found that the rate of endocytosis was not affected by regulating the vesicular glutamate content. Endocytosis is critically dependent on intracellular Ca2+, and the activity of Na+/Ca2+ exchanger (NCX) may be altered when the Na+ gradient is changed. However, neither NCX inhibitor nor change of extracellular Na+ concentration affected the endocytosis rate. Moreover, two-photon Ca2+ imaging showed that presynaptic Na+ did not affect the action potential-evoked intracellular Ca2+ transient and decay. Therefore, we revealed a novel mechanism of cytosolic Na+ in accelerating vesicle endocytosis. During high-frequency synaptic transmission, when large numbers of synaptic vesicles were fused, the rapid buildup of presynaptic cytosolic Na+ promoted vesicle recycling and sustained synaptic transmission.SIGNIFICANCE STATEMENT High-frequency firing neurons are widely distributed in the CNS. A large number of synaptic vesicles are released during high-frequency synaptic transmission; accordingly, synaptic vesicles need to be recycled rapidly to replenish the vesicle pool. Synaptic vesicle exocytosis and endocytosis are tightly coupled, and their coupling is essential for synaptic function and structural stability. We showed here that intracellular Na+ concentration at the calyx of Held terminal increased rapidly during spike activity and the increased Na+ accelerated endocytosis. Thus, when large numbers of synaptic vesicles are released during high-frequency synaptic transmission, Na+ accumulated in terminals and facilitated vesicle recycling. These findings represent a novel cellular mechanism that supports reliable synaptic transmission at high frequency in the CNS.
Collapse
|
19
|
Majumder R, Roy S, Okamoto K, Nagao S, Matsuo T, Parui PP. Porphyrin-Based Probe for Simultaneous Detection of Interface Acidity and Polarity during Lipid-Phase Transition of Vesicles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:426-434. [PMID: 31820997 DOI: 10.1021/acs.langmuir.9b02781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Biochemical activities at a membrane interface are affected by local pH/polarity related to membrane lipid properties including lipid dynamics. pH and polarity at the interface are two highly interdependent parameters, depending on various locations from the water-exposed outer surface to the less polar inner surface. The optical response of common pH or polarity probes is affected by both the local pH and polarity; therefore, estimation of these values using two separate probes localized at different interface depths can be erroneous. To estimate interface pH and polarity at an identical interface depth, we synthesized a glucose-pendant porphyrin (GPP) molecule for simultaneous pH and polarity detection by a single optical probe. pH-induced protonation equilibrium and polarity-dependent π-π stacking aggregation for GPP are exploited to measure pH and polarity changes at the 1,2-dimyristoyl-sn-glycero-3-phospho-(1'-rac-glycerol) (DMPG) membrane interface during DMPG phase transition. An NMR study confirmed that GPP is located at the interface Stern layer of DMPG large unilamellar vesicle (LUV). Using UV-vis absorption studies with an adapted analysis protocol, we estimated interface pH, or its deviation from the bulk phase value (ΔpH), and the interface polarity simultaneously using the same spectra for sodium dodecyl sulfate micelle and DMPG LUV. During temperature-dependent gel to liquid-crystalline phase transition of DMPG, there was ∼0.5 unit increase in ΔpH from approximately -0.6 to -1.1, with a small increase in the interface dielectric constant from ∼60 to 63. A series of spectroscopic data indicate the utility of GPP for evaluation of local pH/polarity change during lipid phase transition of vesicles.
Collapse
Affiliation(s)
- Rini Majumder
- Department of Chemistry , Jadavpur University , Kolkata 700032 , India
| | - Snigdha Roy
- Department of Chemistry , Jadavpur University , Kolkata 700032 , India
| | - Kentaro Okamoto
- Division of Materials Science, Graduate School of Science and Technology , Nara Institute of Science and Technology (NAIST) , 8916-5 Takayama-cho , Ikoma , Nara 630-0192 , Japan
| | - Satoshi Nagao
- Division of Materials Science, Graduate School of Science and Technology , Nara Institute of Science and Technology (NAIST) , 8916-5 Takayama-cho , Ikoma , Nara 630-0192 , Japan
| | - Takashi Matsuo
- Division of Materials Science, Graduate School of Science and Technology , Nara Institute of Science and Technology (NAIST) , 8916-5 Takayama-cho , Ikoma , Nara 630-0192 , Japan
| | | |
Collapse
|
20
|
Expression of plasma membrane calcium ATPases confers Ca 2+/H + exchange in rodent synaptic vesicles. Sci Rep 2019; 9:4289. [PMID: 30862855 PMCID: PMC6414521 DOI: 10.1038/s41598-019-40557-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 02/19/2019] [Indexed: 02/07/2023] Open
Abstract
Ca2+ transport into synaptic vesicles (SVs) at the presynaptic terminals has been proposed to be an important process for regulating presynaptic [Ca2+] during stimulation as well as at rest. However, the molecular identity of the transport system remains elusive. Previous studies have demonstrated that isolated SVs exhibit two distinct Ca2+ transport systems depending on extra-vesicular (cytosolic) pH; one is mediated by a high affinity Ca2+ transporter which is active at neutral pH and the other is mediated by a low affinity Ca2+/H+ antiporter which is maximally active at alkaline pH of 8.5. In addition, synaptic vesicle glycoprotein 2 s (SV2s), a major SV component, have been proposed to contribute to Ca2+ clearance from the presynaptic cytoplasm. Here, we show that at physiological pH, the plasma membrane Ca2+ ATPases (PMCAs) are responsible for both the Ca2+/H+ exchange activity and Ca2+ uptake into SVs. The Ca2+/H+ exchange activity monitored by acidification assay exhibited high affinity for Ca2+ (Km ~ 400 nM) and characteristic divalent cation selectivity for the PMCAs. Both activities were remarkably reduced by PMCA blockers, but not by a blocker of the ATPase that transfers Ca2+ from the cytosol to the lumen of sarcoplasmic endoplasmic reticulum (SERCA) at physiological pH. Furthermore, we rule out the contribution of SV2s, putative Ca2+ transporters on SVs, since both Ca2+/H+ exchange activity and Ca2+ transport were unaffected in isolated vesicles derived from SV2-deficient brains. Finally, using a PMCA1-pHluorin construct that enabled us to monitor cellular distribution and recycling properties in living neurons, we demonstrated that PMCA1-pHluorin localized to intracellular acidic compartments and recycled at presynaptic terminals in an activity-dependent manner. Collectively, our results imply that vesicular PMCAs may play pivotal roles in both presynaptic Ca2+ homeostasis and the modulation of H+ gradient in SVs.
Collapse
|
21
|
Affiliation(s)
- Kartik Venkatachalam
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth), Houston, TX, 77030, USA.,Programs in Cell and Regulatory Biology and Neuroscience at the Graduate School of Biomedical Sciences, University of Texas School of Medicine, Houston, TX, 77030, USA
| |
Collapse
|
22
|
Chromophore-Assisted Light Inactivation of the V-ATPase V0c Subunit Inhibits Neurotransmitter Release Downstream of Synaptic Vesicle Acidification. Mol Neurobiol 2018; 56:3591-3602. [PMID: 30155790 DOI: 10.1007/s12035-018-1324-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 08/17/2018] [Indexed: 10/28/2022]
Abstract
Synaptic vesicle proton V-ATPase is an essential component in synaptic vesicle function. Active acidification of synaptic vesicles, triggered by the V-ATPase, is necessary for neurotransmitter storage. Independently from its proton transport activity, an additional important function of the membrane-embedded sector of the V-ATPase has been uncovered over recent years. Subunits a and c of the membrane sector of this multi-molecular complex have been shown to interact with SNARE proteins and to be involved in modulating neurotransmitter release. The c-subunit interacts with the v-SNARE VAMP2 and facilitates neurotransmission. In this study, we used chromophore-assisted light inactivation and monitored the consequences on neurotransmission on line in CA3 pyramidal neurons. We show that V-ATPase c-subunit V0c is a key element in modulating neurotransmission and that its specific inactivation rapidly inhibited neurotransmission.
Collapse
|
23
|
Burke SRA, Reed EJ, Romer SH, Voss AA. Levator Auris Longus Preparation for Examination of Mammalian Neuromuscular Transmission Under Voltage Clamp Conditions. J Vis Exp 2018. [PMID: 29782004 DOI: 10.3791/57482] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
This protocol describes a technique to record synaptic transmission from the neuromuscular junction under current-clamp and voltage-clamp conditions. An ex vivo preparation of the levator auris longus (LAL) is used because it is a thin muscle that provides easy visualization of the neuromuscular junction for microelectrode impalement at the motor endplate. This method allows for the recording of spontaneous miniature endplate potentials and currents (mEPPs and mEPCs), nerve-evoked endplate potentials and currents (EPPs and EPCs), as well as the membrane properties of the motor endplate. Results obtained from this method include the quantal content (QC), number of vesicle release sites (n), probability of vesicle release (prel), synaptic facilitation and depression, as well as the muscle membrane time constant (τm) and input resistance. Application of this technique to mouse models of human disease can highlight key pathologies in disease states and help identify novel treatment strategies. By fully voltage-clamping a single synapse, this method provides one of the most detailed analyses of synaptic transmission currently available.
Collapse
Affiliation(s)
| | - Eric J Reed
- Department of Biological Sciences, Wright State University
| | | | - Andrew A Voss
- Department of Biological Sciences, Wright State University;
| |
Collapse
|
24
|
A mouse model of autism implicates endosome pH in the regulation of presynaptic calcium entry. Nat Commun 2018; 9:330. [PMID: 29362376 PMCID: PMC5780507 DOI: 10.1038/s41467-017-02716-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 12/19/2017] [Indexed: 11/21/2022] Open
Abstract
Psychoactive compounds such as chloroquine and amphetamine act by dissipating the pH gradient across intracellular membranes, but the physiological mechanisms that normally regulate organelle pH remain poorly understood. Interestingly, recent human genetic studies have implicated the endosomal Na+/H+ exchanger NHE9 in both autism spectrum disorders (ASD) and attention deficit hyperactivity disorder (ADHD). Plasma membrane NHEs regulate cytosolic pH, but the role of intracellular isoforms has remained unclear. We now find that inactivation of NHE9 in mice reproduces behavioral features of ASD including impaired social interaction, repetitive behaviors, and altered sensory processing. Physiological characterization reveals hyperacidic endosomes, a cell-autonomous defect in glutamate receptor expression and impaired neurotransmitter release due to a defect in presynaptic Ca2+ entry. Acute inhibition of synaptic vesicle acidification rescues release but without affecting the primary defect due to loss of NHE9. The Na+/H+ exchanger NHE9 is proposed to regulate the H+ electrochemical gradient across endosomal membranes. Here, the authors find that NHE9 knockout mice show autism spectrum disorder-like behaviors and disrupted synaptic vesicle exocytosis due to impaired presynaptic calcium entry.
Collapse
|
25
|
Martineau M, Guzman RE, Fahlke C, Klingauf J. VGLUT1 functions as a glutamate/proton exchanger with chloride channel activity in hippocampal glutamatergic synapses. Nat Commun 2017; 8:2279. [PMID: 29273736 PMCID: PMC5741633 DOI: 10.1038/s41467-017-02367-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 11/24/2017] [Indexed: 12/18/2022] Open
Abstract
Glutamate is the major excitatory transmitter in the vertebrate nervous system. To maintain synaptic efficacy, recycling synaptic vesicles (SV) are refilled with glutamate by vesicular glutamate transporters (VGLUTs). The dynamics and mechanism of glutamate uptake in intact neurons are still largely unknown. Here, we show by live-cell imaging with pH- and chloride-sensitive fluorescent probes in cultured hippocampal neurons of wild-type and VGLUT1-deficient mice that in SVs VGLUT functions as a glutamate/proton exchanger associated with a channel-like chloride conductance. After endocytosis most internalized Cl− is substituted by glutamate in an electrically, and presumably osmotically, neutral manner, and this process is driven by both the Cl− gradient itself and the proton motive force provided by the vacuolar H+-ATPase. Our results shed light on the transport mechanism of VGLUT under physiological conditions and provide a framework for how modulation of glutamate transport via Cl− and pH can change synaptic strength. During neurotransmission synaptic vesicles are filled with glutamate by vesicular glutamate transporters (VGLUTs). Here, authors image intact neurons and show that in synaptic vesicles VGLUT functions as a glutamate/proton exchanger associated with a channel-like chloride conductance.
Collapse
Affiliation(s)
- Magalie Martineau
- Department of Cellular Biophysics, Institute for Medical Physics and Biophysics, University of Muenster, 48149, Muenster, Germany. .,University of Bordeaux and Centre National de la Recherche Scientifique, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000, Bordeaux, France.
| | - Raul E Guzman
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Christoph Fahlke
- Institute of Complex Systems, Zelluläre Biophysik (ICS-4), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Jürgen Klingauf
- Department of Cellular Biophysics, Institute for Medical Physics and Biophysics, University of Muenster, 48149, Muenster, Germany. .,IZKF Münster and Cluster of Excellence EXC 1003, Cells in Motion (CiM), 48149, Muenster, Germany.
| |
Collapse
|
26
|
Ventimiglia D, Bargmann CI. Diverse modes of synaptic signaling, regulation, and plasticity distinguish two classes of C. elegans glutamatergic neurons. eLife 2017; 6:e31234. [PMID: 29160768 PMCID: PMC5705214 DOI: 10.7554/elife.31234] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/20/2017] [Indexed: 11/13/2022] Open
Abstract
Synaptic vesicle release properties vary between neuronal cell types, but in most cases the molecular basis of this heterogeneity is unknown. Here, we compare in vivo synaptic properties of two neuronal classes in the C. elegans central nervous system, using VGLUT-pHluorin to monitor synaptic vesicle exocytosis and retrieval in intact animals. We show that the glutamatergic sensory neurons AWCON and ASH have distinct synaptic dynamics associated with tonic and phasic synaptic properties, respectively. Exocytosis in ASH and AWCON is differentially affected by SNARE-complex regulators that are present in both neurons: phasic ASH release is strongly dependent on UNC-13, whereas tonic AWCON release relies upon UNC-18 and on the protein kinase C homolog PKC-1. Strong stimuli that elicit high calcium levels increase exocytosis and retrieval rates in AWCON, generating distinct tonic and evoked synaptic modes. These results highlight the differential deployment of shared presynaptic proteins in neuronal cell type-specific functions.
Collapse
Affiliation(s)
- Donovan Ventimiglia
- Lulu and Anthony Wang Laboratory of Neural Circuits and BehaviorThe Rockefeller UniversityNew YorkUnited States
| | - Cornelia I Bargmann
- Lulu and Anthony Wang Laboratory of Neural Circuits and BehaviorThe Rockefeller UniversityNew YorkUnited States
| |
Collapse
|
27
|
Bertone NI, Groisman AI, Mazzone GL, Cano R, Tabares L, Uchitel OD. Carbonic anhydrase inhibitor acetazolamide shifts synaptic vesicle recycling to a fast mode at the mouse neuromuscular junction. Synapse 2017; 71. [PMID: 28873252 DOI: 10.1002/syn.22009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 08/25/2017] [Accepted: 08/29/2017] [Indexed: 01/17/2023]
Abstract
Acetazolamide (AZ), a molecule frequently used to treat different neurological syndromes, is an inhibitor of the carbonic anhydrase (CA), an enzyme that regulates pH inside and outside cells. We combined fluorescent FM styryl dyes and electrophysiological techniques at ex vivo levator auris longus neuromuscular junctions (NMJs) from mice to investigate the modulation of synaptic transmission and vesicle recycling by AZ. Transmitter release was minimally affected by AZ, as evidenced by evoked and spontaneous end-plate potential measurements. However, optical evaluation with FM-styryl dyes of vesicle exocytosis elicited by 50 Hz stimuli showed a strong reduction in fluorescence loss in AZ treated NMJ, an effect that was abolished by bathing the NMJ in Hepes. The remaining dye was quenched by bromophenol, a small molecule capable of diffusing inside vesicles. Furthermore, in transgenic mice expressing Synaptophysin-pHluorin (SypHy), the fluorescence responses of motor nerve terminals to a 50 Hz train of stimuli was decrease to a 50% of controls in the presence of AZ. Immunohistochemistry experiments to evaluate the state of the Myosin light chain kinase (MLCK), an enzyme involved in vesicle recycling, demonstrated that MLCK phosphorylation was much stronger in the presence than AZ than in its absence in 50 Hz stimulated NMJs. We postulate that AZ, via cytosol acidification and activation of MLCK, shifts synaptic vesicle recycling to a fast (kiss-and-run) mode, which changes synaptic performance. These changes may contribute to the therapeutic action reported in many neurological syndromes like ataxia, epilepsy, and migraine.
Collapse
Affiliation(s)
- Nicolas Ivan Bertone
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET) and Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires C1428EHA, Argentina
| | - Ayelén Ivana Groisman
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET) and Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires C1428EHA, Argentina
| | - Graciela Lujan Mazzone
- Laboratorios de Investigación aplicada en Neurociencias (LIAN)-Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), CONICET, Buenos Aires, Argentina
| | - Raquel Cano
- Department of Medical Physiology and Biophysics, School of Medicine, University of Seville, Seville 41009, Spain
| | - Lucia Tabares
- Department of Medical Physiology and Biophysics, School of Medicine, University of Seville, Seville 41009, Spain
| | - Osvaldo Daniel Uchitel
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET) and Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires C1428EHA, Argentina
| |
Collapse
|
28
|
Bodzęta A, Kahms M, Klingauf J. The Presynaptic v-ATPase Reversibly Disassembles and Thereby Modulates Exocytosis but Is Not Part of the Fusion Machinery. Cell Rep 2017; 20:1348-1359. [DOI: 10.1016/j.celrep.2017.07.040] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/23/2017] [Accepted: 07/14/2017] [Indexed: 11/15/2022] Open
|
29
|
Fatty acid synthase cooperates with protrudin to facilitate membrane outgrowth of cellular protrusions. Sci Rep 2017; 7:46569. [PMID: 28429738 PMCID: PMC5399442 DOI: 10.1038/srep46569] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 03/17/2017] [Indexed: 01/02/2023] Open
Abstract
Cellular protrusion formation capacity is a key feature of developing neurons and many eukaryotic cells. However, the mechanisms underlying membrane growth in protrusion formation are largely unclear. In this study, photo-reactive unnatural amino acid 3-(3-methyl-3H-diazirin-3-yl)-propamino-carbonyl-Nε-l-lysine was incorporated by a genetic code expansion strategy into protrudin, a protein localized in acidic endosomes and in the endoplasmic reticulum, that induces cellular protrusion and neurite formation. The modified protrudin was used for covalent trapping of protrudin-interacting proteins in living cells. Fatty acid synthase (FASN), which synthesizes free fatty acids, was identified to transiently interact with protrudin. Further characterization revealed a unique cooperation mechanism in which protrudin cooperates with FASN to facilitate cellular protrusion formation. This work reveals a novel mechanism involved in protrusion formation that is dependent on transient interaction between FASN and protrudin, and establishes a creative strategy to investigate transient protein-protein interactions in mammalian cells.
Collapse
|
30
|
Distinct Actions of Voltage-Activated Ca 2+ Channel Block on Spontaneous Release at Excitatory and Inhibitory Central Synapses. J Neurosci 2017; 37:4301-4310. [PMID: 28320843 DOI: 10.1523/jneurosci.3488-16.2017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 03/08/2017] [Accepted: 03/13/2017] [Indexed: 11/21/2022] Open
Abstract
At chemical synapses, voltage-activated calcium channels (VACCs) mediate Ca2+ influx to trigger action potential-evoked neurotransmitter release. However, the mechanisms by which Ca2+ regulates spontaneous transmission have not been fully determined. We have shown that VACCs are a major trigger of spontaneous release at neocortical inhibitory synapses but not at excitatory synapses, suggesting fundamental differences in spontaneous neurotransmission at GABAergic and glutamatergic synapses. Recently, VACC blockers were reported to reduce spontaneous release of glutamate and it was proposed that there was conservation of underlying mechanisms of neurotransmission at excitatory and inhibitory synapses. Furthermore, it was hypothesized that the different effects on excitatory and inhibitory synapses may have resulted from off-target actions of Cd2+, a nonselective VACC blocker, or other variations in experimental conditions. Here we report that in mouse neocortical neurons, selective and nonselective VACC blockers inhibit spontaneous release at inhibitory but not at excitatory terminals, and that this pattern is observed in culture and slice preparations as well as in synapses from acute slices of the auditory brainstem. The voltage dependence of Cd2+ block of VACCs accounts for the apparent lower potency of Cd2+ on spontaneous release of GABA than on VACC current amplitudes. Our findings indicate fundamental differences in the regulation of spontaneous release at inhibitory and excitatory synapses by stochastic VACC activity that extend beyond the cortex to the brainstem.SIGNIFICANCE STATEMENT Presynaptic Ca2+ entry via voltage-activated calcium channels (VACCs) is the major trigger of action potential-evoked synaptic release. However, the role of VACCs in the regulation of spontaneous neurotransmitter release (in the absence of a synchronizing action potential) remains controversial. We show that spontaneous release is affected differently by VACCs at excitatory and inhibitory synapses. At inhibitory synapses, stochastic openings of VACCs trigger the majority of spontaneous release, whereas they do not affect spontaneous release at excitatory synapses. We find this pattern to be wide ranging, holding for large and small synapses in the neocortex and brainstem. These findings indicate fundamental differences of the Ca2+ dependence of spontaneous release at excitatory and inhibitory synapses and heterogeneity of the mechanisms of release across the CNS.
Collapse
|
31
|
Chiacchiaretta M, Latifi S, Bramini M, Fadda M, Fassio A, Benfenati F, Cesca F. Neuronal hyperactivity causes Na +/H + exchanger-induced extracellular acidification at active synapses. J Cell Sci 2017; 130:1435-1449. [PMID: 28254883 DOI: 10.1242/jcs.198564] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/28/2017] [Indexed: 12/12/2022] Open
Abstract
Extracellular pH impacts on neuronal activity, which is in turn an important determinant of extracellular H+ concentration. The aim of this study was to describe the spatio-temporal dynamics of extracellular pH at synaptic sites during neuronal hyperexcitability. To address this issue we created ex.E2GFP, a membrane-targeted extracellular ratiometric pH indicator that is exquisitely sensitive to acidic shifts. By monitoring ex.E2GFP fluorescence in real time in primary cortical neurons, we were able to quantify pH fluctuations during network hyperexcitability induced by convulsant drugs or high-frequency electrical stimulation. Sustained hyperactivity caused a pH decrease that was reversible upon silencing of neuronal activity and located at active synapses. This acidic shift was not attributable to the outflow of synaptic vesicle H+ into the cleft nor to the activity of membrane-exposed H+ V-ATPase, but rather to the activity of the Na+/H+-exchanger. Our data demonstrate that extracellular synaptic pH shifts take place during epileptic-like activity of neural cultures, emphasizing the strict links existing between synaptic activity and synaptic pH. This evidence may contribute to the understanding of the physio-pathological mechanisms associated with hyperexcitability in the epileptic brain.
Collapse
Affiliation(s)
- Martina Chiacchiaretta
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova 16132, Italy.,Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova 16132, Italy
| | - Shahrzad Latifi
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova 16132, Italy
| | - Mattia Bramini
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova 16132, Italy
| | - Manuela Fadda
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova 16132, Italy
| | - Anna Fassio
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova 16132, Italy.,Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova 16132, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova 16132, Italy.,Department of Experimental Medicine, University of Genova, Viale Benedetto XV 3, Genova 16132, Italy
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience and Technology, Fondazione Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, Genova 16132, Italy
| |
Collapse
|
32
|
Bueno-Junior LS, Ruggiero RN, Rossignoli MT, Del Bel EA, Leite JP, Uchitel OD. Acetazolamide potentiates the afferent drive to prefrontal cortex in vivo. Physiol Rep 2017; 5:5/1/e13066. [PMID: 28087816 PMCID: PMC5256155 DOI: 10.14814/phy2.13066] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 11/14/2016] [Indexed: 11/24/2022] Open
Abstract
The knowledge on real-time neurophysiological effects of acetazolamide is still far behind the wide clinical use of this drug. Acetazolamide - a carbonic anhydrase inhibitor - has been shown to affect the neuromuscular transmission, implying a pH-mediated influence on the central synaptic transmission. To start filling such a gap, we chose a central substrate: hippocampal-prefrontal cortical projections; and a synaptic phenomenon: paired-pulse facilitation (a form of synaptic plasticity) to probe this drug's effects on interareal brain communication in chronically implanted rats. We observed that systemic acetazolamide potentiates the hippocampal-prefrontal paired-pulse facilitation. In addition to this field electrophysiology data, we found that acetazolamide exerts a net inhibitory effect on prefrontal cortical single-unit firing. We propose that systemic acetazolamide reduces the basal neuronal activity of the prefrontal cortex, whereas increasing the afferent drive it receives from the hippocampus. In addition to being relevant to the clinical and side effects of acetazolamide, these results suggest that exogenous pH regulation can have diverse impacts on afferent signaling across the neocortex.
Collapse
Affiliation(s)
- Lezio S Bueno-Junior
- Department of Neuroscience and Behavioral Sciences, Ribeirao Preto Medical School University of Sao Paulo, Ribeirao Preto, Brazil
| | - Rafael N Ruggiero
- Department of Neuroscience and Behavioral Sciences, Ribeirao Preto Medical School University of Sao Paulo, Ribeirao Preto, Brazil
| | - Matheus T Rossignoli
- Department of Neuroscience and Behavioral Sciences, Ribeirao Preto Medical School University of Sao Paulo, Ribeirao Preto, Brazil
| | - Elaine A Del Bel
- Department of Morphology, Physiology and Stomatology, Dentistry School of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Joao P Leite
- Department of Neuroscience and Behavioral Sciences, Ribeirao Preto Medical School University of Sao Paulo, Ribeirao Preto, Brazil
| | - Osvaldo D Uchitel
- Department of Physiology, Molecular and Cell Biology, Institute of Physiology Molecular Biology and Neuroscience University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
33
|
Rossano AJ, Kato A, Minard KI, Romero MF, Macleod GT. Na + /H + exchange via the Drosophila vesicular glutamate transporter mediates activity-induced acid efflux from presynaptic terminals. J Physiol 2016; 595:805-824. [PMID: 27641622 DOI: 10.1113/jp273105] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 09/14/2016] [Indexed: 01/26/2023] Open
Abstract
KEY POINTS Intracellular pH regulation is vital to neurons as nerve activity produces large and rapid acid loads in presynaptic terminals. Rapid clearance of acid loads is necessary to maintain control of neurotransmission, but neuronal acid clearance mechanisms remain poorly understood. Glutamate is loaded into synaptic vesicles via the vesicular glutamate transporter (VGLUT), a mechanism conserved across phyla, and this study reports a previously unknown role for VGLUT as an acid-extruding protein when deposited in the plasmamembrane during exocytosis. The finding was made in Drosophila (fruit fly) larval motor neurons through a combined pharamacological and genetic dissection of presynaptic pH homeostatic mechanisms. A dual role for VGLUT serves to integrate neuronal activity and pH regulation in presynaptic nerve terminals. ABSTRACT Neuronal activity can result in transient acidification of presynaptic terminals, and such shifts in cytosolic pH (pHcyto ) probably influence mechanisms underlying forms of synaptic plasticity with a presynaptic locus. As neuronal activity drives acid loading in presynaptic terminals, we hypothesized that the same activity might drive acid efflux mechanisms to maintain pHcyto homeostasis. To better understand the integration of neuronal activity and pHcyto regulation we investigated the acid extrusion mechanisms at Drosophila glutamatergic motorneuron terminals. Expression of a fluorescent genetically encoded pH indicator, named 'pHerry', in the presynaptic cytosol revealed acid efflux following nerve activity to be greater than that predicted from measurements of the intrinsic rate of acid efflux. Analysis of activity-induced acid transients in terminals deficient in either endocytosis or exocytosis revealed an acid efflux mechanism reliant upon synaptic vesicle exocytosis. Pharmacological and genetic dissection in situ and in a heterologous expression system indicate that this acid efflux is mediated by conventional plasmamembrane acid transporters, and also by previously unrecognized intrinsic H+ /Na+ exchange via the Drosophila vesicular glutamate transporter (DVGLUT). DVGLUT functions not only as a vesicular glutamate transporter but also serves as an acid-extruding protein when deposited on the plasmamembrane.
Collapse
Affiliation(s)
- Adam J Rossano
- School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Akira Kato
- Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, 226-8503, Japan.,Physiology & Biomedical Engineering and Nephrology & Hypertension, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
| | - Karyl I Minard
- Biological Sciences & Wilkes Honors College, Florida Atlantic University, Jupiter, FL, 33431, USA
| | - Michael F Romero
- Physiology & Biomedical Engineering and Nephrology & Hypertension, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
| | - Gregory T Macleod
- Biological Sciences & Wilkes Honors College, Florida Atlantic University, Jupiter, FL, 33431, USA
| |
Collapse
|
34
|
Expression of Carbonic Anhydrase I in Motor Neurons and Alterations in ALS. Int J Mol Sci 2016; 17:ijms17111820. [PMID: 27809276 PMCID: PMC5133821 DOI: 10.3390/ijms17111820] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/20/2016] [Accepted: 10/24/2016] [Indexed: 12/27/2022] Open
Abstract
Carbonic anhydrase I (CA1) is the cytosolic isoform of mammalian α-CA family members which are responsible for maintaining pH homeostasis in the physiology and pathology of organisms. A subset of CA isoforms are known to be expressed and function in the central nervous system (CNS). CA1 has not been extensively characterized in the CNS. In this study, we demonstrate that CA1 is expressed in the motor neurons in human spinal cord. Unexpectedly, a subpopulation of CA1 appears to be associated with endoplasmic reticulum (ER) membranes. In addition, the membrane-associated CA1s are preferentially upregulated in amyotrophic lateral sclerosis (ALS) and exhibit altered distribution in motor neurons. Furthermore, long-term expression of CA1 in mammalian cells activates apoptosis. Our results suggest a previously unknown role for CA1 function in the CNS and its potential involvement in motor neuron degeneration in ALS.
Collapse
|
35
|
Warren TJ, Van Hook MJ, Supuran CT, Thoreson WB. Sources of protons and a role for bicarbonate in inhibitory feedback from horizontal cells to cones in Ambystoma tigrinum retina. J Physiol 2016; 594:6661-6677. [PMID: 27345444 DOI: 10.1113/jp272533] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 06/16/2016] [Indexed: 12/13/2022] Open
Abstract
KEY POINTS In the vertebrate retina, photoreceptors influence the signalling of neighbouring photoreceptors through lateral-inhibitory interactions mediated by horizontal cells (HCs). These interactions create antagonistic centre-surround receptive fields important for detecting edges and generating chromatically opponent responses in colour vision. The mechanisms responsible for inhibitory feedback from HCs involve changes in synaptic cleft pH that modulate photoreceptor calcium currents. However, the sources of synaptic protons involved in feedback and the mechanisms for their removal from the cleft when HCs hyperpolarize to light remain unknown. Our results indicate that Na+ -H+ exchangers are the principal source of synaptic cleft protons involved in HC feedback but that synaptic cleft alkalization during light-evoked hyperpolarization of HCs also involves changes in bicarbonate transport across the HC membrane. In addition to delineating processes that establish lateral inhibition in the retina, these results contribute to other evidence showing the key role for pH in regulating synaptic signalling throughout the nervous system. ABSTRACT Lateral-inhibitory feedback from horizontal cells (HCs) to photoreceptors involves changes in synaptic cleft pH accompanying light-evoked changes in HC membrane potential. We analysed HC to cone feedback by studying surround-evoked light responses of cones and by obtaining paired whole cell recordings from cones and HCs in salamander retina. We tested three potential sources for synaptic cleft protons: (1) generation by extracellular carbonic anhydrase (CA), (2) release from acidic synaptic vesicles and (3) Na+ /H+ exchangers (NHEs). Neither antagonizing extracellular CA nor blocking loading of protons into synaptic vesicles eliminated feedback. However, feedback was eliminated when extracellular Na+ was replaced with choline and significantly reduced by an NHE inhibitor, cariporide. Depriving NHEs of intracellular protons by buffering HC cytosol with a pH 9.2 pipette solution eliminated feedback, whereas alkalinizing the cone cytosol did not, suggesting that HCs are a major source for protons in feedback. We also examined mechanisms for changing synaptic cleft pH in response to changes in HC membrane potential. Increasing the trans-membrane proton gradient by lowering the extracellular pH from 7.8 to 7.4 to 7.1 strengthened feedback. While maintaining constant extracellular pH with 1 mm HEPES, removal of bicarbonate abolished feedback. Elevating intracellular bicarbonate levels within HCs prevented this loss of feedback. A bicarbonate transport inhibitor, 4,4'-diisothiocyano-2,2'-stilbenedisulfonic acid (DIDS), also blocked feedback. Together, these results suggest that NHEs are the primary source of extracellular protons in HC feedback but that changes in cleft pH accompanying changes in HC membrane voltage also require bicarbonate flux across the HC membrane.
Collapse
Affiliation(s)
- Ted J Warren
- Department of Pharmacology & Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA.,Truhlsen Eye Institute and Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Matthew J Van Hook
- Truhlsen Eye Institute and Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Claudiu T Supuran
- University of Florence, Neurofarba Department, Sesto Fiorentino, Italy
| | - Wallace B Thoreson
- Department of Pharmacology & Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA.,Truhlsen Eye Institute and Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
36
|
Zhao H, Carney KE, Falgoust L, Pan JW, Sun D, Zhang Z. Emerging roles of Na⁺/H⁺ exchangers in epilepsy and developmental brain disorders. Prog Neurobiol 2016; 138-140:19-35. [PMID: 26965387 DOI: 10.1016/j.pneurobio.2016.02.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/03/2016] [Accepted: 02/27/2016] [Indexed: 12/15/2022]
Abstract
Epilepsy is a common central nervous system (CNS) disease characterized by recurrent transient neurological events occurring due to abnormally excessive or synchronous neuronal activity in the brain. The CNS is affected by systemic acid-base disorders, and epileptic seizures are sensitive indicators of underlying imbalances in cellular pH regulation. Na(+)/H(+) exchangers (NHEs) are a family of membrane transporter proteins actively involved in regulating intracellular and organellar pH by extruding H(+) in exchange for Na(+) influx. Altering NHE function significantly influences neuronal excitability and plays a role in epilepsy. This review gives an overview of pH regulatory mechanisms in the brain with a special focus on the NHE family and the relationship between epilepsy and dysfunction of NHE isoforms. We first discuss how cells translocate acids and bases across the membrane and establish pH homeostasis as a result of the concerted effort of enzymes and ion transporters. We focus on the specific roles of the NHE family by detailing how the loss of NHE1 in two NHE mutant mice results in enhanced neuronal excitability in these animals. Furthermore, we highlight new findings on the link between mutations of NHE6 and NHE9 and developmental brain disorders including epilepsy, autism, and attention deficit hyperactivity disorder (ADHD). These studies demonstrate the importance of NHE proteins in maintaining H(+) homeostasis and their intricate roles in the regulation of neuronal function. A better understanding of the mechanisms underlying NHE1, 6, and 9 dysfunctions in epilepsy formation may advance the development of new epilepsy treatment strategies.
Collapse
Affiliation(s)
- Hanshu Zhao
- Department of Neurology, The First Affiliated Hospital of the Harbin Medical University, Harbin, China.,Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Karen E Carney
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Lindsay Falgoust
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jullie W Pan
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational and Clinical Center, Pittsburgh, PA 15213, USA
| | - Zhongling Zhang
- Department of Neurology, The First Affiliated Hospital of the Harbin Medical University, Harbin, China
| |
Collapse
|
37
|
Deranieh RM, Shi Y, Tarsio M, Chen Y, McCaffery JM, Kane PM, Greenberg ML. Perturbation of the Vacuolar ATPase: A NOVEL CONSEQUENCE OF INOSITOL DEPLETION. J Biol Chem 2015; 290:27460-72. [PMID: 26324718 DOI: 10.1074/jbc.m115.683706] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Indexed: 11/06/2022] Open
Abstract
Depletion of inositol has profound effects on cell function and has been implicated in the therapeutic effects of drugs used to treat epilepsy and bipolar disorder. We have previously shown that the anticonvulsant drug valproate (VPA) depletes inositol by inhibiting myo-inositol-3-phosphate synthase, the enzyme that catalyzes the first and rate-limiting step of inositol biosynthesis. To elucidate the cellular consequences of inositol depletion, we screened the yeast deletion collection for VPA-sensitive mutants and identified mutants in vacuolar sorting and the vacuolar ATPase (V-ATPase). Inositol depletion caused by starvation of ino1Δ cells perturbed the vacuolar structure and decreased V-ATPase activity and proton pumping in isolated vacuolar vesicles. VPA compromised the dynamics of phosphatidylinositol 3,5-bisphosphate (PI3,5P2) and greatly reduced V-ATPase proton transport in inositol-deprived wild-type cells. Osmotic stress, known to increase PI3,5P2 levels, did not restore PI3,5P2 homeostasis nor did it induce vacuolar fragmentation in VPA-treated cells, suggesting that perturbation of the V-ATPase is a consequence of altered PI3,5P2 homeostasis under inositol-limiting conditions. This study is the first to demonstrate that inositol depletion caused by starvation of an inositol synthesis mutant or by the inositol-depleting drug VPA leads to perturbation of the V-ATPase.
Collapse
Affiliation(s)
- Rania M Deranieh
- From the Department of Biological Sciences, Wayne State University, Detroit, Michigan 48202
| | - Yihui Shi
- From the Department of Biological Sciences, Wayne State University, Detroit, Michigan 48202
| | - Maureen Tarsio
- the Department of Biochemistry and Molecular Biology, State University of New York Upstate Medical University, Syracuse, New York 13210, and
| | - Yan Chen
- From the Department of Biological Sciences, Wayne State University, Detroit, Michigan 48202
| | - J Michael McCaffery
- the Integrated Imaging Center, Department of Biology, Johns Hopkins University, Baltimore, Maryland 21218
| | - Patricia M Kane
- the Department of Biochemistry and Molecular Biology, State University of New York Upstate Medical University, Syracuse, New York 13210, and
| | - Miriam L Greenberg
- From the Department of Biological Sciences, Wayne State University, Detroit, Michigan 48202,
| |
Collapse
|
38
|
Ivannikov MV, Van Remmen H. Sod1 gene ablation in adult mice leads to physiological changes at the neuromuscular junction similar to changes that occur in old wild-type mice. Free Radic Biol Med 2015; 84:254-262. [PMID: 25841780 PMCID: PMC4762177 DOI: 10.1016/j.freeradbiomed.2015.03.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Revised: 03/16/2015] [Accepted: 03/18/2015] [Indexed: 12/27/2022]
Abstract
Reactive oxygen species (ROS) are believed to be important mediators of muscle atrophy and weakness in aging and many degenerative conditions. However, the mechanisms and physiological processes specifically affected by elevated ROS in neuromuscular units that contribute to muscle weakness during aging are not well defined. Here we investigate the effects of chronic oxidative stress on neurotransmission and excitation-contraction (EC) coupling mechanisms in the levator auris longus (LAL) muscle from young (4-8 months) and old (22-28 months) wild-type mice and young adult Cu-Zn superoxide dismutase 1 knockout (Sod1(-/-)) mice. The frequency of spontaneous neurotransmitter release and the amplitude of evoked neurotransmitter release in young Sod1(-/-) and old wild-type LAL neuromuscular junctions were significantly reduced from the young wild-type values, and those declines were mirrored by decreases in synaptic vesicle pool size. Presynaptic cytosolic calcium concentration and mitochondrial calcium uptake amplitudes showed substantial increases in stimulated young Sod1(-/-) and old axon terminals. Surprisingly, LAL muscle fibers from old mice showed a greater excitability than fibers from either young wild-type or young Sod1(-/-) LAL. Both evoked excitatory junction potential (EJP) and spontaneous mini EJP amplitudes were considerably higher in LAL muscles from old mice than in fibers from young Sod1(-/-) LAL muscle. Despite a greater excitability, sarcoplasmic calcium influx in both old wild-type and young Sod1(-/-) LAL muscle fibers was significantly less. Sarcoplasmic reticulum calcium levels were also reduced in both old wild-type and young Sod1(-/-) mice, but the difference was not statistically significant in muscle fibers from old wild-type mice. The protein ratio of triad calcium channels RyR1/DHPR was not different in all groups. However, fibers from both young Sod1(-/-) and old mice had substantially elevated levels of protein carbonylation and S-nitrosylation modifications. Overall, our results suggest that young Sod1(-/-) recapitulate many neuromuscular and muscle fiber changes seen in old mice. We also conclude that muscle weakness in old mice might in part be driven by ROS-mediated EC uncoupling, while both EC uncoupling and reduced neurotransmitter release contribute to muscle weakness in Sod1(-/-) mice.
Collapse
Affiliation(s)
- Maxim V Ivannikov
- Department of Neuroscience and Physiology, NYU School of Medicine, 550 First Avenue, New York, NY 10016, USA.
| | - Holly Van Remmen
- Free Radical Biology and Aging Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA.
| |
Collapse
|
39
|
Linares-Clemente P, Rozas JL, Mircheski J, García-Junco-Clemente P, Martínez-López JA, Nieto-González JL, Vázquez ME, Pintado CO, Fernández-Chacón R. Different dynamin blockers interfere with distinct phases of synaptic endocytosis during stimulation in motoneurones. J Physiol 2015; 593:2867-88. [PMID: 25981717 DOI: 10.1113/jp270112] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Accepted: 04/29/2015] [Indexed: 01/05/2023] Open
Abstract
KEY POINTS Neurotransmitter release requires a tight coupling between synaptic vesicle exocytosis and endocytosis with dynamin being a key protein in that process. We used imaging techniques to examine the time course of endocytosis at mouse motor nerve terminals expressing synaptopHluorin, a genetically encoded reporter of the synaptic vesicle cycle. We separated two sequential phases of endocytosis taking place during the stimulation train: early and late endocytosis. Freshly released synaptic vesicle proteins are preferentially retrieved during the early phase, which is very sensitive to dynasore, an inhibitor of dynamin GTPase activity. Synaptic vesicle proteins pre-existing at the plasma membrane before the stimulation are preferentially retrieved during the late phase, which is very sensitive to myristyl trimethyl ammonium bromide (MitMAB), an inhibitor of the dynamin-phospholipid interaction. ABSTRACT Synaptic endocytosis is essential at nerve terminals to maintain neurotransmitter release by exocytosis. Here, at the neuromuscular junction of synaptopHluorin (spH) transgenic mice, we have used imaging to study exo- and endocytosis occurring simultaneously during nerve stimulation. We observed two endocytosis components, which occur sequentially during stimulation. The early component of endocytosis apparently internalizes spH molecules freshly exocytosed. This component was sensitive to dynasore, a blocker of dynamin 1 GTPase activity. In contrast, this early component was resistant to myristyl trimethyl ammonium bromide (MiTMAB), a competitive agent that blocks dynamin binding to phospholipid membranes. The late component of endocytosis is likely to internalize spH molecules that pre-exist at the plasma membrane before stimulation starts. This component was blocked by MiTMAB, perhaps by impairing the binding of dynamin or other key endocytic proteins to phospholipid membranes. Our study suggests the co-existence of two sequential synaptic endocytosis steps taking place during stimulation that are susceptible to pharmacological dissection: an initial step, preferentially sensitive to dynasore, that internalizes vesicular components immediately after they are released, and a MiTMAB-sensitive step that internalizes vesicular components pre-existing at the plasma membrane surface. In addition, we report that post-stimulus endocytosis also has several components with different sensitivities to dynasore and MiTMAB.
Collapse
Affiliation(s)
- Pedro Linares-Clemente
- Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, and CIBERNED, Seville, Spain
| | - José L Rozas
- Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, and CIBERNED, Seville, Spain
| | - Josif Mircheski
- Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, and CIBERNED, Seville, Spain
| | - Pablo García-Junco-Clemente
- Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, and CIBERNED, Seville, Spain
| | - José A Martínez-López
- Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, and CIBERNED, Seville, Spain
| | | | - M Eugenio Vázquez
- Departamento Química Orgánica y Centro Singular de Investigación en Química Biolóxica y Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - C Oscar Pintado
- Centro Producción y Experimentación Animal, Universidad de Sevilla, Seville, Spain
| | - Rafael Fernández-Chacón
- Instituto de Biomedicina de Sevilla, IBiS, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, and CIBERNED, Seville, Spain
| |
Collapse
|
40
|
ATP binding to synaspsin IIa regulates usage and clustering of vesicles in terminals of hippocampal neurons. J Neurosci 2015; 35:985-98. [PMID: 25609616 DOI: 10.1523/jneurosci.0944-14.2015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Synaptic transmission is expensive in terms of its energy demands and was recently shown to decrease the ATP concentration within presynaptic terminals transiently, an observation that we confirm. We hypothesized that, in addition to being an energy source, ATP may modulate the synapsins directly. Synapsins are abundant neuronal proteins that associate with the surface of synaptic vesicles and possess a well defined ATP-binding site of undetermined function. To examine our hypothesis, we produced a mutation (K270Q) in synapsin IIa that prevents ATP binding and reintroduced the mutant into cultured mouse hippocampal neurons devoid of all synapsins. Remarkably, staining for synaptic vesicle markers was enhanced in these neurons compared with neurons expressing wild-type synapsin IIa, suggesting overly efficient clustering of vesicles. In contrast, the mutation completely disrupted the capability of synapsin IIa to slow synaptic depression during sustained 10 Hz stimulation, indicating that it interfered with synapsin-dependent vesicle recruitment. Finally, we found that the K270Q mutation attenuated the phosphorylation of synapsin IIa on a distant PKA/CaMKI consensus site known to be essential for vesicle recruitment. We conclude that ATP binding to synapsin IIa plays a key role in modulating its function and in defining its contribution to hippocampal short-term synaptic plasticity.
Collapse
|
41
|
Zhang JW, Rangan AV. A reduction for spiking integrate-and-fire network dynamics ranging from homogeneity to synchrony. J Comput Neurosci 2015; 38:355-404. [PMID: 25601481 DOI: 10.1007/s10827-014-0543-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 11/29/2014] [Accepted: 12/09/2014] [Indexed: 10/24/2022]
Abstract
In this paper we provide a general methodology for systematically reducing the dynamics of a class of integrate-and-fire networks down to an augmented 4-dimensional system of ordinary-differential-equations. The class of integrate-and-fire networks we focus on are homogeneously-structured, strongly coupled, and fluctuation-driven. Our reduction succeeds where most current firing-rate and population-dynamics models fail because we account for the emergence of 'multiple-firing-events' involving the semi-synchronous firing of many neurons. These multiple-firing-events are largely responsible for the fluctuations generated by the network and, as a result, our reduction faithfully describes many dynamic regimes ranging from homogeneous to synchronous. Our reduction is based on first principles, and provides an analyzable link between the integrate-and-fire network parameters and the relatively low-dimensional dynamics underlying the 4-dimensional augmented ODE.
Collapse
Affiliation(s)
- J W Zhang
- Courant Institute of Mathematical Sciences, New York University, New York, NY, USA
| | | |
Collapse
|
42
|
Rangaraju V, Calloway N, Ryan TA. Activity-driven local ATP synthesis is required for synaptic function. Cell 2014; 156:825-35. [PMID: 24529383 DOI: 10.1016/j.cell.2013.12.042] [Citation(s) in RCA: 559] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 11/01/2013] [Accepted: 12/31/2013] [Indexed: 01/20/2023]
Abstract
Cognitive function is tightly related to metabolic state, but the locus of this control is not well understood. Synapses are thought to present large ATP demands; however, it is unclear how fuel availability and electrical activity impact synaptic ATP levels and how ATP availability controls synaptic function. We developed a quantitative genetically encoded optical reporter of presynaptic ATP, Syn-ATP, and find that electrical activity imposes large metabolic demands that are met via activity-driven control of both glycolysis and mitochondrial function. We discovered that the primary source of activity-driven metabolic demand is the synaptic vesicle cycle. In metabolically intact synapses, activity-driven ATP synthesis is well matched to the energetic needs of synaptic function, which, at steady state, results in ∼10(6) free ATPs per nerve terminal. Despite this large reservoir of ATP, we find that several key aspects of presynaptic function are severely impaired following even brief interruptions in activity-stimulated ATP synthesis.
Collapse
Affiliation(s)
- Vidhya Rangaraju
- Rockefeller/Sloan-Kettering/Weill Cornell Tri-Institutional Training Program in Chemical Biology, New York, NY 10065, USA; Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Nathaniel Calloway
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Timothy A Ryan
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
43
|
Beppu K, Sasaki T, Tanaka KF, Yamanaka A, Fukazawa Y, Shigemoto R, Matsui K. Optogenetic countering of glial acidosis suppresses glial glutamate release and ischemic brain damage. Neuron 2014; 81:314-20. [PMID: 24462096 DOI: 10.1016/j.neuron.2013.11.011] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2013] [Indexed: 12/31/2022]
Abstract
The brain demands high-energy supply and obstruction of blood flow causes rapid deterioration of the healthiness of brain cells. Two major events occur upon ischemia: acidosis and liberation of excess glutamate, which leads to excitotoxicity. However, cellular source of glutamate and its release mechanism upon ischemia remained unknown. Here we show a causal relationship between glial acidosis and neuronal excitotoxicity. As the major cation that flows through channelrhodopsin-2 (ChR2) is proton, this could be regarded as an optogenetic tool for instant intracellular acidification. Optical activation of ChR2 expressed in glial cells led to glial acidification and to release of glutamate. On the other hand, glial alkalization via optogenetic activation of a proton pump, archaerhodopsin (ArchT), led to cessation of glutamate release and to the relief of ischemic brain damage in vivo. Our results suggest that controlling glial pH may be an effective therapeutic strategy for intervention of ischemic brain damage.
Collapse
Affiliation(s)
- Kaoru Beppu
- Division of Cerebral Structure, National Institute for Physiological Sciences, Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan; Department of Physiological Sciences, Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan
| | - Takuya Sasaki
- Division of Cerebral Structure, National Institute for Physiological Sciences, Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan
| | - Kenji F Tanaka
- Department of Neuropsychiatry, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
| | - Yugo Fukazawa
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Ryuichi Shigemoto
- Division of Cerebral Structure, National Institute for Physiological Sciences, Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan; Department of Physiological Sciences, Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan; IST Austria, 3400 Klosterneuburg, Austria
| | - Ko Matsui
- Division of Cerebral Structure, National Institute for Physiological Sciences, Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan; Department of Physiological Sciences, Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan; Division of Interdisciplinary Medical Science, Center for Neuroscience, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan.
| |
Collapse
|
44
|
Abstract
Synaptic vesicle recycling is one of the best-studied cellular pathways. Many of the proteins involved are known, and their interactions are becoming increasingly clear. However, as for many other pathways, it is still difficult to understand synaptic vesicle recycling as a whole. While it is generally possible to point out how synaptic reactions take place, it is not always easy to understand what triggers or controls them. Also, it is often difficult to understand how the availability of the reaction partners is controlled: how the reaction partners manage to find each other in the right place, at the right time. I present here an overview of synaptic vesicle recycling, discussing the mechanisms that trigger different reactions, and those that ensure the availability of reaction partners. A central argument is that synaptic vesicles bind soluble cofactor proteins, with low affinity, and thus control their availability in the synapse, forming a buffer for cofactor proteins. The availability of cofactor proteins, in turn, regulates the different synaptic reactions. Similar mechanisms, in which one of the reaction partners buffers another, may apply to many other processes, from the biogenesis to the degradation of the synaptic vesicle.
Collapse
Affiliation(s)
- Silvio O Rizzoli
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen European Neuroscience Institute, Göttingen, Germany
| |
Collapse
|
45
|
Proton pump inhibition increases rapid eye movement sleep in the rat. BIOMED RESEARCH INTERNATIONAL 2014; 2014:162314. [PMID: 24701564 PMCID: PMC3950396 DOI: 10.1155/2014/162314] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 01/08/2014] [Accepted: 01/14/2014] [Indexed: 12/14/2022]
Abstract
Increased bodily CO2 concentration alters cellular pH as well as sleep. The proton pump, which plays an important role in the homeostatic regulation of cellular pH, therefore, may modulate sleep. We investigated the effects of the proton pump inhibitor “lansoprazole” on sleep-wakefulness. Male Wistar rats were surgically prepared for chronic polysomnographic recordings. Two different doses of lansoprazole (low: 1 mg/kg; high: 10 mg/kg) were injected intraperitoneally in the same animal (n = 7) and sleep-wakefulness was recorded for 6 hrs. The changes in sleep-wakefulness were compared statistically. Percent REM sleep amount in the vehicle and lansoprazole low dose groups was 9.26 ± 1.03 and 9.09 ± 0.54, respectively, which increased significantly in the lansoprazole high dose group by 31.75% (from vehicle) and 34.21% (from low dose). Also, REM sleep episode numbers significantly increased in lansoprazole high dose group. Further, the sodium-hydrogen exchanger blocker “amiloride” (10 mg/kg; i.p.) (n = 5) did not alter sleep-wake architecture. Our results suggest that the proton pump plays an important role in REM sleep modulation and supports our view that REM sleep might act as a sentinel to help maintain normal CO2 level for unperturbed sleep.
Collapse
|
46
|
Urbano FJ, Lino NG, González-Inchauspe CMF, González LE, Colettis N, Vattino LG, Wunsch AM, Wemmie JA, Uchitel OD. Acid-sensing ion channels 1a (ASIC1a) inhibit neuromuscular transmission in female mice. Am J Physiol Cell Physiol 2013; 306:C396-406. [PMID: 24336653 PMCID: PMC3919981 DOI: 10.1152/ajpcell.00301.2013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Acid-sensing ion channels (ASIC) open in response to extracellular acidosis. ASIC1a, a particular subtype of these channels, has been described to have a postsynaptic distribution in the brain, being involved not only in ischemia and epilepsy, but also in fear and psychiatric pathologies. High-frequency stimulation of skeletal motor nerve terminals (MNTs) can induce presynaptic pH changes in combination with an acidification of the synaptic cleft, known to contribute to muscle fatigue. Here, we studied the role of ASIC1a channels on neuromuscular transmission. We combined a behavioral wire hanging test with electrophysiology, pharmacological, and immunofluorescence techniques to compare wild-type and ASIC1a lacking mice (ASIC1a −/− knockout). Our results showed that 1) ASIC1a −/− female mice were weaker than wild type, presenting shorter times during the wire hanging test; 2) spontaneous neurotransmitter release was reduced by ASIC1a activation, suggesting a presynaptic location of these channels at individual MNTs; 3) ASIC1a-mediated effects were emulated by extracellular local application of acid saline solutions (pH = 6.0; HEPES/MES-based solution); and 4) immunofluorescence techniques revealed the presence of ASIC1a antigens on MNTs. These results suggest that ASIC1a channels might be involved in controlling neuromuscular transmission, muscle contraction and fatigue in female mice.
Collapse
Affiliation(s)
- Francisco J Urbano
- Laboratorio de Fisiología y Biología Molecular (LFBM), Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE, UBA-CONICET), Intendente Güiraldes 2160, Ciudad Universitaria, Buenos Aires, Argentina; and
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Rahmati N, Kunzelmann K, Xu J, Barone S, Sirianant L, De Zeeuw CI, Soleimani M. Slc26a11 is prominently expressed in the brain and functions as a chloride channel: expression in Purkinje cells and stimulation of V H⁺-ATPase. Pflugers Arch 2013; 465:1583-97. [PMID: 23733100 PMCID: PMC11708839 DOI: 10.1007/s00424-013-1300-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/13/2013] [Accepted: 05/14/2013] [Indexed: 02/07/2023]
Abstract
SLC26A11 (human)/Slc26a11 (mouse), also known as kidney brain anion transporter (KBAT), is a member of the SLC26 anion transporter family and shows abundant mRNA expression in the brain. However, its exact cellular distribution and subcellular localization in the brain and its functional identity and possible physiological roles remain unknown. Expression and immunostaining studies demonstrated that Slc26a11 is abundantly expressed in the cerebellum, with a predominant expression in Purkinje cells. Lower expression levels were detected in hippocampus, olfactory bulb, cerebral cortex, and subcortical structures. Patch clamp studies in HEK293 cells transfected with mouse cDNA demonstrated that Slc26a11 can function as a chloride channel that is active under basal conditions and is not regulated by calcium, forskolin, or co-expression with cystic fibrosis transmembrane regulator. Single and double immunofluorescent labeling studies demonstrated the localization of vacuolar (V) H⁺-ATPase and Slc26a11 (KBAT) in the plasma membrane in Purkinje cells. Functional studies in HEK293 cells indicated that transfection with Slc26a11 stimulated acid transport via endogenous V H⁺-ATPase. We conclude that Slc26a11 (KBAT) is prominently distributed in output neurons of various subcortical and cortical structures in the central nervous system, with specific expression in Purkinje cells and that it may operate as a chloride channel regulating acid translocation by H⁺-ATPase across the plasma membrane and in intracellular compartments.
Collapse
Affiliation(s)
- Negah Rahmati
- Department of Neuroscience, Erasmus University, Rotterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
48
|
Sakai H, Li G, Hino Y, Moriura Y, Kawawaki J, Sawada M, Kuno M. Increases in intracellular pH facilitate endocytosis and decrease availability of voltage-gated proton channels in osteoclasts and microglia. J Physiol 2013; 591:5851-66. [PMID: 24081153 DOI: 10.1113/jphysiol.2013.263558] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Voltage-gated proton channels (H(+) channels) are highly proton-selective transmembrane pathways. Although the primary determinants for activation are the pH and voltage gradients across the membrane, the current amplitudes fluctuate often when these gradients are constant. The aim of this study was to investigate the role of the intracellular pH (pHi) in regulating the availability of H(+) channels in osteoclasts and microglia. In whole-cell clamp recordings, the pHi was elevated after exposure to NH4Cl and returned to the control level after washout. However, the H(+) channel conductance did not recover fully when the exposure was prolonged (>5 min). Similar results were observed in osteoclasts and microglia, but not in COS7 cells expressing a murine H(+) channel gene (mVSOP). As other electrophysiological properties, like the gating kinetics and voltage dependence for activation, were unchanged, the decreases in the H(+) channel conductance were probably due to the decreases in H(+) channels available at the plasma membrane. The decreases in the H(+) channel conductances were accompanied by reductions in the cell capacitance. Exposure to NH4Cl increased the uptake of the endocytosis marker FM1-43, substantiating the idea that pHi increases facilitated endocytosis. In osteoclasts, whose plasma membrane expresses V-ATPases and H(+) channels, pHi increases by these H(+)-transferring molecules in part facilitated endocytosis. The endocytosis and decreases in the H(+) channel conductance were reduced by dynasore, a dynamin blocker. These results suggest that pHi increases in osteoclasts and microglia decrease the numbers of H(+) channels available at the plasma membrane through facilitation of dynamin-dependent endocytosis.
Collapse
Affiliation(s)
- Hiromu Sakai
- M. Kuno: Department of Physiology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan.
| | | | | | | | | | | | | |
Collapse
|
49
|
Caldwell L, Harries P, Sydlik S, Schwiening CJ. Presynaptic pH and vesicle fusion in Drosophila larvae neurones. Synapse 2013; 67:729-40. [PMID: 23649934 PMCID: PMC4282566 DOI: 10.1002/syn.21678] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 04/22/2013] [Indexed: 11/11/2022]
Abstract
Both intracellular pH (pHi) and synaptic cleft pH change during neuronal activity yet little is known about how these pH shifts might affect synaptic transmission by influencing vesicle fusion. To address this we imaged pH- and Ca2+-sensitive fluorescent indicators (HPTS, Oregon green) in boutons at neuromuscular junctions. Electrical stimulation of motor nerves evoked presynaptic Ca2+i rises and pHi falls (∼0.1 pH units) followed by recovery of both Ca2+i and pHi. The plasma-membrane calcium ATPase (PMCA) inhibitor, 5(6)-carboxyeosin diacetate, slowed both the calcium recovery and the acidification. To investigate a possible calcium-independent role for the pHi shifts in modulating vesicle fusion we recorded post-synaptic miniature end-plate potential (mEPP) and current (mEPC) frequency in Ca2+-free solution. Acidification by propionate superfusion, NH4+ withdrawal, or the inhibition of acid extrusion on the Na+/H+ exchanger (NHE) induced a rise in miniature frequency. Furthermore, the inhibition of acid extrusion enhanced the rise induced by propionate addition and NH4+ removal. In the presence of NH4+, 10 out of 23 cells showed, after a delay, one or more rises in miniature frequency. These findings suggest that Ca2+-dependent pHi shifts, caused by the PMCA and regulated by NHE, may stimulate vesicle release. Furthermore, in the presence of membrane permeant buffers, exocytosed acid or its equivalents may enhance release through positive feedback. This hitherto neglected pH signalling, and the potential feedback role of vesicular acid, could explain some important neuronal excitability changes associated with altered pH and its buffering. Synapse 67:729–740, 2013.
Collapse
Affiliation(s)
- Lesley Caldwell
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom
| | | | | | | |
Collapse
|
50
|
Corena-McLeod M, Walss-Bass C, Oliveros A, Gordillo Villegas A, Ceballos C, Charlesworth CM, Madden B, Linser PJ, Van Ekeris L, Smith K, Richelson E. New model of action for mood stabilizers: phosphoproteome from rat pre-frontal cortex synaptoneurosomal preparations. PLoS One 2013; 8:e52147. [PMID: 23690912 PMCID: PMC3653908 DOI: 10.1371/journal.pone.0052147] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 11/09/2012] [Indexed: 01/11/2023] Open
Abstract
Background Mitochondrial short and long-range movements are necessary to generate the energy needed for synaptic signaling and plasticity. Therefore, an effective mechanism to transport and anchor mitochondria to pre- and post-synaptic terminals is as important as functional mitochondria in neuronal firing. Mitochondrial movement range is regulated by phosphorylation of cytoskeletal and motor proteins in addition to changes in mitochondrial membrane potential. Movement direction is regulated by serotonin and dopamine levels. However, data on mitochondrial movement defects and their involvement in defective signaling and neuroplasticity in relationship with mood disorders is scarce. We have previously reported the effects of lithium, valproate and a new antipsychotic, paliperidone on protein expression levels at the synaptic level. Hypothesis Mitochondrial function defects have recently been implicated in schizophrenia and bipolar disorder. We postulate that mood stabilizer treatment has a profound effect on mitochondrial function, synaptic plasticity, mitochondrial migration and direction of movement. Methods Synaptoneurosomal preparations from rat pre-frontal cortex were obtained after 28 daily intraperitoneal injections of lithium, valproate and paliperidone. Phosphorylated proteins were identified using 2D-DIGE and nano LC-ESI tandem mass spectrometry. Results Lithium, valproate and paliperidone had a substantial and common effect on the phosphorylation state of specific actin, tubulin and myosin isoforms as well as other proteins associated with neurofilaments. Furthermore, different subunits from complex III and V of the electron transfer chain were heavily phosphorylated by treatment with these drugs indicating selective phosphorylation. Conclusions Mood stabilizers have an effect on mitochondrial function, mitochondrial movement and the direction of this movement. The implications of these findings will contribute to novel insights regarding clinical treatment and the mode of action of these drugs.
Collapse
|