1
|
Bonsignore G, Martinotti S, Ranzato E. Wound Repair and Ca 2+ Signalling Interplay: The Role of Ca 2+ Channels in Skin. Cells 2024; 13:491. [PMID: 38534335 PMCID: PMC10969298 DOI: 10.3390/cells13060491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/02/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024] Open
Abstract
The process of wound healing is intricate and tightly controlled, involving a number of different cellular and molecular processes. Numerous cellular functions, especially those related to wound healing, depend critically on calcium ions (Ca2+). Ca2+ channels are proteins involved in signal transduction and communication inside cells that allow calcium ions to pass through cell membranes. Key Ca2+ channel types involved in wound repair are described in this review.
Collapse
Affiliation(s)
- Gregorio Bonsignore
- Dipartimento di Scienze e Innovazione Tecnologica (DiSIT), University of Piemonte Orientale, 15121 Alessandria, Italy; (G.B.); (S.M.)
| | - Simona Martinotti
- Dipartimento di Scienze e Innovazione Tecnologica (DiSIT), University of Piemonte Orientale, 15121 Alessandria, Italy; (G.B.); (S.M.)
- SSD Laboratori di Ricerca—DAIRI, Azienda Ospedaliero-Universitaria SS. Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy
| | - Elia Ranzato
- Dipartimento di Scienze e Innovazione Tecnologica (DiSIT), University of Piemonte Orientale, 15121 Alessandria, Italy; (G.B.); (S.M.)
- SSD Laboratori di Ricerca—DAIRI, Azienda Ospedaliero-Universitaria SS. Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy
| |
Collapse
|
2
|
Romagnolo A, Dematteis G, Scheper M, Luinenburg MJ, Mühlebner A, Van Hecke W, Manfredi M, De Giorgis V, Reano S, Filigheddu N, Bortolotto V, Tapella L, Anink JJ, François L, Dedeurwaerdere S, Mills JD, Genazzani AA, Lim D, Aronica E. Astroglial calcium signaling and homeostasis in tuberous sclerosis complex. Acta Neuropathol 2024; 147:48. [PMID: 38418708 PMCID: PMC10901927 DOI: 10.1007/s00401-024-02711-3] [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: 12/06/2023] [Revised: 02/12/2024] [Accepted: 02/20/2024] [Indexed: 03/02/2024]
Abstract
Tuberous Sclerosis Complex (TSC) is a multisystem genetic disorder characterized by the development of benign tumors in various organs, including the brain, and is often accompanied by epilepsy, neurodevelopmental comorbidities including intellectual disability and autism. A key hallmark of TSC is the hyperactivation of the mechanistic target of rapamycin (mTOR) signaling pathway, which induces alterations in cortical development and metabolic processes in astrocytes, among other cellular functions. These changes could modulate seizure susceptibility, contributing to the progression of epilepsy and its associated comorbidities. Epilepsy is characterized by dysregulation of calcium (Ca2+) channels and intracellular Ca2+ dynamics. These factors contribute to hyperexcitability, disrupted synaptogenesis, and altered synchronization of neuronal networks, all of which contribute to seizure activity. This study investigates the intricate interplay between altered Ca2+ dynamics, mTOR pathway dysregulation, and cellular metabolism in astrocytes. The transcriptional profile of TSC patients revealed significant alterations in pathways associated with cellular respiration, ER and mitochondria, and Ca2+ regulation. TSC astrocytes exhibited lack of responsiveness to various stimuli, compromised oxygen consumption rate and reserve respiratory capacity underscoring their reduced capacity to react to environmental changes or cellular stress. Furthermore, our study revealed significant reduction of store operated calcium entry (SOCE) along with strong decrease of basal mitochondrial Ca2+ concentration and Ca2+ influx in TSC astrocytes. In addition, we observed alteration in mitochondrial membrane potential, characterized by increased depolarization in TSC astrocytes. Lastly, we provide initial evidence of structural abnormalities in mitochondria within TSC patient-derived astrocytes, suggesting a potential link between disrupted Ca2+ signaling and mitochondrial dysfunction. Our findings underscore the complexity of the relationship between Ca2+ signaling, mitochondria dynamics, apoptosis, and mTOR hyperactivation. Further exploration is required to shed light on the pathophysiology of TSC and on TSC associated neuropsychiatric disorders offering further potential avenues for therapeutic development.
Collapse
Affiliation(s)
- Alessia Romagnolo
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.
| | - Giulia Dematteis
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Mirte Scheper
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Mark J Luinenburg
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Angelika Mühlebner
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wim Van Hecke
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marcello Manfredi
- Center on Autoimmune and Allergic Diseases (CAAD), UPO, Novara, Italy
- Department of Translational Medicine, UPO, Novara, Italy
| | - Veronica De Giorgis
- Center on Autoimmune and Allergic Diseases (CAAD), UPO, Novara, Italy
- Department of Translational Medicine, UPO, Novara, Italy
| | - Simone Reano
- Center on Autoimmune and Allergic Diseases (CAAD), UPO, Novara, Italy
| | | | - Valeria Bortolotto
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Laura Tapella
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Jasper J Anink
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Liesbeth François
- Neurosciences Therapeutic Area, UCB Pharma, Braine-L'Alleud, Belgium
| | | | - James D Mills
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Clinical and Experimental Epilepsy, UCL, London, UK
- Chalfont Centre for Epilepsy, Chalfont St Peter, UK
| | - Armando A Genazzani
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Eleonora Aronica
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| |
Collapse
|
3
|
Gong X, Ogino N, Leite MF, Chen Z, Nguyen R, Liu R, Kruglov E, Flores K, Cabral A, Mendes GMM, Ehrlich BE, Mak M. Adaptation to volumetric compression drives hepatoblastoma cells to an apoptosis-resistant and invasive phenotype. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.08.561453. [PMID: 37873476 PMCID: PMC10592664 DOI: 10.1101/2023.10.08.561453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Liver cancer involves tumor cells rapidly growing within a packed tissue environment. Patient tumor tissues reveal densely packed and deformed cells, especially at tumor boundaries, indicative of physical crowding and compression. It is not well understood how these physical signals modulate tumor evolution and therapeutic susceptibility. Here we investigate the impact of volumetric compression on liver cancer (HepG2) behavior. We find that conditioning cells under a highly compressed state leads to major transcriptional reprogramming, notably the loss of hepatic markers, the epithelial-to-mesenchymal transition (EMT)-like changes, and altered calcium signaling-related gene expression, over the course of several days. Biophysically, compressed cells exhibit increased Rac1-mediated cell spreading and cell-extracellular matrix interactions, cytoskeletal reorganization, increased YAP and β-catenin nuclear translocation, and dysfunction in cytoplasmic and mitochondrial calcium signaling. Furthermore, compressed cells are resistant to chemotherapeutics and desensitized to apoptosis signaling. Apoptosis sensitivity can be rescued by stimulated calcium signaling. Our study demonstrates that volumetric compression is a key microenvironmental factor that drives tumor evolution in multiple pathological directions and highlights potential countermeasures to re-sensitize therapy-resistant cells. Significance statement Compression can arise as cancer cells grow and navigate within the dense solid tumor microenvironment. It is unclear how compression mediates critical programs that drive tumor progression and therapeutic complications. Here, we take an integrative approach in investigating the impact of compression on liver cancer. We identify and characterize compressed subdomains within patient tumor tissues. Furthermore, using in vitro systems, we induce volumetric compression (primarily via osmotic pressure but also via mechanical force) on liver cancer cells and demonstrate significant molecular and biophysical changes in cell states, including in function, cytoskeletal signaling, proliferation, invasion, and chemoresistance. Importantly, our results show that compressed cells have impaired calcium signaling and acquire resistance to apoptosis, which can be countered via calcium mobilization.
Collapse
|
4
|
Wang M, Sun Y, Li L, Wu P, Dkw O, Shi H. Calcium Channels: Noteworthy Regulators and Therapeutic Targets in Dermatological Diseases. Front Pharmacol 2021; 12:702264. [PMID: 34489697 PMCID: PMC8418299 DOI: 10.3389/fphar.2021.702264] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/02/2021] [Indexed: 02/05/2023] Open
Abstract
Dysfunctional skin barrier and impaired skin homeostasis may lead to or aggravate a series of dermatologic diseases. A large variety of biological events and bioactive molecules are involved in the process of skin wound healing and functional recovery. Calcium ions (Ca2+) released from intracellular stores as well as influx through plasma membrane are essential to skin function. Growing evidence suggests that calcium influx is mainly regulated by calcium-sensing receptors and channels, including voltage-gated, transient potential receptor, store-operated, and receptor-operated calcium channels, which not only maintain cellular Ca2+ homeostasis, but also participate in cell proliferation and skin cell homeostasis through Ca2+-sensitive proteins such as calmodulin (CaM). Furthermore, distinct types of Ca2+ channels not merely work separately, they may work concertedly to regulate cell function. In this review, we discussed different calcium-sensing receptors and channels, including voltage-gated, transient receptor potential, store-operated, and receptor-operated calcium channels, particularly focusing on their regulatory functions and inherent interactions as well as calcium channels-related reagents and drugs, which is expected to bridge basic research and clinical applications in dermatologic diseases.
Collapse
Affiliation(s)
- Min Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Institute of Stem Cell, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yaoxiang Sun
- Department of Clinical Laboratory, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Linli Li
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Institute of Stem Cell, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Peipei Wu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Institute of Stem Cell, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Ocansey Dkw
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Institute of Stem Cell, School of Medicine, Jiangsu University, Zhenjiang, China.,Directorate of University Health Services, University of Cape Coast, Cape Coast, Ghana
| | - Hui Shi
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Institute of Stem Cell, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
5
|
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
|
6
|
Distelhorst CW. Targeting Bcl-2-IP 3 receptor interaction to treat cancer: A novel approach inspired by nearly a century treating cancer with adrenal corticosteroid hormones. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1795-1804. [PMID: 30053503 DOI: 10.1016/j.bbamcr.2018.07.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/18/2018] [Accepted: 07/19/2018] [Indexed: 12/12/2022]
Abstract
Bcl-2 inhibits cell death by at least two different mechanisms. On the one hand, its BH3 domain binds to pro-apoptotic proteins such as Bim and prevents apoptosis induction. On the other hand, the BH4 domain of Bcl-2 binds to the inositol 1,4,5-trisphosphate receptor (IP3R), preventing Ca2+ signals that mediate cell death. In normal T-cells, Bcl-2 levels increase during the immune response, protecting against cell death, and then decline as apoptosis ensues and the immune response dissipates. But in many cancers Bcl-2 is aberrantly expressed and exploited to prevent cell death by inhibiting IP3R-mediated Ca2+ elevation. This review summarizes what is known about the mechanism of Bcl-2's control over IP3R-mediated Ca2+ release and cell death induction. Early insights into the role of Ca2+ elevation in corticosteroid-mediated cell death serves as a model for how targeting IP3R-mediated Ca2+ elevation can be a highly effective therapeutic approach for different types of cancer. Moreover, the successful development of ABT-199 (Venetoclax), a small molecule targeting the BH3 domain of Bcl-2 but without effects on Ca2+, serves as proof of principle that targeting Bcl-2 can be an effective therapeutic approach. BIRD-2, a synthetic peptide that inhibits Bcl-2-IP3R interaction, induces cell death induction in ABT-199 (Venetoclax)-resistant cancer models, attesting to the value of developing therapeutic agents that selectively target Bcl-2-IP3R interaction, inducing Ca2+-mediated cell death.
Collapse
Affiliation(s)
- Clark W Distelhorst
- Case Western University School of Medicine, Case Comprehensive Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, United States of America.
| |
Collapse
|
7
|
Structural basis of dual Ca 2+/pH regulation of the endolysosomal TRPML1 channel. Nat Struct Mol Biol 2017; 24:205-213. [PMID: 28112729 PMCID: PMC5336481 DOI: 10.1038/nsmb.3362] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 12/12/2016] [Indexed: 12/01/2022]
Abstract
Organellar ion channels are essential for cell physiology. Their activities are often regulated by Ca2+ and H+, which are concentrated in many organelles. Here we report a novel structural element critical for Ca2+/pH dual regulation of TRPML1, a Ca2+ release channel crucial for endolysosomal functions. TRPML1 mutations cause mucolipidosis type IV (MLIV), a severe lysosomal storage disorder characterized by neurodegeneration, mental retardation and blindness. We obtained high-resolution crystal structures of a 213-amino acid luminal domain of human TRPML1 that harbors three missense MLIV-causing mutations. This domain forms a tetramer with a highly electronegative central pore formed by a novel luminal pore-loop. Cysteine crosslinking and cryo-EM confirm this structure in the full-length channel. Structure-function studies demonstrate that Ca2+ and H+ interact with the luminal pore to exert physiologically important regulation. The MLIV-causing mutations disrupt the luminal domain structure and cause TRPML1 mislocalization. Our study provides a structural underpinning for TRPML1's regulation, assembly and pathogenesis.
Collapse
|
8
|
Tyul’kova EI, Vataeva LA, Vetrovoi OV, Romanovskii DY. Prenatal hypoxia modifies working memory and the activity of hippocampal polyphosphoinositide system in rats. J EVOL BIOCHEM PHYS+ 2015. [DOI: 10.1134/s0022093015020064] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
9
|
Greenberg EF, Lavik AR, Distelhorst CW. Bcl-2 regulation of the inositol 1,4,5-trisphosphate receptor and calcium signaling in normal and malignant lymphocytes: potential new target for cancer treatment. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1843:2205-10. [PMID: 24642270 PMCID: PMC4119508 DOI: 10.1016/j.bbamcr.2014.03.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 03/07/2014] [Accepted: 03/10/2014] [Indexed: 01/23/2023]
Abstract
The anti-apoptotic protein Bcl-2 is a versatile regulator of cell survival. Its interactions with its own pro-apoptotic family members are widely recognized for their role in promoting the survival of cancer cells. These interactions are thus being targeted for cancer treatment. Less widely recognized is the interaction of Bcl-2 with the inositol 1,4,5-trisphosphate receptor (InsP3R), an InsP3-gated Ca(2+) channel located on the endoplasmic reticulum. The nature of this interaction, the mechanism by which it controls Ca(2+) release from the ER, its role in T-cell development and survival, and the possibility of targeting it as a novel cancer treatment strategy are summarized in this review. This article is part of a Special Issue entitled: Calcium signaling in health and disease. Guest Editors: Geert Bultynck, Jacques Haiech, Claus W. Heizmann, Joachim Krebs, and Marc Moreau.
Collapse
Affiliation(s)
- Edward F Greenberg
- Division of Hematology/Oncology, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, University Hospitals Case Medical Center, USA; MetroHealth Medical Center, USA.
| | - Andrew R Lavik
- Division of Hematology/Oncology, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, University Hospitals Case Medical Center, USA.
| | - Clark W Distelhorst
- Division of Hematology/Oncology, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, University Hospitals Case Medical Center, USA.
| |
Collapse
|
10
|
Vais H, Foskett JK, Ullah G, Pearson JE, Mak DOD. Permeant calcium ion feed-through regulation of single inositol 1,4,5-trisphosphate receptor channel gating. ACTA ACUST UNITED AC 2012; 140:697-716. [PMID: 23148262 PMCID: PMC3514735 DOI: 10.1085/jgp.201210804] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The ubiquitous inositol 1,4,5-trisphosphate (InsP(3)) receptor (InsP(3)R) Ca(2+) release channel plays a central role in the generation and modulation of intracellular Ca(2+) signals, and is intricately regulated by multiple mechanisms including cytoplasmic ligand (InsP(3), free Ca(2+), free ATP(4-)) binding, posttranslational modifications, and interactions with cytoplasmic and endoplasmic reticulum (ER) luminal proteins. However, regulation of InsP(3)R channel activity by free Ca(2+) in the ER lumen ([Ca(2+)](ER)) remains poorly understood because of limitations of Ca(2+) flux measurements and imaging techniques. Here, we used nuclear patch-clamp experiments in excised luminal-side-out configuration with perfusion solution exchange to study the effects of [Ca(2+)](ER) on homotetrameric rat type 3 InsP(3)R channel activity. In optimal [Ca(2+)](i) and subsaturating [InsP(3)], jumps of [Ca(2+)](ER) from 70 nM to 300 µM reduced channel activity significantly. This inhibition was abrogated by saturating InsP(3) but restored when [Ca(2+)](ER) was raised to 1.1 mM. In suboptimal [Ca(2+)](i), jumps of [Ca(2+)](ER) (70 nM to 300 µM) enhanced channel activity. Thus, [Ca(2+)](ER) effects on channel activity exhibited a biphasic dependence on [Ca(2+)](i). In addition, the effect of high [Ca(2+)](ER) was attenuated when a voltage was applied to oppose Ca(2+) flux through the channel. These observations can be accounted for by Ca(2+) flux driven through the open InsP(3)R channel by [Ca(2+)](ER), raising local [Ca(2+)](i) around the channel to regulate its activity through its cytoplasmic regulatory Ca(2+)-binding sites. Importantly, [Ca(2+)](ER) regulation of InsP(3)R channel activity depended on cytoplasmic Ca(2+)-buffering conditions: it was more pronounced when [Ca(2+)](i) was weakly buffered but completely abolished in strong Ca(2+)-buffering conditions. With strong cytoplasmic buffering and Ca(2+) flux sufficiently reduced by applied voltage, both activation and inhibition of InsP(3)R channel gating by physiological levels of [Ca(2+)](ER) were completely abolished. Collectively, these results rule out Ca(2+) regulation of channel activity by direct binding to the luminal aspect of the channel.
Collapse
Affiliation(s)
- Horia Vais
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
11
|
Abstract
Ca2+ transfer from endoplasmic reticulum (ER) to mitochondria can trigger apoptotic pathways by inducing release of mitochondrial pro-apoptotic factors. Three different types of inositol 1,4,5-trisphosphate receptor (IP3R) serve to discharge Ca2+ from ER, but possess some peculiarities, especially in apoptosis induction. The anti-apoptotic protein Akt can phosphorylate all IP3R isoforms and protect cells from apoptosis, reducing ER Ca2+ release. However, it has not been elucidated which IP3R subtypes mediate these effects. Here, we show that Akt activation in COS7 cells, which lack of IP3R I, strongly suppresses IP3-mediated Ca2+ release and apoptosis. Conversely, in SH-SY 5Y cells, which are type III-deficient, Akt is unable to modulate ER Ca2+ flux, losing its anti-apoptotic activity. In SH-SY 5Y-expressing subtype III, Akt recovers its protective function on cell death, by reduction of Ca2+ release. Moreover, regulating Ca2+ flux to mitochondria, Akt maintains the mitochondrial integrity and delays the trigger of apoptosis, in a type III-dependent mechanism. These results demonstrate a specific activity of Akt on IP3R III, leading to diminished Ca2+ transfer to mitochondria and protection from apoptosis, suggesting an additional level of cell death regulation mediated by Akt.
Collapse
|
12
|
Narayanan D, Adebiyi A, Jaggar JH. Inositol trisphosphate receptors in smooth muscle cells. Am J Physiol Heart Circ Physiol 2012; 302:H2190-210. [PMID: 22447942 DOI: 10.1152/ajpheart.01146.2011] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Inositol 1,4,5-trisphosphate receptors (IP(3)Rs) are a family of tetrameric intracellular calcium (Ca(2+)) release channels that are located on the sarcoplasmic reticulum (SR) membrane of virtually all mammalian cell types, including smooth muscle cells (SMC). Here, we have reviewed literature investigating IP(3)R expression, cellular localization, tissue distribution, activity regulation, communication with ion channels and organelles, generation of Ca(2+) signals, modulation of physiological functions, and alterations in pathologies in SMCs. Three IP(3)R isoforms have been identified, with relative expression and cellular localization of each contributing to signaling differences in diverse SMC types. Several endogenous ligands, kinases, proteins, and other modulators control SMC IP(3)R channel activity. SMC IP(3)Rs communicate with nearby ryanodine-sensitive Ca(2+) channels and mitochondria to influence SR Ca(2+) release and reactive oxygen species generation. IP(3)R-mediated Ca(2+) release can stimulate plasma membrane-localized channels, including transient receptor potential (TRP) channels and store-operated Ca(2+) channels. SMC IP(3)Rs also signal to other proteins via SR Ca(2+) release-independent mechanisms through physical coupling to TRP channels and local communication with large-conductance Ca(2+)-activated potassium channels. IP(3)R-mediated Ca(2+) release generates a wide variety of intracellular Ca(2+) signals, which vary with respect to frequency, amplitude, spatial, and temporal properties. IP(3)R signaling controls multiple SMC functions, including contraction, gene expression, migration, and proliferation. IP(3)R expression and cellular signaling are altered in several SMC diseases, notably asthma, atherosclerosis, diabetes, and hypertension. In summary, IP(3)R-mediated pathways control diverse SMC physiological functions, with pathological alterations in IP(3)R signaling contributing to disease.
Collapse
Affiliation(s)
- Damodaran Narayanan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, 38163, USA
| | | | | |
Collapse
|
13
|
Sensory neurons derived from diabetic rats have diminished internal Ca2+ stores linked to impaired re-uptake by the endoplasmic reticulum. ASN Neuro 2012; 4:AN20110038. [PMID: 22168362 PMCID: PMC3260471 DOI: 10.1042/an20110038] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Distal symmetrical sensory neuropathy in diabetes involves the dying back of axons, and the pathology equates with axonal dystrophy generated under conditions of aberrant Ca2+ signalling. Previous work has described abnormalities in Ca2+ homoeostasis in sensory and dorsal horn neurons acutely isolated from diabetic rodents. We extended this work by testing the hypothesis that sensory neurons exposed to long-term Type 1 diabetes in vivo would exhibit abnormal axonal Ca2+ homoeostasis and focused on the role of SERCA (sarcoplasmic/endoplasmic reticulum Ca2+-ATPase). DRG (dorsal root ganglia) sensory neurons from age-matched normal and 3-5-month-old STZ (streptozotocin)-diabetic rats (an experimental model of Type 1 diabetes) were cultured. At 1-2 days in vitro an array of parameters were measured to investigate Ca2+ homoeostasis including (i) axonal levels of intracellular Ca2+, (ii) Ca2+ uptake by the ER (endoplasmic reticulum), (iii) assessment of Ca2+ signalling following a long-term thapsigargin-induced blockade of SERCA and (iv) determination of expression of ER mass and stress markers using immunocytochemistry and Western blotting. KCl- and caffeine-induced Ca2+ transients in axons were 2-fold lower in cultures of diabetic neurons compared with normal neurons indicative of reduced ER calcium loading. The rate of uptake of Ca2+ into the ER was reduced by 2-fold (P<0.05) in diabetic neurons, while markers for ER mass and ER stress were unchanged. Abnormalities in Ca2+ homoeostasis in diabetic neurons could be mimicked via long-term inhibition of SERCA in normal neurons. In summary, axons of neurons from diabetic rats exhibited aberrant Ca2+ homoeostasis possibly triggered by sub-optimal SERCA activity that could contribute to the distal axonopathy observed in diabetes.
Collapse
|
14
|
Ondrias K, Lencesova L, Sirova M, Labudova M, Pastorekova S, Kopacek J, Krizanova O. Apoptosis induced clustering of IP(3)R1 in nuclei of non-differentiated PC12 cells. J Cell Physiol 2011; 226:3147-55. [PMID: 21302308 DOI: 10.1002/jcp.22665] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Inositol 1,4,5-trisphosphate (IP(3)) receptors are emerging as key sites for regulation by pro- and anti-apoptotic factors. Induction of apoptosis for 3 h increased mRNA and protein levels of type 1 IP(3) receptors in non-differentiated (ND), but not in differentiated (D) PC12 cells. Inhibitors of the IP(3) R's calcium release-2-aminoethoxydiphenyl borate (2-APB) and xestospongin-completely prevented Bax and caspase-3 mRNA increase after treatment with the apoptosis inducer set (AIK), and this reinforces the importance of IP(3) R1 in the apoptosis of ND PC12 cells. Apoptosis induction not only increases the IP(3) R1 protein, but it also causes formation of IP(3) R1 clusters in the nucleus which most likely result from fusion of the nucleoplasmic reticulum and/or IP(3) R1 translocation to the nucleus. This is quite similar to the observations noted after overexpression of IP(3) R1 in PC12 cells. The amount of IP(3) induced calcium release was higher in control than in AIK-treated cells. From our results we propose that after the apoptosis induction the amount of intranuclear calcium decreased dramatically due to the increase of calcium permeability of the nuclear calcium store vesicles. Therefore, increase of the calcium permeability may result from IP(3) receptors translocation to nuclei that can boost the calcium transport through IP(3) receptors.
Collapse
Affiliation(s)
- Karol Ondrias
- Institute of Molecular Physiology and Genetics, Centre of Excellence for Cardiovascular Research, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | | | | | | | | | | | | |
Collapse
|
15
|
Stutzmann GE, Mattson MP. Endoplasmic reticulum Ca(2+) handling in excitable cells in health and disease. Pharmacol Rev 2011; 63:700-27. [PMID: 21737534 PMCID: PMC3141879 DOI: 10.1124/pr.110.003814] [Citation(s) in RCA: 195] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The endoplasmic reticulum (ER) is a morphologically and functionally diverse organelle capable of integrating multiple extracellular and internal signals and generating adaptive cellular responses. It plays fundamental roles in protein synthesis and folding and in cellular responses to metabolic and proteotoxic stress. In addition, the ER stores and releases Ca(2+) in sophisticated scenarios that regulate a range of processes in excitable cells throughout the body, including muscle contraction and relaxation, endocrine regulation of metabolism, learning and memory, and cell death. One or more Ca(2+) ATPases and two types of ER membrane Ca(2+) channels (inositol trisphosphate and ryanodine receptors) are the major proteins involved in ER Ca(2+) uptake and release, respectively. There are also direct and indirect interactions of ER Ca(2+) stores with plasma membrane and mitochondrial Ca(2+)-regulating systems. Pharmacological agents that selectively modify ER Ca(2+) release or uptake have enabled studies that revealed many different physiological roles for ER Ca(2+) signaling. Several inherited diseases are caused by mutations in ER Ca(2+)-regulating proteins, and perturbed ER Ca(2+) homeostasis is implicated in a range of acquired disorders. Preclinical investigations suggest a therapeutic potential for use of agents that target ER Ca(2+) handling systems of excitable cells in disorders ranging from cardiac arrhythmias and skeletal muscle myopathies to Alzheimer disease.
Collapse
Affiliation(s)
- Grace E Stutzmann
- Department of Neuroscience, Rosalind Franklin University/The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL 60064, USA.
| | | |
Collapse
|
16
|
Bcl-2 interaction with the inositol 1,4,5-trisphosphate receptor: role in Ca(2+) signaling and disease. Cell Calcium 2011; 50:234-41. [PMID: 21628070 DOI: 10.1016/j.ceca.2011.05.011] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 05/09/2011] [Accepted: 05/10/2011] [Indexed: 01/02/2023]
Abstract
The Bcl-2 protein, best known for its ability to inhibit apoptosis, interacts with the inositol 1,4,5-trisphosphate receptor (IP(3)R) Ca(2+) channel to regulate IP(3)-mediated Ca(2+) release from the endoplasmic reticulum. This review summarizes the current state of knowledge regarding the interaction of Bcl-2, and also its homologue Bcl-xl, with the IP(3)R and how these interactions regulate Ca(2+) signaling. The dual role of these interactions in promoting prosurvival Ca(2+) signals, while at the same time inhibiting proapoptotic Ca(2+) signals, is discussed. Moreover, this review will elucidate the recently recognized importance of the Bcl-2-IP(3)R interaction in human disease.
Collapse
|
17
|
Higo T, Hamada K, Hisatsune C, Nukina N, Hashikawa T, Hattori M, Nakamura T, Mikoshiba K. Mechanism of ER Stress-Induced Brain Damage by IP3 Receptor. Neuron 2010; 68:865-78. [DOI: 10.1016/j.neuron.2010.11.010] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2010] [Indexed: 11/25/2022]
|
18
|
Zhao G, Neeb ZP, Leo MD, Pachuau J, Adebiyi A, Ouyang K, Chen J, Jaggar JH. Type 1 IP3 receptors activate BKCa channels via local molecular coupling in arterial smooth muscle cells. J Gen Physiol 2010; 136:283-91. [PMID: 20713546 PMCID: PMC2931145 DOI: 10.1085/jgp.201010453] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Accepted: 07/28/2010] [Indexed: 11/20/2022] Open
Abstract
Plasma membrane large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels and sarcoplasmic reticulum inositol 1,4,5-trisphosphate (IP(3)) receptors (IP(3)Rs) are expressed in a wide variety of cell types, including arterial smooth muscle cells. Here, we studied BK(Ca) channel regulation by IP(3) and IP(3)Rs in rat and mouse cerebral artery smooth muscle cells. IP(3) activated BK(Ca) channels both in intact cells and in excised inside-out membrane patches. IP(3) caused concentration-dependent BK(Ca) channel activation with an apparent dissociation constant (K(d)) of approximately 4 microM at physiological voltage (-40 mV) and intracellular Ca(2+) concentration ([Ca(2+)](i); 10 microM). IP(3) also caused a leftward-shift in BK(Ca) channel apparent Ca(2+) sensitivity and reduced the K(d) for free [Ca(2+)](i) from approximately 20 to 12 microM, but did not alter the slope or maximal P(o). BAPTA, a fast Ca(2+) buffer, or an elevation in extracellular Ca(2+) concentration did not alter IP(3)-induced BK(Ca) channel activation. Heparin, an IP(3)R inhibitor, and a monoclonal type 1 IP(3)R (IP(3)R1) antibody blocked IP(3)-induced BK(Ca) channel activation. Adenophostin A, an IP(3)R agonist, also activated BK(Ca) channels. IP(3) activated BK(Ca) channels in inside-out patches from wild-type (IP(3)R1(+/+)) mouse arterial smooth muscle cells, but had no effect on BK(Ca) channels of IP(3)R1-deficient (IP(3)R1(-/-)) mice. Immunofluorescence resonance energy transfer microscopy indicated that IP(3)R1 is located in close spatial proximity to BK(Ca) alpha subunits. The IP(3)R1 monoclonal antibody coimmunoprecipitated IP(3)R1 and BK(Ca) channel alpha and beta1 subunits from cerebral arteries. In summary, data indicate that IP(3)R1 activation elevates BK(Ca) channel apparent Ca(2+) sensitivity through local molecular coupling in arterial smooth muscle cells.
Collapse
MESH Headings
- Adenosine/analogs & derivatives
- Adenosine/pharmacology
- Animals
- Antibodies, Monoclonal/pharmacology
- Aorta/metabolism
- Calcium Channels/deficiency
- Calcium Channels/drug effects
- Calcium Channels/genetics
- Calcium Channels/metabolism
- Calcium Signaling/drug effects
- Cell Membrane/metabolism
- Cerebral Arteries/metabolism
- Chelating Agents/pharmacology
- Egtazic Acid/analogs & derivatives
- Egtazic Acid/pharmacology
- Fluorescence Resonance Energy Transfer
- Fluorescent Antibody Technique
- Heparin/pharmacology
- Immunoprecipitation
- Inositol 1,4,5-Trisphosphate Receptors
- Ion Channel Gating
- Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects
- Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism
- Male
- Membrane Potentials
- Mice
- Mice, Knockout
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Patch-Clamp Techniques
- Potassium Channels/drug effects
- Potassium Channels/metabolism
- Protein Binding
- Rats
- Rats, Sprague-Dawley
- Receptors, Cytoplasmic and Nuclear/deficiency
- Receptors, Cytoplasmic and Nuclear/drug effects
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Sarcoplasmic Reticulum/metabolism
- Time Factors
Collapse
Affiliation(s)
- Guiling Zhao
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Zachary P. Neeb
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - M. Dennis Leo
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Judith Pachuau
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Adebowale Adebiyi
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Kunfu Ouyang
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Ju Chen
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Jonathan H. Jaggar
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| |
Collapse
|
19
|
Abstract
The versatility of Ca(2+) as an intracellular messenger derives largely from the spatial organization of cytosolic Ca(2+) signals, most of which are generated by regulated openings of Ca(2+)-permeable channels. Most Ca(2+) channels are expressed in the plasma membrane (PM). Others, including the almost ubiquitous inositol 1,4,5-trisphosphate receptors (IP(3)R) and their relatives, the ryanodine receptors (RyR), are predominantly expressed in membranes of the sarcoplasmic or endoplasmic reticulum (ER). Targeting of these channels to appropriate destinations underpins their ability to generate spatially organized Ca(2+) signals. All Ca(2+) channels begin life in the cytosol, and the vast majority are then functionally assembled in the ER, where they may either remain or be dispatched to other membranes. Here, by means of selective examples, we review two issues related to this trafficking of Ca(2+) channels via the ER. How do cells avoid wayward activity of Ca(2+) channels in transit as they pass from the ER via other membranes to their final destination? How and why do some cells express small numbers of the archetypal intracellular Ca(2+) channels, IP(3)R and RyR, in the PM?
Collapse
Affiliation(s)
- Colin W Taylor
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, UK.
| | | | | |
Collapse
|
20
|
Cohen R, Torres A, Ma HT, Holowka D, Baird B. Ca2+ waves initiate antigen-stimulated Ca2+ responses in mast cells. THE JOURNAL OF IMMUNOLOGY 2009; 183:6478-88. [PMID: 19864608 DOI: 10.4049/jimmunol.0901615] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ca(2+) mobilization is central to many cellular processes, including stimulated exocytosis and cytokine production in mast cells. Using single cell stimulation by IgE-specific Ag and high-speed imaging of conventional or genetically encoded Ca(2+) sensors in rat basophilic leukemia and bone marrow-derived rat mast cells, we observe Ca(2+) waves that originate most frequently from the tips of extended cell protrusions, as well as Ca(2+) oscillations throughout the cell that usually follow the initiating Ca(2+) wave. In contrast, Ag conjugated to the tip of a micropipette stimulates local, repetitive Ca(2+) puffs at the region of cell contact. Initiating Ca(2+) waves are observed in most rat basophilic leukemia cells stimulated with soluble Ag and are sensitive to inhibitors of Ca(2+) release from endoplasmic reticulum stores and to extracellular Ca(2+), but they do not depend on store-operated Ca(2+) entry. Knockdown of transient receptor potential channel (TRPC)1 and TRPC3 channel proteins by short hairpin RNA reduces the sensitivity of these cells to Ag and shifts the wave initiation site from protrusions to the cell body. Our results reveal spatially encoded Ca(2+) signaling in response to immunoreceptor activation that utilizes TRPC channels to specify the initiation site of the Ca(2+) response.
Collapse
Affiliation(s)
- Roy Cohen
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14850, USA
| | | | | | | | | |
Collapse
|
21
|
Yehuda-Shnaidman E, Kalderon B, Azazmeh N, Bar-Tana J. Gating of the mitochondrial permeability transition pore by thyroid hormone. FASEB J 2009; 24:93-104. [PMID: 19723706 DOI: 10.1096/fj.09-133538] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The calorigenic-thermogenic activity of thyroid hormone (T3) has long been ascribed to uncoupling of mitochondrial oxidative phosphorylation. However, the mode of action of T3 in promoting mitochondrial proton leak is still unresolved. Mitochondrial uncoupling by T3 is reported here to be transduced in vivo in rats and in cultured Jurkat cells by gating of the mitochondrial permeability transition pore (PTP). T3-induced PTP gating is shown here to be abrogated in inositol 1,4,5-trisphosphate (IP(3)) receptor 1 (IP(3)R1)(-/-) cells, indicating that the endoplasmic reticulum IP(3)R1 may serve as upstream target for the mitochondrial activity of T3. IP(3)R1 gating by T3 is due to its increased expression and truncation into channel-only peptides, resulting in IP(3)-independent Ca(2+) efflux. Increased cytosolic Ca(2+) results in activation of protein phosphatase 2B, dephosphorylation and depletion of mitochondrial Bcl2 (S70), and increase in mitochondrial free Bax leading to low-conductance PTP gating. The T3 transduction pathway integrates genomic and nongenomic activities of T3 in regulating mitochondrial energetics and may offer novel targets for thyromimetics designed to modulate energy expenditure.
Collapse
Affiliation(s)
- Einav Yehuda-Shnaidman
- Department of Human Nutrition and Metabolism, Hebrew University Medical School, Jerusalem, Israel
| | | | | | | |
Collapse
|
22
|
Cooper Z, Greenwood M, Mazzag B. A computational analysis of localized Ca2+-dynamics generated by heterogeneous release sites. Bull Math Biol 2009; 71:1543-79. [PMID: 19440797 DOI: 10.1007/s11538-009-9413-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2007] [Accepted: 02/10/2009] [Indexed: 11/30/2022]
Abstract
We investigate the role of heterogeneous expression of IP3R and RyR in generating diverse elementary Ca2+ signals. It has been shown empirically (Wojcikiewicz and Luo in Mol. Pharmacol. 53(4):656-662, 1998; Newton et al. in J. Biol. Chem. 269(46):28613-28619, 1994; Smedt et al. in Biochem. J. 322(Pt. 2):575-583, 1997) that tissues express various proportions of IP3 and RyR isoforms and this expression is dynamically regulated (Parrington et al. in Dev. Biol. 203(2):451-461, 1998; Fissore et al. in Biol. Reprod. 60(1):49-57, 1999; Tovey et al. in J. Cell Sci. 114(Pt. 22):3979-3989, 2001). Although many previous theoretical studies have investigated the dynamics of localized calcium release sites (Swillens et al. in Proc. Natl. Acad. Sci. U.S.A. 96(24):13750-13755, 1999; Shuai and Jung in Proc. Natl. Acad. Sci. U.S.A. 100(2):506-510, 2003a; Shuai and Jung in Phys. Rev. E, Stat. Nonlinear Soft Matter Phys. 67(3 Pt. 1):031905, 2003b; Thul and Falcke in Biophys. J. 86(5):2660-2673, 2004; DeRemigio and Smith in Cell Calcium 38(2):73-86, 2005; Nguyen et al. in Bull. Math. Biol. 67(3):393-432, 2005), so far all such studies focused on release sites consisting of identical channel types. We have extended an existing mathematical model (Nguyen et al. in Bull. Math. Biol. 67(3):393-432, 2005) to release sites with two (or more) receptor types, each with its distinct channel kinetics. Mathematically, the release site is represented by a transition probability matrix for a collection of nonidentical stochastically gating channels coupled through a shared Ca2+ domain. We demonstrate that under certain conditions a previously defined mean-field approximation of the coupling strength does not accurately reproduce the release site dynamics. We develop a novel approximation and establish that its performance in these instances is superior. We use this mathematical framework to study the effect of heterogeneity in the Ca2+-regulation of two colocalized channel types on the release site dynamics. We consider release sites consisting of channels with both Ca2+-activation and inactivation ("four-state channels") and channels with Ca2+-activation only ("two-state channels") and show that for the appropriate parameter values, synchronous channel openings within a release site with any proportion of two-state to four-state channels are possible, however, the larger the proportion of two-state channels, the more sensitive the dynamics are to the exact spatial positioning of the channels and the distance between channels. Specifically, the clustering of even a small number of two-state channels interferes with puff/spark termination and increases puff durations or leads to a tonic response.
Collapse
Affiliation(s)
- Zachary Cooper
- Department of Mathematics, Humboldt State University, Arcata, CA 95521, USA
| | | | | |
Collapse
|
23
|
Antigny F, Norez C, Cantereau A, Becq F, Vandebrouck C. Abnormal spatial diffusion of Ca2+ in F508del-CFTR airway epithelial cells. Respir Res 2008; 9:70. [PMID: 18973672 PMCID: PMC2584091 DOI: 10.1186/1465-9921-9-70] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Accepted: 10/30/2008] [Indexed: 11/25/2022] Open
Abstract
Background In airway epithelial cells, calcium mobilization can be elicited by selective autocrine and/or paracrine activation of apical or basolateral membrane heterotrimeric G protein-coupled receptors linked to phospholipase C (PLC) stimulation, which generates inositol 1,4,5-trisphosphate (IP3) and 1,2-diacylglycerol (DAG) and induces Ca2+ release from endoplasmic reticulum (ER) stores. Methods In the present study, we monitored the cytosolic Ca2+ transients using the UV light photolysis technique to uncage caged Ca2+ or caged IP3 into the cytosol of loaded airway epithelial cells of cystic fibrosis (CF) and non-CF origin. We compared in these cells the types of Ca2+ receptors present in the ER, and measured their Ca2+ dependent activity before and after correction of F508del-CFTR abnormal trafficking either by low temperature or by the pharmacological corrector miglustat (N-butyldeoxynojirimycin). Results We showed reduction of the inositol 1,4,5-trisphosphate receptors (IP3R) dependent-Ca2+ response following both correcting treatments compared to uncorrected cells in such a way that Ca2+ responses (CF+treatment vs wild-type cells) were normalized. This normalization of the Ca2+ rate does not affect the activity of Ca2+-dependent chloride channel in miglustat-treated CF cells. Using two inhibitors of IP3R1, we observed a decrease of the implication of IP3R1 in the Ca2+ response in CF corrected cells. We observed a similar Ca2+ mobilization between CF-KM4 cells and CFTR-cDNA transfected CF cells (CF-KM4-reverted). When we restored the F508del-CFTR trafficking in CFTR-reverted cells, the specific IP3R activity was also reduced to a similar level as in non CF cells. At the structural level, the ER morphology of CF cells was highly condensed around the nucleus while in non CF cells or corrected CF cells the ER was extended at the totality of cell. Conclusion These results suggest reversal of the IP3R dysfunction in F508del-CFTR epithelial cells by correction of the abnormal trafficking of F508del-CFTR in cystic fibrosis cells. Moreover, using CFTR cDNA-transfected CF cells, we demonstrated that abnormal increase of IP3R Ca2+ release in CF human epithelial cells could be the consequence of F508del-CFTR retention in ER compartment.
Collapse
Affiliation(s)
- Fabrice Antigny
- Institut de Physiologie et Biologie Cellulaires, Université de Poitiers, CNRS, 86022 Poitiers, France.
| | | | | | | | | |
Collapse
|
24
|
Paredes-Gamero EJ, Leon CMMP, Borojevic R, Oshiro MEM, Ferreira AT. Changes in intracellular Ca2+ levels induced by cytokines and P2 agonists differentially modulate proliferation or commitment with macrophage differentiation in murine hematopoietic cells. J Biol Chem 2008; 283:31909-19. [PMID: 18775989 DOI: 10.1074/jbc.m801990200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The role of intracellular Ca2+ (Ca2+i) on hematopoiesis was investigated in long term bone marrow cultures using cytokines and agonists of P2 receptors. Cytokines interleukin 3 and granulocyte/macrophage colony stimulator factor promoted a modest increase in Ca2+i concentration ([Ca2+]i) with activation of phospholipase Cgamma, MEK1/2, and Ca2+/calmodulin kinase II. Involvement of protein kinase C was restricted to stimulation with interleukin 3. In addition, these cytokines promoted proliferation (20 times) and an increase in the Gr-1(-)Mac-1+ population with participation of gap junctions (GJ). Nevertheless ATP, ADP, and UTP promoted a large increase in [Ca2+]i, moderate proliferation (6 times), a reduction in the primitive Gr-1(-)Mac-1(-)c-Kit+ population, and differentiation into macrophages without participation of GJ. It is likely that Ca2+i participates as a regulator of hematopoietic signaling: moderate increases in [Ca2+]i would be related to cytokine-dependent proliferation with participation of GJ, whereas high increases in [Ca2+]i would be related to macrophage differentiation without maintenance of the primitive population.
Collapse
Affiliation(s)
- Edgar J Paredes-Gamero
- Department of Biophysics, Federal University of São Paulo, Rua Botucatu 862, 04023-062 São Paulo, Brazil.
| | | | | | | | | |
Collapse
|
25
|
Heidrich FM, Zhang K, Estrada M, Huang Y, Giordano FJ, Ehrlich BE. Chromogranin B regulates calcium signaling, nuclear factor kappaB activity, and brain natriuretic peptide production in cardiomyocytes. Circ Res 2008; 102:1230-8. [PMID: 18420944 PMCID: PMC2952358 DOI: 10.1161/circresaha.107.166033] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Altered regulation of signaling pathways can lead to pathologies including cardiac hypertrophy and heart failure. We report that neonatal and adult cardiomyocytes express chromogranin B (CGB), a Ca(2+) binding protein that modulates Ca(2+) release by the inositol 1,4,5-trisphosphate receptor (InsP(3)R). Using fluorescent Ca(2+) indicator dyes, we found that CGB regulates InsP(3)-dependent Ca(2+) release in response to angiotensin II, an octapeptide hormone that promotes cardiac hypertrophy. ELISA experiments and luciferase reporter assays identified angiotensin II as a potent inducer of brain natriuretic peptide (BNP), a hormone that recently emerged as an important biomarker in cardiovascular disease. CGB was found to regulate angiotensin II-stimulated and basal secretion, expression and promoter activity of BNP that depend on the InsP(3)R. Moreover, we provide evidence that CGB acts via the transcription activity of nuclear factor kappaB in an InsP(3)/Ca(2+)-dependent manner but independent of nuclear factor of activated T cells. In vivo experiments further showed that cardiac hypertrophy induced by angiotensin II, a condition characterized by increased ventricular BNP production, is associated with upregulation of ventricular CGB expression. Overexpression of CGB in cardiomyocytes, in turn, induced the BNP promoter. The evidence presented in this study identifies CGB as a novel regulator of cardiomyocyte InsP(3)/Ca(2+)-dependent signaling, nuclear factor kappaB activity, and BNP production.
Collapse
Affiliation(s)
- Felix M. Heidrich
- Department of Pharmacology, Yale University, New Haven, CT, 06520, USA
- Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, 01307, Germany
| | - Kun Zhang
- Department of Pharmacology, Yale University, New Haven, CT, 06520, USA
- Neuroscience Research Centre, Charité Universitätsmedizin Berlin, Berlin, 10117, Germany
| | - Manuel Estrada
- Department of Physiology and Biophysics, University of Chile, Santiago, Chile
| | - Yan Huang
- Department of Medicine/Cardiology, Yale University, New Haven, CT, 06520, USA
| | - Frank J. Giordano
- Department of Medicine/Cardiology, Yale University, New Haven, CT, 06520, USA
| | | |
Collapse
|
26
|
Rong Y, Distelhorst CW. Bcl-2 protein family members: versatile regulators of calcium signaling in cell survival and apoptosis. Annu Rev Physiol 2008; 70:73-91. [PMID: 17680735 DOI: 10.1146/annurev.physiol.70.021507.105852] [Citation(s) in RCA: 282] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Bcl-2 family members are important regulators of cell survival and cell death. Researchers have focused mainly on mitochondria, where both proapoptotic and antiapoptotic family members function to regulate the release of cytochrome c and other mediators of apoptosis. However, as reviewed here, Bcl-2 family members also operate on another front, the endoplasmic reticulum (ER), to both positively and negatively regulate the release of Ca2+. There is abundant evidence that Ca2+ signals trigger apoptosis in response to a wide variety of agents and conditions. Conversely, Ca2+ signals can also mediate cell survival. Recent findings indicate that Bcl-2 interacts with inositol 1,4,5-trisphosphate (IP3) receptor Ca2+ channels on the ER, regulating their opening in response to IP3- and thus inhibiting IP3-mediated Ca2+ signals that induce apoptosis while enhancing Ca2+ signals that support cell survival.
Collapse
Affiliation(s)
- Yiping Rong
- Department of Medicine and Pharmacology, Comprehensive Cancer Center and University Hospitals of Cleveland, Case Western Reserve University, Cleveland, OH 44106, USA.
| | | |
Collapse
|
27
|
Acetylcholine-induced Ca2+ oscillations are modulated by a Ca2+ regulation of InsP3R2 in rat portal vein myocytes. Pflugers Arch 2007; 456:277-83. [PMID: 18026983 DOI: 10.1007/s00424-007-0379-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2007] [Revised: 10/20/2007] [Accepted: 10/25/2007] [Indexed: 12/14/2022]
Abstract
Oscillations of cytosolic Ca2+ levels are believed to have important roles in various metabolic and signalling processes in many cell types. Previously, we have demonstrated that acetylcholine (ACh) evokes Ca2+ oscillations in vascular myocytes expressing InsP3R1 and InsP3R2, whereas transient responses are activated in vascular myocytes expressing InsP3R1 alone. The molecular mechanisms underlying oscillations remain to be described in these native smooth muscle cells. Two major hypotheses are proposed to explain this crucial signalling activity: (1) Ca2+ oscillations are activated by InsP3 oscillations; and (2) Ca2+ oscillations depend on the regulation of the InsP3R by both InsP3 and Ca2+. In the present study, we used a fluorescent InsP3 biosensor and revealed that ACh induced a transient InsP3 production in all myocytes. Moreover, steady concentrations of 3F-InsP3, a poorly hydrolysable analogue of InsP3, and pharmacological activation of PLC evoked Ca2+ oscillations. Increasing cytosolic Ca2+ inhibited the ACh-induced calcium oscillations but not the transient responses and strongly reduced the 3F-InsP3-evoked Ca2+ response in oscillating cells but not in non-oscillating cells. These results suggest that, in native vascular myocytes, ACh-induced InsP3 production is transient and Ca2+ oscillations depend on a Ca2+ modulation of InsP3R2.
Collapse
|
28
|
Abstract
Ca2+ is a ubiquitous cellular signal. Altered expression of specific Ca2+ channels and pumps are characterizing features of some cancers. The ability of Ca2+ to regulate both cell death and proliferation, combined with the potential for pharmacological modulation, offers the opportunity for a set of new drug targets in cancer. However, the ubiquity of the Ca2+ signal is often mistakenly presumed to thwart the specific therapeutic targeting of proteins that transport Ca2+. This Review presents evidence to the contrary and addresses the question: which Ca2+ channels and pumps should be targeted?
Collapse
Affiliation(s)
- Gregory R Monteith
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia, 4072.
| | | | | | | |
Collapse
|
29
|
Nicolay NH, Hertle D, Boehmerle W, Heidrich FM, Yeckel M, Ehrlich BE. Inositol 1,4,5 trisphosphate receptor and chromogranin B are concentrated in different regions of the hippocampus. J Neurosci Res 2007; 85:2026-36. [PMID: 17471556 PMCID: PMC2945619 DOI: 10.1002/jnr.21328] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Calcium (Ca(2+)) release from intracellular stores plays a crucial role in many cellular functions in the brain. These intracellular signals have been shown to be transmitted within and between cells. We report a non-uniform distribution of proteins essential for Ca(2+) signaling in acutely prepared brain slice preparations and organotypic slice cultures, both made from rat hippocampus. The Type I inositol-1,4,5 trisphosphate receptor (InsP(3)R1) is the main InsP(3)R subtype in neurons. Immunohistochemistry experiments showed a prominent expression of InsP(3)R1 in the CA1 region of the hippocampus whereas the CA3 region and dentate gyrus (DG) showed only moderate immunoreactivity. In contrast, chromogranin B (CGB), a protein binding to the InsP(3)R1 on the luminal side of the endoplasmic reticular membrane was enriched in the CA3 region whereas DG and the CA1 region showed only faint CGB signals. The neuronal kinases leading to the formation of inositol-1,4,5 trisphosphate (InsP(3)), phosphatidylinositol-4-kinase (PI4K), and phosphatidylinositol-4-phosphate-5-kinase (PIPK), showed strong immunoreactivity throughout all hippocampal cell fields with differences in the subcellular distribution. Moreover, a distinct band of strong CGB and PIPK immunoreactivity was observed in the CA3 region that coincides with the mossy fiber tract (stratum lucidum). These data show differential expression of the components of the signaling toolkit leading to InsP(3)-mediated Ca(2+) release in cells of the hippocampus. The regulation of these differences may play an important role in various neuropathologic conditions such as Alzheimer's disease, epilepsy, or schizophrenia.
Collapse
Affiliation(s)
- Nils H. Nicolay
- Departments of Pharmacology, Cellular and Molecular Physiology, Yale University, New Haven, Connecticut
- Department of Pharmacology, University of Heidelberg, Heidelberg, Germany
| | - Daniel Hertle
- Department of Neurobiology, Yale University, New Haven, Connecticut
| | - Wolfgang Boehmerle
- Departments of Pharmacology, Cellular and Molecular Physiology, Yale University, New Haven, Connecticut
- Neuroscience Research Centre, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Felix M. Heidrich
- Departments of Pharmacology, Cellular and Molecular Physiology, Yale University, New Haven, Connecticut
- Department of Pharmacology and Toxicology, University of Technology Dresden, Dresden, Germany
| | - Mark Yeckel
- Department of Neurobiology, Yale University, New Haven, Connecticut
| | - Barbara E. Ehrlich
- Departments of Pharmacology, Cellular and Molecular Physiology, Yale University, New Haven, Connecticut
| |
Collapse
|
30
|
Siefjediers A, Hardt M, Prinz G, Diener M. Characterization of inositol 1,4,5-trisphosphate (IP3) receptor subtypes at rat colonic epithelium. Cell Calcium 2007; 41:303-15. [PMID: 16950509 DOI: 10.1016/j.ceca.2006.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2006] [Revised: 05/15/2006] [Accepted: 07/05/2006] [Indexed: 10/24/2022]
Abstract
The aim of the present study was the characterization of the subtypes of inositol 1,4,5-trisphosphate receptors (IP3R) in rat colonic epithelium. A monoclonal antibody against IP3R1 did not stain the colonic epithelial cells. In contrast, IP3R2 and IP3R3 were found within the epithelium; however, with a distinct intracellular localization and differences in their distribution along the crypt axis. IP3R2 immunoreactivity was found within the nuclei of the epithelial cells. The signal was distributed all over the nucleus and not restricted to the nuclear envelope as demonstrated by counterstaining with lamin B1 and electron microscopical examination after immunogold labelling. In contrast, an antibody against IP3R3 stained the epithelial cells mostly in their apical half in accordance with the typical localization of IP3R in organelles such as the endoplasmic reticulum. In addition, there was a gradient from the surface region towards the crypt fundus, where the IP3R3 signal could not be detected. Despite the strong IP3R3-gradient, in saponin-permeabilized colonic crypts exogenously administered IP3 or adenophostin A evoked a similar depletion of mag-fura-2-loaded intracellular Ca2+ stores in crypt and surface cells suggesting a contribution of the nuclear IP3R2 to the Ca2+ release. This conclusion was confirmed by experiments with isolated nuclei from colonic epithelium, at which IP3 was able to induce changes in the Ca2+ concentration, which were inhibited by 2-aminoethoxy-diphenylborate (2-APB), a blocker of IP3 receptors. These results demonstrate that the colonic epithelial cells undergo changes in IP3R subtype expression during differentiation.
Collapse
Affiliation(s)
- Anne Siefjediers
- Institute for Veterinary Physiology, University of Giessen, Frankfurter Str. 100, D-35392 Giessen, Germany
| | | | | | | |
Collapse
|
31
|
Choe CU, Ehrlich BE. The inositol 1,4,5-trisphosphate receptor (IP3R) and its regulators: sometimes good and sometimes bad teamwork. ACTA ACUST UNITED AC 2006; 2006:re15. [PMID: 17132820 DOI: 10.1126/stke.3632006re15] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In both nonexcitable and excitable cells, the inositol 1,4,5-trisphosphate receptor (IP(3)R) is the primary cytosolic target responsible for the initiation of intracellular calcium (Ca(2+)) signaling. To fulfill this function, the IP(3)R depends on interaction with accessory subunits and regulatory proteins. These include proteins that reside in the lumen of the endoplasmic reticulum (ER), such as chromogranin A and B and ERp44, and cytosolic proteins, such as neuronal Ca(2+) sensor 1, huntingtin, cytochrome c, IP(3)R-binding protein released with inositol 1,4,5-trisphosphate, Homer, and 4.1N. Specific interactions between these modulatory proteins and the IP(3)R have been described, making it clear that the controlled modulation of the IP(3)R by its binding partners is necessary for physiological cell regulation. The functional coupling of these modulators with the IP(3)R can control apoptosis, intracellular pH, the initiation and regulation of neuronal Ca(2+) signaling, exocytosis, and gene expression. The pathophysiological relevance of IP(3)R modulation is apparent when the functional interaction of these proteins is enhanced or abolished by mutation or overexpression. The subsequent deregulation of the IP(3)R leads to pathological changes in Ca(2+) signaling, signal initiation, the amplitude and frequency of Ca(2+) signals, and the duration of the Ca(2+) elevation. Consequences of this deregulation include abnormal growth and apoptosis. Complex regulation of Ca(2+) signaling is required for the cell to live and function, and this difficult task can only be managed when the IP(3)R teams up and acts properly with its numerous binding partners.
Collapse
Affiliation(s)
- Chi-Un Choe
- Department of Pharmacology, Yale University, New Haven, CT 06520, USA
| | | |
Collapse
|
32
|
McCarron JG, Chalmers S, Bradley KN, MacMillan D, Muir TC. Ca2+ microdomains in smooth muscle. Cell Calcium 2006; 40:461-93. [PMID: 17069885 DOI: 10.1016/j.ceca.2006.08.010] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2006] [Accepted: 08/23/2006] [Indexed: 02/03/2023]
Abstract
In smooth muscle, Ca(2+) controls diverse activities including cell division, contraction and cell death. Of particular significance in enabling Ca(2+) to perform these multiple functions is the cell's ability to localize Ca(2+) signals to certain regions by creating high local concentrations of Ca(2+) (microdomains), which differ from the cytoplasmic average. Microdomains arise from Ca(2+) influx across the plasma membrane or release from the sarcoplasmic reticulum (SR) Ca(2+) store. A single Ca(2+) channel can create a microdomain of several micromolar near (approximately 200 nm) the channel. This concentration declines quickly with peak rates of several thousand micromolar per second when influx ends. The high [Ca(2+)] and the rapid rates of decline target Ca(2+) signals to effectors in the microdomain with rapid kinetics and enable the selective activation of cellular processes. Several elements within the cell combine to enable microdomains to develop. These include the brief open time of ion channels, localization of Ca(2+) by buffering, the clustering of ion channels to certain regions of the cell and the presence of membrane barriers, which restrict the free diffusion of Ca(2+). In this review, the generation of microdomains arising from Ca(2+) influx across the plasma membrane and the release of the ion from the SR Ca(2+) store will be discussed and the contribution of mitochondria and the Golgi apparatus as well as endogenous modulators (e.g. cADPR and channel binding proteins) will be considered.
Collapse
Affiliation(s)
- John G McCarron
- Department of Physiology and Pharmacology, University of Strathclyde, SIPBS, Glasgow, UK.
| | | | | | | | | |
Collapse
|
33
|
Cooper BJ, Key B, Carter A, Angel NZ, Hart DNJ, Kato M. Suppression and overexpression of adenosylhomocysteine hydrolase-like protein 1 (AHCYL1) influences zebrafish embryo development: a possible role for AHCYL1 in inositol phospholipid signaling. J Biol Chem 2006; 281:22471-84. [PMID: 16754674 DOI: 10.1074/jbc.m602520200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Adenosylhomocysteine hydrolase-like protein 1 (AHCYL1) is a novel intracellular protein with approximately 50% protein identity to adenosylhomocysteine hydrolase (AHCY), an important enzyme for metabolizing S-adenosyl-l-homocysteine, the by-product of S-adenosyl-l-homomethionine-dependent methylation. AHCYL1 binds to the inositol 1,4,5-trisphosphate receptor, suggesting that AHCYL1 is involved in intracellular calcium release. We identified two zebrafish AHCYL1 orthologs (zAHCYL1A and -B) by bioinformatics and reverse transcription-PCR. Unlike the ubiquitously present AHCY genes, AHCYL1 genes were only detected in segmented animals, and AHCYL1 proteins were highly conserved among species. Phylogenic analysis suggested that the AHCYL1 gene diverged early from AHCY and evolved independently. Quantitative reverse transcription-PCR showed that zAHCYL1A and -B mRNA expression was regulated differently from the other AHCY-like protein zAHCYL2 and zAHCY during zebrafish embryogenesis. Injection of morpholino antisense oligonucleotides against zAHCYL1A and -B into zebrafish embryos inhibited zAHCYL1A and -B mRNA translation specifically and induced ventralized morphologies. Conversely, human and zebrafish AHCYL1A mRNA injection into zebrafish embryos induced dorsalized morphologies that were similar to those obtained by depleting intracellular calcium with thapsigargin. Human AHCY mRNA injection showed little effect on the embryos. These data suggest that AHCYL1 has a different function from AHCY and plays an important role in embryogenesis by modulating inositol 1,4,5-trisphosphate receptor function for the intracellular calcium release.
Collapse
Affiliation(s)
- Benjamine J Cooper
- Dendritic Cell Program, Mater Medical Research Institute, Brisbane, Queensland 4101, Australia
| | | | | | | | | | | |
Collapse
|
34
|
Devogelaere B, Nadif Kasri N, Derua R, Waelkens E, Callewaert G, Missiaen L, Parys JB, De Smedt H. Binding of IRBIT to the IP3 receptor: Determinants and functional effects. Biochem Biophys Res Commun 2006; 343:49-56. [PMID: 16527252 DOI: 10.1016/j.bbrc.2006.02.119] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2006] [Accepted: 02/21/2006] [Indexed: 11/26/2022]
Abstract
IRBIT has previously been shown to interact with the inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) in an IP3-sensitive way. So far it remained to be elucidated whether this interaction was direct or indirect, and whether it was functionally relevant. We now show that IRBIT can directly interact with the IP3R, and that both the suppressor domain and the IP3-binding core of the IP3R are essential for a strong interaction. Moreover, we identified a PEST motif and a PDZ-ligand on IRBIT which were critical for the interaction with the IP3R. Furthermore, we identified Asp-73 as a critical residue for this interaction. Finally, we demonstrated that this interaction functionally affects the IP3R: IRBIT inhibits both IP3 binding and IP3-induced Ca2+ release.
Collapse
Affiliation(s)
- Benoit Devogelaere
- Laboratorium voor Fysiologie, Katholieke Universiteit Leuven Campus Gasthuisberg O/N1, Leuven, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Camello-Almaraz MC, Pozo MJ, Murphy MP, Camello PJ. Mitochondrial production of oxidants is necessary for physiological calcium oscillations. J Cell Physiol 2006; 206:487-94. [PMID: 16206242 DOI: 10.1002/jcp.20498] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Mitochondrial involvement in Ca2+ signaling is thought to be due to the effect of mitochondrial Ca2+ removal from and Ca2+ release to cytosolic domains close to ryanodine and IP3 Ca2+ channels. However, mitochondria are a source of low levels of endogenous reactive oxygen species, and Ca2+ release channels are known to be redox-sensitive. In the present work, we studied the role of mitochondrial production of oxygen species in Ca2+ oscillations during physiological stimulation. Mitochondria-targeted antioxidants and mitochondrial inhibitors quickly inhibited calcium oscillations in pancreatic acinar cells stimulated by postprandial levels of the gut hormone cholecystokinin. Confocal microscopy using different redox-sensitive dyes showed that cholecystokinin-induced oscillations are associated with mitochondrial production of reactive oxygen species. This production is inhibited by application of mitochondria-targeted antioxidants and mitochondrial inhibitors. In addition, we found no correlation between inhibition of oscillations and mitochondrial depolarization. We conclude that low level production of reactive oxygen species by mitochondria is a necessary element in the development of Ca2+ oscillations during physiological stimulation. This study unveils a new and unexplored aspect of the participation of mitochondria in calcium signals.
Collapse
|
36
|
Watras J, Fink CC, Loew LM. Endogenous inhibitors of InsP3-induced Ca2+ release in neuroblastoma cells. Brain Res 2006; 1055:60-72. [PMID: 16095574 DOI: 10.1016/j.brainres.2005.06.091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2005] [Revised: 06/25/2005] [Accepted: 06/28/2005] [Indexed: 10/25/2022]
Abstract
Cerebellar Purkinje neurons and neuroblastoma N1E-115 cells require 10-50 times more InsP3 to induce Ca2+ release than do a variety of non-neuronal cells (including astrocytes, hepatocytes, endothelial cells, or smooth muscle cells). Given the importance of InsP3-induced Ca2+ release for the development of synaptic plasticity in Purkinje neurons, a low InsP3 sensitivity may facilitate the integration of numerous synaptic inputs before initiating a change in synaptic strength. In the present study, attention is directed at the mechanism underlying this low InsP3 sensitivity of Ca2+ release. We show that permeabilization of neuroblastoma cells with saponin increased InsP3 sensitivity of Ca2+ release, indicating the presence of a diffusible, cytosolic inhibitor(s) of Ca2+ release. Consistent with this hypothesis, gel filtration of the neuroblastoma cytosol yielded three peaks that inhibited InsP3-induced Ca2+ release from permeabilized cells. The prominent inhibitory peak decreased the InsP3 sensitivity of Ca2+ release from permeabilized cells, did not bind 3H-InsP3, and was present in sufficient levels to account for the low InsP3 sensitivity of Ca2+ release in intact neuroblastoma cells. Purification of this prominent inhibitory fraction yielded a protein band that was identified by mass spectrometry as stress-induced phosphoprotein 1 (mSTI1). Furthermore, immunoprecipitation of mSTI1 decreased the inhibitory activity of N1E-115 cytosol, indicating that mSTI1 contributes to the inhibition of InsP3-induced Ca2+ release. Thus, the low InsP3 sensitivity of Ca2+ release in neuroblastoma cells can be explained by the presence of cytosolic inhibitors of Ca2+ release and include stress-induced phosphoprotein 1.
Collapse
Affiliation(s)
- James Watras
- Department of Pharmacology, University of Connecticut Health Center, Farmington, CT 06032, USA.
| | | | | |
Collapse
|
37
|
Woodcock EA, Matkovich SJ. Ins(1,4,5)P3 receptors and inositol phosphates in the heart-evolutionary artefacts or active signal transducers? Pharmacol Ther 2005; 107:240-51. [PMID: 15908009 DOI: 10.1016/j.pharmthera.2005.04.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The generation of the second messenger inositol 1,4,5-trisphosphate (Ins(1,4,5)P(3)) and its associated release of Ca(2+) from internal stores is a highly conserved module in intracellular signaling from Drosophila to mammals. Many cell types, often nonexcitable cells, depend on this pathway to couple external signals to intracellular Ca(2+) release. However, despite the presence of the requisite Ins(1,4,5)P(3) signaling machinery, excitable cells such as cardiac myocytes employ a robust alternate system of intracellular Ca(2+) release, namely, a coupled system of Ca(2+) influx, followed by Ca(2+) release via the IP(3)R-related ryanodine receptors. In these systems, Ins(1,4,5)P(3) signaling pathways appear to be largely dormant. In this review, we consider the general features of inositol phosphate (InsP) responses in cardiac myocytes and the molecules mediating these responses. The spatial localization of Ins(1,4,5)P(3) generation and Ins(1,4,5)P(3) receptor (IP(3)Rs) is likely of key importance, and we examine the state of knowledge in atrial, ventricular, and Purkinje myocytes. Several studies have implicated Ins(1,4,5)P(3) generation in both arrhythmogenic and hypertrophic responses, and possible mechanisms involving Ins(1,4,5)P(3) are discussed. While Ins(1,4,5)P(3) is unlikely to be a key player in cardiac excitation-contraction (EC) coupling, its potential role in an alternate Ca(2+) release system to signal changes in gene transcription warrants further investigation. Such studies will help to determine whether cardiac Ins(1,4,5)P(3) generation represents a vestigial pathway or plays an active role in cardiac signaling.
Collapse
Affiliation(s)
- Elizabeth A Woodcock
- Cellular Biochemistry Laboratory, Baker Heart Research Institute, Commercial Road, Melbourne, Australia.
| | | |
Collapse
|
38
|
Samways DSK, Henderson G. Opioid elevation of intracellular free calcium: possible mechanisms and physiological relevance. Cell Signal 2005; 18:151-61. [PMID: 16199136 DOI: 10.1016/j.cellsig.2005.08.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2005] [Accepted: 08/19/2005] [Indexed: 01/02/2023]
Abstract
Opioid receptors are seven transmembrane domain Gi/G0 protein-coupled receptors, the activation of which stimulates a variety of intracellular signalling mechanisms including activation of inwardly rectifying potassium channels, and inhibition of both voltage-operated N-type Ca2+ channels and adenylyl cyclase activity. It is now apparent that like many other Gi/G0-coupled receptors, opioid receptor activation can significantly elevate intracellular free Ca2+ ([Ca2+]i), although the mechanism underlying this phenomenon is not well understood. In some cases opioid receptor activation alone appears to elevate [Ca2+]i, but in many cases it requires concomitant activation of Gq-coupled receptors, which themselves stimulate Ca2+ release from intracellular stores via the inositol phosphate pathway. Given the number of Ca2+-sensitive processes known to occur in cells, there are therefore a myriad of situations in which opioid receptor-mediated elevations of [Ca2+](i) may be important. Here, we review the literature documenting opioid receptor-mediated elevations of [Ca2+]i, discussing both the possible mechanisms underlying this phenomenon and its potential physiological relevance.
Collapse
Affiliation(s)
- Damien S K Samways
- Department of Pharmacological and Physiological Science, Health Science Center, School of Medicine, Saint Louis University, MO, USA.
| | | |
Collapse
|
39
|
Przyklenk K, Maynard M, Darling CE, Whittaker P. Pretreatment with D-myo-inositol trisphosphate reduces infarct size in rabbit hearts: role of inositol trisphosphate receptors and gap junctions in triggering protection. J Pharmacol Exp Ther 2005; 314:1386-92. [PMID: 15919762 DOI: 10.1124/jpet.105.087742] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pretreatment with D-myo-inositol-1,4,5-trisphosphate hexasodium (D-myo-IP(3)), the sodium salt of the second messenger inositol 1,4,5-trisphosphate (IP(3)), is cardioprotective and triggers a reduction of infarct size comparable in magnitude to that obtained with ischemic preconditioning. However, this observation is enigmatic; whereas IP(3) signaling is conventionally initiated by receptor binding, IP(3) receptors are typically considered to be intracellular, and D-myo-IP(3) is membrane-impermeable. We propose that this paradox is explained by the presence of poorly characterized external IP(3) receptors and hypothesize that: 1) infarct size reduction with D-myo-IP(3) is receptor-mediated; and 2) communication via gap junctions and/or hemichannels is required to initiate this protection. To investigate the role of receptor binding, isolated buffer-perfused rabbit hearts underwent 30 min of coronary occlusion (CO) and 2 h of reflow. Prior to CO, hearts received no treatment (controls), D-myo-IP(3), L-myo-IP(3) (enantiomer not recognized by the IP(3) receptor), D-myo-IP(3) + the IP(3) receptor inhibitor xestospongin C (XeC), or XeC alone. Infarct size, assessed by tetrazolium staining, was reduced with D-myo-IP(3) treatment, whereas hearts that received L-myo-IP(3) or D-myo-IP(3) + XeC showed no protection. To evaluate the contribution of gap junctions/hemichannels, additional control and D-myo-IP(3)-treated cohorts received a 5-min infusion of heptanol or Gap 27, two structurally distinct gap junction inhibitors, administered at doses confirmed to attenuate intercellular transmission of a gap junction-permeable fluorescent dye. There was no infarct-sparing effect of D-myo-IP(3) in inhibitor-treated hearts. These data support the concepts that infarct size reduction with D-myo-IP(3) is triggered by receptor binding and that communication via gap junctions/hemichannels is involved in initiating this protection.
Collapse
Affiliation(s)
- Karin Przyklenk
- Department of Emergency Medicine, University of Massachusetts Medical School, Worcester, 01655, USA.
| | | | | | | |
Collapse
|
40
|
Thorneloe KS, Nelson MT. Ion channels in smooth muscle: regulators of intracellular calcium and contractility. Can J Physiol Pharmacol 2005; 83:215-42. [PMID: 15870837 DOI: 10.1139/y05-016] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Smooth muscle (SM) is essential to all aspects of human physiology and, therefore, key to the maintenance of life. Ion channels expressed within SM cells regulate the membrane potential, intracellular Ca2+ concentration, and contractility of SM. Excitatory ion channels function to depolarize the membrane potential. These include nonselective cation channels that allow Na+ and Ca2+ to permeate into SM cells. The nonselective cation channel family includes tonically active channels (Icat), as well as channels activated by agonists, pressure-stretch, and intracellular Ca2+ store depletion. Cl--selective channels, activated by intracellular Ca2+ or stretch, also mediate SM depolarization. Plasma membrane depolarization in SM activates voltage-dependent Ca2+ channels that demonstrate a high Ca2+ selectivity and provide influx of contractile Ca2+. Ca2+ is also released from SM intracellular Ca2+ stores of the sarcoplasmic reticulum (SR) through ryanodine and inositol trisphosphate receptor Ca2+ channels. This is part of a negative feedback mechanism limiting contraction that occurs by the Ca2+-dependent activation of large-conductance K+ channels, which hyper polarize the plasma membrane. Unlike the well-defined contractile role of SR-released Ca2+ in skeletal and cardiac muscle, the literature suggests that in SM Ca2+ released from the SR functions to limit contractility. Depolarization-activated K+ chan nels, ATP-sensitive K+ channels, and inward rectifier K+ channels also hyperpolarize SM, favouring relaxation. The expression pattern, density, and biophysical properties of ion channels vary among SM types and are key determinants of electrical activity, contractility, and SM function.
Collapse
Affiliation(s)
- Kevin S Thorneloe
- Department of Pharmacology, College of Medicine, University of Vermont, Burlington 05405, USA.
| | | |
Collapse
|
41
|
Hur EM, Park YS, Huh YH, Yoo SH, Woo KC, Choi BH, Kim KT. Junctional membrane inositol 1,4,5-trisphosphate receptor complex coordinates sensitization of the silent EGF-induced Ca2+ signaling. ACTA ACUST UNITED AC 2005; 169:657-67. [PMID: 15911880 PMCID: PMC2171708 DOI: 10.1083/jcb.200411034] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ca(2+) is a highly versatile intracellular signal that regulates many different cellular processes, and cells have developed mechanisms to have exquisite control over Ca(2+) signaling. Epidermal growth factor (EGF), which fails to mobilize intracellular Ca(2+) when administrated alone, becomes capable of evoking [Ca(2+)](i) increase and exocytosis after bradykinin (BK) stimulation in chromaffin cells. Here, we provide evidence that this sensitization process is coordinated by a macromolecular signaling complex comprised of inositol 1,4,5-trisphosphate receptor type I (IP(3)R1), cAMP-dependent protein kinase (PKA), EGF receptor (EGFR), and an A-kinase anchoring protein, yotiao. The IP(3)R complex functions as a focal point to promote Ca(2+) release in two ways: (1) it facilitates PKA-dependent phosphorylation of IP(3)R1 in response to BK-induced elevation of cAMP, and (2) it couples the plasmalemmal EGFR with IP(3)R1 at the Ca(2+) store located juxtaposed to the plasma membrane. Our study illustrates how the junctional membrane IP(3)R complex connects different signaling pathways to define the fidelity and specificity of Ca(2+) signaling.
Collapse
Affiliation(s)
- Eun-Mi Hur
- National Research Laboratory of Molecular Neurophysiology, Pohang University of Science and Technology, Hyo-ja dong, San31, Pohang, 790-784, South Korea
| | | | | | | | | | | | | |
Collapse
|
42
|
Estevez AY, Strange K. Calcium feedback mechanisms regulate oscillatory activity of a TRP-like Ca2+ conductance in C. elegans intestinal cells. J Physiol 2005; 567:239-51. [PMID: 15961418 PMCID: PMC1474156 DOI: 10.1113/jphysiol.2005.091900] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Inositol-1,4,5-trisphosphate (IP3)-dependent Ca2+ oscillations in Caenorhabditis elegans intestinal epithelial cells regulate the nematode defecation cycle. The role of plasma membrane ion channels in intestinal cell oscillatory Ca2+ signalling is unknown. We have shown previously that cultured intestinal cells express a Ca2+-selective conductance, I(ORCa), that is biophysically similar to TRPM7 currents. I(ORCa) activates slowly and stabilizes when cells are patch clamped with pipette solutions containing 10 mm BAPTA and free Ca2+ concentrations of approximately 17 nm. However, when BAPTA concentration is lowered to 1 mm, I(ORCa) oscillates. Oscillations in channel activity induced simultaneous oscillations in cytoplasmic Ca2+ levels. Removal of extracellular Ca2+ inhibited I(ORCa) oscillations, whereas readdition of Ca2+ to the bath caused a rapid and transient reactivation of the current. Experimental manoeuvres that elevated intracellular Ca2+ blocked current oscillations. Elevation of intracellular Ca2+ in the presence of 10 mm BAPTA to block I(ORCa) oscillations led to a dose-dependent increase in the rate of current activation. At intracellular Ca2+ concentrations of 250 nm, current activation was transient. Patch pipette solutions buffered with 1-4 mm of either BAPTA or EGTA gave rise to similar patterns of I(ORCa) oscillations. We conclude that changes in Ca2+ concentration close to the intracellular opening of the channel pore regulate channel activity. Low concentrations of Ca2+ activate the channel. As Ca2+ enters and accumulates near the pore mouth, channel activity is inhibited. Oscillating plasma membrane Ca2+ entry may play a role in generating intracellular Ca2+ oscillations that regulate the C. elegans defecation rhythm.
Collapse
Affiliation(s)
- Ana Y Estevez
- Vanderbilt University Medical Center, T-4202 Medical Center North, Nashville, TN 37232-2520, USA
| | | |
Collapse
|
43
|
Fellner SK, Rybczynski R, Gilbert LI. Ca2+ signaling in prothoracicotropic hormone-stimulated prothoracic gland cells of Manduca sexta: evidence for mobilization and entry mechanisms. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2005; 35:263-275. [PMID: 15763463 DOI: 10.1016/j.ibmb.2004.11.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2004] [Revised: 11/15/2004] [Accepted: 11/16/2004] [Indexed: 05/24/2023]
Abstract
Prothoracicotropic hormone (PTTH) stimulates ecdysteroidogenesis in lepidopteran prothoracic glands (PGs), thus indirectly controlling molting and metamorphosis. PTTH triggers a signal transduction cascade in PGs that involves an early influx of Ca2+. Although the importance of Ca2+ has been long known, the mechanism(s) of PTTH-stimulated changes in cytoplasmic Ca2+ [Ca2+]i are not yet well understood. PGs from the fifth instar of Manduca sexta were exposed to PTTH in vitro. The resultant changes in [Ca2+]i were measured using ratiometric analysis of a fura-2 fluorescence signal in the presence and absence of inhibitors of specific cellular signaling mechanisms. The phospholipase C (PLC) inhibitor U-73122 nearly abolished the PTTH-stimulated increase in [Ca2+]i, as well as PTTH-stimulated ecdysteroidogenesis and extracellular-signal regulated kinase phosphorylation, thus establishing a role for PLC and implicating inositol trisphosphate (IP3) in PTTH signal transduction. Two antagonists of the IP3 receptor, 2-APB and TMB-8, likewise blocked the [Ca2+]i response by a mean of 92%. We describe for the first time the presence of Ca2+ oscillations in PTTH-stimulated cells in Ca2+-free medium. External Ca2+ entered PG cells via at least two routes: store-operated (capacitative) Ca2+ entry channels and L-type voltage-gated Ca2+ channels. We propose that PTTH initiates a transductory cascade typical of many G-protein coupled receptors, involving both Ca2+ mobilization and entry pathways.
Collapse
Affiliation(s)
- Susan K Fellner
- Department of Cell and Molecular Physiology (CB# 7545), University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | |
Collapse
|
44
|
Marchenko SM, Yarotskyy VV, Kovalenko TN, Kostyuk PG, Thomas RC. Spontaneously active and InsP3-activated ion channels in cell nuclei from rat cerebellar Purkinje and granule neurones. J Physiol 2005; 565:897-910. [PMID: 15774532 PMCID: PMC1464565 DOI: 10.1113/jphysiol.2004.081299] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Increases in Ca(2+) concentration in the nucleus of neurones modulate gene transcription and may be involved in activity-dependent long-term plasticity, apoptosis, and neurotoxicity. Little is currently known about the regulation of Ca(2+) in the nuclei of neurones. Investigation of neuronal nuclei is hampered by the cellular heterogeneity of the brain where neurones comprise no more than 10% of the cells. The situation is further complicated by large differences in properties of different neurones. Here we report a method for isolating nuclei from identified central neurones. We employed this technique to study nuclei from rat cerebellar Purkinje and granule neurones. Patch-clamp recording from the nuclear membrane of Purkinje neurones revealed numerous large-conductance channels selective for monovalent cations. The nuclear membrane of Purkinje neurones also contained multiple InsP(3)- activated ion channels localized exclusively in the inner nuclear membrane with their receptor loci facing the nucleoplasm. In contrast, the nuclear membrane of granule neurones contained only a small number of mainly anion channels. Nuclear InsP(3) receptors (InsP(3)Rs) were activated by InsP(3) with EC(50) = 0.67 microm and a Hill coefficient of 2.5. Ca(2+) exhibited a biphasic effect on the receptors elevating its activity at low concentrations and inhibiting it at micromolar concentrations. InsP(3) in saturating concentrations did not prevent the inhibitory effect of Ca(2+), but strongly increased InsP(3)R activity at resting Ca(2+) concentrations. These data are the first evidence for the presence of intranuclear sources of Ca(2+) in neurones. Ca(2+) release from the nuclear envelope may amplify Ca(2+) transients penetrating the nucleus from the cytoplasm or generate Ca(2+) transients in the nucleus independently of the cytoplasm.
Collapse
Affiliation(s)
- Sergey M Marchenko
- Bogomoletz Institute of Physiology, 4 Bogomoletz Street, Kiev, 01024, Ukraine.
| | | | | | | | | |
Collapse
|
45
|
Johenning F, Wenk M, Uhlén P, DeGray B, Lee E, de Camilli P, Ehrlich B. InsP3-mediated intracellular calcium signalling is altered by expression of synaptojanin-1. Biochem J 2005; 382:687-94. [PMID: 15080793 PMCID: PMC1133826 DOI: 10.1042/bj20040418] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2004] [Accepted: 04/14/2004] [Indexed: 11/17/2022]
Abstract
Phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P2] plays an important physiological role as a precursor for the InsP3-mediated intracellular calcium (Ca2+) signalling cascade. It also regulates membrane trafficking, actin function and transmembrane proteins. SJ-1 (synaptojanin-1), a phosphoinositide phosphatase, regulates the turnover of a PtdIns(4,5)P2 pool involved in clathrin and actin dynamics at the cell surface. We tested the interrelationship of this pool with PtdIns(4,5)P2 pools involved in Ca2+ signalling by expressing in Chinese-hamster ovary cells full-length SJ-1 or its 5-Pase (inositol 5-phosphatase) domain. SJ-1 significantly attenuated the generation of Ca2+ oscillations induced by ATP and the 5-Pase domain mimicked this effect. These changes correlated with increased PtdIns(4,5)P2 phosphatase activity of cellular extracts. Overexpression of the endoplasmic reticulum-anchored PtdIns(4)P phosphatase Sac1 did not affect Ca2+ oscillations, although it increased the Ca2+ efflux rate from intracellular stores. The ability of SJ-1 to alter intracellular Ca2+ signalling indicates a close functional interrelationship between plasma membrane PtdIns(4,5)P2 pools that control actin and endocytosis and those involved in the regulation of specific spatio-temporal Ca2+ signals.
Collapse
Affiliation(s)
- Friedrich W. Johenning
- *Departments of Pharmacology, Cell and Molecular Physiology, Yale University School of Medicine, New Haven, CT, U.S.A
- †Institute for Anatomy, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Markus R. Wenk
- ‡Departments of Cell Biology and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, U.S.A
| | - Per Uhlén
- *Departments of Pharmacology, Cell and Molecular Physiology, Yale University School of Medicine, New Haven, CT, U.S.A
| | - Brenda DeGray
- *Departments of Pharmacology, Cell and Molecular Physiology, Yale University School of Medicine, New Haven, CT, U.S.A
| | - Eunkyung Lee
- ‡Departments of Cell Biology and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, U.S.A
| | - Pietro de Camilli
- ‡Departments of Cell Biology and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, U.S.A
| | - Barbara E. Ehrlich
- *Departments of Pharmacology, Cell and Molecular Physiology, Yale University School of Medicine, New Haven, CT, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|
46
|
Srikanth S, Wang Z, Tu H, Nair S, Mathew MK, Hasan G, Bezprozvanny I. Functional properties of the Drosophila melanogaster inositol 1,4,5-trisphosphate receptor mutants. Biophys J 2005; 86:3634-46. [PMID: 15189860 PMCID: PMC1304265 DOI: 10.1529/biophysj.104.040121] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The inositol (1,4,5)-trisphosphate receptor (InsP(3)R) is an intracellular calcium (Ca(2+)) release channel that plays a crucial role in cell signaling. In Drosophila melanogaster a single InsP(3)R gene (itpr) encodes a protein (DmInsP(3)R) that is approximately 60% conserved with mammalian InsP(3)Rs. A number of itpr mutant alleles have been identified in genetic screens and studied for their effect on development and physiology. However, the functional properties of wild-type or mutant DmInsP(3)Rs have never been described. Here we use the planar lipid bilayer reconstitution technique to describe single-channel properties of embryonic and adult head DmInsP(3)R splice variants. The three mutants chosen in this study reside in each of the three structural domains of the DmInsP(3)R-the amino-terminal ligand binding domain (ug3), the middle-coupling domain (wc703), and the channel-forming region (ka901). We discovered that 1), the major functional properties of DmInsP(3)R (conductance, gating, and sensitivity to InsP(3) and Ca(2+)) are remarkably conserved with the mammalian InsP(3)R1; 2), single-channel conductance of the adult head DmInsP(3)R isoform is 89 pS and the embryonic DmInsP(3)R isoform is 70 pS; 3), ug3 mutation affects sensitivity of the DmInsP(3)Rs to activation by InsP(3), but not their InsP(3)-binding properties; 4), wc703 channels have increased sensitivity to modulation by Ca(2+); and 5), homomeric ka901 channels are not functional. We correlated the results obtained in planar lipid bilayer experiments with measurements of InsP(3)-induced Ca(2+) fluxes in microsomes isolated from wild-type and heterozygous itpr mutants. Our study validates the use of D. melanogaster as an appropriate model for InsP(3)R structure-function studies and provides novel insights into the fundamental mechanisms of the InsP(3)R function.
Collapse
Affiliation(s)
- Sonal Srikanth
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Li L, Wice BM. Bombesin and nutrients independently and additively regulate hormone release from GIP/Ins cells. Am J Physiol Endocrinol Metab 2005; 288:E208-15. [PMID: 15383372 DOI: 10.1152/ajpendo.00346.2004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) regulates glucose homeostasis and high-fat diet-induced obesity and insulin resistance. Therefore, elucidating the mechanisms that regulate GIP release is important. GIP is produced by K cells, a specific subtype of small intestinal enteroendocrine (EE) cell. Bombesin-like peptides produced by enteric neurons and luminal nutrients stimulate GIP release in vivo. We previously showed that PMA, bombesin, meat hydrolysate, glyceraldehyde, and methylpyruvate increase hormone release from a GIP-producing EE cell line (GIP/Ins cells). Here we demonstrate that bombesin and nutrients additively stimulate hormone release from GIP/Ins cells. In various cell systems, bombesin and PMA regulate cell physiology by activating PKD signaling in a PKC-dependent fashion, whereas nutrients regulate cell physiology by inhibiting AMPK signaling. Western blot analyses of GIP/Ins cells using antibodies specific for activated and/or phosphorylated forms of PKD and AMPK and one substrate for each kinase revealed that bombesin and PMA, but not nutrients, activated PKC, but not PKD. Conversely, nutrients, but not bombesin or PMA, inhibited AMPK activity. Pharmacological studies showed that PKC inhibition blocked bombesin- and PMA-stimulated hormone release, but AMPK activation failed to suppress nutrient-stimulated hormone secretion. Forced expression of constitutively active vs. dominant negative PKDs or AMPKs failed to perturb bombesin- or nutrient-stimulated hormone release. Thus, in GIP/Ins cells, PKC regulates bombesin-stimulated hormone release, whereas nutrients may control hormone release by regulating the activity of AMPK-related kinases, rather than AMPK itself. These results strongly suggest that K cells in vivo independently respond to neuronal vs. nutritional stimuli via two distinct signaling pathways.
Collapse
Affiliation(s)
- Lin Li
- Department of Internal Medicine, Division of Endocrinology, Diabetes & Metabolism, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | |
Collapse
|
48
|
Grayson TH, Haddock RE, Murray TP, Wojcikiewicz RJH, Hill CE. Inositol 1,4,5-trisphosphate receptor subtypes are differentially distributed between smooth muscle and endothelial layers of rat arteries. Cell Calcium 2004; 36:447-58. [PMID: 15488594 DOI: 10.1016/j.ceca.2004.04.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2004] [Revised: 03/29/2004] [Accepted: 04/20/2004] [Indexed: 11/22/2022]
Abstract
In blood vessels, the ability to control vascular tone depends on extracellular calcium entry and the release of calcium from inositol 1,4,5-trisphosphate receptor (IP3R)-gated stores located in both the endothelial and smooth muscle cells of the vascular wall. Therefore, we examined mRNA expression and protein distribution of IP3R subtypes in intact aorta, basilar and mesenteric arteries of the rat. IP3R1 mRNA was predominantly expressed in all three arteries. Immunohistochemistry showed that IP3R1 was present in both the muscle and endothelial cell layers, while IP3R2 and IP3R3 were largely restricted to the endothelium. Weak expression of IP3R2 was observed in the smooth muscle of the basilar artery. Co-localisation studies of IP3R subtypes with known cellular elements showed no association of any of the three subtypes with the endothelial cell plasma membrane, but a close association between the subtypes and actin filaments was observed in all cell layers. IP3R2 was found to be present near the endothelial cell nucleus. We are the first to demonstrate differential IP3R subtype distribution between the cell layers of the intact vascular wall and hypothesise that this may underlie the diversity of IP3R-dependent responses, such as vasoconstriction, vasodilation and vasomotion, displayed by arteries.
Collapse
MESH Headings
- Animals
- Arteries/chemistry
- Arteries/metabolism
- Calcium Channels/analysis
- Calcium Channels/biosynthesis
- Endothelium, Vascular/chemistry
- Endothelium, Vascular/metabolism
- Inositol 1,4,5-Trisphosphate Receptors
- Muscle, Smooth, Vascular/chemistry
- Muscle, Smooth, Vascular/metabolism
- Protein Subunits/analysis
- Protein Subunits/biosynthesis
- Rats
- Rats, Wistar
- Receptors, Cytoplasmic and Nuclear/analysis
- Receptors, Cytoplasmic and Nuclear/biosynthesis
Collapse
Affiliation(s)
- T Hilton Grayson
- Division of Neuroscience, John Curtin School of Medical Research, Australian National University, Canberra, ACT 0200, Australia.
| | | | | | | | | |
Collapse
|
49
|
Tu H, Wang Z, Nosyreva E, De Smedt H, Bezprozvanny I. Functional characterization of mammalian inositol 1,4,5-trisphosphate receptor isoforms. Biophys J 2004; 88:1046-55. [PMID: 15533917 PMCID: PMC1305111 DOI: 10.1529/biophysj.104.049593] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Inositol 1,4,5-trisphosphate receptors (InsP3R) play a key role in intracellular calcium (Ca2+) signaling. Three mammalian InsP3R isoforms--InsP3R type 1 (InsP3R1), InsP3R type 2 (InsP3R2), and InsP3R type 3 (InsP3R3) are expressed in mammals, but the functional differences between the three mammalian InsP3R isoforms are poorly understood. Here we compared single-channel behavior of the recombinant rat InsP3R1, InsP3R2, and InsP3R3 expressed in Sf9 cells, reconstituted into planar lipid bilayers and recorded with 50 mM Ba2+ as a current carrier. We found that: 1), for all three mammalian InsP3R isoforms the size of the unitary current is 1.9 pA and single-channel conductance is 74-80 pS; 2), in optimal recording conditions the maximal single-channel open probability for all three mammalian InsP3R isoforms is in the range 30-40%; 3), in optimal recording conditions the mean open dwell time for all three mammalian InsP3R isoforms is 7-8 ms, the mean closed dwell time is approximately 10 ms; 4), InsP3R2 has the highest apparent affinity for InsP(3) (0.10 microM), followed by InsP3R1 (0.27 microM), and then by InsP3R3 (0.40 microM); 5), InsP3R1 has a high-affinity (0.13 mM) ATP modulatory site, InsP3R2 gating is ATP independent, and InsP3R3 has a low-affinity (2 mM) ATP modulatory site; 6), ATP modulates InsP3R1 gating in a noncooperative manner (n(Hill) = 1.3); 7), ATP modulates InsP3R3 gating in a highly cooperative manner (n(Hill) = 4.1). Obtained results provide novel information about functional properties of mammalian InsP3R isoforms.
Collapse
Affiliation(s)
- Huiping Tu
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, USA
| | | | | | | | | |
Collapse
|
50
|
Abstract
The function of Bcl-2 family members on the endoplasmic reticulum has received increasing attention in recent years. The endoplasmic reticulum is the major organelle involved in intracellular calcium homeostasis and calcium signaling, including calcium signals that mediate apoptosis induction by anticancer drugs. But currently a controversy exists regarding reported effects of Bcl-2 on the calcium concentration within the lumen of the endoplasmic reticulum. Although several prominent reports indicate that Bcl-2 overexpression is associated with a decrease in luminal calcium, there are a large number of reports indicating that Bcl-2 either does not decrease luminal calcium or actually increases luminal calcium. This review summarizes this vast array of conflicting findings, and analyses potential reasons why different conclusions have been reached by different laboratories. Future directions are emphasized that might bring clarity to this important area of apoptosis biology.
Collapse
Affiliation(s)
- Clark W Distelhorst
- Division of Hematology/Oncology, Comprehensive Cancer Center, University Hospitals of Cleveland and Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106-4937, USA.
| | | |
Collapse
|