1
|
Chen B, Yu P, Chan WN, Xie F, Zhang Y, Liang L, Leung KT, Lo KW, Yu J, Tse GMK, Kang W, To KF. Cellular zinc metabolism and zinc signaling: from biological functions to diseases and therapeutic targets. Signal Transduct Target Ther 2024; 9:6. [PMID: 38169461 PMCID: PMC10761908 DOI: 10.1038/s41392-023-01679-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 09/15/2023] [Accepted: 10/10/2023] [Indexed: 01/05/2024] Open
Abstract
Zinc metabolism at the cellular level is critical for many biological processes in the body. A key observation is the disruption of cellular homeostasis, often coinciding with disease progression. As an essential factor in maintaining cellular equilibrium, cellular zinc has been increasingly spotlighted in the context of disease development. Extensive research suggests zinc's involvement in promoting malignancy and invasion in cancer cells, despite its low tissue concentration. This has led to a growing body of literature investigating zinc's cellular metabolism, particularly the functions of zinc transporters and storage mechanisms during cancer progression. Zinc transportation is under the control of two major transporter families: SLC30 (ZnT) for the excretion of zinc and SLC39 (ZIP) for the zinc intake. Additionally, the storage of this essential element is predominantly mediated by metallothioneins (MTs). This review consolidates knowledge on the critical functions of cellular zinc signaling and underscores potential molecular pathways linking zinc metabolism to disease progression, with a special focus on cancer. We also compile a summary of clinical trials involving zinc ions. Given the main localization of zinc transporters at the cell membrane, the potential for targeted therapies, including small molecules and monoclonal antibodies, offers promising avenues for future exploration.
Collapse
Affiliation(s)
- Bonan Chen
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Peiyao Yu
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Wai Nok Chan
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Fuda Xie
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Yigan Zhang
- Institute of Biomedical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Li Liang
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Kam Tong Leung
- Department of Pediatrics, The Chinese University of Hong Kong, Hong Kong, China
| | - Kwok Wai Lo
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Gary M K Tse
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
2
|
Peng C, Shao X, Tian X, Li Y, Liu D, Yan C, Han Y. CREG ameliorates embryonic stem cell differentiation into smooth muscle cells by modulation of TGF-β expression. Differentiation 2022; 125:9-17. [DOI: 10.1016/j.diff.2022.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 02/19/2022] [Accepted: 03/14/2022] [Indexed: 11/27/2022]
|
3
|
Tworig JM, Coate C, Feller MB. Excitatory neurotransmission activates compartmentalized calcium transients in Müller glia without affecting lateral process motility. eLife 2021; 10:73202. [PMID: 34913435 PMCID: PMC8806189 DOI: 10.7554/elife.73202] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/15/2021] [Indexed: 11/13/2022] Open
Abstract
Neural activity has been implicated in the motility and outgrowth of glial cell processes throughout the central nervous system. Here, we explore this phenomenon in Müller glia, which are specialized radial astroglia that are the predominant glial type of the vertebrate retina. Müller glia extend fine filopodia-like processes into retinal synaptic layers, in similar fashion to brain astrocytes and radial glia that exhibit perisynaptic processes. Using two-photon volumetric imaging, we found that during the second postnatal week, Müller glial processes were highly dynamic, with rapid extensions and retractions that were mediated by cytoskeletal rearrangements. During this same stage of development, retinal waves led to increases in cytosolic calcium within Müller glial lateral processes and stalks. These regions comprised distinct calcium compartments, distinguished by variable participation in waves, timing, and sensitivity to an M1 muscarinic acetylcholine receptor antagonist. However, we found that motility of lateral processes was unaffected by the presence of pharmacological agents that enhanced or blocked wave-associated calcium transients. Finally, we found that mice lacking normal cholinergic waves in the first postnatal week also exhibited normal Müller glial process morphology. Hence, outgrowth of Müller glial lateral processes into synaptic layers is determined by factors that are independent of neuronal activity. When it comes to studying the nervous system, neurons often steal the limelight; yet, they can only work properly thanks to an ensemble cast of cell types whose roles are only just emerging. For example, ‘glial cells’ – their name derives from the Greek word for glue – were once thought to play only a passive, supporting function in nervous tissues. Now, growing evidence shows that they are, in fact, integrated into neural circuits: their activity is influenced by neurons, and, in turn, they help neurons to function properly. The role of glial cells is becoming clear in the retina, the thin, light-sensitive layer that lines the back of the eye and relays visual information to the brain. There, beautifully intricate Müller glial cells display fine protrusions (or ‘processes') that intermingle with synapses, the busy space between neurons where chemical messengers are exchanged. These messengers can act on Müller cells, triggering cascades of molecular events that may influence the structure and function of glia. This is of particular interest during development: as Müller cells mature, they are exposed to chemicals released by more fully formed retinal neurons. Tworig et al. explored how neuronal messengers can influence the way Müller cells grow their processes. To do so, they tracked mouse retinal glial cells ‘live’ during development, showing that they were growing fine, highly dynamic processes in a region rich in synapses just as neurons and glia increased their communication. However, using drugs to disrupt this messaging for a short period did not seem to impact how the processes grew. Extending the blockade over a longer timeframe also did not change the way Müller cells developed, with the cells still acquiring their characteristic elaborate process networks. Taken together, these results suggest that the structural maturation of Müller glial cells is not impacted by neuronal signaling, giving a more refined understanding of how glia form in the retina and potentially in the brain.
Collapse
Affiliation(s)
- Joshua M Tworig
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Chandler Coate
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Marla B Feller
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
4
|
Kang KR, Kim J, Ryu B, Lee SG, Oh MS, Baek J, Ren X, Canavero S, Kim CY, Chung HM. BAPTA, a calcium chelator, neuroprotects injured neurons in vitro and promotes motor recovery after spinal cord transection in vivo. CNS Neurosci Ther 2021; 27:919-929. [PMID: 33942993 PMCID: PMC8265943 DOI: 10.1111/cns.13651] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/25/2021] [Accepted: 04/11/2021] [Indexed: 12/27/2022] Open
Abstract
Aim Despite animal evidence of a role of calcium in the pathogenesis of spinal cord injury, several studies conducted in the past found calcium blockade ineffective. However, those studies involved oral or parenteral administration of Ca++ antagonists. We hypothesized that Ca++ blockade might be effective with local/immediate application (LIA) at the time of neural injury. Methods In this study, we assessed the effects of LIA of BAPTA (1,2‐bis (o‐aminophenoxy) ethane‐N, N, N′, N'‐tetraacetic acid), a cell‐permeable highly selective Ca++ chelator, after spinal cord transection (SCT) in mice over 4 weeks. Effects of BAPTA were assessed behaviorally and with immunohistochemistry. Concurrently, BAPTA was submitted for the first time to multimodality assessment in an in vitro model of neural damage as a possible spinal neuroprotectant. Results We demonstrate that BAPTA alleviates neuronal apoptosis caused by physical damage by inhibition of neuronal apoptosis and reactive oxygen species (ROS) generation. This translates to enhanced preservation of electrophysiological function and superior behavioral recovery. Conclusion This study shows for the first time that local/immediate application of Ca++ chelator BAPTA is strongly neuroprotective after severe spinal cord injury.
Collapse
Affiliation(s)
- Kyu-Ree Kang
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Jin Kim
- Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Bokyeong Ryu
- Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Seul-Gi Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Min-Seok Oh
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Jieun Baek
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Xiaoping Ren
- Department of Orthopedics, Ruikang Hospital, Nanning, China.,GICUP-Global Initiative to Cure Paralysis, Columbus, Ohio, USA
| | - Sergio Canavero
- GICUP-Global Initiative to Cure Paralysis, Columbus, Ohio, USA.,HEAVEN/GEMINI International Collaborative Group, Turin, Italy
| | - C-Yoon Kim
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea.,GICUP-Global Initiative to Cure Paralysis, Columbus, Ohio, USA.,Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul, Korea
| | - Hyung Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| |
Collapse
|
5
|
Semyanov A, Henneberger C, Agarwal A. Making sense of astrocytic calcium signals — from acquisition to interpretation. Nat Rev Neurosci 2020; 21:551-564. [DOI: 10.1038/s41583-020-0361-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2020] [Indexed: 12/31/2022]
|
6
|
Denizot A, Arizono M, Nägerl UV, Soula H, Berry H. Simulation of calcium signaling in fine astrocytic processes: Effect of spatial properties on spontaneous activity. PLoS Comput Biol 2019; 15:e1006795. [PMID: 31425510 PMCID: PMC6726244 DOI: 10.1371/journal.pcbi.1006795] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 09/04/2019] [Accepted: 07/08/2019] [Indexed: 12/20/2022] Open
Abstract
Astrocytes, a glial cell type of the central nervous system, have emerged as detectors and regulators of neuronal information processing. Astrocyte excitability resides in transient variations of free cytosolic calcium concentration over a range of temporal and spatial scales, from sub-microdomains to waves propagating throughout the cell. Despite extensive experimental approaches, it is not clear how these signals are transmitted to and integrated within an astrocyte. The localization of the main molecular actors and the geometry of the system, including the spatial organization of calcium channels IP3R, are deemed essential. However, as most calcium signals occur in astrocytic ramifications that are too fine to be resolved by conventional light microscopy, most of those spatial data are unknown and computational modeling remains the only methodology to study this issue. Here, we propose an IP3R-mediated calcium signaling model for dynamics in such small sub-cellular volumes. To account for the expected stochasticity and low copy numbers, our model is both spatially explicit and particle-based. Extensive simulations show that spontaneous calcium signals arise in the model via the interplay between excitability and stochasticity. The model reproduces the main forms of calcium signals and indicates that their frequency crucially depends on the spatial organization of the IP3R channels. Importantly, we show that two processes expressing exactly the same calcium channels can display different types of calcium signals depending on the spatial organization of the channels. Our model with realistic process volume and calcium concentrations successfully reproduces spontaneous calcium signals that we measured in calcium micro-domains with confocal microscopy and predicts that local variations of calcium indicators might contribute to the diversity of calcium signals observed in astrocytes. To our knowledge, this model is the first model suited to investigate calcium dynamics in fine astrocytic processes and to propose plausible mechanisms responsible for their variability.
Collapse
Affiliation(s)
- Audrey Denizot
- INRIA, F-69603, Villeurbanne, France
- Univ Lyon, LIRIS, UMR5205 CNRS, F-69621, Villeurbanne, France
| | - Misa Arizono
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, Bordeaux, France
| | - U. Valentin Nägerl
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, Bordeaux, France
| | - Hédi Soula
- INRIA, F-69603, Villeurbanne, France
- Univ P&M Curie, CRC, INSERM UMRS 1138, F-75006, Paris, France
| | - Hugues Berry
- INRIA, F-69603, Villeurbanne, France
- Univ Lyon, LIRIS, UMR5205 CNRS, F-69621, Villeurbanne, France
| |
Collapse
|
7
|
Greenwald EC, Mehta S, Zhang J. Genetically Encoded Fluorescent Biosensors Illuminate the Spatiotemporal Regulation of Signaling Networks. Chem Rev 2018; 118:11707-11794. [PMID: 30550275 PMCID: PMC7462118 DOI: 10.1021/acs.chemrev.8b00333] [Citation(s) in RCA: 353] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cellular signaling networks are the foundation which determines the fate and function of cells as they respond to various cues and stimuli. The discovery of fluorescent proteins over 25 years ago enabled the development of a diverse array of genetically encodable fluorescent biosensors that are capable of measuring the spatiotemporal dynamics of signal transduction pathways in live cells. In an effort to encapsulate the breadth over which fluorescent biosensors have expanded, we endeavored to assemble a comprehensive list of published engineered biosensors, and we discuss many of the molecular designs utilized in their development. Then, we review how the high temporal and spatial resolution afforded by fluorescent biosensors has aided our understanding of the spatiotemporal regulation of signaling networks at the cellular and subcellular level. Finally, we highlight some emerging areas of research in both biosensor design and applications that are on the forefront of biosensor development.
Collapse
Affiliation(s)
- Eric C Greenwald
- University of California , San Diego, 9500 Gilman Drive, BRFII , La Jolla , CA 92093-0702 , United States
| | - Sohum Mehta
- University of California , San Diego, 9500 Gilman Drive, BRFII , La Jolla , CA 92093-0702 , United States
| | - Jin Zhang
- University of California , San Diego, 9500 Gilman Drive, BRFII , La Jolla , CA 92093-0702 , United States
| |
Collapse
|
8
|
Savtchenko LP, Bard L, Jensen TP, Reynolds JP, Kraev I, Medvedev N, Stewart MG, Henneberger C, Rusakov DA. Disentangling astroglial physiology with a realistic cell model in silico. Nat Commun 2018; 9:3554. [PMID: 30177844 PMCID: PMC6120909 DOI: 10.1038/s41467-018-05896-w] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 07/10/2018] [Indexed: 12/05/2022] Open
Abstract
Electrically non-excitable astroglia take up neurotransmitters, buffer extracellular K+ and generate Ca2+ signals that release molecular regulators of neural circuitry. The underlying machinery remains enigmatic, mainly because the sponge-like astrocyte morphology has been difficult to access experimentally or explore theoretically. Here, we systematically incorporate multi-scale, tri-dimensional astroglial architecture into a realistic multi-compartmental cell model, which we constrain by empirical tests and integrate into the NEURON computational biophysical environment. This approach is implemented as a flexible astrocyte-model builder ASTRO. As a proof-of-concept, we explore an in silico astrocyte to evaluate basic cell physiology features inaccessible experimentally. Our simulations suggest that currents generated by glutamate transporters or K+ channels have negligible distant effects on membrane voltage and that individual astrocytes can successfully handle extracellular K+ hotspots. We show how intracellular Ca2+ buffers affect Ca2+ waves and why the classical Ca2+ sparks-and-puffs mechanism is theoretically compatible with common readouts of astroglial Ca2+ imaging.
Collapse
Affiliation(s)
- Leonid P Savtchenko
- UCL Institute of Neurology, University College London, London, WC1N 3BG, UK.
| | - Lucie Bard
- UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Thomas P Jensen
- UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - James P Reynolds
- UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Igor Kraev
- The Open University, Milton Keynes, MK7 6AA, UK
| | | | | | - Christian Henneberger
- UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
- German Center of Neurodegenerative Diseases (DZNE), Bonn, 53127, Germany
- Institute of Cellular Neurosciences, University of Bonn Medical School, Bonn, 53127, Germany
| | - Dmitri A Rusakov
- UCL Institute of Neurology, University College London, London, WC1N 3BG, UK.
| |
Collapse
|
9
|
Fujii Y, Maekawa S, Morita M. Astrocyte calcium waves propagate proximally by gap junction and distally by extracellular diffusion of ATP released from volume-regulated anion channels. Sci Rep 2017; 7:13115. [PMID: 29030562 PMCID: PMC5640625 DOI: 10.1038/s41598-017-13243-0] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 09/21/2017] [Indexed: 11/09/2022] Open
Abstract
Wave-like propagation of [Ca2+]i increases is a remarkable intercellular communication characteristic in astrocyte networks, intercalating neural circuits and vasculature. Mechanically-induced [Ca2+]i increases and their subsequent propagation to neighboring astrocytes in culture is a classical model of astrocyte calcium wave and is known to be mediated by gap junction and extracellular ATP, but the role of each pathway remains unclear. Pharmacologic analysis of time-dependent distribution of [Ca2+]i revealed three distinct [Ca2+]i increases, the largest being in stimulated cells independent of extracellular Ca2+ and inositol 1,4,5-trisphosphate-induced Ca2+ release. In addition, persistent [Ca2+]i increases were found to propagate rapidly via gap junctions in the proximal region, and transient [Ca2+]i increases were found to propagate slowly via extracellular ATP in the distal region. Simultaneous imaging of astrocyte [Ca2+]i and extracellular ATP, the latter of which was measured by an ATP sniffing cell, revealed that ATP was released within the proximal region by volume-regulated anion channel in a [Ca2+]i independent manner. This detailed analysis of a classical model is the first to address the different contributions of two major pathways of calcium waves, gap junctions and extracellular ATP.
Collapse
Affiliation(s)
- Yuki Fujii
- Kobe University Graduate School of Science, Department of Biology, Kobe, 657-8501, Japan
| | - Shohei Maekawa
- Kobe University Graduate School of Science, Department of Biology, Kobe, 657-8501, Japan
| | - Mitsuhiro Morita
- Kobe University Graduate School of Science, Department of Biology, Kobe, 657-8501, Japan.
| |
Collapse
|
10
|
Liang X, Dempski RE, Burdette SC. Zn(2+) at a cellular crossroads. Curr Opin Chem Biol 2016; 31:120-5. [PMID: 27010344 PMCID: PMC4870122 DOI: 10.1016/j.cbpa.2016.02.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 02/18/2016] [Indexed: 11/27/2022]
Abstract
Zinc is an essential micronutrient for cellular homeostasis. Initially proposed to only contribute to cellular viability through structural roles and non-redox catalysis, advances in quantifying changes in nM and pM quantities of Zn(2+) have elucidated increasing functions as an important signaling molecule. This includes Zn(2+)-mediated regulation of transcription factors and subsequent protein expression, storage and release of intracellular compartments of zinc quanta into the extracellular space which modulates plasma membrane protein function, as well as intracellular signaling pathways which contribute to the immune response. This review highlights some recent advances in our understanding of zinc signaling.
Collapse
Affiliation(s)
- Xiaomeng Liang
- Worcester Polytechnic Institute, Department of Chemistry and Biochemistry, Worcester, MA 01609-2280, United States
| | - Robert E Dempski
- Worcester Polytechnic Institute, Department of Chemistry and Biochemistry, Worcester, MA 01609-2280, United States
| | - Shawn C Burdette
- Worcester Polytechnic Institute, Department of Chemistry and Biochemistry, Worcester, MA 01609-2280, United States.
| |
Collapse
|
11
|
Wolf IMA, Diercks BP, Gattkowski E, Czarniak F, Kempski J, Werner R, Schetelig D, Mittrücker HW, Schumacher V, von Osten M, Lodygin D, Flügel A, Fliegert R, Guse AH. Frontrunners of T cell activation: Initial, localized Ca2+ signals mediated by NAADP and the type 1 ryanodine receptor. Sci Signal 2015; 8:ra102. [PMID: 26462735 DOI: 10.1126/scisignal.aab0863] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The activation of T cells is the fundamental on switch for the adaptive immune system. Ca(2+) signaling is essential for T cell activation and starts as initial, short-lived, localized Ca(2+) signals. The second messenger nicotinic acid adenine dinucleotide phosphate (NAADP) forms rapidly upon T cell activation and stimulates early Ca(2+) signaling. We developed a high-resolution imaging technique using multiple fluorescent Ca(2+) indicator dyes to characterize these early signaling events and investigate the channels involved in NAADP-dependent Ca(2+) signals. In the first seconds of activation of either primary murine T cells or human Jurkat cells with beads coated with an antibody against CD3, we detected Ca(2+) signals with diameters close to the limit of detection and that were close to the activation site at the plasma membrane. In Jurkat cells in which the ryanodine receptor (RyR) was knocked down or in primary T cells from RyR1(-/-) mice, either these early Ca(2+) signals were not detected or the number of signals was markedly reduced. Local Ca(2+) signals observed within 20 ms upon microinjection of Jurkat cells with NAADP were also sensitive to RyR knockdown. In contrast, TRPM2 (transient receptor potential channel, subtype melastatin 2), a potential NAADP target channel, was not required for the formation of initial Ca(2+) signals in primary T cells. Thus, through our high-resolution imaging method, we characterized early Ca(2+) release events in T cells and obtained evidence for the involvement of RyR and NAADP in such signals.
Collapse
Affiliation(s)
- Insa M A Wolf
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Björn-Philipp Diercks
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Ellen Gattkowski
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Frederik Czarniak
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Jan Kempski
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - René Werner
- Department of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Daniel Schetelig
- Department of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Hans-Willi Mittrücker
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Valéa Schumacher
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Manuel von Osten
- Institute for Multiple Sclerosis Research, Department of Neuroimmunology, Gemeinnützige Hertie-Stiftung and University Medical Center Göttingen, Waldweg 33, 37073 Göttingen, Germany. Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Dimitri Lodygin
- Institute for Multiple Sclerosis Research, Department of Neuroimmunology, Gemeinnützige Hertie-Stiftung and University Medical Center Göttingen, Waldweg 33, 37073 Göttingen, Germany. Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Alexander Flügel
- Institute for Multiple Sclerosis Research, Department of Neuroimmunology, Gemeinnützige Hertie-Stiftung and University Medical Center Göttingen, Waldweg 33, 37073 Göttingen, Germany. Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Ralf Fliegert
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Andreas H Guse
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| |
Collapse
|
12
|
Modeling secondary messenger pathways in neurovascular coupling. Bull Math Biol 2013; 75:428-43. [PMID: 23358799 DOI: 10.1007/s11538-013-9813-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 01/08/2013] [Indexed: 10/27/2022]
Abstract
Neurovascular coupling is the well-documented link between neural stimulation and constriction or dilation of the surrounding vasculature. Glial cells mediate this response via their unique anatomy, which connects neurons to arterioles. It is believed that calcium transients and the release of secondary messengers by these cells influence the vascular response. We present a model of intracellular calcium dynamics in an astrocyte (glial cell) and show that stable oscillatory behaviour is possible under certain conditions. We then couple this to a novel model for the relationship between calcium concentration and the production of vasoactive secondary messengers through a fatty-acid intermediate. The two secondary messengers modelled are epoxyeicosatrienoic and 20-hydroxyeicosatetraenoic acids (EET and 20-HETE, respectively). These secondary messengers are produced on different time scales, and we show how this supports the observation that the vasculature dilates rapidly in response to neural stimulation, before returning to baseline levels on a slower time scale.
Collapse
|
13
|
Parys B, Côté A, Gallo V, De Koninck P, Sík A. Intercellular calcium signaling between astrocytes and oligodendrocytes via gap junctions in culture. Neuroscience 2010; 167:1032-43. [PMID: 20211698 DOI: 10.1016/j.neuroscience.2010.03.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Revised: 02/23/2010] [Accepted: 03/02/2010] [Indexed: 11/16/2022]
Abstract
To understand further how oligodendrocytes regulate brain function, the mechanism of communication between oligodendrocytes and other cell types needs to be explored. An important mode of communication between various cell types in the nervous system involves gap junctions. Astroglial cells are extensively connected through gap junctions forming the glial syncytium. Although the presence of gap junctions between oligodendrocytes and astrocytes have been well documented, evidence for gap junction-mediated calcium transfer between these two glial populations is still missing. To measure functional coupling between astrocytes and oligodendrocytes and to test whether this coupling is mediated by gap junctions we used laser photostimulation and monitored Ca(2+) propagation in cultures from transgenic animals in which oligodendrocytes express enhanced green fluorescent protein (eGFP). We show that waves of Ca(2+) spread from astrocytes to oligodendrocytes and that these waves are blocked by the broad-spectrum gap junction blocker carbenoxolone, but not the neuron-specific gap junction blocker quinine. We also show that the spread of Ca(2+) waves between astrocytes and oligodendrocytes is bi-directional. Thus, increase of Ca(2+) concentration in astrocytes triggered by surrounding neuronal activity may feed back onto different neuronal populations through oligodendrocytes.
Collapse
Affiliation(s)
- B Parys
- Centre de Recherche Université Laval Robert-Giffard; 2601, chemin de la Canardière, Quebec, QC, G1J 2G3 Canada
| | | | | | | | | |
Collapse
|
14
|
Mann ZF, Duchen MR, Gale JE. Mitochondria modulate the spatio-temporal properties of intra- and intercellular Ca2+ signals in cochlear supporting cells. Cell Calcium 2009; 46:136-46. [PMID: 19631380 DOI: 10.1016/j.ceca.2009.06.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2009] [Revised: 06/12/2009] [Accepted: 06/28/2009] [Indexed: 10/20/2022]
Abstract
In the cochlea, cell damage triggers intercellular Ca2+ waves that propagate through the glial-like supporting cells that surround receptor hair cells. These Ca2+ waves are thought to convey information about sensory hair cell-damage to the surrounding supporting cells within the cochlear epithelium. Mitochondria are key regulators of cytoplasmic Ca2+ concentration ([Ca2+](cyt)), and yet little is known about their role during the propagation of such intercellular Ca2+ signalling. Using neonatal rat cochlear explants and fluorescence imaging techniques, we explore how mitochondria modulate supporting cell [Ca2+](cyt) signals that are triggered by ATP or by hair cell damage. ATP application (0.1-50 microM) caused a dose dependent increase in [Ca2+](cyt) which was accompanied by an increase in mitochondrial calcium. Blocking mitochondrial Ca2+ uptake by dissipating the mitochondrial membrane potential using CCCP and oligomycin or using Ru360, an inhibitor of the mitochondrial Ca2+ uniporter, enhanced the peak amplitude and duration of ATP-induced [Ca2+](cyt) transients. In the presence of Ru360, the mean propagation velocity, amplitude and extent of spread of damage-induced intercellular Ca2+ waves was significantly increased. Thus, mitochondria function as spatial Ca2+ buffers during agonist-evoked [Ca2+](cyt) signalling in cochlear supporting cells and play a significant role in regulating the spatio-temporal properties of intercellular Ca2+ waves.
Collapse
Affiliation(s)
- Zoë F Mann
- UCL Ear Institute, 332 Gray's Inn Road, London WC1X 8EE, UK; Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | | | | |
Collapse
|
15
|
Smith JM, James MF, Fraser JA, Huang CLH. Translational imaging studies of cortical spreading depression in experimental models for migraine aura. Expert Rev Neurother 2008; 8:759-68. [PMID: 18457533 DOI: 10.1586/14737175.8.5.759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
This perspective discusses cortical spreading depression (CSD) phenomena and their translational significance for human migraine aura and the peri-infarct events following cerebral ischemia and injury. They begin with interstitial K(+) release and accumulation following neuronal stimulation, and a buffering astrocytic K(+) influx and remote liberation propagating waves of neuronal hyperexcitability and depression. Diffusion-weighted echoplanar MRI demonstrates CSD features in gyrencephalic brains recapitulating human migraine aura, spatial and temporal features of single primary events and multiple secondary events, their stimulus dependence, pharmacological properties, and their relationship to blood oxygenation level-dependent signals and late cerebrovascular changes. The article finally explores prospects for physiological studies of CSD gaining fuller insights both into mechanisms underlying the pathology of the corresponding human condition and possible approaches to management.
Collapse
Affiliation(s)
- Justin M Smith
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK.
| | | | | | | |
Collapse
|
16
|
Smith JM, Bradley DP, James MF, Huang CLH. Physiological studies of cortical spreading depression. Biol Rev Camb Philos Soc 2007. [DOI: 10.1111/j.1469-185x.2006.tb00214.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
17
|
Abstract
Several lines of evidence indicate that the elaborated calcium signals and the occurrence of calcium waves in astrocytes provide these cells with a specific form of excitability. The identification of the cellular and molecular steps involved in the triggering and transmission of Ca(2+) waves between astrocytes resulted in the identification of two pathways mediating this form of intercellular communication. One of them involves the direct communication between the cytosols of two adjoining cells through gap junction channels, while the other depends upon the release of "gliotransmitters" that activates membrane receptors on neighboring cells. In this review we summarize evidence in favor of these two mechanisms of Ca(2+) wave transmission and we discuss that they may not be mutually exclusive, but are likely to work in conjunction to coordinate the activity of a group of cells. To address a key question regarding the functional consequences following the passage of a Ca(2+) wave, we list, in this review, some of the potential intracellular targets of these Ca(2+) transients in astrocytes, and discuss the functional consequences of the activation of these targets for the interactions that astrocytes maintain with themselves and with other cellular partners, including those at the glial/vasculature interface and at perisynaptic sites where astrocytic processes tightly interact with neurons.
Collapse
Affiliation(s)
- Eliana Scemes
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York
| | | |
Collapse
|
18
|
Benediktsson AM, Schachtele SJ, Green SH, Dailey ME. Ballistic labeling and dynamic imaging of astrocytes in organotypic hippocampal slice cultures. J Neurosci Methods 2005; 141:41-53. [PMID: 15585287 DOI: 10.1016/j.jneumeth.2004.05.013] [Citation(s) in RCA: 143] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2004] [Revised: 05/19/2004] [Accepted: 05/19/2004] [Indexed: 10/26/2022]
Abstract
Protoplasmic astrocytes in mammalian CNS tissues in vivo have a highly complex 3D morphology, but in dissociated cell cultures they often assume a flattened, fibroblast-like morphology bearing only a few, simple processes. By fluorescent labeling and confocal reconstruction we show that many astrocytes in organotypic hippocampal slice cultures exhibit a more native complex cytoarchitecture. Although astrocytes at the surface of slice cultures show a reactive form with several thick glial fibrillary acidic protein (GFAP)-positive processes, astrocytes situated in deeper portions of tissue slices retain a highly complex 3D morphology with many fine spine- or veil-like protrusions. Dozens of astrocytes can be labeled in single slice cultures by gene gun-mediated ballistic delivery of gold or tungsten particles carrying cDNAs (Biolistics), lipophilic dyes (DiOlistics), or fluorescent intracellular calcium indicators (Calistics). Expression of a membrane-targeted form of eGFP (Lck-GFP) is superior to soluble eGFP for resolving fine astrocytic processes. Time-lapse confocal imaging of Lck-GFP transfected astrocytes or "calistically" labeled astrocytes show structural remodeling and calcium transients, respectively. This approach provides an in vitro system for investigating the functional architecture, development and dynamic remodeling of astrocytes and their relationships to neurons and glia in live mammalian brain tissues.
Collapse
Affiliation(s)
- Adrienne M Benediktsson
- Program in Neuroscience and Department of Biological Sciences, 369 Biology Building, The University of Iowa, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
19
|
Wang X, Arcuino G, Takano T, Lin J, Peng WG, Wan P, Li P, Xu Q, Liu QS, Goldman SA, Nedergaard M. P2X7 receptor inhibition improves recovery after spinal cord injury. Nat Med 2004; 10:821-7. [PMID: 15258577 DOI: 10.1038/nm1082] [Citation(s) in RCA: 409] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2004] [Accepted: 06/25/2004] [Indexed: 01/29/2023]
Abstract
Secondary injury exacerbates the extent of spinal cord insults, yet the mechanistic basis of this phenomenon has largely been unexplored. Here we report that broad regions of the peritraumatic zone are characterized by a sustained process of pathologic, high ATP release. Spinal cord neurons expressed P2X7 purine receptors (P2X7R), and exposure to ATP led to high-frequency spiking, irreversible increases in cytosolic calcium and cell death. To assess the potential effect of P2X7R blockade in ameliorating acute spinal cord injury (SCI), we delivered P2X7R antagonists OxATP or PPADS to rats after acute impact injury. We found that both OxATP and PPADS significantly improved functional recovery and diminished cell death in the peritraumatic zone. These observations demonstrate that SCI is associated with prolonged purinergic receptor activation, which results in excitotoxicity-based neuronal degeneration. P2X7R antagonists inhibit this process, reducing both the histological extent and functional sequelae of acute SCI.
Collapse
Affiliation(s)
- Xiaohai Wang
- Department of Neurosurgery, Center for Aging and Developmental Biology, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Rael LT, Thomas GW, Bar-Or R, Craun ML, Bar-Or D. An anti-inflammatory role for N-acetyl aspartate in stimulated human astroglial cells. Biochem Biophys Res Commun 2004; 319:847-53. [PMID: 15184060 DOI: 10.1016/j.bbrc.2004.04.200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2004] [Indexed: 10/26/2022]
Abstract
Although N-acetyl-L-aspartate (NAA) has been shown to be important to myelin synthesis and osmotic regulation, the biological rationale for the high levels of NAA found in the brain remains unknown. Here, a human astroglial cell line (STTG) was treated with NAA and stimulated with ionomycin, ionomycin/PMA, or IL-1beta. PGE(2) levels in ionomycin-stimulated STTG cells decreased by 76% and > 95% at NAA concentrations of 10 and 20mM, respectively. NAA also decreased the levels of COX-2 protein and activated NF-kappaB in IL-1beta-stimulated STTG cells but had little effect on unstimulated cells. Also, NAA significantly decreased intracellular calcium levels in ionomycin/PMA-stimulated cells. NAA had no effect on total COX-2 activity or COX-2 mRNA. Acetylation of IkappaBalpha kinase, an acetylation target of aspirin, was not observed when NAA was present. These results demonstrate that NAA appears to be important in the modulation of inflammation in the human STTG astroglial cell line. The results of these findings are discussed in relation to neuronal pathologies that exhibit abnormal NAA levels within the brain.
Collapse
Affiliation(s)
- Leonard T Rael
- Department of Trauma Research, Swedish Medical Center, 501 E. Hampden Avenue, Englewood, CO 80113, USA
| | | | | | | | | |
Collapse
|
21
|
Abstract
Astrocytes in the rat thalamus display spontaneous [Ca(2+)](i) oscillations that are due to intracellular release, but are not dependent on neuronal activity. In this study we have investigated the mechanisms involved in these spontaneous [Ca(2+)](i) oscillations using slices loaded with Fluo-4 AM (5 microM) and confocal microscopy. Bafilomycin A1 incubation had no effect on the number of spontaneous [Ca(2+)](i) oscillations indicating that they were not dependent on vesicular neurotransmitter release. Oscillations were also unaffected by ryanodine. Phospholipase C (PLC) inhibition decreased the number of astrocytes responding to metabotropic glutamate receptor (mGluR) activation but did not reduce the number of spontaneously active astrocytes, indicating that [Ca(2+)](i) increases are not due to membrane-coupled PLC activation. Spontaneous [Ca(2+)](i) increases were abolished by an IP3 receptor antagonist, whilst the protein kinase C (PKC) inhibitor chelerythrine chloride prolonged their duration, indicating a role for PKC and inositol 1,4,5,-triphosphate receptor activation. BayK8644 increased the number of astrocytes exhibiting [Ca(2+)](i) oscillations, and prolonged the responses to mGluR activation, indicating a possible effect on store-operated Ca(2+) entry. Increasing [Ca(2+)](o) increased the number of spontaneously active astrocytes and the number of transients exhibited by each astrocyte. Inhibition of the endoplasmic reticulum Ca(2+) ATPase by cyclopiazonic acid also induced [Ca(2+)](i) transients in astrocytes indicating a role for cytoplasmic Ca(2+) in the induction of spontaneous oscillations. Incubation with 20 microM Fluo-4 reduced the number of astrocytes exhibiting spontaneous increases. This study indicates that Ca(2+) has a role in triggering Ca(2+) release from an inositol 1,4,5,-triphosphate sensitive store in astrocytes during the generation of spontaneous [Ca(2+)](i) oscillations.
Collapse
Affiliation(s)
- H R Parri
- School of Biosciences, Cardiff University, Museum Avenue, PO Box 911, CF10 3US, Wales, Cardiff, UK.
| | | |
Collapse
|
22
|
Rintoul GL, Baimbridge KG. Effects of calcium buffers and calbindin-D28k upon histamine-induced calcium oscillations and calcium waves in HeLa cells. Cell Calcium 2003; 34:131-44. [PMID: 12810055 DOI: 10.1016/s0143-4160(03)00041-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The effects of the artificial Ca(2+) buffers EGTA and BAPTA upon histamine-induced Ca(2+) oscillations and calcium waves were studied in HeLa cells. These events were also examined in HeLa cell lines transfected with the intracellular calcium-binding protein calbindin-D28k (CaBP; HeLa-CaBP) or the pCINeo vector alone (HeLa-pCINeo). High concentrations of the Ca(2+) indicators fluo-3 and fura-2 significantly influenced the oscillatory pattern of intracellular Ca(2+) in HeLa-pCINeo cells exposed to 1 microM histamine. Loading cells with low concentrations of the cell-permeant esters of the artificial Ca(2+)-buffers EGTA or BAPTA, resulted in fewer cells with a distinct "baseline" oscillatory pattern, and loading with higher concentrations of BAPTA almost completely abolished them. In HeLa-CaBP cells, stimulation with 1 microM histamine resulted in individual Ca(2+) spikes that had a flattened profile when compared to control cells; peak [Ca(2+)](i) was lowered, the rate of increase in [Ca(2+)](i) was slower and transients were prolonged. When compared to HeLa-pCINeo cells, loading with EGTA or BAPTA, or transfection of CaBP, significantly reduced the propagation velocity (by up to 60%) of Ca(2+) waves induced by exposure to 100 microM histamine. We conclude that intracellular Ca(2+) buffering exerts a significant influence on global Ca(2+) responses in HeLa cells and the propagation of Ca(2+) waves that underlie them. The relative effectiveness of different Ca(2+) buffers, including CaBP, appears to be particularly dependent upon the rapidity of their binding kinetics, with BAPTA being the most effective.
Collapse
Affiliation(s)
- Gordon L Rintoul
- Department of Physiology, University of British Columbia, 2146 Health Sciences Mall, Vancouver, BC, Canada V6T 1Z3
| | | |
Collapse
|
23
|
Vigh J, Solessio E, Morgans CW, Lasater EM. Ionic mechanisms mediating oscillatory membrane potentials in wide-field retinal amacrine cells. J Neurophysiol 2003; 90:431-43. [PMID: 12649310 DOI: 10.1152/jn.00092.2003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Particular types of amacrine cells of the vertebrate retina show oscillatory membrane potentials (OMPs) in response to light stimulation. Historically it has been thought the oscillations arose as a result of circuit properties. In a previous study we found that in some amacrine cells, the ability to oscillate was an intrinsic property of the cell. Here we characterized the ionic mechanisms responsible for the oscillations in wide-field amacrine cells (WFACs) in an effort to better understand the functional properties of the cell. The OMPs were found to be calcium (Ca2+) dependent; blocking voltage-gated Ca2+ channels eliminated the oscillations, whereas elevating extracellular Ca2+ enhanced them. Strong intracellular Ca2+ buffering (10 mM EGTA or bis-(o-aminophenoxy)-N,N,N',N'-tetraacetic acid) eliminated any attenuation in the OMPs as well as a Ca2+-dependent inactivation of the voltage-gated Ca2+ channels. Pharmacological and immunohistochemical characterization revealed that WFACs express L- and N-type voltage-sensitive Ca2+ channels. Block of the L-type channels eliminated the OMPs, but omega-conotoxin GVIA did not, suggesting a different function for the N-type channels. The L-type channels in WFACs are functionally coupled to a set of calcium-dependent potassium (K(Ca)) channels to mediate OMPs. The initiation of OMPs depended on penitrem-A-sensitive (BK) K(Ca) channels, whereas their duration is under apamin-sensitive (SK) K(Ca) channel control. The Ca2+ current is essential to evoke the OMPs and triggering the K(Ca) currents, which here act as resonant currents, enhances the resonance as an amplifying current, influences the filtering characteristics of the cell membrane, and attenuates the OMPs via CDI of the L-type Ca2+ channel.
Collapse
Affiliation(s)
- Jozsef Vigh
- Department of Ophthalmology and Visual Sciences, John Moran Eye Center, University of Utah, Health Sciences Center, Salt Lake City, Utah 84132, USA
| | | | | | | |
Collapse
|
24
|
Vigh J, Lasater EM. Intracellular calcium release resulting from mGluR1 receptor activation modulates GABAA currents in wide-field retinal amacrine cells: a study with caffeine. Eur J Neurosci 2003; 17:2237-48. [PMID: 12814357 DOI: 10.1046/j.1460-9568.2003.02652.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The modulatory action of calcium (Ca2+) released from intracellular stores on GABAA receptor-mediated current was investigated in wide-field amacrine cells isolated from the teleost, Morone chrysops, retina. Caffeine, ryanodine or inositol 1,4,5-triphosphate (IP3) markedly inhibited the GABAA current by elevating [Ca2+]i. The inhibition resulted from the activation of a Ca2+--> Ca2+/calmodulin --> calcineurin cascade. Long (>12 s) exposure to glutamate mimicked the caffeine effect, i.e. it inhibited the GABAA current by elevating [Ca2+]i through mGluR1 receptor activation and consequent IP3 generation. This pathway provides a 'timed' disinhibitory mechanism to potentiate excitatory postsynaptic potentials in wide-field amacrine cells. It occurs as a result of the suppression of GABA-mediated conductances as a function of the duration of presynaptic excitatory input activity. This is much like some forms of long-term potentiation in the central nervous system. In a local retinal circuit this will selectively accentuate particular excitatory inputs to the wide-field amacrine cell. Similar to other neural systems, we suggest that activity-dependent postsynaptic disinhibition is an important feature of the signal processing in the inner retina.
Collapse
MESH Headings
- Amacrine Cells/drug effects
- Amacrine Cells/physiology
- Animals
- Anticoagulants/pharmacology
- Bicuculline/pharmacology
- Caffeine/pharmacology
- Calcium/metabolism
- Calcium Channels/metabolism
- Carps
- Cells, Cultured
- Central Nervous System Stimulants/pharmacology
- Chelating Agents/pharmacology
- Dose-Response Relationship, Drug
- Drug Interactions
- Egtazic Acid/analogs & derivatives
- Egtazic Acid/pharmacology
- Electric Conductivity
- Enzyme Inhibitors/pharmacology
- Excitatory Amino Acid Agonists/pharmacology
- Extracellular Space/metabolism
- GABA Antagonists/pharmacology
- Glutamic Acid/physiology
- Heparin/pharmacology
- Immunohistochemistry
- Inositol 1,4,5-Trisphosphate/pharmacology
- Inositol 1,4,5-Trisphosphate Receptors
- Kainic Acid/pharmacology
- Membrane Potentials/drug effects
- Methoxyhydroxyphenylglycol/analogs & derivatives
- Methoxyhydroxyphenylglycol/pharmacology
- Organophosphorus Compounds/pharmacology
- Patch-Clamp Techniques/methods
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, GABA-A/physiology
- Receptors, Metabotropic Glutamate/antagonists & inhibitors
- Receptors, Metabotropic Glutamate/metabolism
- Retina/cytology
- Ryanodine/pharmacology
- alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
- gamma-Aminobutyric Acid/pharmacology
Collapse
Affiliation(s)
- Jozsef Vigh
- Dept. of Ophthalmology and Visual Sciences, John Moran Eye Center, University of Utah, Health Sciences Center, Salt Lake City, UT 84132, USA
| | | |
Collapse
|
25
|
Zhang W, DeMattia JA, Song H, Couldwell WT. Communication between malignant glioma cells and vascular endothelial cells through gap junctions. J Neurosurg 2003; 98:846-53. [PMID: 12691411 DOI: 10.3171/jns.2003.98.4.0846] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Extensive invasion and angiogenesis are hallmark features of malignant gliomas. Communication between malignant glioma cells and surrounding astrocytes occurs, resulting in transformation of the astrocytic phenotype. In the present study, the authors examined whether malignant glioma cells and vascular endothelial cells (VECs) communicate through the formation of gap junctions and whether this communication influences angiogenesis. METHODS Connexin43 (Cx43), a gap junction protein expressed in glioma cells, was identified in human umbilical VECs (HUVECs). Immunocytochemical staining for Cx43 demonstrated immunoreactive plaques at areas of cell-cell contact among HUVECs as well as between HUVECs and Cx43-expressing malignant glioma cells. Dye transfer, performed using the gap junction-permeable dye dicarboxy-dichlorofluorescein diacetate (CDCF), among these cocultures indicated that these were functional communications. Calcium signaling also occurred from malignant glioma cells to HUVECs. Tube formation by HUVECs cocultured with Cx43-transfected T98G malignant glioma cells (T98G-Cx43 cells) or with U87MG malignant glioma cells, which naturally express Cx43, was significantly increased compared with tube formation by HUVECs alone. The difference in tube formation by HUVECs cocultured with empty vector-transfected T98G glioma cells (T98G-mock cells) or with Cx43-deficient U373MG malignant glioma cells and tube formation by HUVECs alone was not statistically significant. Furthermore, the concentration of vascular endothelial growth factor (VEGF), an angiogenic factor important for the induction of angiogenesis and blood vessel formation, was significantly higher in medium harvested from cultures of T98G-Cx43 cells than in that harvested from cultures of control T98G-mock cells. Human malignant glioma U87MG cells also secreted increased concentrations of VEGF as compared with HUVECs alone. Nevertheless, there was no statistically significant difference in tube formation by HUVECs cultured in medium conditioned by either Cx43-expressing or Cx43-deficient glioma cells, suggesting that the direct gap junction communication between glioma cells and HUVECs may play a much more significant role than the increased VEGF secretion in vascular tube formation in this assay. CONCLUSIONS These results indicate that functional gap junction formation between human malignant glioma cells and VECs occurs. This communication appears to influence tumor angiogenesis. Targeting gap junction signaling may offer a potential mechanism for therapy in patients with these tumors.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Neurosurgery, New York Medical College, Valhalla, New York, USA
| | | | | | | |
Collapse
|
26
|
Noguchi T, Mabuchi I. Localized calcium signals along the cleavage furrow of the Xenopus egg are not involved in cytokinesis. Mol Biol Cell 2002; 13:1263-73. [PMID: 11950937 PMCID: PMC102267 DOI: 10.1091/mbc.01-10-0501] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2001] [Revised: 12/19/2001] [Accepted: 12/24/2001] [Indexed: 11/11/2022] Open
Abstract
It has been proposed that a localized calcium (Ca) signal at the growing end of the cleavage furrow triggers cleavage furrow formation in large eggs. We have examined the possible role of a Ca signal in cleavage furrow formation in the Xenopus laevis egg during the first cleavage. We were able to detect two kinds of Ca waves along the cleavage furrow. However, the Ca waves appeared after cleavage furrow formation in late stages of the first cleavage. In addition, cleavage was not affected by injection of dibromoBAPTA or EGTA into the eggs at a concentration sufficient to suppress the Ca waves. Furthermore, even smaller classes of Ca release such as Ca puffs and Ca blips do not occur at the growing end of the cleavage furrow. These observations demonstrate that localized Ca signals in the cleavage furrow are not involved in cytokinesis. The two Ca waves have unique characteristics. The first wave propagates only in the region of newly inserted membrane along the cleavage furrow. On the other hand, the second wave propagates along the border of new and old membranes, suggesting that this wave might be involved in adhesion between two blastomeres.
Collapse
Affiliation(s)
- Tatsuhiko Noguchi
- Division of Biology, School of Arts and Sciences, University of Tokyo, Tokyo 153-8902, Japan.
| | | |
Collapse
|
27
|
Noguchi T, Arai R, Motegi F, Nakano K, Mabuchi I. Contractile ring formation in Xenopus egg and fission yeast. Cell Struct Funct 2001; 26:545-54. [PMID: 11942608 DOI: 10.1247/csf.26.545] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
How actin filaments (F-actin) and myosin II (myosin) assemble to form the contractile ring was investigated with fission yeast and Xenopus egg. In fission yeast cells, an aster-like structure composed of F-actin cables is formed at the medial cortex of the cell during prophase to metaphase, and a single F-actin cable(s) extends from this structure, which seems to be a structural basis of the contractile ring. In early mitosis, myosin localizes as dots in the medial cortex independently of F-actin. Then they fuse with each other and are packed into a thin contractile ring. At the growing ends of the cleavage furrow of Xenopus eggs, F-actin at first assembles to form patches. Next they fuse with each other to form short F-actin bundles. The short bundles then form long bundles. Myosin seems to be transported by the cortical movement to the growing end and assembles there as spots earlier than F-actin. Actin polymerization into the patches is likely to occur after accumulation of myosin. The myosin spots and the F-actin patches are simultaneously reorganized to form the contractile ring bundles. The idea that a Ca signal triggers cleavage furrow formation was tested with Xenopus eggs during the first cleavage. We could not detect any Ca signals such as a Ca wave, Ca puffs or even Ca blips at the growing end of the cleavage furrow. Furthermore, cleavages are not affected by Ca-chelators injected into the eggs at concentrations sufficient to suppress the Ca waves. Thus we conclude that formation of the contractile ring is not induced by a Ca signal at the growing end of the cleavage furrow.
Collapse
Affiliation(s)
- T Noguchi
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Japan
| | | | | | | | | |
Collapse
|
28
|
Abstract
It is often proposed that quinolinic acid (QUIN) contributes to the pathophysiology of neuroinflammation because this kynurenine pathway metabolite is a selective agonist of N-methyl-D-aspartate (NMDA) receptors, and both its brain tissue and cerebrospinal fluid concentrations increase markedly with inflammation. However, whether or not the extracellular levels of QUIN reached during neuroinflammation are high enough to promote excitotoxicity, remains unclear because QUIN is a weak NMDA receptor agonist. We have addressed this issue by evaluating the extracellular concentrations of QUIN that must be reached to initiate potentially excitotoxic changes in the cerebral cortex of rats, under normal conditions, and when superimposed on another insult. We have also examined the increase in extracellular lactate associated with the perfusion of increasing concentrations of QUIN through a microdialysis probe. The extracellular EC50 for induction of local depolarisation was 228 microM with QUIN alone; that is, about 30 times the levels of QUIN measured previously in immune activated brain. Furthermore, at least 20 microM extracellular QUIN needed to be reached to reduce K+ induced spreading depression, an unexpected effect since spreading depression is inhibited by NMDA receptor antagonists. Our data suggest that, although synthesis of QUIN from activated microglia and invading macrophages can increase its extracellular concentration 10-100-fold, the levels that are reached in these conditions remain far below the concentrations of QUIN that are necessary for excessive NMDA receptor activation. However, the possibility that QUIN accumulation may be a deleterious feature of neuroinflammation cannot be ruled out at this stage.
Collapse
Affiliation(s)
- T P Obrenovitch
- School of Pharmacy, University of Bradford, Bradford BD7 1DP, U.K.
| |
Collapse
|
29
|
Höfer T, Politi A, Heinrich R. Intercellular Ca2+ wave propagation through gap-junctional Ca2+ diffusion: a theoretical study. Biophys J 2001; 80:75-87. [PMID: 11159384 PMCID: PMC1301215 DOI: 10.1016/s0006-3495(01)75996-6] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Intercellular regenerative calcium waves in systems such as the liver and the blowfly salivary gland have been hypothesized to spread through calcium-induced calcium release (CICR) and gap-junctional calcium diffusion. A simple mathematical model of this mechanism is developed. It includes CICR and calcium removal from the cytoplasm, cytoplasmic and gap-junctional calcium diffusion, and calcium buffering. For a piecewise linear approximation of the calcium kinetics, expressions in terms of the cellular parameters are derived for 1) the condition for the propagation of intercellular waves, and 2) the characteristic time of the delay of a wave encountered at the gap junctions. Intercellular propagation relies on the local excitation of CICR in the perijunctional space by gap-junctional calcium influx. This mechanism is compatible with low effective calcium diffusivity, and necessitates that CICR can be excited in every cell along the path of a wave. The gap-junctional calcium permeability required for intercellular waves in the model falls in the range of reported gap-junctional permeability values. The concentration of diffusive cytoplasmic calcium buffers and the maximal rate of CICR, in the case of inositol 1,4,5-trisphosphate (IP3) receptor calcium release channels set by the IP(3) concentration, are shown to be further determinants of wave behavior.
Collapse
Affiliation(s)
- T Höfer
- Theoretical Biophysics, Institute of Biology, Humboldt University-Berlin, D-10115 Berlin, Germany.
| | | | | |
Collapse
|
30
|
Cotrina ML, Lin JH, López-García JC, Naus CC, Nedergaard M. ATP-mediated glia signaling. J Neurosci 2000; 20:2835-44. [PMID: 10751435 PMCID: PMC6772203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
Glia calcium signaling has recently been identified as a potent modulator of synaptic transmission. We show here that the spatial expansion of calcium waves is mediated by ATP and subsequent activation of purinergic receptors. Ectopic expression of gap junction proteins, connexins (Cxs), leads to an increase in both ATP release and the radius of calcium wave propagation. Cx expression was also associated with a phenotypic transformation, and cortical neurons extended longer neurites when co-cultured with Cx-expressing than with Cx-deficient cells. Purinergic receptor activation mediated both these effects, because treatment with receptor antagonists restored the glia phenotype and slowed neurite outgrowth. These results identify a key role of ATP in both short-term calcium signaling events and in long-term differentiation regulated by glia.
Collapse
Affiliation(s)
- M L Cotrina
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York 10595, USA
| | | | | | | | | |
Collapse
|
31
|
Paemeleire K, Leybaert L. Ionic changes accompanying astrocytic intercellular calcium waves triggered by mechanical cell damaging stimulation. Brain Res 2000; 857:235-45. [PMID: 10700572 DOI: 10.1016/s0006-8993(99)02436-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mechanically poking or damaging a single cell within a confluent astrocyte culture produces the so-called intercellular calcium (Ca(2+)) waves, that is, cell-to-cell propagating changes of intracellular free Ca(2+). We were interested whether intercellular Ca(2+) waves are also associated with changes in other intra- or extracellular ions. To that purpose, we investigated spatiotemporal changes of intracellular Ca(2+) (Ca(i)2+), sodium (Na(i)+) and protons (H(i)+) in primary cultures of rat cortical astrocytes using microfluorescence imaging with fura-2, SBFI and BCECF, respectively; changes of extracellular potassium (K(e)+) were monitored with K(+)-sensitive microelectrodes. Mechanical damage to a single cell by stimulation with a piezo-electrically driven micropipette initiated intercellular Ca(2+) waves that propagated to about 160 microm away from the stimulation point. Na(i)(+) increases could be detected in cells located 2-3 cell diameters from the stimulated cell, acidification was observed 1-2 cell diameters away and Ke(+) increases were measured up to 75 microm away. Kinetic analysis suggests that the Na(i)(+) and H(i)(+) changes occur after, and thus secondary to the Ca(i)(2+) changes. In contrast, K(e)(+) changes occurred very fast, even before the Ca(i)(2+) changes, but their propagation speed was too fast to implicate them as a trigger of Ca(i)(2+) changes. As Na(i)(+) is an important regulator of glycolysis in astrocytes, we hypothesize that astrocytic Na(i)(+) changes in cells located remotely from a damaged cell might be a signal that activates glycolysis thereby producing more lactate that is transferred to the neurons and increases their energy potential to survive the inflicted damage.
Collapse
Affiliation(s)
- K Paemeleire
- Department of Physiology and Pathophysiology, University of Ghent, De Pintelaan 185 (Blok B), B-9000, Ghent, Belgium
| | | |
Collapse
|
32
|
Nagy JI, Dermietzel R. Gap junctions and connexins in the mammalian central nervous system. GAP JUNCTIONS 2000. [DOI: 10.1016/s1569-2558(00)30009-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
33
|
Simard M, Couldwell WT, Zhang W, Song H, Liu S, Cotrina ML, Goldman S, Nedergaard M. Glucocorticoids-potent modulators of astrocytic calcium signaling. Glia 1999; 28:1-12. [PMID: 10498817 DOI: 10.1002/(sici)1098-1136(199910)28:1<1::aid-glia1>3.0.co;2-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Glucocorticoids are the first line of choice in the treatment of cerebral edema associated with brain tumors. High-dose glucocorticoids reduce the extent of edema within hours, often relieving critical increases in intracranial pressure, but the mechanisms by which glucocorticoids modulate brain water content are not well-understood. A possible target of action may be glucocorticoid receptor-expressing astrocytes, which are the primary regulators of interstitial ion homeostasis in brain. In this study, we demonstrate that two glucocorticoids, methylprednisolone and dexamethasone, potentiate astrocytic signaling, via long-range calcium waves. Glucocorticoid treatment increased both resting cytosolic calcium (Ca2+i) level and the extent and amplitude of Ca2+ wave propagation two-fold, compared to matched controls. RU-486, a potent steroid receptor antagonist, inhibited the effects of methylprednisolone. The glucocorticoid-associated potentiation of Ca2+ signaling may result from upregulation of the cellular ability to mobilize Ca2+ and release ATP, because both agonist-induced Ca2+i increments (via ATP and bradykinin) and ATP release were proportionally enhanced by glucocorticoids. In contrast, neither gap junction expression (as manifested connexin 43 immunoreactivity) nor functional coupling was significantly affected by methylprednisolone. Confocal microscopy revealed both the expression of glucocorticoid receptors and nuclear translocation of these receptors when exposed to methylprednisolone. We postulate that the edemolytic effects of glucocorticoids may result from enhanced astrocytic calcium signaling.
Collapse
Affiliation(s)
- M Simard
- Department of Neurosurgery, New York Medical College, Valhalla, New York 10595, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Boitier E, Rea R, Duchen MR. Mitochondria exert a negative feedback on the propagation of intracellular Ca2+ waves in rat cortical astrocytes. J Cell Biol 1999; 145:795-808. [PMID: 10330407 PMCID: PMC2133193 DOI: 10.1083/jcb.145.4.795] [Citation(s) in RCA: 232] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/1998] [Revised: 03/19/1999] [Indexed: 11/22/2022] Open
Abstract
We have used digital fluorescence imaging techniques to explore the interplay between mitochondrial Ca2+ uptake and physiological Ca2+ signaling in rat cortical astrocytes. A rise in cytosolic Ca2+ ([Ca2+]cyt), resulting from mobilization of ER Ca2+ stores was followed by a rise in mitochondrial Ca2+ ([Ca2+]m, monitored using rhod-2). Whereas [Ca2+]cyt recovered within approximately 1 min, the time to recovery for [Ca2+]m was approximately 30 min. Dissipating the mitochondrial membrane potential (Deltapsim, using the mitochondrial uncoupler carbonyl cyanide p-trifluoromethoxy-phenyl-hydrazone [FCCP] with oligomycin) prevented mitochondrial Ca2+ uptake and slowed the rate of decay of [Ca2+]cyt transients, suggesting that mitochondrial Ca2+ uptake plays a significant role in the clearance of physiological [Ca2+]cyt loads in astrocytes. Ca2+ signals in these cells initiated either by receptor-mediated ER Ca2+ release or mechanical stimulation often consisted of propagating waves (measured using fluo-3). In response to either stimulus, the wave traveled at a mean speed of 22.9 +/- 11.2 micrometer/s (n = 262). This was followed by a wave of mitochondrial depolarization (measured using tetramethylrhodamine ethyl ester [TMRE]), consistent with Ca2+ uptake into mitochondria as the Ca2+ wave traveled across the cell. Collapse of Deltapsim to prevent mitochondrial Ca2+ uptake significantly increased the rate of propagation of the Ca2+ waves by 50%. Taken together, these data suggest that cytosolic Ca2+ buffering by mitochondria provides a potent mechanism to regulate the localized spread of astrocytic Ca2+ signals.
Collapse
Affiliation(s)
- E Boitier
- Department of Physiology, University College London, London, WC1E 6BT, United Kingdom
| | | | | |
Collapse
|
35
|
Guthrie PB, Knappenberger J, Segal M, Bennett MV, Charles AC, Kater SB. ATP released from astrocytes mediates glial calcium waves. J Neurosci 1999; 19:520-8. [PMID: 9880572 PMCID: PMC6782195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Calcium waves represent a widespread form of intercellular communication. Although they have been thought for a long time to require gap junctions, we recently demonstrated that mouse cortical astrocytes use an extracellular messenger for calcium wave propagation. The present experiments identify ATP as a major extracellular messenger in this system. Medium collected from astrocyte cultures during (but not before) calcium wave stimulation contains ATP. The excitatory effects of medium samples and of ATP are blocked by purinergic receptor antagonists and by pretreatment with apyrase; these same purinergic receptor antagonists block propagation of electrically evoked calcium waves. ATP, applied at the concentration measured in medium samples, evokes responses that are qualitatively and quantitatively similar to those evoked by those medium samples. These data implicate ATP as an important transmitter between CNS astrocytes.
Collapse
Affiliation(s)
- P B Guthrie
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah 84132, USA
| | | | | | | | | | | |
Collapse
|
36
|
Cotrina ML, Lin JH, Alves-Rodrigues A, Liu S, Li J, Azmi-Ghadimi H, Kang J, Naus CC, Nedergaard M. Connexins regulate calcium signaling by controlling ATP release. Proc Natl Acad Sci U S A 1998; 95:15735-40. [PMID: 9861039 PMCID: PMC28113 DOI: 10.1073/pnas.95.26.15735] [Citation(s) in RCA: 624] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/1998] [Accepted: 10/26/1998] [Indexed: 01/07/2023] Open
Abstract
Forced expression of gap junction proteins, connexins, enables gap junction-deficient cell lines to propagate intercellular calcium waves. Here, we show that ATP secretion from the poorly coupled cell lines, C6 glioma, HeLa, and U373 glioblastoma, is potentiated 5- to 15-fold by connexin expression. ATP release required purinergic receptor-activated intracellular Ca2+ mobilization and was inhibited by Cl- channel blockers. Calcium wave propagation also was reduced by purinergic receptor antagonists and by Cl- channel blockers but insensitive to gap junction inhibitors. These observations suggest that cell-to-cell signaling associated with connexin expression results from enhanced ATP release and not, as previously believed, from an increase in intercellular coupling.
Collapse
Affiliation(s)
- M L Cotrina
- Departments of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 10595, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kodavanti PR, Derr-Yellin EC, Mundy WR, Shafer TJ, Herr DW, Barone S, Choksi NY, MacPhail RC, Tilson HA. Repeated exposure of adult rats to Aroclor 1254 causes brain region-specific changes in intracellular Ca2+ buffering and protein kinase C activity in the absence of changes in tyrosine hydroxylase. Toxicol Appl Pharmacol 1998; 153:186-98. [PMID: 9878590 DOI: 10.1006/taap.1998.8533] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Polychlorinated biphenyls (PCBs) are ubiquitous environmental contaminants, some of which may be neurotoxic. In vitro studies from this laboratory indicated that noncoplanar PCBs perturbed intracellular signal transduction mechanisms including Ca2+ homeostasis, receptor-mediated inositol phosphate production, and translocation of protein kinase C (PKC). In the present study, we examined the effects of PCBs in vivo by dosing adult male Long-Evans rats orally with Aroclor 1254 (0, 10, or 30 mg/kg/day; 5 days/week for 4 weeks) in corn oil. At 24 h after the last dose, rats were tested for motor activity in a photocell device for 30 min. Immediately, the rats were euthanized, blood was collected for thyroid hormone analysis, and brains were removed, dissected into regions (cerebellum, frontal cortex, and striatum), and subcellular fractions were obtained for neurochemical analysis. Following Aroclor 1254 treatment, body weight gain in the high-dose group was significantly lower than the control and low-dose groups. Horizontal motor activity was significantly lower in rats dosed with 30 mg/kg Aroclor 1254. Ca2+ buffering by microsomes was significantly lower in all three brain regions from the 30 mg/kg group. In the same dose group, mitochondrial Ca2+ buffering was affected in cerebellum but not in cortex or striatum. Similarly, total cerebellar PKC activity was decreased significantly while membrane-bound PKC activity was significantly elevated at 10 and 30 mg/kg. PKC activity was not altered either in cortex or the striatum. Neurotransmitter levels in striatum or cortex were slightly altered in PCB-exposed rats compared to controls. Furthermore, repeated oral administration of Aroclor 1254 to rats did not significantly alter forebrain tyrosine hydroxylase immunoreactivity or enzymatic activity. Circulating T4 (total and free) concentrations were severely depressed at both doses in Aroclor 1254-exposed rats compared to control rats, suggesting a severe hypothyroid state. These results indicate that (1) in vivo exposure to a PCB mixture can produce changes in second messenger systems that are similar to those observed after in vitro exposure of neuronal cell cultures; (2) second messenger systems seem to be more sensitive than alterations in neurotransmitter levels or tyrosine hydroxylase involved in dopamine synthesis during repeated exposure to PCBs; and (3) the observed motor activity changes were independent of changes in striatal dopamine levels.
Collapse
Affiliation(s)
- P R Kodavanti
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, 27711, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
|
39
|
Kunkler PE, Kraig RP. Calcium waves precede electrophysiological changes of spreading depression in hippocampal organ cultures. J Neurosci 1998; 18:3416-25. [PMID: 9547248 PMCID: PMC2699599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/1997] [Revised: 01/27/1998] [Accepted: 02/23/1998] [Indexed: 02/07/2023] Open
Abstract
Although intercellular Ca2+ waves resemble spreading depression (SD) and occur in hippocampal organ cultures (HOTCs), SD has not been reported in these cultures. Accordingly, electrophysiological and Ca2+ imaging techniques were used to examine potential interrelations between Ca2+ waves and electrophysiological changes of SD. Our results show, for the first time, that HOTCs can support SD. Furthermore, two distinct Ca2+ waves were found to precede SD. The first traveled >100 micron/sec along the pyramidal cell dendritic layer. The second subsequently traveled mostly perpendicular to the pyramidal cell layer from CA3 (or CA1) but also in all directions from its area of initiation. This second, slower wave spread with the interstitial DC change of SD at millimeters per minute but always ahead of it by 6-16 sec. Heptanol, which uncouples gap junctions, blocked both of these Ca2+ waves and SD. Thus, two types of Ca2+ waves occur with the initiation and propagation of SD. The first might reflect interneuronal changes linked by gap junctions, whereas the second might stem from interastrocyte changes linked via similar connections. Because individual cells can be followed in space and time for protracted periods in HOTCs, this preparation may be ideal for studies designed to explore not only the mechanisms of SD but also the long-term consequences of SD, such as ischemic tolerance.
Collapse
Affiliation(s)
- P E Kunkler
- Department of Neurology, The University of Chicago, Chicago, Illinois 60637, USA
| | | |
Collapse
|
40
|
Cotrina ML, Kang J, Lin JH, Bueno E, Hansen TW, He L, Liu Y, Nedergaard M. Astrocytic gap junctions remain open during ischemic conditions. J Neurosci 1998; 18:2520-37. [PMID: 9502812 PMCID: PMC6793088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/1997] [Revised: 12/22/1997] [Accepted: 12/29/1997] [Indexed: 02/06/2023] Open
Abstract
Gap junctions are highly conductive channels that allow the direct transfer of intracellular messengers such as Ca2+ and inositol triphosphate (IP3) between interconnected cells. In brain, astrocytes are coupled extensively by gap junctions. We found here that gap junctions among astrocytes in acutely prepared brain slices as well as in culture remained open during ischemic conditions. Uncoupling first occurred after the terminal loss of plasma membrane integrity. Gap junctions therefore may link ischemic astrocytes in an evolving infarct with the surroundings. The free exchange of intracellular messengers between dying and potentially viable astrocytes might contribute to secondary expansion of ischemic lesions.
Collapse
Affiliation(s)
- M L Cotrina
- Department of Cell Biology, New York Medical College, Valhalla, New York 10595, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Tymianski M, Bernstein GM, Abdel-Hamid KM, Sattler R, Velumian A, Carlen PL, Razavi H, Jones OT. A novel use for a carbodiimide compound for the fixation of fluorescent and non-fluorescent calcium indicators in situ following physiological experiments. Cell Calcium 1997; 21:175-83. [PMID: 9105727 DOI: 10.1016/s0143-4160(97)90042-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The inability to determine the precise intracellular location of non-fluorescent organic calcium chelators such as BAPTA is a persistent problem which has precluded much detailed analysis of the chelators' spatial or temporal dynamics in live cells. Similarly, following physiological experiments with fluorescent indicators like Fura-2, it has often been desirable to maintain the dye within the cell for later analysis by additional histological techniques. Based on chemical considerations, and its prior use in tissue fixation, we examined the water soluble reagent 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC) as a potential fixative for diverse calcium chelators. The utility of EDC, but not other common fixatives, was confirmed through electrophysiological means, through a novel ELISA, which exploits anti-BAPTA antibodies to assess the extent and kinetics of fixation; by autoradiography of neurons loaded with [14C]-BAPTA, and by immunocytochemistry and imaging of intracellular BAPTA or Calcium Green in neurons. At concentrations > 0.1 mg/ml, EDC caused virtually instantaneous, irreversible, fixation of > 95% of BAPTA free acid. Fixation of intracellular BAPTA was confirmed in hippocampal brain slices loaded with BAPTA/AM ester, and showed biphasic kinetics consistent with rapid loading and subsequent extrusion of the chelator. Immunocytochemistry on neurons microinjected with BAPTA free acid and the dye Lucifer Yellow showed BAPTA-specific staining which was distributed in the cell similarly to that of the accompanying marker dye. Application of EDC also efficiently fixed in situ analogs of BAPTA such as Calcium Green (a fluorescent Ca2+ indicator) as shown by confocal imaging of EDC-fixed brain slices loaded with this indicator. Taken together, these data show that EDC is an effective, inexpensive and versatile fixative for calcium chelators in diverse cells. The availability of a suitable fixative now makes it possible to determine the distributions of such chelators at both the light and, possibly, the electron microscope level. Two important features of EDC, arise from its specificity for free carboxyl groups. First, the ability to fix, selectively, the chelators but not their AM esters; and, second, its enormous potential as a fixative for the numerous other carboxyl-containing chelators, dyes and pH indicators currently available.
Collapse
Affiliation(s)
- M Tymianski
- Playfair Neuroscience Unit, Toronto Hospital Research Institute, Toronto Western Hospital, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|