51
|
Barak P, Parekh AB. Signaling through Ca 2+ Microdomains from Store-Operated CRAC Channels. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035097. [PMID: 31358516 DOI: 10.1101/cshperspect.a035097] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Calcium (Ca2+) ion microdomains are subcellular regions of high Ca2+ concentration that develop rapidly near open Ca2+ channels in the plasma membrane or internal stores and generate local regions of high Ca2+ concentration. These microdomains are remarkably versatile in that they activate a range of responses that differ enormously in both their temporal and spatial profile. In this review, we describe how Ca2+ microdomains generated by store-operated calcium channels, a widespread and conserved Ca2+ entry pathway, stimulate different signaling pathways, and how the spatial extent of a Ca2+ microdomain can be influenced by Ca2+ ATPase pumps.
Collapse
Affiliation(s)
- Pradeep Barak
- Department of Physiology, Anatomy, and Genetics, Oxford University, Oxford OX1 3PT, United Kingdom
| | - Anant B Parekh
- Department of Physiology, Anatomy, and Genetics, Oxford University, Oxford OX1 3PT, United Kingdom
| |
Collapse
|
52
|
Shah SI, Ullah G. The Function of Mitochondrial Calcium Uniporter at the Whole-Cell and Single Mitochondrion Levels in WT, MICU1 KO, and MICU2 KO Cells. Cells 2020; 9:E1520. [PMID: 32580385 PMCID: PMC7349584 DOI: 10.3390/cells9061520] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 01/05/2023] Open
Abstract
Mitochondrial Ca2+ ([Ca2+]M) uptake through its Ca2+ uniporter (MCU) is central to many cell functions such as bioenergetics, spatiotemporal organization of Ca2+ signals, and apoptosis. MCU activity is regulated by several intrinsic proteins including MICU1, MICU2, and EMRE. While significant details about the role of MICU1, MICU2, and EMRE in MCU function have emerged recently, a key challenge for the future experiments is to investigate how these regulatory proteins modulate mitochondrial Ca2+ influx through MCU in intact cells under pathophysiological conditions. This is further complicated by the fact that several variables affecting MCU function change dynamically as cell functions. To overcome this void, we develop a data-driven model that closely replicates the behavior of MCU under a wide range of cytosolic Ca2+ ([Ca2+]C), [Ca2+]M, and mitochondrial membrane potential values in WT, MICU1 knockout (KO), and MICU2 KO cells at the single mitochondrion and whole-cell levels. The model is extended to investigate how MICU1 or MICU2 KO affect mitochondrial function. Moreover, we show how Ca2+ buffering proteins, the separation between mitochondrion and Ca2+-releasing stores, and the duration of opening of Ca2+-releasing channels affect mitochondrial function under different conditions. Finally, we demonstrate an easy extension of the model to single channel function of MCU.
Collapse
Affiliation(s)
| | - Ghanim Ullah
- Department of Physics, University of South Florida, Tampa, FL 33647, USA;
| |
Collapse
|
53
|
Chubinskiy-Nadezhdin VI, Vasileva VY, Negulyaev YA, Morachevskaya EA. Functional clustering and coupling of ion channels in cellular mechanosensing is independent on lipid raft integrity in plasma membrane. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118764. [PMID: 32479769 DOI: 10.1016/j.bbamcr.2020.118764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/23/2020] [Accepted: 05/27/2020] [Indexed: 10/24/2022]
|
54
|
Functional Postnatal Maturation of the Medial Olivocochlear Efferent-Outer Hair Cell Synapse. J Neurosci 2020; 40:4842-4857. [PMID: 32430293 DOI: 10.1523/jneurosci.2409-19.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 04/19/2020] [Accepted: 05/11/2020] [Indexed: 01/07/2023] Open
Abstract
The organ of Corti, the auditory mammalian sensory epithelium, contains two types of mechanotransducer cells, inner hair cells (IHCs) and outer hair cells (OHCs). IHCs are involved in conveying acoustic stimuli to the CNS, while OHCs are implicated in the fine tuning and amplification of sounds. OHCs are innervated by medial olivocochlear (MOC) cholinergic efferent fibers. The functional characteristics of the MOC-OHC synapse during maturation were assessed by electrophysiological and pharmacological methods in mouse organs of Corti at postnatal day 11 (P11)-P13, hearing onset in altricial rodents, and at P20-P22 when the OHCs are morphologically and functionally mature. Synaptic currents were recorded in whole-cell voltage-clamped OHCs while electrically stimulating the MOC fibers. A progressive increase in the number of functional MOC-OHC synapses, as well as in their strength and efficacy, was observed between P11-13 and P20-22. At hearing onset, the MOC-OHC synapse presented facilitation during MOC fibers high-frequency stimulation that disappeared at mature stages. In addition, important changes were found in the VGCC that are coupled to transmitter release. Ca2+ flowing in through L-type VGCCs contribute to trigger ACh release together with P/Q- and R-type VGCCs at P11-P13, but not at P20-P22. Interestingly, N-type VGCCs were found to be involved in this process at P20-P22, but not at hearing onset. Moreover, the degree of compartmentalization of calcium channels with respect to BK channels and presynaptic release components significantly increased from P11-P13 to P20-P22. These results suggest that the MOC-OHC synapse is immature at the onset of hearing.SIGNIFICANCE STATEMENT The functional expression of both VGCCs and BK channels, as well as their localization with respect to the presynaptic components involved in transmitter release, are key elements in determining synaptic efficacy. In this work, we show dynamic changes in the expression of VGCCs and Ca2+-dependent BK K+ channels coupled to ACh release at the MOC-OHC synapse and their shift in compartmentalization during postnatal maturation. These processes most likely set the short-term plasticity pattern and reliability of the MOC-OHC synapse on high-frequency activity.
Collapse
|
55
|
Omar-Hmeadi M, Lund PE, Gandasi NR, Tengholm A, Barg S. Paracrine control of α-cell glucagon exocytosis is compromised in human type-2 diabetes. Nat Commun 2020; 11:1896. [PMID: 32312960 PMCID: PMC7171169 DOI: 10.1038/s41467-020-15717-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/23/2020] [Indexed: 01/05/2023] Open
Abstract
Glucagon is released from pancreatic α-cells to activate pathways that raise blood glucose. Its secretion is regulated by α-cell-intrinsic glucose sensing and paracrine control through insulin and somatostatin. To understand the inadequately high glucagon levels that contribute to hyperglycemia in type-2 diabetes (T2D), we analyzed granule behavior, exocytosis and membrane excitability in α-cells of 68 non-diabetic and 21 T2D human donors. We report that exocytosis is moderately reduced in α-cells of T2D donors, without changes in voltage-dependent ion currents or granule trafficking. Dispersed α-cells have a non-physiological V-shaped dose response to glucose, with maximal exocytosis at hyperglycemia. Within intact islets, hyperglycemia instead inhibits α-cell exocytosis, but not in T2D or when paracrine inhibition by insulin or somatostatin is blocked. Surface expression of somatostatin-receptor-2 is reduced in T2D, suggesting a mechanism for the observed somatostatin resistance. Thus, elevated glucagon in human T2D may reflect α-cell insensitivity to paracrine inhibition at hyperglycemia. Glucagon is elevated Type-2 diabetes, which contributes to poor glucose control in patients with the disease. Here the authors report that secretion of the hormone is controlled by paracrine inhibition, and that resistance of α-cells to somatostatin can explain hyperglucagonemia in type-2 diabetes.
Collapse
Affiliation(s)
- Muhmmad Omar-Hmeadi
- Medical Cell Biology, Uppsala University, Box 571, BMC, 751 23, Uppsala, Sweden
| | - Per-Eric Lund
- Medical Cell Biology, Uppsala University, Box 571, BMC, 751 23, Uppsala, Sweden
| | - Nikhil R Gandasi
- Medical Cell Biology, Uppsala University, Box 571, BMC, 751 23, Uppsala, Sweden
| | - Anders Tengholm
- Medical Cell Biology, Uppsala University, Box 571, BMC, 751 23, Uppsala, Sweden
| | - Sebastian Barg
- Medical Cell Biology, Uppsala University, Box 571, BMC, 751 23, Uppsala, Sweden.
| |
Collapse
|
56
|
Zhang Y, Wang J, Xing S, Li L, Zhao S, Zhu W, Liang K, Liu Y, Chen L. Mitochondria determine the sequential propagation of the calcium macrodomains revealed by the super-resolution calcium lantern imaging. SCIENCE CHINA-LIFE SCIENCES 2020; 63:1543-1551. [PMID: 32279282 DOI: 10.1007/s11427-019-1659-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 02/27/2020] [Indexed: 01/02/2023]
Abstract
Despite the wide application of super-resolution (SR) microscopy in biological studies of cells, the technology is rarely used to monitor functional changes in live cells. By combining fast spinning disc-confocal structured illumination microscopy (SD-SIM) with loading of cytosolic fluorescent Ca2+ indicators, we have developed an SR method for visualization of regional Ca2+ dynamics and related cellular organelle morphology and dynamics, termed SR calcium lantern imaging. In COS-7 cells stimulated with ATP, we have identified various calcium macrodomains characterized by different types of Ca2+ release from endoplasmic reticulum (ER) stores. Finally, we demonstrated various roles of mitochondria in mediating calcium signals from different sources; while mitochondria can globally potentiate the Ca2+ entry associated with store release, mitochondria also locally control Ca2+ release from the neighboring ER stores and assist in their refilling processes.
Collapse
Affiliation(s)
- Yulin Zhang
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, 100871, China
| | - Jianyong Wang
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, 100871, China
| | - Shijia Xing
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, 100871, China
| | - Liuju Li
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, 100871, China
| | - Shiqun Zhao
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, 100871, China
| | - Wenzhen Zhu
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, 100871, China
| | - Kuo Liang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yanmei Liu
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, 100871, China.,Institute for Brain Research and Rehabilitation (IBRR), Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, 200062, China
| | - Liangyi Chen
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, 100871, China. .,PKU-IDG/McGovern Institute for Brain Research, Beijing, 100871, China.
| |
Collapse
|
57
|
Zhang H, Liu Y, Men H, Zhang F, Zhang H. LRRCA8A and ANO1 contribute to serum-induced VRAC in a Ca 2+-dependent manners. J Pharmacol Sci 2020; 143:176-181. [PMID: 32386905 DOI: 10.1016/j.jphs.2020.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 03/05/2020] [Accepted: 03/23/2020] [Indexed: 10/24/2022] Open
Abstract
The volume-regulated anion channel (VRAC) plays a central role in maintaining cell volume in response to osmotic stress. Leucine-rich repeat-containing 8A (LRRC8A) was recently identified as an essential component of VRAC although other Cl- channels were also suggested to contribute to VRAC. VRAC is activated when a cell is challenged with a hypotonic environment or even in isotonic conditions challenged with different stimuli. It is not clear how VRAC is activated and whether activation of VRAC in hypotonic and isotonic conditions share the same mechanism. In this present study, we investigated relative contribution of LRRC8A and anoctamin 1(ANO1) to VRAC currents activated by fetal bovine serum (FBS) in isotonic condition, and studied the role of intracellular Ca2+ in this activation. We used CRISPR/Cas9 gene editing approach, electrophysiology, and pharmacology approaches to show that VRAC currents induced by FBS is mostly mediated by LRRC8A in HEK293 cells, but also with significant contribution from ANO1. FBS induces Ca2+ transients and these Ca2+ signals are required for the activation of VRAC by serum. These findings will help to further understand the mechanism in activation of VRAC.
Collapse
Affiliation(s)
- Huiran Zhang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, Hebei, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, Shijiazhuang, Hebei, China; Department of Biopharmacy, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yani Liu
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, Hebei, China; Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, Shandong, China
| | - Hongchao Men
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, Hebei, China
| | - Fan Zhang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, Hebei, China
| | - Hailin Zhang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, Hebei, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province, Shijiazhuang, Hebei, China.
| |
Collapse
|
58
|
de Diego AMG, Ortega-Cruz D, García AG. Disruption of Exocytosis in Sympathoadrenal Chromaffin Cells from Mouse Models of Neurodegenerative Diseases. Int J Mol Sci 2020; 21:ijms21061946. [PMID: 32178443 PMCID: PMC7139653 DOI: 10.3390/ijms21061946] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/05/2020] [Accepted: 03/10/2020] [Indexed: 11/16/2022] Open
Abstract
Synaptic disruption and altered neurotransmitter release occurs in the brains of patients and in murine models of neurodegenerative diseases (NDDs). During the last few years, evidence has accumulated suggesting that the sympathoadrenal axis is also affected as disease progresses. Here, we review a few studies done in adrenal medullary chromaffin cells (CCs), that are considered as the amplifying arm of the sympathetic nervous system; the sudden fast exocytotic release of their catecholamines—stored in noradrenergic and adrenergic cells—plays a fundamental role in the stress fight-or-flight response. Bulk exocytosis and the fine kinetics of single-vesicle exocytotic events have been studied in mouse models carrying a mutation linked to NDDs. For instance, in R6/1 mouse models of Huntington’s disease (HD), mutated huntingtin is overexpressed in CCs; this causes decreased quantal secretion, smaller quantal size and faster kinetics of the exocytotic fusion pore, pore expansion, and closure. This was accompanied by decreased sodium current, decreased acetylcholine-evoked action potentials, and attenuated [Ca2+]c transients with faster Ca2+ clearance. In the SOD1G93A mouse model of amyotrophic lateral sclerosis (ALS), CCs exhibited secretory single-vesicle spikes with a slower release rate but higher exocytosis. Finally, in the APP/PS1 mouse model of Alzheimer’s disease (AD), the stabilization, expansion, and closure of the fusion pore was faster, but the secretion was attenuated. Additionally, α-synuclein that is associated with Parkinson’s disease (PD) decreases exocytosis and promotes fusion pore dilation in adrenal CCs. Furthermore, Huntington-associated protein 1 (HAP1) interacts with the huntingtin that, when mutated, causes Huntington’s disease (HD); HAP1 reduces full fusion exocytosis by affecting vesicle docking and controlling fusion pore stabilization. The alterations described here are consistent with the hypothesis that central alterations undergone in various NDDs are also manifested at the peripheral sympathoadrenal axis to impair the stress fight-or-flight response in patients suffering from those diseases. Such alterations may occur: (i) primarily by the expression of mutated disease proteins in CCs; (ii) secondarily to stress adaptation imposed by disease progression and the limitations of patient autonomy.
Collapse
Affiliation(s)
- Antonio M. G. de Diego
- Instituto Teófilo Hernando, Departamento. de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (A.M.G.d.D.); (D.O.-C.)
- Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, 28006 Madrid, Spain
| | - Diana Ortega-Cruz
- Instituto Teófilo Hernando, Departamento. de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (A.M.G.d.D.); (D.O.-C.)
- Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, 28006 Madrid, Spain
| | - Antonio G. García
- Instituto Teófilo Hernando, Departamento. de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (A.M.G.d.D.); (D.O.-C.)
- Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, 28006 Madrid, Spain
- Correspondence: ; Tel.: +34-91-497-5384
| |
Collapse
|
59
|
Glasgow SD, Ruthazer ES, Kennedy TE. Guiding synaptic plasticity: Novel roles for netrin-1 in synaptic plasticity and memory formation in the adult brain. J Physiol 2020; 599:493-505. [PMID: 32017127 DOI: 10.1113/jp278704] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/14/2020] [Indexed: 12/12/2022] Open
Abstract
Adult neural plasticity engages mechanisms that change synapse structure and function, yet many of the underlying events bear a striking similarity to processes that occur during the initial establishment of neural circuits during development. It is a long-standing hypothesis that the molecular mechanisms critical for neural development may also regulate synaptic plasticity related to learning and memory in adults. Netrins were initially described as chemoattractant guidance cues that direct cell and axon migration during embryonic development, yet they continue to be expressed by neurons in the adult brain. Recent findings have identified roles for netrin-1 in synaptogenesis during postnatal maturation, and in synaptic plasticity in the adult mammalian brain, regulating AMPA glutamate receptor trafficking at excitatory synapses. These findings provide an example of a conserved developmental guidance cue that is expressed by neurons in the adult brain and functions as a key regulator of activity-dependent synaptic plasticity. Notably, in humans, genetic polymorphisms in netrin-1 and its receptors have been linked to neurodevelopmental and neurodegenerative disorders. The molecular mechanisms associated with the synaptic function of netrin-1 therefore present new therapeutic targets for neuropathologies associated with memory dysfunction. Here, we summarize recent findings that link netrin-1 signalling to synaptic plasticity, and discuss the implications of these discoveries for the neurobiological basis of memory consolidation.
Collapse
Affiliation(s)
- Stephen D Glasgow
- Department of Neurology & Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Edward S Ruthazer
- Department of Neurology & Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Timothy E Kennedy
- Department of Neurology & Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, H3A 2B4, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, H3A 0C7, Canada
| |
Collapse
|
60
|
Moser T, Grabner CP, Schmitz F. Sensory Processing at Ribbon Synapses in the Retina and the Cochlea. Physiol Rev 2020; 100:103-144. [DOI: 10.1152/physrev.00026.2018] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In recent years, sensory neuroscientists have made major efforts to dissect the structure and function of ribbon synapses which process sensory information in the eye and ear. This review aims to summarize our current understanding of two key aspects of ribbon synapses: 1) their mechanisms of exocytosis and endocytosis and 2) their molecular anatomy and physiology. Our comparison of ribbon synapses in the cochlea and the retina reveals convergent signaling mechanisms, as well as divergent strategies in different sensory systems.
Collapse
Affiliation(s)
- Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany; Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, Göttingen, Germany; Synaptic Nanophysiology Group, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany; and Institute for Anatomy and Cell Biology, Department of Neuroanatomy, Medical School, Saarland University, Homburg, Germany
| | - Chad P. Grabner
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany; Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, Göttingen, Germany; Synaptic Nanophysiology Group, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany; and Institute for Anatomy and Cell Biology, Department of Neuroanatomy, Medical School, Saarland University, Homburg, Germany
| | - Frank Schmitz
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany; Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, Göttingen, Germany; Synaptic Nanophysiology Group, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany; and Institute for Anatomy and Cell Biology, Department of Neuroanatomy, Medical School, Saarland University, Homburg, Germany
| |
Collapse
|
61
|
Wesseling JF. Considerations for Measuring Activity-Dependence of Recruitment of Synaptic Vesicles to the Readily Releasable Pool. Front Synaptic Neurosci 2019; 11:32. [PMID: 31824292 PMCID: PMC6879548 DOI: 10.3389/fnsyn.2019.00032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 11/06/2019] [Indexed: 11/29/2022] Open
Abstract
The connection strength of most chemical synapses changes dynamically during normal use as a function of the recent history of activity. The phenomenon is known as short-term synaptic plasticity or synaptic dynamics, and is thought to be involved in processing and filtering information as it is transmitted across the synaptic cleft. Multiple presynaptic mechanisms have been implicated, but large gaps remain in our understanding of how the mechanisms are modulated and how they interact. One important factor is the timing of recruitment of synaptic vesicles to a readily-releasable pool. A number of studies have concluded that activity and/or residual Ca2+ can accelerate the mechanism, but alternative explanations for some of the evidence have emerged. Here I review the methodology that we have developed for isolating the recruitment and the dependence on activity from other kinds of mechanisms that are activated concurrently.
Collapse
Affiliation(s)
- John F Wesseling
- CSIC/Instituto de Neurociencias, Universidad Miguel Hernández, Alicante, Spain
| |
Collapse
|
62
|
Johnson SL, Safieddine S, Mustapha M, Marcotti W. Hair Cell Afferent Synapses: Function and Dysfunction. Cold Spring Harb Perspect Med 2019; 9:a033175. [PMID: 30617058 PMCID: PMC6886459 DOI: 10.1101/cshperspect.a033175] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
To provide a meaningful representation of the auditory landscape, mammalian cochlear hair cells are optimized to detect sounds over an incredibly broad range of frequencies and intensities with unparalleled accuracy. This ability is largely conferred by specialized ribbon synapses that continuously transmit acoustic information with high fidelity and sub-millisecond precision to the afferent dendrites of the spiral ganglion neurons. To achieve this extraordinary task, ribbon synapses employ a unique combination of molecules and mechanisms that are tailored to sounds of different frequencies. Here we review the current understanding of how the hair cell's presynaptic machinery and its postsynaptic afferent connections are formed, how they mature, and how their function is adapted for an accurate perception of sound.
Collapse
Affiliation(s)
- Stuart L Johnson
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Saaid Safieddine
- UMRS 1120, Institut Pasteur, Paris, France
- Sorbonne Universités, UPMC Université Paris 06, Complexité du Vivant, Paris, France
| | - Mirna Mustapha
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, United Kingdom
- Department of Otolaryngology-Head & Neck Surgery, Stanford University, Stanford, California 94035
| | - Walter Marcotti
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, United Kingdom
| |
Collapse
|
63
|
Martínez San Segundo P, Terni B, Burgueño J, Monroy X, Dordal A, Merlos M, Llobet A. Outside-in regulation of the readily releasable pool of synaptic vesicles by α2δ-1. FASEB J 2019; 34:1362-1377. [PMID: 31914622 DOI: 10.1096/fj.201901551r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 10/09/2019] [Accepted: 10/23/2019] [Indexed: 12/17/2022]
Abstract
The readily releasable pool (RRP) of synaptic vesicles is a key determinant of phasic neurotransmission. Although the size of the RRP is tightly regulated by intracellular factors, there is little evidence for its modification by extracellular signals. By studying the homogeneous population of synapses present in autaptic microcultures, we show that pregabalin, a prototypical gabapentinoid, decreases the effective RRP size. Simultaneous imaging of presynaptic calcium influx and recording of postsynaptic responses shows that the effect is not related to a reduction of calcium entry. The main cause is the impairment of the functional coupling among N-type calcium channels and the RRP, resembling an increase of intracellular mobile calcium buffers. The ectodomain of neurexin-1α shows a similar action to pregabalin, acting as an endogenous ligand of α2δ-1 that reduces the RRP size without affecting presynaptic calcium influx. The regulatory actions described for pregabalin and the ectodomain of neurexin-1α are mutually exclusive. The overexpression of α2δ-1 enhances the effect of pregabalin and the ectodomain of neurexin-1α on neurotransmission by decreasing their effective concentration. In contrast, knockdown of α2δ-1 causes a profound inhibition of synaptic transmission. These observations prompt to consider α2δ-1 as an outside-in signaling platform that binds exogenous and endogenous cues for regulating the coupling of voltage-gated calcium channels to synaptic vesicles.
Collapse
Affiliation(s)
- Pablo Martínez San Segundo
- Laboratory of Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain.,Department of Pathology and Experimental Therapeutics, School of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Beatrice Terni
- Laboratory of Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain.,Department of Pathology and Experimental Therapeutics, School of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Javier Burgueño
- Department of Pharmacology, Drug Discovery and Preclinical Development, ESTEVE, Barcelona, Spain
| | - Xavier Monroy
- Department of Pharmacology, Drug Discovery and Preclinical Development, ESTEVE, Barcelona, Spain
| | - Albert Dordal
- Department of Pharmacology, Drug Discovery and Preclinical Development, ESTEVE, Barcelona, Spain
| | - Manuel Merlos
- Department of Pharmacology, Drug Discovery and Preclinical Development, ESTEVE, Barcelona, Spain
| | - Artur Llobet
- Laboratory of Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain.,Department of Pathology and Experimental Therapeutics, School of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| |
Collapse
|
64
|
From Local to Global Modeling for Characterizing Calcium Dynamics and Their Effects on Electrical Activity and Exocytosis in Excitable Cells. Int J Mol Sci 2019; 20:ijms20236057. [PMID: 31801305 PMCID: PMC6928823 DOI: 10.3390/ijms20236057] [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: 09/04/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 12/25/2022] Open
Abstract
Electrical activity in neurons and other excitable cells is a result of complex interactions between the system of ion channels, involving both global coupling (e.g., via voltage or bulk cytosolic Ca2+ concentration) of the channels, and local coupling in ion channel complexes (e.g., via local Ca2+ concentration surrounding Ca2+ channels (CaVs), the so-called Ca2+ nanodomains). We recently devised a model of large-conductance BKCa potassium currents, and hence BKCa–CaV complexes controlled locally by CaVs via Ca2+ nanodomains. We showed how different CaV types and BKCa–CaV stoichiometries affect whole-cell electrical behavior. Ca2+ nanodomains are also important for triggering exocytosis of hormone-containing granules, and in this regard, we implemented a strategy to characterize the local interactions between granules and CaVs. In this study, we coupled electrical and exocytosis models respecting the local effects via Ca2+ nanodomains. By simulating scenarios with BKCa–CaV complexes with different stoichiometries in pituitary cells, we achieved two main electrophysiological responses (continuous spiking or bursting) and investigated their effects on the downstream exocytosis process. By varying the number and distance of CaVs coupled with the granules, we found that bursting promotes exocytosis with faster rates than spiking. However, by normalizing to Ca2+ influx, we found that bursting is only slightly more efficient than spiking when CaVs are far away from granules, whereas no difference in efficiency between bursting and spiking is observed with close granule-CaV coupling.
Collapse
|
65
|
Noh J, Chung JM. Modulation of Dopaminergic Neuronal Excitability by Zinc through the Regulation of Calcium-related Channels. Exp Neurobiol 2019; 28:578-592. [PMID: 31698550 PMCID: PMC6844832 DOI: 10.5607/en.2019.28.5.578] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 10/28/2019] [Accepted: 10/28/2019] [Indexed: 11/19/2022] Open
Abstract
Depending on the intracellular buffering of calcium by chelation, zinc has the following two apparent effects on neuronal excitability: enhancement or reduction. Zinc increased tonic activity in the depolarized state when neurons were intracellularly dialyzed with EGTA but attenuated the neuronal activity when BAPTA was used as an intracellular calcium buffer. This suggests that neuronal excitability can be modulated by zinc, depending on the internal calcium buffering capacity. In this study, we elucidated the mechanisms of zinc-mediated alterations in neuronal excitability and determined the effect of calcium-related channels on zinc-mediated alterations in excitability. The zinc-induced augmentation of firing activity was mediated via the inhibition of small-conductance calcium-activated potassium (SK) channels with not only the contribution of voltage-gated L-type calcium channels (VGCCs) and ryanodine receptors (RyRs), but also through the activation of VGCCs via melastatin-like transient receptor potential channels. We suggest that zinc modulates the dopaminergic neuronal activity by regulating not only SK channels as calcium sensors, but also VGCCs or RyRs as calcium sources. Our results suggest that the cytosolic calcium-buffering capacity can tightly regulate zinc-induced neuronal firing patterns and that local calcium-signaling domains can determine the physiological and pathological state of synaptic activity in the dopaminergic system.
Collapse
Affiliation(s)
- Jihyun Noh
- Department of Science Education, Dankook University, Yongin 16890, Korea
| | - Jun-Mo Chung
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
66
|
Nakamura Y. EGTA Can Inhibit Vesicular Release in the Nanodomain of Single Ca 2+ Channels. Front Synaptic Neurosci 2019; 11:26. [PMID: 31632263 PMCID: PMC6779814 DOI: 10.3389/fnsyn.2019.00026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/02/2019] [Indexed: 01/21/2023] Open
Abstract
The exogenous Ca2+ chelator EGTA (ethylene glycol tetraacetic acid) has been widely used to probe the coupling distance between Ca2+ channels and vesicular Ca2+ sensors for neurotransmitter release. Because of its slow forward rate for binding, EGTA is thought to not capture calcium ions in very proximity to a channel, whereas it does capture calcium ions at the remote distance. However, in this study, our reaction diffusion simulations (RDSs) of Ca2+ combined with a release calculation using vesicular sensor models indicate that a high concentration of EGTA decreases Ca2+ and vesicular release in the nanodomain of single channels. We found that a key determinant of the effect of EGTA on neurotransmitter release is the saturation of the vesicular sensor. When the sensor is saturated, the reduction in the Ca2+ concentration by EGTA is masked. By contrast, when the sensor is in a linear range, even a small reduction in Ca2+ by EGTA can decrease vesicular release. In proximity to a channel, the vesicular sensor is often saturated for a long voltage step, but not for a brief Ca2+ influx typically evoked by an action potential. Therefore, when EGTA is used as a diagnostic tool to probe the coupling distance, care must be taken regarding the presynaptic Ca2+ entry duration as well as the property of the vesicular Ca2+ sensor.
Collapse
Affiliation(s)
- Yukihiro Nakamura
- Department of Pharmacology, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
67
|
Carbone E, Borges R, Eiden LE, García AG, Hernández-Cruz A. Chromaffin Cells of the Adrenal Medulla: Physiology, Pharmacology, and Disease. Compr Physiol 2019; 9:1443-1502. [PMID: 31688964 DOI: 10.1002/cphy.c190003] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Chromaffin cells (CCs) of the adrenal gland and the sympathetic nervous system produce the catecholamines (epinephrine and norepinephrine; EPI and NE) needed to coordinate the bodily "fight-or-flight" response to fear, stress, exercise, or conflict. EPI and NE release from CCs is regulated both neurogenically by splanchnic nerve fibers and nonneurogenically by hormones (histamine, corticosteroids, angiotensin, and others) and paracrine messengers [EPI, NE, adenosine triphosphate, opioids, γ-aminobutyric acid (GABA), etc.]. The "stimulus-secretion" coupling of CCs is a Ca2+ -dependent process regulated by Ca2+ entry through voltage-gated Ca2+ channels, Ca2+ pumps, and exchangers and intracellular organelles (RE and mitochondria) and diffusible buffers that provide both Ca2+ -homeostasis and Ca2+ -signaling that ultimately trigger exocytosis. CCs also express Na+ and K+ channels and ionotropic (nAChR and GABAA ) and metabotropic receptors (mACh, PACAP, β-AR, 5-HT, histamine, angiotensin, and others) that make CCs excitable and responsive to autocrine and paracrine stimuli. To maintain high rates of E/NE secretion during stressful conditions, CCs possess a large number of secretory chromaffin granules (CGs) and members of the soluble NSF-attachment receptor complex protein family that allow docking, fusion, and exocytosis of CGs at the cell membrane, and their recycling. This article attempts to provide an updated account of well-established features of the molecular processes regulating CC function, and a survey of the as-yet-unsolved but important questions relating to CC function and dysfunction that have been the subject of intense research over the past 15 years. Examples of CCs as a model system to understand the molecular mechanisms associated with neurodegenerative diseases are also provided. Published 2019. Compr Physiol 9:1443-1502, 2019.
Collapse
Affiliation(s)
- Emilio Carbone
- Laboratory of Cellular and Molecular Neuroscience, Department of Drug Science, N.I.S. Centre, University of Torino, Torino, Italy
| | - Ricardo Borges
- Unidad de Farmacología, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | - Lee E Eiden
- Section on Molecular Neuroscience, National Institute of Mental Health, Bethesda, Maryland, USA
| | - Antonio G García
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Instituto Teófilo Hernando, Universidad Autónoma de Madrid, Madrid, Spain
| | - Arturo Hernández-Cruz
- Departamento de Neurociencia Cognitiva and Laboratorio Nacional de Canalopatías, Instituto de Fisiología Celular, Universidad Nacional Autonoma de México, Ciudad Universitaria, CDMX, México
| |
Collapse
|
68
|
Liu Y, Zhang H, Men H, Du Y, Xiao Z, Zhang F, Huang D, Du X, Gamper N, Zhang H. Volume-regulated Cl - current: contributions of distinct Cl - channels and localized Ca 2+ signals. Am J Physiol Cell Physiol 2019; 317:C466-C480. [PMID: 31242393 DOI: 10.1152/ajpcell.00507.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The swelling-activated chloride current (ICl,swell) is induced when a cell swells and plays a central role in maintaining cell volume in response to osmotic stress. The major contributor of ICl,swell is the volume-regulated anion channel (VRAC). Leucine-rich repeat containing 8A (LRRC8A; SWELL1) was recently identified as an essential component of VRAC, but the mechanisms of VRAC activation are still largely unknown; moreover, other Cl- channels, such as anoctamin 1 (ANO1), were also suggested to contribute to ICl,swell. In this present study, we investigated the roles of LRRC8A and ANO1 in activation of ICl,swell; we also explored the role of intracellular Ca2+ in ICl,swell activation. We used a CRISPR/Cas9 gene editing approach, electrophysiology, live fluorescent imaging, selective pharmacology, and other approaches to show that both LRRC8A and ANO1 can be activated by cell swelling in HEK293 cells. Yet, both channels contribute biophysically and pharmacologically distinct components to ICl,swell, with LRRC8A being the major component. Cell swelling induced oscillatory Ca2+ transients, and these Ca2+ signals were required to activate both the LRRC8A- and ANO1-dependent components of ICl,swell. Both ICl,swell components required localized rather than global Ca2+ for activation. Interestingly, while intracellular Ca2+ was necessary and sufficient to activate ANO1, it was necessary but not sufficient to activate LRRC8A-mediated currents. Finally, Ca2+ transients linked to the ICl,swell activation were mediated by the G protein-coupled receptor-independent PLC isoforms.
Collapse
Affiliation(s)
- Yani Liu
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China.,Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| | - Huiran Zhang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China.,Department of Pulmonary Medicine, the Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hongchao Men
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| | - Yuwei Du
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| | - Ziqian Xiao
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| | - Fan Zhang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| | - Dongyang Huang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| | - Xiaona Du
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| | - Nikita Gamper
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China.,School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Hailin Zhang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China.,The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, China
| |
Collapse
|
69
|
de Pascual R, Álvarez-Ortego N, de Los Ríos C, Jacob-Mazariego G, García AG. Tetrabenazine Facilitates Exocytosis by Enhancing Calcium-Induced Calcium Release through Ryanodine Receptors. J Pharmacol Exp Ther 2019; 371:219-230. [PMID: 31209099 DOI: 10.1124/jpet.119.256560] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/04/2019] [Indexed: 11/22/2022] Open
Abstract
Vesicular monoamine transporter-2 is expressed in the presynaptic secretory vesicles membrane in the brain. Its blockade by tetrabenazine (TBZ) causes depletion of dopamine at striatal basal ganglia; this is the mechanism underlying its long-standing use in the treatment of Huntington's disease. In the frame of a project aimed at investigating the kinetics of exocytosis from vesicles with partial emptying of their neurotransmitter, we unexpectedly found that TBZ facilitates exocytosis; thus, we decided to characterize such effect. We used bovine chromaffin cells (BCCs) challenged with repeated pulses of high K+ Upon repeated K+ pulsing, the exocytotic catecholamine release responses were gradually decaying. However, when cells were exposed to TBZ, responses were mildly augmented and decay rate delayed. Facilitation of exocytosis was not due to Ca2+ entry blockade through voltage-activated calcium channels (VACCs) because, in fact, TBZ mildly blocked the whole-cell Ca2+ current. However, TBZ mimicked the facilitatory effects of exocytosis elicited by BayK8644 (L-subtype VACC agonist), an effect blocked by nifedipine (VACC antagonist). On the basis that TBZ augmented the secretory responses to caffeine (but not those of histamine), we monitored its effects on cytosolic Ca2+ elevations ([Ca2+]c) triggered by caffeine or histamine. While the responses to caffeine were augmented twice by TBZ, those of histamine were unaffected; the same happened in rat cortical neurons. Hence, we hypothesize that TBZ facilitates exocytosis by increasing Ca2+ release through the endoplasmic reticulum ryanodine receptor channel (RyR). Confirming this hypothesis are docking results, showing an interaction of TBZ with RyRs. This is consonant with the existence of a healthy Ca2+-induced-Ca2+-release mechanism in BCCs. SIGNIFICANCE STATEMENT: A novel mechanism of action for tetrabenazine (TBZ), a drug used in the therapy of Huntington's disease (HD), is described here. Such mechanism consists of facilitation by combining TBZ with the ryanodine receptor of the endoplasmic reticulum, thereby increasing Ca2+-induced Ca2+ release. This novel mechanism should be taken into account when considering the efficacy and/or safety of TBZ in the treatment of chorea associated with HD and other disorders. Additionally, it could be of interest in the development of novel medicines to treat these pathological conditions.
Collapse
Affiliation(s)
- Ricardo de Pascual
- Instituto Teófilo Hernando, Madrid, Spain (R.d.P., N.Á.-O., C.d.l.R., G.J.-M., A.G.G.); and Departamento de Farmacología y Terapéutica (R.d.P., N.Á.-O., G.J.-M., A.G.G.) and Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa (C.d.l.R., A.G.G.), Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Nuria Álvarez-Ortego
- Instituto Teófilo Hernando, Madrid, Spain (R.d.P., N.Á.-O., C.d.l.R., G.J.-M., A.G.G.); and Departamento de Farmacología y Terapéutica (R.d.P., N.Á.-O., G.J.-M., A.G.G.) and Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa (C.d.l.R., A.G.G.), Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Cristóbal de Los Ríos
- Instituto Teófilo Hernando, Madrid, Spain (R.d.P., N.Á.-O., C.d.l.R., G.J.-M., A.G.G.); and Departamento de Farmacología y Terapéutica (R.d.P., N.Á.-O., G.J.-M., A.G.G.) and Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa (C.d.l.R., A.G.G.), Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Gema Jacob-Mazariego
- Instituto Teófilo Hernando, Madrid, Spain (R.d.P., N.Á.-O., C.d.l.R., G.J.-M., A.G.G.); and Departamento de Farmacología y Terapéutica (R.d.P., N.Á.-O., G.J.-M., A.G.G.) and Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa (C.d.l.R., A.G.G.), Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Antonio G García
- Instituto Teófilo Hernando, Madrid, Spain (R.d.P., N.Á.-O., C.d.l.R., G.J.-M., A.G.G.); and Departamento de Farmacología y Terapéutica (R.d.P., N.Á.-O., G.J.-M., A.G.G.) and Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa (C.d.l.R., A.G.G.), Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
70
|
Affiliation(s)
- Anant B Parekh
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford, OX1 3PT, UK.
| |
Collapse
|
71
|
Kawaguchi SY. Dynamic Factors for Transmitter Release at Small Presynaptic Boutons Revealed by Direct Patch-Clamp Recordings. Front Cell Neurosci 2019; 13:269. [PMID: 31249514 PMCID: PMC6582627 DOI: 10.3389/fncel.2019.00269] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 05/29/2019] [Indexed: 12/29/2022] Open
Abstract
Small size of an axon and presynaptic structures have hindered direct functional analysis of axonal signaling and transmitter release at presynaptic boutons in the central nervous system. However, recent technical advances in subcellular patch-clamp recordings and in fluorescent imagings are shedding light on the dynamic nature of axonal and presynaptic mechanisms. Here I summarize the functional design of an axon and presynaptic boutons, such as diversity and activity-dependent changes of action potential (AP) waveforms, Ca2+ influx, and kinetics of transmitter release, revealed by the technical tour de force of direct patch-clamp recordings and the leading-edge fluorescent imagings. I highlight the critical factors for dynamic modulation of transmitter release and presynaptic short-term plasticity.
Collapse
Affiliation(s)
- Shin-Ya Kawaguchi
- Society-Academia Collaboration for Innovation, Kyoto University, Kyoto, Japan.,Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan.,Institute for Advanced Study, Kyoto University, Kyoto, Japan
| |
Collapse
|
72
|
Chakrabarti R, Wichmann C. Nanomachinery Organizing Release at Neuronal and Ribbon Synapses. Int J Mol Sci 2019; 20:E2147. [PMID: 31052288 PMCID: PMC6539712 DOI: 10.3390/ijms20092147] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/26/2019] [Accepted: 04/26/2019] [Indexed: 11/17/2022] Open
Abstract
A critical aim in neuroscience is to obtain a comprehensive view of how regulated neurotransmission is achieved. Our current understanding of synapses relies mainly on data from electrophysiological recordings, imaging, and molecular biology. Based on these methodologies, proteins involved in a synaptic vesicle (SV) formation, mobility, and fusion at the active zone (AZ) membrane have been identified. In the last decade, electron tomography (ET) combined with a rapid freezing immobilization of neuronal samples opened a window for understanding the structural machinery with the highest spatial resolution in situ. ET provides significant insights into the molecular architecture of the AZ and the organelles within the presynaptic nerve terminal. The specialized sensory ribbon synapses exhibit a distinct architecture from neuronal synapses due to the presence of the electron-dense synaptic ribbon. However, both synapse types share the filamentous structures, also commonly termed as tethers that are proposed to contribute to different steps of SV recruitment and exocytosis. In this review, we discuss the emerging views on the role of filamentous structures in SV exocytosis gained from ultrastructural studies of excitatory, mainly central neuronal compared to ribbon-type synapses with a focus on inner hair cell (IHC) ribbon synapses. Moreover, we will speculate on the molecular entities that may be involved in filament formation and hence play a crucial role in the SV cycle.
Collapse
Affiliation(s)
- Rituparna Chakrabarti
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany.
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany.
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing", 37099 Göttingen, Germany.
| | - Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany.
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany.
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing", 37099 Göttingen, Germany.
- Collaborative Research Center 1286 "Quantitative Synaptology", 37099 Göttingen, Germany.
- Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany.
| |
Collapse
|
73
|
Brockhaus J, Brüggen B, Missler M. Imaging and Analysis of Presynaptic Calcium Influx in Cultured Neurons Using synGCaMP6f. Front Synaptic Neurosci 2019; 11:12. [PMID: 31057389 PMCID: PMC6477507 DOI: 10.3389/fnsyn.2019.00012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/26/2019] [Indexed: 12/12/2022] Open
Abstract
Presynaptic Ca2+ influx through voltage-gated calcium channels (VGCCs) is a key step in synaptic transmission that links action potential (AP)-derived depolarization to vesicle release. However, investigation of presynaptic Ca2+ influx by patch clamp recordings is difficult due to the small size of the majority of synaptic boutons along thin axons that hamper clamp control. Genetically encoded calcium indicators (GECIs) in combination with live cell imaging provide an alternative method to study Ca2+ transients in individual presynaptic terminals. The indicator GCaMP6f was developed for fast speed and high sensitivity in detecting Ca2+ transients even in subcellular compartments. We fused GCaMP6f to synaptophysin (synGCaMP6f) to enrich the calcium indicator in presynaptic boutons of transfected primary hippocampal neurons to study presynaptic Ca2+ changes in response to individual APs or short bursts. Changes in fluorescence intensity were evaluated by normalization to control level or, alternatively, by normalization to maximal fluorescence using the calcium ionophore ionomycin. Measurements revealed robust Ca2+ transients with amplitudes that depend on parameters like the number of APs, stimulation frequency or external calcium concentration. Our findings indicate an appropriate sensitivity of synGCaMP6f for studying total presynaptic Ca2+ transients induced by single APs or short bursts that showed little rundown of the response after repeated bursts. Moreover, these recordings are fast enough to even study short-term plasticity like paired pulse facilitation (PPF) and frequency dependence of Ca2+ transients. In addition, synGCaMP6f could be used to dissect the contribution of different subtypes of VGCCs to presynaptic Ca2+ influx. Our results demonstrate that synGCaMP6f allows the reliable analysis of changes in presynaptic calcium concentration at many individual synaptic boutons in parallel and provides the possibility to study the regulation of this important step in synaptic transmission.
Collapse
Affiliation(s)
- Johannes Brockhaus
- Institute of Anatomy and Molecular Neurobiology, Westfälische Wilhelms-University, Münster, Germany
| | - Bianca Brüggen
- Institute of Anatomy and Molecular Neurobiology, Westfälische Wilhelms-University, Münster, Germany
| | - Markus Missler
- Institute of Anatomy and Molecular Neurobiology, Westfälische Wilhelms-University, Münster, Germany
| |
Collapse
|
74
|
Archer CR, Enslow BT, Taylor AB, De la Rosa V, Bhattacharya A, Shapiro MS. A mutually induced conformational fit underlies Ca 2+-directed interactions between calmodulin and the proximal C terminus of KCNQ4 K + channels. J Biol Chem 2019; 294:6094-6112. [PMID: 30808708 PMCID: PMC6463706 DOI: 10.1074/jbc.ra118.006857] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/24/2019] [Indexed: 11/06/2022] Open
Abstract
Calmodulin (CaM) conveys intracellular Ca2+ signals to KCNQ (Kv7, "M-type") K+ channels and many other ion channels. Whether this "calmodulation" involves a dramatic structural rearrangement or only slight perturbations of the CaM/KCNQ complex is as yet unclear. A consensus structural model of conformational shifts occurring between low nanomolar and physiologically high intracellular [Ca2+] is still under debate. Here, we used various techniques of biophysical chemical analyses to investigate the interactions between CaM and synthetic peptides corresponding to the A and B domains of the KCNQ4 subtype. We found that in the absence of CaM, the peptides are disordered, whereas Ca2+/CaM imposed helical structure on both KCNQ A and B domains. Isothermal titration calorimetry revealed that Ca2+/CaM has higher affinity for the B domain than for the A domain of KCNQ2-4 and much higher affinity for the B domain when prebound with the A domain. X-ray crystallography confirmed that these discrete peptides spontaneously form a complex with Ca2+/CaM, similar to previous reports of CaM binding KCNQ-AB domains that are linked together. Microscale thermophoresis and heteronuclear single-quantum coherence NMR spectroscopy indicated the C-lobe of Ca2+-free CaM to interact with the KCNQ4 B domain (Kd ∼10-20 μm), with increasing Ca2+ molar ratios shifting the CaM-B domain interactions via only the CaM C-lobe to also include the N-lobe. Our findings suggest that in response to increased Ca2+, CaM undergoes lobe switching that imposes a dramatic mutually induced conformational fit to both the proximal C terminus of KCNQ4 channels and CaM, likely underlying Ca2+-dependent regulation of KCNQ gating.
Collapse
Affiliation(s)
- Crystal R Archer
- From the Departments of Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas 78229; Departments of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Benjamin T Enslow
- the Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Alexander B Taylor
- Departments of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Victor De la Rosa
- From the Departments of Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Akash Bhattacharya
- Departments of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Mark S Shapiro
- From the Departments of Cell and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas 78229.
| |
Collapse
|
75
|
Bolshakov AP, Kolleker A, Volkova EP, Valiullina-Rakhmatullina F, Kolosov PM, Rozov A. Overexpression of Calretinin Enhances Short-Term Synaptic Depression. Front Cell Neurosci 2019; 13:91. [PMID: 30930749 PMCID: PMC6425694 DOI: 10.3389/fncel.2019.00091] [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: 12/02/2018] [Accepted: 02/22/2019] [Indexed: 12/03/2022] Open
Abstract
Analysis of the effects of various proteins on short-term synaptic plasticity is a difficult task, which may require the use of knockout animals. Here, we propose an alternative experimental approach for studying the roles of desired proteins in synaptic plasticity. We packed the Ca2+-binding protein calretinin and the fluorescent protein Venus into AAV and injected the concentrated viral suspension into the neocortex of newborn rats. The infected layer 2/3 pyramidal cells were identified in rat cortical slices using Venus fluorescence. Analysis of short-term synaptic plasticity using paired patch clamp recordings between layer 2/3 pyramidal cells (presynaptic cell) and fast-spiking (FS) interneurons (post-synaptic cell) showed that calretinin expression in the pyramidal cells did not change the failure rate in this synapse but did decrease synaptic delay. Analysis of the parameters of short-term synaptic plasticity showed that the amplitude of the first EPSP in the train was not affected by calretinin, however, calretinin strongly enhanced short-term depression. In addition, we found that the effect of calretinin depended on the presynaptic firing frequency: an increase in frequency resulted in enhancement of synaptic depression.
Collapse
Affiliation(s)
- Alexey P Bolshakov
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences (RAS), Moscow, Russia.,Research Laboratory of Electrophysiology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Alexander Kolleker
- Max Planck Institute for Medical Research, Department of Molecular Neurobiology, Heidelberg, Germany.,Buryat State University, Medical Institute, Ulan-Ude, Russia
| | - Evgenia P Volkova
- Research Laboratory of Electrophysiology, Pirogov Russian National Research Medical University, Moscow, Russia
| | | | - Peter M Kolosov
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences (RAS), Moscow, Russia
| | - Andrei Rozov
- Laboratory of Neurobiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Department of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
76
|
Imaging of Somatic Ca 2+ Transients in Differentiated Human Neurons. Methods Mol Biol 2019. [PMID: 30900180 DOI: 10.1007/978-1-4939-9080-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Calcium is a major regulator of neuronal activity and calcium signaling is critically important for normal neuronal function. Ca imaging is a well-established tool for studying neuronal function and ongoing spontaneous Ca2+ transients are a good indicator of neuronal maturity. There are various indicators available today, differing by their sensitivity, spectra, and loading method. Here we present a method for measurement of Ca2+ transients in neurons using two different Ca2+ indicators, Oregon Green BAPTA-1 and GCaMP6.
Collapse
|
77
|
Okubo Y, Iino M. Visualization of astrocytic intracellular Ca 2+ mobilization. J Physiol 2019; 598:1671-1681. [PMID: 30825213 DOI: 10.1113/jp277609] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 02/06/2019] [Indexed: 11/08/2022] Open
Abstract
Astrocytes generate robust intracellular Ca2+ concentration changes (Ca2+ signals), which are assumed to regulate astrocytic functions that play crucial roles in the regulation of brain functions. One frequently used strategy for exploring the role of astrocytic Ca2+ signalling is the use of mice deficient in the type 2 inositol 1,4,5-trisphosphate receptor (IP3 R2). These IP3 R2-knockout (KO) mice are reportedly devoid of Ca2+ mobilization from the endoplasmic reticulum (ER) in astrocytes. However, they have shown no functional deficits in several studies, causing a heated debate as to the functional relevance of ER-mediated Ca2+ signalling in astrocytes. Recently, the assumption that Ca2+ mobilization from the ER is absent in IP3 R2-KO astrocytes has been re-evaluated using intraorganellar Ca2+ imaging techniques. The new results indicated that IP3 R2-independent Ca2+ release may generate Ca2+ nanodomains around the ER, which may help explain the absence of functional deficits in IP3 R2-KO mice.
Collapse
Affiliation(s)
- Yohei Okubo
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, 133-0033, Japan
| | - Masamitsu Iino
- Division of Cellular and Molecular Pharmacology, Nihon University School of Medicine, Tokyo, 173-8610, Japan
| |
Collapse
|
78
|
Abstract
Fundamental cell processes such as synaptic neurotransmitter release, endocrine hormone secretion, and myocyte contraction are controlled by highly localized calcium (Ca2+) signals resulting from brief openings of trans-membrane Ca2+ channels. On short temporal and spatial scales, the corresponding local Ca2+ nanodomains formed in the vicinity of a single or several open Ca2+ channels can be effectively approximated by quasi-stationary solutions. The rapid buffering approximation (RBA) is one of the most powerful of such approximations, and is based on the assumption of instantaneous equilibration of the bimolecular Ca2+ buffering reaction, combined with the conservation condition for the total Ca2+ and buffer molecule numbers. Previously, RBA has been generalized to an arbitrary arrangement of Ca2+ channels on a flat membrane, in the presence of any number of simple Ca2+ buffers with one-to-one Ca2+ binding stoichiometry. However, many biological buffers have multiple binding sites. For example, buffers and sensors phylogenetically related to calmodulin consist of two Ca2+-binding domains (lobes), with each domain binding two Ca2+ ions in a cooperative manner. Here we consider an extension of RBA to such buffers with two interdependent Ca2+ binding sites. We show that in the presence of such buffers, RBA solution is given by the solution to a cubic equation, analogous to the quadratic equation describing RBA in the case of a simple, one-to-one Ca2+ buffer. We examine in detail the dependence of RBA accuracy on buffering parameters, to reveal conditions under which RBA provides sufficient precision.
Collapse
Affiliation(s)
- Victor Matveev
- Department of Mathematical Sciences, New Jersey Institute of Technology, Newark, New Jersey.
| |
Collapse
|
79
|
Rozov A, Bolshakov AP, Valiullina-Rakhmatullina F. The Ever-Growing Puzzle of Asynchronous Release. Front Cell Neurosci 2019; 13:28. [PMID: 30809127 PMCID: PMC6379310 DOI: 10.3389/fncel.2019.00028] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/22/2019] [Indexed: 11/13/2022] Open
Abstract
Invasion of an action potential (AP) to presynaptic terminals triggers calcium dependent vesicle fusion in a relatively short time window, about a millisecond, after the onset of the AP. This allows fast and precise information transfer from neuron to neuron by means of synaptic transmission and phasic mediator release. However, at some synapses a single AP or a short burst of APs can generate delayed or asynchronous synaptic release lasting for tens or hundreds of milliseconds. Understanding the mechanisms underlying asynchronous release (AR) is important, since AR can better recruit extrasynaptic metabotropic receptors and maintain a high level of neurotransmitter in the extracellular space for a substantially longer period of time after presynaptic activity. Over the last decade substantial work has been done to identify the presynaptic calcium sensor that may be involved in AR. Several models have been suggested which may explain the long lasting presynaptic calcium elevation a prerequisite for prolonged delayed release. However, the presynaptic mechanisms underlying asynchronous vesicle release are still not well understood. In this review article, we provide an overview of the current state of knowledge on the molecular components involved in delayed vesicle fusion and in the maintenance of sufficient calcium concentration to trigger AR. In addition, we discuss possible alternative models that may explain intraterminal calcium dynamics underlying AR.
Collapse
Affiliation(s)
- Andrei Rozov
- Laboratory of Neurobiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Department of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Alexey P Bolshakov
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences (RAS), Moscow, Russia.,Laboratory of Electrophysiology, Pirogov Russian National Research Medical University, Moscow, Russia
| | | |
Collapse
|
80
|
Bornschein G, Schmidt H. Synaptotagmin Ca 2+ Sensors and Their Spatial Coupling to Presynaptic Ca v Channels in Central Cortical Synapses. Front Mol Neurosci 2019; 11:494. [PMID: 30697148 PMCID: PMC6341215 DOI: 10.3389/fnmol.2018.00494] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/21/2018] [Indexed: 11/21/2022] Open
Abstract
Ca2+ concentrations drop rapidly over a distance of a few tens of nanometers from an open voltage-gated Ca2+ channel (Cav), thereby, generating a spatially steep and temporally short-lived Ca2+ gradient that triggers exocytosis of a neurotransmitter filled synaptic vesicle. These non-steady state conditions make the Ca2+-binding kinetics of the Ca2+ sensors for release and their spatial coupling to the Cavs important parameters of synaptic efficacy. In the mammalian central nervous system, the main release sensors linking action potential mediated Ca2+ influx to synchronous release are Synaptotagmin (Syt) 1 and 2. We review here quantitative work focusing on the Ca2+ kinetics of Syt2-mediated release. At present similar quantitative detail is lacking for Syt1-mediated release. In addition to triggering release, Ca2+ remaining bound to Syt after the first of two successive high-frequency activations was found to be capable of facilitating release during the second activation. More recently, the Ca2+ sensor Syt7 was identified as additional facilitation sensor. We further review how several recent functional studies provided quantitative insights into the spatial topographical relationships between Syts and Cavs and identified mechanisms regulating the sensor-to-channel coupling distances at presynaptic active zones. Most synapses analyzed in matured cortical structures were found to operate at tight, nanodomain coupling. For fast signaling synapses a developmental switch from loose, microdomain to tight, nanodomain coupling was found. The protein Septin5 has been known for some time as a developmentally down-regulated “inhibitor” of tight coupling, while Munc13-3 was found only recently to function as a developmentally up-regulated mediator of tight coupling. On the other hand, a highly plastic synapse was found to operate at loose coupling in the matured hippocampus. Together these findings suggest that the coupling topography and its regulation is a specificity of the type of synapse. However, to definitely draw such conclusion our knowledge of functional active zone topographies of different types of synapses in different areas of the mammalian brain is too incomplete.
Collapse
Affiliation(s)
- Grit Bornschein
- Carl-Ludwig Institute for Physiology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Hartmut Schmidt
- Carl-Ludwig Institute for Physiology, Medical Faculty, University of Leipzig, Leipzig, Germany
| |
Collapse
|
81
|
|
82
|
Sforna L, Franciolini F, Catacuzzeno L. Ca 2+ -dependent and Ca 2+ -independent somatic release from trigeminal neurons. J Cell Physiol 2018; 234:10977-10989. [PMID: 30536400 DOI: 10.1002/jcp.27901] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/25/2018] [Indexed: 01/03/2023]
Abstract
Besides the nerve endings, the soma of trigeminal neurons also respond to membrane depolarizations with the release of neurotransmitters and neuromodulators in the extracellular space within the ganglion, a process potentially important for the cross-communication between neighboring sensory neurons. In this study, we addressed the dependence of somatic release on Ca2+ influx in trigeminal neurons and the involvement of the different types of voltage-gated Ca2+ (Cav) channels in the process. Similar to the closely related dorsal root ganglion neurons, we found two kinetically distinct components of somatic release, a faster component stimulated by voltage but independent of the Ca2+ influx, and a slower component triggered by Ca2+ influx. The Ca2+ -dependent component was inhibited 80% by ω-conotoxin-MVIIC, an inhibitor of both N- and P/Q-type Cav channels, and 55% by the P/Q-type selective inhibitor ω-agatoxin-IVA. The selective L-type Ca2+ channel inhibitor nimodipine was instead without effect. These results suggest a major involvement of N- and P/Q-, but not L-type Cav channels in the somatic release of trigeminal neurons. Thus antinociceptive Cav channel antagonists acting on the N- and P/Q-type channels may exert their function by also modulating the somatic release and cross-communication between sensory neurons.
Collapse
Affiliation(s)
- Luigi Sforna
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Fabio Franciolini
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy
| | - Luigi Catacuzzeno
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy
| |
Collapse
|
83
|
Samigullin DV, Zhilyakov NV, Khaziev EF, Bukharaeva EA, Nikolsky EE. Calcium Transient and Quantal Release in Mouse Neuromuscular Junction Under Extracellular Calcium Concentration Change. BIONANOSCIENCE 2018. [DOI: 10.1007/s12668-018-0558-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
84
|
Caffeine chelates calcium in the lumen of the endoplasmic reticulum. Biochem J 2018; 475:3639-3649. [PMID: 30389846 DOI: 10.1042/bcj20180532] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 10/14/2018] [Accepted: 11/01/2018] [Indexed: 12/29/2022]
Abstract
Cytosolic Ca2+ signals are often amplified by massive calcium release from the endoplasmic reticulum (ER). This calcium-induced calcium release (CICR) occurs by activation of an ER Ca2+ channel, the ryanodine receptor (RyR), which is facilitated by both cytosolic- and ER Ca2+ levels. Caffeine sensitizes RyR to Ca2+ and promotes ER Ca2+ release at basal cytosolic Ca2+ levels. This outcome is frequently used as a readout for the presence of CICR. By monitoring ER luminal Ca2+ with the low-affinity genetic Ca2+ probe erGAP3, we find here that application of 50 mM caffeine rapidly reduces the Ca2+ content of the ER in HeLa cells by ∼50%. Interestingly, this apparent ER Ca2+ release does not go along with the expected cytosolic Ca2+ increase. These results can be explained by Ca2+ chelation by caffeine inside the ER. Ca2+-overloaded mitochondria also display a drop of the matrix Ca2+ concentration upon caffeine addition. In contrast, in the cytosol, with a low free Ca2+ concentration (10-7 M), no chelation is observed. Expression of RyR3 sensitizes the responses to caffeine with effects both in the ER (increase in Ca2+ release) and in the cytosol (increase in Ca2+ peak) at low caffeine concentrations (0.3-1 mM) that have no effects in control cells. Our results illustrate the fact that simultaneous monitoring of both cytosolic- and ER Ca2+ are necessary to understand the action of caffeine and raise concerns against the use of high concentrations of caffeine as a readout of the presence of CICR.
Collapse
|
85
|
Limpitikul WB, Greenstein JL, Yue DT, Dick IE, Winslow RL. A bilobal model of Ca 2+-dependent inactivation to probe the physiology of L-type Ca 2+ channels. J Gen Physiol 2018; 150:1688-1701. [PMID: 30470716 PMCID: PMC6279366 DOI: 10.1085/jgp.201812115] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 08/01/2018] [Accepted: 10/26/2018] [Indexed: 12/20/2022] Open
Abstract
L-type calcium channels undergo Ca2+-dependent inactivation (CDI) in order to precisely control the entry of Ca2+ into cells such as cardiomyocytes. Limpitikul et al. develop a bilobal model of CDI and use it to understand the pathogenesis of arrhythmias associated with mutations in CaM. L-type calcium channels (LTCCs) are critical elements of normal cardiac function, playing a major role in orchestrating cardiac electrical activity and initiating downstream signaling processes. LTCCs thus use feedback mechanisms to precisely control calcium (Ca2+) entry into cells. Of these, Ca2+-dependent inactivation (CDI) is significant because it shapes cardiac action potential duration and is essential for normal cardiac rhythm. This important form of regulation is mediated by a resident Ca2+ sensor, calmodulin (CaM), which is comprised of two lobes that are each capable of responding to spatially distinct Ca2+ sources. Disruption of CaM-mediated CDI leads to severe forms of long-QT syndrome (LQTS) and life-threatening arrhythmias. Thus, a model capable of capturing the nuances of CaM-mediated CDI would facilitate increased understanding of cardiac (patho)physiology. However, one critical barrier to achieving a detailed kinetic model of CDI has been the lack of quantitative data characterizing CDI as a function of Ca2+. This data deficit stems from the experimental challenge of uncoupling the effect of channel gating on Ca2+ entry. To overcome this obstacle, we use photo-uncaging of Ca2+ to deliver a measurable Ca2+ input to CaM/LTCCs, while simultaneously recording CDI. Moreover, we use engineered CaMs with Ca2+ binding restricted to a single lobe, to isolate the kinetic response of each lobe. These high-resolution measurements enable us to build mathematical models for each lobe of CaM, which we use as building blocks for a full-scale bilobal model of CDI. Finally, we use this model to probe the pathogenesis of LQTS associated with mutations in CaM (calmodulinopathies). Each of these models accurately recapitulates the kinetics and steady-state properties of CDI in both physiological and pathological states, thus offering powerful new insights into the mechanistic alterations underlying cardiac arrhythmias.
Collapse
Affiliation(s)
- Worawan B Limpitikul
- Calcium Signals Laboratory, Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Joseph L Greenstein
- Institute for Computational Medicine, Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD
| | - David T Yue
- Calcium Signals Laboratory, Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ivy E Dick
- Calcium Signals Laboratory, Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD .,Department of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - Raimond L Winslow
- Institute for Computational Medicine, Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD
| |
Collapse
|
86
|
Explicit Theoretical Analysis of How the Rate of Exocytosis Depends on Local Control by Ca 2+ Channels. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2018; 2018:5721097. [PMID: 30607171 PMCID: PMC6261245 DOI: 10.1155/2018/5721097] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 09/19/2018] [Indexed: 12/11/2022]
Abstract
Hormones and neurotransmitters are released from cells by calcium-regulated exocytosis, and local coupling between Ca2+ channels (CaVs) and secretory granules is a key factor determining the exocytosis rate. Here, we devise a methodology based on Markov chain models that allows us to obtain analytic results for the expected rate. First, we analyze the property of the secretory complex obtained by coupling a single granule with one CaV. Then, we extend our results to a more general case where the granule is coupled with n CaVs. We investigate how the exocytosis rate is affected by varying the location of granules and CaVs. Moreover, we assume that the single granule can form complexes with inactivating or non-inactivating CaVs. We find that increasing the number of CaVs coupled with the granule determines a much higher rise of the exocytosis rate that, in case of inactivating CaVs, is more pronounced when the granule is close to CaVs, while, surprisingly, in case of non-inactivating CaVs, the highest relative increase in rate is obtained when the granule is far from the CaVs. Finally, we exploit the devised model to investigate the relation between exocytosis and calcium influx. We find that the quantities are typically linearly related, as observed experimentally. For the case of inactivating CaVs, our simulations show a change of the linear relation due to near-complete inactivation of CaVs.
Collapse
|
87
|
Mojard Kalkhoran S, Chow SHJ, Walia JS, Gershome C, Saraev N, Kim B, Poburko D. VNUT and VMAT2 segregate within sympathetic varicosities and localize near preferred Cav2 isoforms in the rat tail artery. Am J Physiol Heart Circ Physiol 2018; 316:H89-H105. [PMID: 30311774 DOI: 10.1152/ajpheart.00560.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
ATP and norepinephrine (NE) are coreleased from peripheral sympathetic nerve terminals. Whether they are stored in the same vesicles has been debated for decades. Preferential dependence of NE or ATP release on Ca2+ influx through specific voltage-gated Ca2+ channel (Cav2) isoforms suggests that NE and ATP are stored in separate vesicle pools, but simultaneous imaging of NE and ATP containing vesicles within single varicosities has not been reported. We conducted an immunohistochemical study of vesicular monoamine transporter 2 (VMAT2/SLC18A2) and vesicular nucleotide translocase (VNUT/SLC17A9) as markers of vesicles containing NE and ATP in sympathetic nerves of the rat tail artery. A large fraction of varicosities exhibited neighboring, rather than overlapping, VNUT and VMAT2 fluorescent puncta. VMAT2, but not VNUT, colocalized with synaptotagmin 1. Cav2.1, Cav2.2, and Cav2.3 are expressed in nerves in the tunica adventitia. VMAT2 preferentially localized adjacent to Cav2.2 and Cav2.3 rather than Cav2.1. VNUT preferentially localized adjacent to Cav2.3 > Cav2.2 >> Cav2.1. With the use of wire myography, inhibition of field-stimulated vasoconstriction with the Cav2.3 blocker SNX-482 (0.25 µM) mimicked the effects of the P2X inhibitor suramin (100 µM) rather than the α-adrenergic inhibitor phentolamine (10 µM). Variable sensitivity to SNX-482 and suramin between animals closely correlated with Cav2.3 staining. We concluded that a majority of ATP and NE stores localize to separate vesicle pools that use different synaptotagmin isoforms and that localize near different Cav2 isoforms to mediate vesicle release. Cav2.3 appears to play a previously unrecognized role in mediating ATP release in the rat tail artery. NEW & NOTEWORTHY Immunofluorescence imaging of vesicular nucleotide translocase and vesicular monoamine transporter 2 in rat tail arteries revealed that ATP and norepinephrine, classical cotransmitters, localize to well-segregated vesicle pools. Furthermore, vesicular nucleotide translocase and vesicular monoamine transporter 2 exhibit preferential localization with specific Cav2 isoforms. These novel observations address long-standing debates regarding the mechanism(s) of sympathetic neurotransmitter corelease.
Collapse
Affiliation(s)
- Somayeh Mojard Kalkhoran
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University , Burnaby, British Columbia , Canada.,Centre for Cell Biology, Development and Disease, Simon Fraser University , Burnaby, British Columbia , Canada
| | - Sarah Heather Jane Chow
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University , Burnaby, British Columbia , Canada
| | - Jagdeep Singh Walia
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University , Burnaby, British Columbia , Canada
| | - Cynthia Gershome
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University , Burnaby, British Columbia , Canada
| | - Nickolas Saraev
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University , Burnaby, British Columbia , Canada
| | - BaRun Kim
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University , Burnaby, British Columbia , Canada
| | - Damon Poburko
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University , Burnaby, British Columbia , Canada.,Centre for Cell Biology, Development and Disease, Simon Fraser University , Burnaby, British Columbia , Canada
| |
Collapse
|
88
|
de Diego AMG, García AG. Altered exocytosis in chromaffin cells from mouse models of neurodegenerative diseases. Acta Physiol (Oxf) 2018; 224:e13090. [PMID: 29742321 DOI: 10.1111/apha.13090] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 04/19/2018] [Accepted: 04/25/2018] [Indexed: 12/26/2022]
Abstract
Chromaffin cells from the adrenal gland (CCs) have extensively been used to explore the molecular structure and function of the exocytotic machinery, neurotransmitter release and synaptic transmission. The CC is integrated in the sympathoadrenal axis that helps the body maintain homoeostasis during both routine life and in acute stress conditions. This function is exquisitely controlled by the cerebral cortex and the hypothalamus. We propose the hypothesis that damage undergone by the brain during neurodegenerative diseases is also affecting the neurosecretory function of adrenal medullary CCs. In this context, we review here the following themes: (i) How the discharge of catecholamines is centrally and peripherally regulated at the sympathoadrenal axis; (ii) which are the intricacies of the amperometric techniques used to study the quantal release of single-vesicle exocytotic events; (iii) which are the alterations of the exocytotic fusion pore so far reported, in CCs of mouse models of neurodegenerative diseases; (iv) how some proteins linked to neurodegenerative pathologies affect the kinetics of exocytotic events; (v) finally, we try to integrate available data into a hypothesis to explain how the centrally originated neurodegenerative diseases may alter the kinetics of single-vesicle exocytotic events in peripheral adrenal medullary CCs.
Collapse
Affiliation(s)
- A. M. García de Diego
- Instituto Teófilo Hernando; Universidad Autónoma de Madrid; Madrid Spain
- Instituto de Investigación Sanitaria; Hospital Universitario de la Princesa; Universidad Autónoma de Madrid; Madrid Spain
- DNS Neuroscience; Parque Científico de Madrid; Madrid Spain
| | - A. García García
- Instituto Teófilo Hernando; Universidad Autónoma de Madrid; Madrid Spain
- Instituto de Investigación Sanitaria; Hospital Universitario de la Princesa; Universidad Autónoma de Madrid; Madrid Spain
- DNS Neuroscience; Parque Científico de Madrid; Madrid Spain
- Departamento de Farmacología y Terapéutica; Facultad de Medicina; Universidad Autónoma de Madrid; Madrid Spain
| |
Collapse
|
89
|
Two-component latency distributions indicate two-step vesicular release at simple glutamatergic synapses. Nat Commun 2018; 9:3943. [PMID: 30258069 PMCID: PMC6158186 DOI: 10.1038/s41467-018-06336-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 07/31/2018] [Indexed: 01/03/2023] Open
Abstract
It is often assumed that only stably docked synaptic vesicles can fuse following presynaptic action potential stimulation. However, during action potential trains docking sites are increasingly depleted, raising the question of the source of synaptic vesicles during sustained release. We have recently developed methods to reliably measure release latencies during high frequency trains at single synapses between parallel fibers and molecular layer interneurons. The latency distribution exhibits a single fast component at train onset but contains both a fast and a slow component later in the train. The contribution of the slow component increases with stimulation frequency and with release probability and decreases when blocking the docking step with latrunculin. These results suggest that the slow component reflects sequential docking and release in immediate succession. The transition from fast to slow component, as well as a later transition to asynchronous release, appear as successive adaptations of the synapse to maintain fidelity at the expense of time accuracy.
Collapse
|
90
|
Luján R, Aguado C, Ciruela F, Arus XM, Martín-Belmonte A, Alfaro-Ruiz R, Martínez-Gómez J, de la Ossa L, Watanabe M, Adelman JP, Shigemoto R, Fukazawa Y. SK2 Channels Associate With mGlu 1α Receptors and Ca V2.1 Channels in Purkinje Cells. Front Cell Neurosci 2018; 12:311. [PMID: 30283304 PMCID: PMC6156379 DOI: 10.3389/fncel.2018.00311] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/28/2018] [Indexed: 11/13/2022] Open
Abstract
The small-conductance, Ca2+-activated K+ (SK) channel subtype SK2 regulates the spike rate and firing frequency, as well as Ca2+ transients in Purkinje cells (PCs). To understand the molecular basis by which SK2 channels mediate these functions, we analyzed the exact location and densities of SK2 channels along the neuronal surface of the mouse cerebellar PCs using SDS-digested freeze-fracture replica labeling (SDS-FRL) of high sensitivity combined with quantitative analyses. Immunogold particles for SK2 were observed on post- and pre-synaptic compartments showing both scattered and clustered distribution patterns. We found an axo-somato-dendritic gradient of the SK2 particle density increasing 12-fold from soma to dendritic spines. Using two different immunogold approaches, we also found that SK2 immunoparticles were frequently adjacent to, but never overlap with, the postsynaptic density of excitatory synapses in PC spines. Co-immunoprecipitation analysis demonstrated that SK2 channels form macromolecular complexes with two types of proteins that mobilize Ca2+: CaV2.1 channels and mGlu1α receptors in the cerebellum. Freeze-fracture replica double-labeling showed significant co-clustering of particles for SK2 with those for CaV2.1 channels and mGlu1α receptors. SK2 channels were also detected at presynaptic sites, mostly at the presynaptic active zone (AZ), where they are close to CaV2.1 channels, though they are not significantly co-clustered. These data demonstrate that SK2 channels located in different neuronal compartments can associate with distinct proteins mobilizing Ca2+, and suggest that the ultrastructural association of SK2 with CaV2.1 and mGlu1α provides the mechanism that ensures voltage (excitability) regulation by distinct intracellular Ca2+ transients in PCs.
Collapse
Affiliation(s)
- Rafael Luján
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Campus Biosanitario, Universidad Castilla-La Mancha, Albacete, Spain
| | - Carolina Aguado
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Campus Biosanitario, Universidad Castilla-La Mancha, Albacete, Spain
| | - Francisco Ciruela
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina i Ciències de la Salut, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Department of Biochemistry and Microbiology, Faculty of Sciences, University of Ghent, Ghent, Belgium
| | - Xavier Morató Arus
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina i Ciències de la Salut, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Alejandro Martín-Belmonte
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Campus Biosanitario, Universidad Castilla-La Mancha, Albacete, Spain
| | - Rocío Alfaro-Ruiz
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Campus Biosanitario, Universidad Castilla-La Mancha, Albacete, Spain
| | - Jesús Martínez-Gómez
- Departamento de Sistemas Informáticos, Escuela Superior de Ingeniería Informática, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Luis de la Ossa
- Departamento de Sistemas Informáticos, Escuela Superior de Ingeniería Informática, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Masahiko Watanabe
- Department of Anatomy, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - John P Adelman
- Vollum Institute, Oregon Health and Science University, Portland, OR, United States
| | - Ryuichi Shigemoto
- Institute of Science and Technology (IST Austria), Klosterneuburg, Austria
| | - Yugo Fukazawa
- Division of Brain Structure and Function, Research Center for Child Mental Development, Life Science Advancement Program, Faculty of Medical Science, University of Fukui, Fukui, Japan
| |
Collapse
|
91
|
Nunes D, Kuner T. Axonal sodium channel NaV1.2 drives granule cell dendritic GABA release and rapid odor discrimination. PLoS Biol 2018; 16:e2003816. [PMID: 30125271 PMCID: PMC6117082 DOI: 10.1371/journal.pbio.2003816] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 08/30/2018] [Accepted: 08/06/2018] [Indexed: 12/19/2022] Open
Abstract
Dendrodendritic synaptic interactions between olfactory bulb mitral and granule cells represent a key neuronal mechanism of odor discrimination. Dendritic release of gamma-aminobutyric acid (GABA) from granule cells contributes to stimulus-dependent, rapid, and accurate odor discrimination, yet the physiological mechanisms governing this release and its behavioral relevance are unknown. Here, we show that granule cells express the voltage-gated sodium channel α-subunit NaV1.2 in clusters distributed throughout the cell surface including dendritic spines. Deletion of NaV1.2 in granule cells abolished spiking and GABA release as well as inhibition of synaptically connected mitral cells (MCs). As a consequence, mice required more time to discriminate highly similar odorant mixtures, while odor discrimination learning remained unaffected. In conclusion, we show that expression of NaV1.2 in granule cells is crucial for physiological dendritic GABA release and rapid discrimination of similar odorants with high accuracy. Hence, our data indicate that neurotransmitter-releasing dendritic spines function just like axon terminals. In axonal nerve terminals, neurotransmitter release is triggered by a localized Ca2+ nanodomain generated by voltage-gated calcium channels in response to an action potential, which in turn is mediated by voltage-gated sodium channels. Dendritic neurotransmitter release has been thought to work differently, mainly depending on Ca2+ entering directly through N-methyl-D-aspartate (NMDA) receptors, a subtype of ligand-gated ion channel. To further investigate how dendritic neurotransmitter is released, we studied granule cells in the olfactory bulb of mice, which establish inhibitory dendrodendritic synapses with mitral cells. We show that granule cells express voltage-gated sodium channels predominantly localized in dendrites and spines. Down-regulation of these channels precludes action potential firing in granule cells and strongly reduces mitral cell inhibition. Behaviorally, these mice require more time to discriminate highly similar odorants at maximal accuracy. Therefore, the inhibition of mitral cells relies on neurotransmitter released from the dendrites of granule cells by a mechanism that resembles axonal neurotransmitter release much more than previously thought.
Collapse
Affiliation(s)
- Daniel Nunes
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
- Functional Neuroanatomy Department, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
- * E-mail: (DN); (TK)
| | - Thomas Kuner
- Functional Neuroanatomy Department, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
- * E-mail: (DN); (TK)
| |
Collapse
|
92
|
Kirchner MK, Foehring RC, Callaway J, Armstrong WE. Specificity in the interaction of high-voltage-activated Ca 2+ channel types with Ca 2+-dependent afterhyperpolarizations in magnocellular supraoptic neurons. J Neurophysiol 2018; 120:1728-1739. [PMID: 30020842 DOI: 10.1152/jn.00285.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Magnocellular oxytocin (OT) and vasopressin (VP) neurons express an afterhyperpolarization (AHP) following spike trains that attenuates firing rate and contributes to burst patterning. This AHP includes contributions from an apamin-sensitive, medium-duration AHP (mAHP) and from an apamin-insensitive, slow-duration AHP (sAHP). These AHPs are Ca2+ dependent and activated by Ca2+ influx through voltage-gated Ca2+ channels. Across central nervous system neurons that generate Ca2+-dependent AHPs, the Ca2+ channels that couple to the mAHP and sAHP differ greatly, but for magnocellular neurosecretory cells this relationship is unknown. Using simultaneous whole cell recording and Ca2+ imaging, we evaluated the effect of specific high-voltage-activated (HVA) Ca2+ channel blockers on the mAHP and sAHP. Block of all HVA channels via 400 μM Cd2+ inhibited almost the entire AHP. We tested nifedipine, conotoxin GVIA, agatoxin IVA, and SNX-482, specific blockers of L-, N-, P/Q-, and R-type channels, respectively. The N-type channel blocker conotoxin GVIA (1 μM) was the only toxin that inhibited the mAHP in either OT or VP neurons although the effect on VP neurons was weaker by comparison. The sAHP was significantly inhibited by N-type block in OT neurons and by R-type block in VP neurons although neither accounted for the entirety of the sAHP. Thus the mAHP appears to be elicited by Ca2+ from mostly N-type channels in both OT and VP neurons, but the contributions of specific Ca2+ channel types to the sAHP in each cell type are different. Alternative sources to HVA channels may contribute Ca2+ for the sAHP. NEW & NOTEWORTHY Despite the importance of afterhyperpolarization (AHP) mechanisms for regulating firing behavior of oxytocin (OT) and vasopressin (VP) neurons of supraoptic nucleus, which types of high-voltage-activated Ca2+ channels elicit AHPs in these cells was unknown. We found that N-type channels couple to the medium AHP in both cell types. For the slow AHP, N-type channels contribute in OT neurons, whereas R-type contribute in VP neurons. No single Ca2+ channel blocker abolished the entire AHP, suggesting that additional Ca2+ sources are involved.
Collapse
Affiliation(s)
- Matthew K Kirchner
- University of Tennessee Health Science Center, Department of Anatomy & Neurobiology
| | - Robert C Foehring
- University of Tennessee Health Science Center, Department of Anatomy & Neurobiology
| | - Joseph Callaway
- University of Tennessee Health Science Center, Department of Anatomy & Neurobiology
| | - William E Armstrong
- University of Tennessee Health Science Center, Department of Anatomy & Neurobiology
| |
Collapse
|
93
|
Xiong Y, Sun S, Teng S, Jin M, Zhou Z. Ca 2+-Dependent and Ca 2+-Independent ATP Release in Astrocytes. Front Mol Neurosci 2018; 11:224. [PMID: 30079012 PMCID: PMC6063199 DOI: 10.3389/fnmol.2018.00224] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/07/2018] [Indexed: 01/19/2023] Open
Affiliation(s)
- Yingfei Xiong
- State Key Laboratory of Biomembrane and Membrane Biotechnology and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Institute of Molecular Medicine, Peking University, Beijing, China.,Department of Neurosurgery, Affiliated Hospital of The Air Force Institute of Aeromedicine, Beijing, China
| | - Suhua Sun
- State Key Laboratory of Biomembrane and Membrane Biotechnology and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Sasa Teng
- State Key Laboratory of Biomembrane and Membrane Biotechnology and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Mu Jin
- State Key Laboratory of Biomembrane and Membrane Biotechnology and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Institute of Molecular Medicine, Peking University, Beijing, China.,Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhuan Zhou
- State Key Laboratory of Biomembrane and Membrane Biotechnology and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Institute of Molecular Medicine, Peking University, Beijing, China
| |
Collapse
|
94
|
Durán E, Montes MÁ, Jemal I, Satterfield R, Young S, Álvarez de Toledo G. Synaptotagmin-7 controls the size of the reserve and resting pools of synaptic vesicles in hippocampal neurons. Cell Calcium 2018; 74:53-60. [PMID: 29957297 DOI: 10.1016/j.ceca.2018.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/04/2018] [Accepted: 06/18/2018] [Indexed: 02/07/2023]
Abstract
Continuous neurotransmitter release is subjected to synaptic vesicle availability, which in turn depends on vesicle recycling and the traffic of vesicles between pools. We studied the role of Synaptotagmin-7 (Syt-7) in synaptic vesicle accessibility for release in hippocampal neurons in culture. Synaptic boutons from Syt-7 knockout (KO) mice displayed normal basal secretion with no alteration in the RRP size or the probability of release. However, stronger stimuli revealed an increase in the size of the reserve and resting vesicle pools in Syt-7 KO boutons compared with WT. These data suggest that Syt-7 plays a significant role in the vesicle pool homeostasis and, consequently, in the availability of vesicles for synaptic transmission during strong stimulation, probably, by facilitating advancing synaptic vesicles to the readily releasable pool.
Collapse
Affiliation(s)
- Elisa Durán
- Dpto. Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| | - María Ángeles Montes
- Dpto. Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| | - Imane Jemal
- Dpto. Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| | - Rachel Satterfield
- Research Group Molecular Mechanisms of Synaptic Function, Max Planck Florida Institute, Jupiter, FL 33458. USA
| | - Samuel Young
- Research Group Molecular Mechanisms of Synaptic Function, Max Planck Florida Institute, Jupiter, FL 33458. USA
| | | |
Collapse
|
95
|
Csordás G, Weaver D, Hajnóczky G. Endoplasmic Reticulum-Mitochondrial Contactology: Structure and Signaling Functions. Trends Cell Biol 2018; 28:523-540. [PMID: 29588129 DOI: 10.1016/j.tcb.2018.02.009] [Citation(s) in RCA: 397] [Impact Index Per Article: 56.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 02/23/2018] [Accepted: 02/23/2018] [Indexed: 02/08/2023]
Abstract
Interorganellar contacts are increasingly recognized as central to the control of cellular behavior. These contacts, which typically involve a small fraction of the endomembrane surface, are local communication hubs that resemble synapses. We propose the term contactology to denote the analysis of interorganellar contacts. Endoplasmic reticulum (ER) contacts with mitochondria were recognized several decades ago; major roles in ion and lipid transfer, signaling, and membrane dynamics have been established, while others continue to emerge. The functional diversity of ER-mitochondrial (ER-mito) contacts is mirrored in their structural heterogeneity, with subspecialization likely supported by multiple, different linker-forming protein structures. The nanoscale size of the contacts has made studying their structure, function, and dynamics difficult. This review focuses on the structure of the ER-mito contacts, methods for studying them, and the roles of contacts in Ca2+ and reactive oxygen species (ROS) signaling.
Collapse
Affiliation(s)
- György Csordás
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - David Weaver
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - György Hajnóczky
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
96
|
Variations in Ca 2+ Influx Can Alter Chelator-Based Estimates of Ca 2+ Channel-Synaptic Vesicle Coupling Distance. J Neurosci 2018; 38:3971-3987. [PMID: 29563180 DOI: 10.1523/jneurosci.2061-17.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 02/23/2018] [Accepted: 02/28/2018] [Indexed: 12/20/2022] Open
Abstract
The timing and probability of synaptic vesicle fusion from presynaptic terminals is governed by the distance between voltage-gated Ca2+ channels (VGCCs) and Ca2+ sensors for exocytosis. This VGCC-sensor coupling distance can be determined from the fractional block of vesicular release by exogenous Ca2+ chelators, which depends on biophysical factors that have not been thoroughly explored. Using numerical simulations of Ca2+ reaction and diffusion, as well as vesicular release, we examined the contributions of conductance, density, and open duration of VGCCs, and the influence of endogenous Ca2+ buffers on the inhibition of exocytosis by EGTA. We found that estimates of coupling distance are critically influenced by the duration and amplitude of Ca2+ influx at active zones, but relatively insensitive to variations of mobile endogenous buffer. High concentrations of EGTA strongly inhibit vesicular release in close proximity (20-30 nm) to VGCCs if the flux duration is brief, but have little influence for longer flux durations that saturate the Ca2+ sensor. Therefore, the diversity in presynaptic action potential duration is sufficient to alter EGTA inhibition, resulting in errors potentially as large as 300% if Ca2+ entry durations are not considered when estimating VGCC-sensor coupling distances.SIGNIFICANT STATEMENT The coupling distance between voltage-gated Ca2+ channels and Ca2+ sensors for exocytosis critically determines the timing and probability of neurotransmitter release. Perfusion of presynaptic terminals with the exogenous Ca2+ chelator EGTA has been widely used for both qualitative and quantitative estimates of this distance. However, other presynaptic terminal parameters such as the amplitude and duration of Ca2+ entry can also influence EGTA inhibition of exocytosis, thus confounding conclusions based on EGTA alone. Here, we performed reaction-diffusion simulations of Ca2+-driven synaptic vesicle fusion, which delineate the critical parameters influencing an accurate prediction of coupling distance. Our study provides guidelines for characterizing and understanding how variability in coupling distance across chemical synapses could be estimated accurately.
Collapse
|
97
|
Coupling of SK channels, L-type Ca 2+ channels, and ryanodine receptors in cardiomyocytes. Sci Rep 2018; 8:4670. [PMID: 29549309 PMCID: PMC5856806 DOI: 10.1038/s41598-018-22843-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 03/01/2018] [Indexed: 01/01/2023] Open
Abstract
Small-conductance Ca2+-activated K+ (SK) channels regulate the excitability of cardiomyocytes by integrating intracellular Ca2+ and membrane potentials on a beat-to-beat basis. The inextricable interplay between activation of SK channels and Ca2+ dynamics suggests the pathology of one begets another. Yet, the exact mechanistic underpinning for the activation of cardiac SK channels remains unaddressed. Here, we investigated the intracellular Ca2+ microdomains necessary for SK channel activation. SK currents coupled with Ca2+ influx via L-type Ca2+ channels (LTCCs) continued to be elicited after application of caffeine, ryanodine or thapsigargin to deplete SR Ca2+ store, suggesting that LTCCs provide the immediate Ca2+ microdomain for the activation of SK channels in cardiomyocytes. Super-resolution imaging of SK2, Cav1.2 Ca2+ channel, and ryanodine receptor 2 (RyR2) was performed to quantify the nearest neighbor distances (NND) and localized the three molecules within hundreds of nanometers. The distribution of NND between SK2 and RyR2 as well as SK2 and Cav1.2 was bimodal, suggesting a spatial relationship between the channels. The activation mechanism revealed by our study paved the way for the understanding of the roles of SK channels on the feedback mechanism to regulate the activities of LTCCs and RyR2 to influence local and global Ca2+ signaling.
Collapse
|
98
|
Mammucari C, Raffaello A, Vecellio Reane D, Gherardi G, De Mario A, Rizzuto R. Mitochondrial calcium uptake in organ physiology: from molecular mechanism to animal models. Pflugers Arch 2018. [PMID: 29541860 PMCID: PMC6060757 DOI: 10.1007/s00424-018-2123-2] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mitochondrial Ca2+ is involved in heterogeneous functions, ranging from the control of metabolism and ATP production to the regulation of cell death. In addition, mitochondrial Ca2+ uptake contributes to cytosolic [Ca2+] shaping thus impinging on specific Ca2+-dependent events. Mitochondrial Ca2+ concentration is controlled by influx and efflux pathways: the former controlled by the activity of the mitochondrial Ca2+ uniporter (MCU), the latter by the Na+/Ca2+ exchanger (NCLX) and the H+/Ca2+ (mHCX) exchanger. The molecular identities of MCU and of NCLX have been recently unraveled, thus allowing genetic studies on their physiopathological relevance. After a general framework on the significance of mitochondrial Ca2+ uptake, this review discusses the structure of the MCU complex and the regulation of its activity, the importance of mitochondrial Ca2+ signaling in different physiological settings, and the consequences of MCU modulation on organ physiology.
Collapse
Affiliation(s)
| | - Anna Raffaello
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | | | - Gaia Gherardi
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Agnese De Mario
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padova, Padua, Italy.
| |
Collapse
|
99
|
Lou X. Sensing Exocytosis and Triggering Endocytosis at Synapses: Synaptic Vesicle Exocytosis-Endocytosis Coupling. Front Cell Neurosci 2018; 12:66. [PMID: 29593500 PMCID: PMC5861208 DOI: 10.3389/fncel.2018.00066] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/26/2018] [Indexed: 12/29/2022] Open
Abstract
The intact synaptic structure is critical for information processing in neural circuits. During synaptic transmission, rapid vesicle exocytosis increases the size of never terminals and endocytosis counteracts the increase. Accumulating evidence suggests that SV exocytosis and endocytosis are tightly connected in time and space during SV recycling, and this process is essential for synaptic function and structural stability. Research in the past has illustrated the molecular details of synaptic vesicle (SV) exocytosis and endocytosis; however, the mechanisms that timely connect these two fundamental events are poorly understood at central synapses. Here we discuss recent progress in SV recycling and summarize several emerging mechanisms by which synapses can “sense” the occurrence of exocytosis and timely initiate compensatory endocytosis. They include Ca2+ sensing, SV proteins sensing, and local membrane stress sensing. In addition, the spatial organization of endocytic zones adjacent to active zones provides a structural basis for efficient coupling between SV exocytosis and endocytosis. Through linking different endocytosis pathways with SV fusion, these mechanisms ensure necessary plasticity and robustness of nerve terminals to meet diverse physiological needs.
Collapse
Affiliation(s)
- Xuelin Lou
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
100
|
Bradford AB, Machamer JB, Russo TM, McNutt PM. 3,4-diaminopyridine reverses paralysis in botulinum neurotoxin-intoxicated diaphragms through two functionally distinct mechanisms. Toxicol Appl Pharmacol 2018; 341:77-86. [PMID: 29366638 DOI: 10.1016/j.taap.2018.01.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 01/15/2018] [Accepted: 01/18/2018] [Indexed: 01/11/2023]
Abstract
Botulinum neurotoxins (BoNTs) are exceedingly potent neurological poisons that prevent neurotransmitter release from peripheral nerve terminals by cleaving presynaptic proteins required for synaptic vesicle fusion. The ensuing neuromuscular paralysis causes death by asphyxiation. Although no antidotal treatments exist to block toxin activity within the nerve terminal, aminopyridine antagonists of voltage-gated potassium channels have been proposed as symptomatic treatments for botulism toxemia. However, clinical evaluation of aminopyridines as symptomatic treatments for botulism has been inconclusive, in part because mechanisms responsible for reversal of paralysis in BoNT-poisoned nerve terminals are not understood. Here we measured the effects of 3,4-diaminopyridine (DAP) on phrenic nerve-elicited diaphragm contraction and end-plate potentials at various times after intoxication with BoNT serotypes A, B, or E. We found that DAP-mediated increases in quantal content promote neurotransmission from intoxicated nerve terminals through two functionally distinguishable mechanisms. First, DAP increases the probability of neurotransmission at non-intoxicated release sites. This mechanism is serotype-independent, becomes less effective as nerve terminals become progressively impaired, and remains susceptible to ongoing intoxication. Second, DAP elicits persistent production of toxin-resistant endplate potentials from nerve terminals fully intoxicated by BoNT/A, but not serotypes B or E. Since this effect appears specific to BoNT/A intoxication, we propose that DAP treatment enables BoNT/A-cleaved SNAP-25 to productively engage in fusogenic release by increasing the opportunity for low-efficiency fusion events. These findings have important implications for DAP as a botulism therapeutic by defining conditions under which DAP may be clinically effective in reversing botulism symptoms.
Collapse
Affiliation(s)
- Aaron B Bradford
- Department of Neuroscience, United States Army Medical Research Institute of Chemical Defense, 2900 Ricketts Point Road, Gunpowder, MD 21010, USA
| | - James B Machamer
- Department of Neuroscience, United States Army Medical Research Institute of Chemical Defense, 2900 Ricketts Point Road, Gunpowder, MD 21010, USA
| | - Trisha M Russo
- Department of Neuroscience, United States Army Medical Research Institute of Chemical Defense, 2900 Ricketts Point Road, Gunpowder, MD 21010, USA
| | - Patrick M McNutt
- Department of Neuroscience, United States Army Medical Research Institute of Chemical Defense, 2900 Ricketts Point Road, Gunpowder, MD 21010, USA.
| |
Collapse
|