1
|
Filadi R, De Mario A, Audano M, Romani P, Pedretti S, Cardenas C, Dupont S, Mammucari C, Mitro N, Pizzo P. Sustained IP3-linked Ca2+ signaling promotes progression of triple negative breast cancer cells by regulating fatty acid metabolism. Front Cell Dev Biol 2023; 11:1071037. [PMID: 36994106 PMCID: PMC10040683 DOI: 10.3389/fcell.2023.1071037] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 03/03/2023] [Indexed: 03/14/2023] Open
Abstract
Rewiring of mitochondrial metabolism has been described in different cancers as a key step for their progression. Calcium (Ca2+) signaling regulates mitochondrial function and is known to be altered in several malignancies, including triple negative breast cancer (TNBC). However, whether and how the alterations in Ca2+ signaling contribute to metabolic changes in TNBC has not been elucidated. Here, we found that TNBC cells display frequent, spontaneous inositol 1,4,5-trisphosphate (IP3)-dependent Ca2+ oscillations, which are sensed by mitochondria. By combining genetic, pharmacologic and metabolomics approaches, we associated this pathway with the regulation of fatty acid (FA) metabolism. Moreover, we demonstrated that these signaling routes promote TNBC cell migration in vitro, suggesting they might be explored to identify potential therapeutic targets.
Collapse
Affiliation(s)
- Riccardo Filadi
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
- Department of Biomedical Sciences, University of Padova, Padua, Italy
- *Correspondence: Riccardo Filadi, ,
| | - Agnese De Mario
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Matteo Audano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Patrizia Romani
- Department of Molecular Medicine (DMM), University of Padova, Padua, Italy
| | - Silvia Pedretti
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Cesar Cardenas
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
- Buck Institute for Research on Aging, Novato, CA, United States
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA, United States
| | - Sirio Dupont
- Department of Molecular Medicine (DMM), University of Padova, Padua, Italy
| | - Cristina Mammucari
- Department of Biomedical Sciences, University of Padova, Padua, Italy
- Myology Center (CIR-Myo), University of Padova, Padua, Italy
| | - Nico Mitro
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Paola Pizzo
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| |
Collapse
|
2
|
Smith BS, Hewitt T, Bakovic M, Lu R. ER stress-associated transcription factor CREB3 is essential for normal Ca 2+, ATP, and ROS homeostasis. Mitochondrion 2023; 69:10-17. [PMID: 36627030 DOI: 10.1016/j.mito.2023.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 12/24/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023]
Abstract
In mammalian cells, mitochondrial respiration produces reactive oxygen species (ROS) such as superoxide (O2-), which is then converted by the SOD1 enzyme into hydrogen peroxide (H2O2), the predominant form of cytosolic ROS. ROS at high levels can be toxic, but below this threshold are important for physiological processes acting as a second messenger similar to Ca2+. Mitochondrial Ca2+ influx from the ER increases ATP and ROS production, while ATP and ROS can regulate Ca2+ homeostasis, leading to an intricate interplay between Ca2+, ROS, and ATP synthesis. The Unfolded Protein Response (UPR) proteins ATF6α and XBP1 contribute to protection from oxidative stress through upregulation of Sod1 and Catalase genes. Here, UPR-associated protein CREB3 is shown to play a role in balancing Ca2+, ROS, and ATP homeostasis. Creb3-deficient mouse embryonic fibroblast cells (MEF-/-) were susceptible to H2O2-induced oxidative stress while having a functioning antioxidant gene expression response compared to MEF+/+. MEF-/- cells also contained elevated basal cytosolic ROS levels, which was attributed to drastically increased basal mitochondrial respiration and spare respiratory capacity relative to MEF+/+. MEF-/- cells also showed an increase in endoplasmic reticulum Ca2+ release and mitochondrial Ca2+ levels hinting at a potential cause for MEF-/- cell mitochondrial dysfunction. These results suggest that CREB3 is essential for maintaining proper Ca2+, ATP, and ROS homeostasis in mammalian cells.
Collapse
Affiliation(s)
- Brandon S Smith
- University of Guelph, Department of Molecular and Cellular Biology, Canada
| | - Tristen Hewitt
- University of Guelph, Department of Molecular and Cellular Biology, Canada
| | - Marica Bakovic
- University of Guelph, Department of Human Health & Nutritional Sciences, Canada.
| | - Ray Lu
- University of Guelph, Department of Molecular and Cellular Biology, Canada.
| |
Collapse
|
3
|
Petrick HL, Brownell S, Vachon B, Brunetta HS, Handy RM, van Loon LJC, Murrant CL, Holloway GP. Dietary nitrate increases submaximal SERCA activity and ADP transfer to mitochondria in slow-twitch muscle of female mice. Am J Physiol Endocrinol Metab 2022; 323:E171-E184. [PMID: 35732003 DOI: 10.1152/ajpendo.00371.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Rapid oscillations in cytosolic calcium (Ca2+) coordinate muscle contraction, relaxation, and physical movement. Intriguingly, dietary nitrate decreases ATP cost of contraction, increases force production, and increases cytosolic Ca2+, which would seemingly necessitate a greater demand for sarcoplasmic reticulum Ca2+ ATPase (SERCA) to sequester Ca2+ within the sarcoplasmic reticulum (SR) during relaxation. As SERCA is highly regulated, we aimed to determine the effect of 7-day nitrate supplementation (1 mM via drinking water) on SERCA enzymatic properties and the functional interaction between SERCA and mitochondrial oxidative phosphorylation. In soleus, we report that dietary nitrate increased force production across all stimulation frequencies tested, and throughout a 25 min fatigue protocol. Mice supplemented with nitrate also displayed an ∼25% increase in submaximal SERCA activity and SERCA efficiency (P = 0.053) in the soleus. To examine a possible link between ATP consumption and production, we established a methodology coupling SERCA and mitochondria in permeabilized muscle fibers. The premise of this experiment is that the addition of Ca2+ in the presence of ATP generates ADP from SERCA to support mitochondrial respiration. Similar to submaximal SERCA activity, mitochondrial respiration supported by SERCA-derived ADP was increased by ∼20% following nitrate in red gastrocnemius. This effect was fully attenuated by the SERCA inhibitor cyclopiazonic acid and was not attributed to differences in mitochondrial oxidative capacity, ADP sensitivity, protein content, or reactive oxygen species emission. Overall, these findings suggest that improvements in submaximal SERCA kinetics may contribute to the effects of nitrate on force production during fatigue.NEW & NOTEWORTHY We show that nitrate supplementation increased force production during fatigue and increased submaximal SERCA activity. This was also evident regarding the high-energy phosphate transfer from SERCA to mitochondria, as nitrate increased mitochondrial respiration supported by SERCA-derived ADP. Surprisingly, these observations were only apparent in muscle primarily expressing type I (soleus) but not type II fibers (EDL). These findings suggest that alterations in SERCA properties are a possible mechanism in which nitrate increases force during fatiguing contractions.
Collapse
Affiliation(s)
- Heather L Petrick
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Stuart Brownell
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Bayley Vachon
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Henver S Brunetta
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
- Department of Physiological Sciences, Federal University of Santa Catarina, Santa Catarina, Brazil
| | - Rachel M Handy
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Luc J C van Loon
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Coral L Murrant
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Graham P Holloway
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
4
|
Reddish FN, Miller CL, Deng X, Dong B, Patel AA, Ghane MA, Mosca B, McBean C, Wu S, Solntsev KM, Zhuo Y, Gadda G, Fang N, Cox DN, Mabb AM, Treves S, Zorzato F, Yang JJ. Rapid subcellular calcium responses and dynamics by calcium sensor G-CatchER . iScience 2021; 24:102129. [PMID: 33665552 PMCID: PMC7900224 DOI: 10.1016/j.isci.2021.102129] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 12/14/2020] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
The precise spatiotemporal characteristics of subcellular calcium (Ca2+) transients are critical for the physiological processes. Here we report a green Ca2+ sensor called "G-CatchER+" using a protein design to report rapid local ER Ca2+ dynamics with significantly improved folding properties. G-CatchER+ exhibits a superior Ca2+ on rate to G-CEPIA1er and has a Ca2+-induced fluorescence lifetimes increase. G-CatchER+ also reports agonist/antagonist triggered Ca2+ dynamics in several cell types including primary neurons that are orchestrated by IP3Rs, RyRs, and SERCAs with an ability to differentiate expression. Upon localization to the lumen of the RyR channel (G-CatchER+-JP45), we report a rapid local Ca2+ release that is likely due to calsequestrin. Transgenic expression of G-CatchER+ in Drosophila muscle demonstrates its utility as an in vivo reporter of stimulus-evoked SR local Ca2+ dynamics. G-CatchER+ will be an invaluable tool to examine local ER/SR Ca2+ dynamics and facilitate drug development associated with ER dysfunction.
Collapse
Affiliation(s)
- Florence N. Reddish
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Cassandra L. Miller
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Xiaonan Deng
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Bin Dong
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Atit A. Patel
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30303, USA
| | - Mohammad A. Ghane
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30303, USA
| | - Barbara Mosca
- Department of Life Sciences, General Pathology, University of Ferrara, Ferrara, Italy
| | - Cheyenne McBean
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Shengnan Wu
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA
| | - Kyril M. Solntsev
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - You Zhuo
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Giovanni Gadda
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Ning Fang
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Daniel N. Cox
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30303, USA
| | - Angela M. Mabb
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30303, USA
| | - Susan Treves
- Department of Life Sciences, General Pathology, University of Ferrara, Ferrara, Italy
- Department of Biomedicine, Basel University, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Francesco Zorzato
- Department of Life Sciences, General Pathology, University of Ferrara, Ferrara, Italy
- Department of Biomedicine, Basel University, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Jenny J. Yang
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
5
|
Rossini M, Pizzo P, Filadi R. Better to keep in touch: investigating inter‐organelle cross‐talk. FEBS J 2020; 288:740-755. [DOI: 10.1111/febs.15451] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/28/2020] [Accepted: 06/11/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Michela Rossini
- Department of Biomedical Sciences University of Padua Padua Italy
| | - Paola Pizzo
- Department of Biomedical Sciences University of Padua Padua Italy
- Neuroscience Institute National Research Council (CNR) Padua Italy
| | - Riccardo Filadi
- Department of Biomedical Sciences University of Padua Padua Italy
- Neuroscience Institute National Research Council (CNR) Padua Italy
| |
Collapse
|
6
|
Zakyrjanova GF, Gilmutdinov AI, Tsentsevitsky AN, Petrov AM. Olesoxime, a cholesterol-like neuroprotectant restrains synaptic vesicle exocytosis in the mice motor nerve terminals: Possible role of VDACs. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158739. [PMID: 32428575 DOI: 10.1016/j.bbalip.2020.158739] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/11/2020] [Accepted: 05/13/2020] [Indexed: 12/13/2022]
Abstract
Olesoxime is a cholesterol-like neuroprotective compound that targets to mitochondrial voltage dependent anion channels (VDACs). VDACs were also found in the plasma membrane and highly expressed in the presynaptic compartment. Here, we studied the effects of olesoxime and VDAC inhibitors on neurotransmission in the mouse neuromuscular junction. Electrophysiological analysis revealed that olesoxime suppressed selectively evoked neurotransmitter release in response to a single stimulus and 20 Hz activity. Also olesoxime decreased the rate of FM1-43 dye loss (an indicator of synaptic vesicle exocytosis) at low frequency stimulation and 20 Hz. Furthermore, an increase in extracellular Cl- enhanced the action of olesoxime on the exocytosis and olesoxime increased intracellular Cl- levels. The effects of olesoxime on the evoked synaptic vesicle exocytosis and [Cl-]i were blocked by membrane-permeable and impermeable VDAC inhibitors. Immunofluorescent labeling pointed on the presence of VDACs on the synaptic membranes. Rotenone-induced mitochondrial dysfunction perturbed the exocytotic release of FM1-43 and cell-permeable VDAC inhibitor (but not olesoxime or impermeable VDAC inhibitor) partially mitigated the rotenone-driven alterations in the FM1-43 unloading and mitochondrial superoxide production. Thus, olesoxime restrains neurotransmission by acting on plasmalemmal VDACs whose activation can limit synaptic vesicle exocytosis probably via increasing anion flux into the nerve terminals.
Collapse
Affiliation(s)
- Guzalia F Zakyrjanova
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, box 30, Kazan 420111, Russia; Institute of Neuroscience, Kazan State Medial University, 49 Butlerova Street, Kazan 420012, Russia
| | - Amir I Gilmutdinov
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, box 30, Kazan 420111, Russia
| | - Andrey N Tsentsevitsky
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, box 30, Kazan 420111, Russia
| | - Alexey M Petrov
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", 2/31 Lobachevsky Street, box 30, Kazan 420111, Russia; Institute of Neuroscience, Kazan State Medial University, 49 Butlerova Street, Kazan 420012, Russia.
| |
Collapse
|
7
|
Abstract
The process of embryonic development is crucial and radically influences preimplantation embryo competence. It involves oocyte maturation, fertilization, cell division and blastulation and is characterized by different key phases that have major influences on embryo quality. Each stage of the process of preimplantation embryonic development is led by important signalling pathways that include very many regulatory molecules, such as primary and secondary messengers. Many studies, both in vivo and in vitro, have shown the importance of the contribution of reactive oxygen species (ROS) as important second messengers in embryo development. ROS may originate from embryo metabolism and/or oocyte/embryo surroundings, and their effect on embryonic development is highly variable, depending on the needs of the embryo at each stage of development and on their environment (in vivo or under in vitro culture conditions). Other studies have also shown the deleterious effects of ROS in embryo development, when cellular tissue production overwhelms antioxidant production, leading to oxidative stress. This stress is known to be the cause of many cellular alterations, such as protein, lipid, and DNA damage. Considering that the same ROS level can have a deleterious effect on the fertilizing oocyte or embryo at certain stages, and a positive effect at another stage of the development process, further studies need to be carried out to determine the rate of ROS that benefits the embryo and from what rate it starts to be harmful, this measured at each key phase of embryonic development.
Collapse
|
8
|
Zhang X, Lee MD, Wilson C, McCarron JG. Hydrogen peroxide depolarizes mitochondria and inhibits IP 3-evoked Ca 2+ release in the endothelium of intact arteries. Cell Calcium 2019; 84:102108. [PMID: 31715384 PMCID: PMC6891240 DOI: 10.1016/j.ceca.2019.102108] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/30/2019] [Accepted: 10/30/2019] [Indexed: 12/30/2022]
Abstract
H2O2 is produced by several cell processes including mitochondria and may act as an intracellular messenger and cell-cell signalling molecule. Spontaneous local Ca2+ signals and IP3-evoked Ca2+ increases were inhibited by H2O2. H2O2 suppression of IP3-evoked Ca2+ signalling may be mediated by mitochondria via a decrease in the mitochondrial membrane potential. H2O2-induced mitochondrial depolarization and inhibition of IP3-evoked Ca2+ release, may protect mitochondria from Ca2+ overload during IP3-linked Ca2+ signals.
Hydrogen peroxide (H2O2) is a mitochondrial-derived reactive oxygen species (ROS) that regulates vascular signalling transduction, vasocontraction and vasodilation. Although the physiological role of ROS in endothelial cells is acknowledged, the mechanisms underlying H2O2 regulation of signalling in native, fully-differentiated endothelial cells is unresolved. In the present study, the effects of H2O2 on Ca2+ signalling were investigated in the endothelium of intact rat mesenteric arteries. Spontaneous local Ca2+ signals and acetylcholine evoked Ca2+ increases were inhibited by H2O2. H2O2 inhibition of acetylcholine-evoked Ca2+ signals was reversed by catalase. H2O2 exerts its inhibition on the IP3 receptor as Ca2+ release evoked by photolysis of caged IP3 was supressed by H2O2. H2O2 suppression of IP3-evoked Ca2+ signalling may be mediated by mitochondria. H2O2 depolarized mitochondria membrane potential. Acetylcholine-evoked Ca2+ release was inhibited by depolarisation of the mitochondrial membrane potential by the uncoupler carbonyl cyanide 3-chlorophenylhydrazone (CCCP) or complex 1 inhibitor, rotenone. We propose that the suppression of IP3-evoked Ca2+ release by H2O2 arises from the decrease in mitochondrial membrane potential. These results suggest that mitochondria may protect themselves against Ca2+ overload during IP3-linked Ca2+ signals by a H2O2 mediated negative feedback depolarization of the organelle and inhibition of IP3-evoked Ca2+ release.
Collapse
Affiliation(s)
- Xun Zhang
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Matthew D Lee
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Calum Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - John G McCarron
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| |
Collapse
|
9
|
Wilson C, Lee MD, Heathcote HR, Zhang X, Buckley C, Girkin JM, Saunter CD, McCarron JG. Mitochondrial ATP production provides long-range control of endothelial inositol trisphosphate-evoked calcium signaling. J Biol Chem 2019; 294:737-758. [PMID: 30498088 PMCID: PMC6341391 DOI: 10.1074/jbc.ra118.005913] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 11/27/2018] [Indexed: 01/09/2023] Open
Abstract
Endothelial cells are reported to be glycolytic and to minimally rely on mitochondria for ATP generation. Rather than providing energy, mitochondria in endothelial cells may act as signaling organelles that control cytosolic Ca2+ signaling or modify reactive oxygen species (ROS). To control Ca2+ signaling, these organelles are often observed close to influx and release sites and may be tethered near Ca2+ transporters. In this study, we used high-resolution, wide-field fluorescence imaging to investigate the regulation of Ca2+ signaling by mitochondria in large numbers of endothelial cells (∼50 per field) in intact arteries from rats. We observed that mitochondria were mostly spherical or short-rod structures and were distributed widely throughout the cytoplasm. The density of these organelles did not increase near contact sites with smooth muscle cells. However, local inositol trisphosphate (IP3)-mediated Ca2+ signaling predominated near these contact sites and required polarized mitochondria. Of note, mitochondrial control of Ca2+ signals occurred even when mitochondria were far from Ca2+ release sites. Indeed, the endothelial mitochondria were mobile and moved throughout the cytoplasm. Mitochondrial control of Ca2+ signaling was mediated by ATP production, which, when reduced by mitochondrial depolarization or ATP synthase inhibition, eliminated local IP3-mediated Ca2+ release events. ROS buffering did not significantly alter local Ca2+ release events. These results highlight the importance of mitochondrial ATP production in providing long-range control of endothelial signaling via IP3-evoked local Ca2+ release in intact endothelium.
Collapse
Affiliation(s)
- Calum Wilson
- From the Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, SIPBS Building, 161 Cathedral Street, Glasgow G4 0RE, Scotland, United Kingdom and
| | - Matthew D Lee
- From the Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, SIPBS Building, 161 Cathedral Street, Glasgow G4 0RE, Scotland, United Kingdom and
| | - Helen R Heathcote
- From the Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, SIPBS Building, 161 Cathedral Street, Glasgow G4 0RE, Scotland, United Kingdom and
| | - Xun Zhang
- From the Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, SIPBS Building, 161 Cathedral Street, Glasgow G4 0RE, Scotland, United Kingdom and
| | - Charlotte Buckley
- From the Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, SIPBS Building, 161 Cathedral Street, Glasgow G4 0RE, Scotland, United Kingdom and
| | - John M Girkin
- the Centre for Advanced Instrumentation, Biophysical Sciences Institute, Department of Physics, Durham University, South Road, Durham DH1 3LE, United Kingdom
| | - Christopher D Saunter
- the Centre for Advanced Instrumentation, Biophysical Sciences Institute, Department of Physics, Durham University, South Road, Durham DH1 3LE, United Kingdom
| | - John G McCarron
- From the Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, SIPBS Building, 161 Cathedral Street, Glasgow G4 0RE, Scotland, United Kingdom and
| |
Collapse
|
10
|
Song J, Yang R, Yang J, Zhou L. Mitochondrial Dysfunction-Associated Arrhythmogenic Substrates in Diabetes Mellitus. Front Physiol 2018; 9:1670. [PMID: 30574091 PMCID: PMC6291470 DOI: 10.3389/fphys.2018.01670] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/07/2018] [Indexed: 12/15/2022] Open
Abstract
There is increasing evidence that diabetic cardiomyopathy increases the risk of cardiac arrhythmia and sudden cardiac death. While the detailed mechanisms remain incompletely understood, the loss of mitochondrial function, which is often observed in the heart of patients with diabetes, has emerged as a key contributor to the arrhythmogenic substrates. In this mini review, the pathophysiology of mitochondrial dysfunction in diabetes mellitus is explored in detail, followed by descriptions of several mechanisms potentially linking mitochondria to arrhythmogenesis in the context of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Jiajia Song
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ruilin Yang
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States.,Key Laboratory of Mechanism Theory and Equipment Design of Ministry of Education, Tianjin University, Tianjin, China
| | - Jing Yang
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Lufang Zhou
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
11
|
Filadi R, Basso E, Lefkimmiatis K, Pozzan T. Beyond Intracellular Signaling: The Ins and Outs of Second Messengers Microdomains. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 981:279-322. [PMID: 29594866 DOI: 10.1007/978-3-319-55858-5_12] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A typical characteristic of eukaryotic cells compared to prokaryotes is represented by the spatial heterogeneity of the different structural and functional components: for example, most of the genetic material is surrounded by a highly specific membrane structure (the nuclear membrane), continuous with, yet largely different from, the endoplasmic reticulum (ER); oxidative phosphorylation is carried out by organelles enclosed by a double membrane, the mitochondria; in addition, distinct domains, enriched in specific proteins, are present in the plasma membrane (PM) of most cells. Less obvious, but now generally accepted, is the notion that even the concentration of small molecules such as second messengers (Ca2+ and cAMP in particular) can be highly heterogeneous within cells. In the case of most organelles, the differences in the luminal levels of second messengers depend either on the existence on their membrane of proteins that allow the accumulation/release of the second messenger (e.g., in the case of Ca2+, pumps, exchangers or channels), or on the synthesis and degradation of the specific molecule within the lumen (the autonomous intramitochondrial cAMP system). It needs stressing that the existence of a surrounding membrane does not necessarily imply the existence of a gradient between the cytosol and the organelle lumen. For example, the nuclear membrane is highly permeable to both Ca2+ and cAMP (nuclear pores are permeable to solutes up to 50 kDa) and differences in [Ca2+] or [cAMP] between cytoplasm and nucleoplasm are not seen in steady state and only very transiently during cell activation. A similar situation has been observed, as far as Ca2+ is concerned, in peroxisomes.
Collapse
Affiliation(s)
- Riccardo Filadi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Emy Basso
- Institute of Neuroscience, Padova Section, National Research Council, Padova, Italy
| | - Konstantinos Lefkimmiatis
- Institute of Neuroscience, Padova Section, National Research Council, Padova, Italy
- Venetian Institute of Molecular Medicine, Padova, Italy
| | - Tullio Pozzan
- Department of Biomedical Sciences, University of Padova, Padova, Italy.
- Institute of Neuroscience, Padova Section, National Research Council, Padova, Italy.
- Venetian Institute of Molecular Medicine, Padova, Italy.
| |
Collapse
|
12
|
Behringer EJ, Segal SS. Impact of Aging on Calcium Signaling and Membrane Potential in Endothelium of Resistance Arteries: A Role for Mitochondria. J Gerontol A Biol Sci Med Sci 2017; 72:1627-1637. [PMID: 28510636 DOI: 10.1093/gerona/glx079] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 04/21/2017] [Indexed: 12/20/2022] Open
Abstract
Impaired blood flow to peripheral tissues during advanced age is associated with endothelial dysfunction and diminished bioavailability of nitric oxide (NO). However, it is unknown whether aging impacts coupling between intracellular calcium ([Ca2+]i) signaling and small- and intermediate K+ channel (SKCa/IKCa) activity during endothelium-derived hyperpolarization (EDH), a signaling pathway integral to dilation of the resistance vasculature. To address the potential impact of aging on EDH, Fura-2 photometry and intracellular recording were applied to evaluate [Ca2+]i and membrane potential of intact endothelial tubes (width, 60 µm; length, 1-3 mm) freshly isolated from superior epigastric arteries of young (4-6 mo) and old (24-26 mo) male C57BL/6 mice. In response to acetylcholine, intracellular release of Ca2+ from the endoplasmic reticulum (ER) was enhanced with aging. Further, treatment with the mitochondrial uncoupler FCCP evoked a significant increase of [Ca2+]i with membrane hyperpolarization in an SKCa/IKCa-dependent manner in the endothelium of old but not young mice. We conclude that the ability of resistance artery endothelium to release Ca2+ from intracellular stores (ie, ER and mitochondria) and hyperpolarize Vm via SKCa/IKCa activation is augmented as compensation for reduced NO bioavailability during advanced age.
Collapse
Affiliation(s)
- Erik J Behringer
- Department of Basic Sciences, Loma Linda University, California.,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia.,Dalton Cardiovascular Research Center, Columbia, Missouri
| |
Collapse
|
13
|
Stefan CJ, Trimble WS, Grinstein S, Drin G, Reinisch K, De Camilli P, Cohen S, Valm AM, Lippincott-Schwartz J, Levine TP, Iaea DB, Maxfield FR, Futter CE, Eden ER, Judith D, van Vliet AR, Agostinis P, Tooze SA, Sugiura A, McBride HM. Membrane dynamics and organelle biogenesis-lipid pipelines and vesicular carriers. BMC Biol 2017; 15:102. [PMID: 29089042 PMCID: PMC5663033 DOI: 10.1186/s12915-017-0432-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Discoveries spanning several decades have pointed to vital membrane lipid trafficking pathways involving both vesicular and non-vesicular carriers. But the relative contributions for distinct membrane delivery pathways in cell growth and organelle biogenesis continue to be a puzzle. This is because lipids flow from many sources and across many paths via transport vesicles, non-vesicular transfer proteins, and dynamic interactions between organelles at membrane contact sites. This forum presents our latest understanding, appreciation, and queries regarding the lipid transport mechanisms necessary to drive membrane expansion during organelle biogenesis and cell growth.
Collapse
Affiliation(s)
- Christopher J. Stefan
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT UK
| | - William S. Trimble
- Cell Biology Program, The Hospital for Sick Children and Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Sergio Grinstein
- Cell Biology Program, The Hospital for Sick Children and Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Guillaume Drin
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Karin Reinisch
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520 USA
| | - Pietro De Camilli
- Department of Neuroscience and Cell Biology, Howard Hughes Medical Institute, Kavli Institute for Neuroscience and Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT 06510 USA
| | | | | | | | - Tim P. Levine
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - David B. Iaea
- Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Frederick R. Maxfield
- Department of Biochemistry, Weill Cornell Medical College, 1300 York Ave, New York, NY 10065 USA
| | - Clare E. Futter
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Emily R. Eden
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - Delphine Judith
- Molecular Cell Biology of Autophagy Laboratory, The Francis Crick Institute, London, UK
| | - Alexander R. van Vliet
- Molecular Cell Biology of Autophagy Laboratory, The Francis Crick Institute, London, UK
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Sharon A. Tooze
- Molecular Cell Biology of Autophagy Laboratory, The Francis Crick Institute, London, UK
| | - Ayumu Sugiura
- Kobe University Graduate School of Medicine, 1-5-6 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047 Japan
| | - Heidi M. McBride
- Montreal Neurological Institute, McGill University, 3801 University Avenue, Montreal, Quebec H3A 2B4 Canada
| |
Collapse
|
14
|
Charles E, Hammadi M, Kischel P, Delcroix V, Demaurex N, Castelbou C, Vacher AM, Devin A, Ducret T, Nunes P, Vacher P. The antidepressant fluoxetine induces necrosis by energy depletion and mitochondrial calcium overload. Oncotarget 2017; 8:3181-3196. [PMID: 27911858 PMCID: PMC5356874 DOI: 10.18632/oncotarget.13689] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 11/21/2016] [Indexed: 11/25/2022] Open
Abstract
Selective Serotonin Reuptake Inhibitor antidepressants, such as fluoxetine (Prozac), have been shown to induce cell death in cancer cells, paving the way for their potential use as cancer therapy. These compounds are able to increase cytosolic calcium concentration ([Ca2+]cyt), but the involved mechanisms and their physiological consequences are still not well understood. Here, we show that fluoxetine induces an increase in [Ca2+]cyt by emptying the endoplasmic reticulum (ER) through the translocon, an ER Ca2+ leakage structure. Our data also show that fluoxetine inhibits oxygen consumption and lowers mitochondrial ATP. This latter is essential for Ca2+ reuptake into the ER, and we postulated therefore that the fluoxetine-induced decrease in mitochondrial ATP production results in the emptying of the ER, leading to capacitative calcium entry. Furthermore, Ca2+ quickly accumulated in the mitochondria, leading to mitochondrial Ca2+ overload and cell death. We found that fluoxetine could induce an early necrosis in human peripheral blood lymphocytes and Jurkat cells, and could also induce late apoptosis, especially in the tumor cell line. These results shed light on fluoxetine-induced cell death and its potential use in cancer treatment.
Collapse
Affiliation(s)
- Emilie Charles
- INSERM U1218, Institut Bergonié, Bordeaux, France
- Université de Bordeaux, Bordeaux, France
| | - Mehdi Hammadi
- INSERM U1218, Institut Bergonié, Bordeaux, France
- Université de Bordeaux, Bordeaux, France
| | - Philippe Kischel
- Laboratory of Cellular and Molecular Physiology EA4667, Université de Picardie Jules Verne, SFR CAP-SANTE (FED 4231), Amiens, France
| | - Vanessa Delcroix
- INSERM U1218, Institut Bergonié, Bordeaux, France
- Université de Bordeaux, Bordeaux, France
| | - Nicolas Demaurex
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Cyril Castelbou
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Anne-Marie Vacher
- INSERM U1218, Institut Bergonié, Bordeaux, France
- Université de Bordeaux, Bordeaux, France
| | - Anne Devin
- Institut de Biochimie et Génétique Cellulaires, UMR 5095, Bordeaux, France
- Université de Bordeaux, Bordeaux, France
| | - Thomas Ducret
- INSERM U1045, Centre de Recherche Cardio-Thoracique, Bordeaux, France
- Université de Bordeaux, Bordeaux, France
| | - Paula Nunes
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Pierre Vacher
- INSERM U1218, Institut Bergonié, Bordeaux, France
- Université de Bordeaux, Bordeaux, France
| |
Collapse
|
15
|
Over Six Decades of Discovery and Characterization of the Architecture at Mitochondria-Associated Membranes (MAMs). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 997:13-31. [PMID: 28815519 DOI: 10.1007/978-981-10-4567-7_2] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The discovery of proteins regulating ER-mitochondria tethering including phosphofurin acidic cluster sorting protein 2 (PACS-2) and mitofusin-2 has pushed contact sites between the endoplasmic reticulum (ER) and mitochondria into the spotlight of cell biology. While the field is developing rapidly and controversies have come and gone multiple times during its history, it is sometimes overlooked that significant research has been done decades ago with the original discovery of these structures in the 1950s and the first characterization of their function (and coining of the term mitochondria-associated membrane, MAM) in 1990. Today, an ever-increasing array of proteins localize to the MAM fraction of the endoplasmic reticulum (ER) to regulate the interaction of this organelle with mitochondria. These mitochondria-ER contacts, sometimes referred to as MERCs, regulate a multitude of biological functions, including lipid metabolism, Ca2+ signaling, bioenergetics, inflammation, autophagy, mitochondrial structure, and apoptosis.
Collapse
|
16
|
Pendin D, Greotti E, Lefkimmiatis K, Pozzan T. Exploring cells with targeted biosensors. J Gen Physiol 2016; 149:1-36. [PMID: 28028123 PMCID: PMC5217087 DOI: 10.1085/jgp.201611654] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 10/26/2016] [Accepted: 12/01/2016] [Indexed: 01/10/2023] Open
Abstract
Cellular signaling networks are composed of multiple pathways, often interconnected, that form complex networks with great potential for cross-talk. Signal decoding depends on the nature of the message as well as its amplitude, temporal pattern, and spatial distribution. In addition, the existence of membrane-bound organelles, which are both targets and generators of messages, add further complexity to the system. The availability of sensors that can localize to specific compartments in live cells and monitor their targets with high spatial and temporal resolution is thus crucial for a better understanding of cell pathophysiology. For this reason, over the last four decades, a variety of strategies have been developed, not only to generate novel and more sensitive probes for ions, metabolites, and enzymatic activity, but also to selectively deliver these sensors to specific intracellular compartments. In this review, we summarize the principles that have been used to target organic or protein sensors to different cellular compartments and their application to cellular signaling.
Collapse
Affiliation(s)
- Diana Pendin
- Neuroscience Institute, National Research Council, Padua Section, 35121 Padua, Italy.,Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
| | - Elisa Greotti
- Neuroscience Institute, National Research Council, Padua Section, 35121 Padua, Italy.,Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
| | - Konstantinos Lefkimmiatis
- Neuroscience Institute, National Research Council, Padua Section, 35121 Padua, Italy.,Venetian Institute of Molecular Medicine, 35129 Padua, Italy
| | - Tullio Pozzan
- Neuroscience Institute, National Research Council, Padua Section, 35121 Padua, Italy.,Venetian Institute of Molecular Medicine, 35129 Padua, Italy.,Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
| |
Collapse
|
17
|
Almukhtar H, Garle M, Smith P, Roberts R. Effect of simvastatin on vascular tone in porcine coronary artery: Potential role of the mitochondria. Toxicol Appl Pharmacol 2016; 305:176-185. [DOI: 10.1016/j.taap.2016.06.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 06/08/2016] [Accepted: 06/21/2016] [Indexed: 12/17/2022]
|
18
|
Abstract
In the last 5 years, most of the molecules that control mitochondrial Ca(2+) homeostasis have been finally identified. Mitochondrial Ca(2+) uptake is mediated by the Mitochondrial Calcium Uniporter (MCU) complex, a macromolecular structure that guarantees Ca(2+) accumulation inside mitochondrial matrix upon increases in cytosolic Ca(2+). Conversely, Ca(2+) release is under the control of the Na(+)/Ca(2+) exchanger, encoded by the NCLX gene, and of a H(+)/Ca(2+) antiporter, whose identity is still debated. The low affinity of the MCU complex, coupled to the activity of the efflux systems, protects cells from continuous futile cycles of Ca(2+) across the inner mitochondrial membrane and consequent massive energy dissipation. In this review, we discuss the basic principles that govern mitochondrial Ca(2+) homeostasis and the methods used to investigate the dynamics of Ca(2+) concentration within the organelles. We discuss the functional and structural role of the different molecules involved in mitochondrial Ca(2+) handling and their pathophysiological role.
Collapse
Affiliation(s)
- Diego De Stefani
- Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy; , ,
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy; , , .,National Research Council (CNR) Neuroscience Institute, 35121 Padova, Italy
| | - Tullio Pozzan
- Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy; , , .,National Research Council (CNR) Neuroscience Institute, 35121 Padova, Italy.,Venetian Institute of Molecular Medicine, 35121 Padova, Italy
| |
Collapse
|
19
|
Xu L, Auzins A, Sun X, Xu Y, Harnischfeger F, Lu Y, Li Z, Chen YH, Zheng W, Liu W. The synaptic recruitment of lipid rafts is dependent on CD19-PI3K module and cytoskeleton remodeling molecules. J Leukoc Biol 2015; 98:223-34. [PMID: 25979433 DOI: 10.1189/jlb.2a0614-287rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 04/15/2015] [Indexed: 11/24/2022] Open
Abstract
Sphingolipid- and cholesterol-rich lipid raft microdomains are important in the initiation of BCR signaling. Although it is known that lipid rafts promote the coclustering of BCR and Lyn kinase microclusters within the B cell IS, the molecular mechanism of the recruitment of lipid rafts into the B cell IS is not understood completely. Here, we report that the synaptic recruitment of lipid rafts is dependent on the cytoskeleton-remodeling proteins, RhoA and Vav. Such an event is also efficiently regulated by motor proteins, myosin IIA and dynein. Further evidence suggests the synaptic recruitment of lipid rafts is, by principle, an event triggered by BCR signaling molecules and second messenger molecules. BCR-activating coreceptor CD19 potently enhances such an event depending on its cytoplasmic Tyr421 and Tyr482 residues. The enhancing function of the CD19-PI3K module in synaptic recruitment of lipid rafts is also confirmed in human peripheral blood B cells. Thus, these results improve our understanding of the molecular mechanism of the recruitment of lipid raft microdomains in B cell IS.
Collapse
Affiliation(s)
- Liling Xu
- *MOE Key Laboratory of Protein Science, School of Life Sciences, and State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China; Collaborative Innovation Center for Infectious Diseases, Hangzhou, China; Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China; Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Arturs Auzins
- *MOE Key Laboratory of Protein Science, School of Life Sciences, and State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China; Collaborative Innovation Center for Infectious Diseases, Hangzhou, China; Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China; Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Xiaolin Sun
- *MOE Key Laboratory of Protein Science, School of Life Sciences, and State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China; Collaborative Innovation Center for Infectious Diseases, Hangzhou, China; Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China; Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Yinsheng Xu
- *MOE Key Laboratory of Protein Science, School of Life Sciences, and State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China; Collaborative Innovation Center for Infectious Diseases, Hangzhou, China; Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China; Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Fiona Harnischfeger
- *MOE Key Laboratory of Protein Science, School of Life Sciences, and State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China; Collaborative Innovation Center for Infectious Diseases, Hangzhou, China; Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China; Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Yun Lu
- *MOE Key Laboratory of Protein Science, School of Life Sciences, and State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China; Collaborative Innovation Center for Infectious Diseases, Hangzhou, China; Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China; Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Zhanguo Li
- *MOE Key Laboratory of Protein Science, School of Life Sciences, and State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China; Collaborative Innovation Center for Infectious Diseases, Hangzhou, China; Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China; Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Ying-Hua Chen
- *MOE Key Laboratory of Protein Science, School of Life Sciences, and State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China; Collaborative Innovation Center for Infectious Diseases, Hangzhou, China; Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China; Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Wenjie Zheng
- *MOE Key Laboratory of Protein Science, School of Life Sciences, and State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China; Collaborative Innovation Center for Infectious Diseases, Hangzhou, China; Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China; Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Wanli Liu
- *MOE Key Laboratory of Protein Science, School of Life Sciences, and State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment, Tsinghua University, Beijing, China; Collaborative Innovation Center for Infectious Diseases, Hangzhou, China; Department of Rheumatology and Immunology, Clinical Immunology Center, Peking University People's Hospital, Beijing, China; Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; and Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| |
Collapse
|
20
|
Moccia F, Zuccolo E, Soda T, Tanzi F, Guerra G, Mapelli L, Lodola F, D'Angelo E. Stim and Orai proteins in neuronal Ca(2+) signaling and excitability. Front Cell Neurosci 2015; 9:153. [PMID: 25964739 PMCID: PMC4408853 DOI: 10.3389/fncel.2015.00153] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/03/2015] [Indexed: 02/01/2023] Open
Abstract
Stim1 and Orai1 are ubiquitous proteins that have long been known to mediate Ca2+ release-activated Ca2+ (CRAC) current (ICRAC) and store-operated Ca2+ entry (SOCE) only in non-excitable cells. SOCE is activated following the depletion of the endogenous Ca2+ stores, which are mainly located within the endoplasmic reticulum (ER), to replete the intracellular Ca2+ reservoir and engage specific Ca2+-dependent processes, such as proliferation, migration, cytoskeletal remodeling, and gene expression. Their paralogs, Stim2, Orai2 and Orai3, support SOCE in heterologous expression systems, but their physiological role is still obscure. Ca2+ inflow in neurons has long been exclusively ascribed to voltage-operated and receptor-operated channels. Nevertheless, recent work has unveiled that Stim1–2 and Orai1-2, but not Orai3, proteins are also expressed and mediate SOCE in neurons. Herein, we survey current knowledge about the neuronal distribution of Stim and Orai proteins in rodent and human brains; we further discuss that Orai2 is the main pore-forming subunit of CRAC channels in central neurons, in which it may be activated by either Stim1 or Stim2 depending on species, brain region and physiological stimuli. We examine the functions regulated by SOCE in neurons, where this pathway is activated under resting conditions to refill the ER, control spinogenesis and regulate gene transcription. Besides, we highlighted the possibility that SOCE also controls neuronal excitation and regulate synaptic plasticity. Finally, we evaluate the involvement of Stim and Orai proteins in severe neurodegenerative and neurological disorders, such as Alzheimer’s disease and epilepsy.
Collapse
Affiliation(s)
- Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia Pavia, Italy
| | - Estella Zuccolo
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia Pavia, Italy
| | - Teresa Soda
- Neurophysiology Unit, Department of Brain and Behavioral Sciences, University of Pavia Pavia, Italy
| | - Franco Tanzi
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia Pavia, Italy
| | - Germano Guerra
- Department of Medicine and Health Sciences, University of Molise Campobasso, Italy
| | - Lisa Mapelli
- Neurophysiology Unit, Department of Brain and Behavioral Sciences, University of Pavia Pavia, Italy ; Museo Storico della Fisica e Centro di Studi e Ricerche Enrico Fermi Roma, Italy
| | - Francesco Lodola
- Laboratory of Molecular Cardiology, IRCCS Fondazione Salvatore Maugeri Pavia, Italy
| | - Egidio D'Angelo
- Neurophysiology Unit, Department of Brain and Behavioral Sciences, University of Pavia Pavia, Italy ; Brain Connectivity Center, C. Mondino National Neurological Institute, Fondazione IRCCS Policlinico San Matteo Pavia Pavia, Italy
| |
Collapse
|
21
|
Filadi R, Pozzan T. Generation and functions of second messengers microdomains. Cell Calcium 2015; 58:405-14. [PMID: 25861743 DOI: 10.1016/j.ceca.2015.03.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 03/17/2015] [Accepted: 03/18/2015] [Indexed: 01/09/2023]
Abstract
A compelling example of the mechanisms by which the cells can organize and decipher complex and different functional activities is the convergence of a multitude of stimuli into signalling cascades, involving only few intracellular second messengers. The possibility of restricting these signalling events in distinct microdomains allows a fine and selective tuning of very different tasks. In this review, we will discuss the mechanisms that control the formation and the spatial distribution of Ca(2+) and cAMP microdomains, providing some examples of their functional consequences.
Collapse
Affiliation(s)
- Riccardo Filadi
- Department of Biomedical Sciences, University of Padova, Italy
| | - Tullio Pozzan
- Department of Biomedical Sciences, University of Padova, Italy; CNR Institute of Neuroscience, Padova Section, Padova, Italy; Venetian Institute of Molecular Medicine (VIMM), Padova, Italy.
| |
Collapse
|
22
|
Wu Y, Rasmussen TP, Koval OM, Joiner MLA, Hall DD, Chen B, Luczak ED, Wang Q, Rokita AG, Wehrens XHT, Song LS, Anderson ME. The mitochondrial uniporter controls fight or flight heart rate increases. Nat Commun 2015; 6:6081. [PMID: 25603276 PMCID: PMC4398998 DOI: 10.1038/ncomms7081] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 12/10/2014] [Indexed: 01/13/2023] Open
Abstract
Heart rate increases are a fundamental adaptation to physiological stress, while inappropriate heart rate increases are resistant to current therapies. However, the metabolic mechanisms driving heart rate acceleration in cardiac pacemaker cells remain incompletely understood. The mitochondrial calcium uniporter (MCU) facilitates calcium entry into the mitochondrial matrix to stimulate metabolism. We developed mice with myocardial MCU inhibition by transgenic expression of a dominant negative (DN) MCU. Here we show that DN-MCU mice had normal resting heart rates but were incapable of physiological fight or flight heart rate acceleration. We found MCU function was essential for rapidly increasing mitochondrial calcium in pacemaker cells and that MCU enhanced oxidative phoshorylation was required to accelerate reloading of an intracellular calcium compartment prior to each heartbeat. Our findings show the MCU is necessary for complete physiological heart rate acceleration and suggest MCU inhibition could reduce inappropriate heart rate increases without affecting resting heart rate.
Collapse
Affiliation(s)
- Yuejin Wu
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA
| | - Tyler P Rasmussen
- 1] Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA [2] Department of Molecular Physiology and Biophysics, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA
| | - Olha M Koval
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA
| | - Mei-Ling A Joiner
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA
| | - Duane D Hall
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA
| | - Biyi Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA
| | - Elizabeth D Luczak
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA
| | - Qiongling Wang
- Cardiovascular Research Institute, Department of Molecular Physiology and Biophysics and Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Adam G Rokita
- 1] Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA [2] Department of Internal Medicine II, University Hospital Regensburg, 93042 Regensburg, Germany
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Department of Molecular Physiology and Biophysics and Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Long-Sheng Song
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA
| | - Mark E Anderson
- 1] Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA [2] Department of Molecular Physiology and Biophysics, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA
| |
Collapse
|
23
|
Abstract
SIGNIFICANCE Store-operated Ca2+ entry (SOCE) is a ubiquitous Ca2+ signaling mechanism triggered by Ca2+ depletion of the endoplasmic reticulum (ER) and by a variety of cellular stresses. Reactive oxygen species (ROS) are often concomitantly produced in response to these stresses, however, the relationship between redox signaling and SOCE is not completely understood. Various cardiovascular, neurological, and immune diseases are associated with alterations in both Ca2+ signaling and ROS production, and thus understanding this relationship has therapeutic implications. RECENT ADVANCES Several reactive cysteine modifications in stromal interaction molecule (STIM) and Orai proteins comprising the core SOCE machinery were recently shown to modulate SOCE in a redox-dependent manner. Moreover, STIM1 and Orai1 expression levels may reciprocally regulate and be affected by responses to oxidative stress. ER proteins involved in oxidative protein folding have gained increased recognition as important sources of ROS, and the recent discovery of their accumulation in contact sites between the ER and mitochondria provides a further link between ROS production and intracellular Ca2+ handling. CRITICAL ISSUES AND FUTURE DIRECTIONS Future research should aim to establish the complete set of SOCE controlling molecules, to determine their redox-sensitive residues, and to understand how intracellular Ca2+ stores dynamically respond to different types of stress. Mapping the precise nature and functional consequence of key redox-sensitive components of the pre- and post-translational control of SOCE machinery and of proteins regulating ER calcium content will be pivotal in advancing our understanding of the complex cross-talk between redox and Ca2+ signaling.
Collapse
Affiliation(s)
- Paula Nunes
- Department of Cell Physiology and Metabolism, University of Geneva , Geneva, Switzerland
| | | |
Collapse
|
24
|
Billaud M, Lohman AW, Johnstone SR, Biwer LA, Mutchler S, Isakson BE. Regulation of cellular communication by signaling microdomains in the blood vessel wall. Pharmacol Rev 2014; 66:513-69. [PMID: 24671377 DOI: 10.1124/pr.112.007351] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
It has become increasingly clear that the accumulation of proteins in specific regions of the plasma membrane can facilitate cellular communication. These regions, termed signaling microdomains, are found throughout the blood vessel wall where cellular communication, both within and between cell types, must be tightly regulated to maintain proper vascular function. We will define a cellular signaling microdomain and apply this definition to the plethora of means by which cellular communication has been hypothesized to occur in the blood vessel wall. To that end, we make a case for three broad areas of cellular communication where signaling microdomains could play an important role: 1) paracrine release of free radicals and gaseous molecules such as nitric oxide and reactive oxygen species; 2) role of ion channels including gap junctions and potassium channels, especially those associated with the endothelium-derived hyperpolarization mediated signaling, and lastly, 3) mechanism of exocytosis that has considerable oversight by signaling microdomains, especially those associated with the release of von Willebrand factor. When summed, we believe that it is clear that the organization and regulation of signaling microdomains is an essential component to vessel wall function.
Collapse
Affiliation(s)
- Marie Billaud
- Dept. of Molecular Physiology and Biophysics, University of Virginia School of Medicine, PO Box 801394, Charlottesville, VA 22902.
| | | | | | | | | | | |
Collapse
|
25
|
Eisner V, Csordás G, Hajnóczky G. Interactions between sarco-endoplasmic reticulum and mitochondria in cardiac and skeletal muscle - pivotal roles in Ca²⁺ and reactive oxygen species signaling. J Cell Sci 2013; 126:2965-78. [PMID: 23843617 DOI: 10.1242/jcs.093609] [Citation(s) in RCA: 161] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mitochondria are strategically and dynamically positioned in the cell to spatially coordinate ATP production with energy needs and to allow the local exchange of material with other organelles. Interactions of mitochondria with the sarco-endoplasmic reticulum (SR/ER) have been receiving much attention owing to emerging evidence on the role these sites have in cell signaling, dynamics and biosynthetic pathways. One of the most important physiological and pathophysiological paradigms for SR/ER-mitochondria interactions is in cardiac and skeletal muscle. The contractile activity of these tissues has to be matched by mitochondrial ATP generation that is achieved, at least in part, by propagation of Ca(2+) signals from SR to mitochondria. However, the muscle has a highly ordered structure, providing only limited opportunity for mitochondrial dynamics and interorganellar interactions. This Commentary focuses on the latest advances in the structure, function and disease relevance of the communication between SR/ER and mitochondria in muscle. In particular, we discuss the recent demonstration of SR/ER-mitochondria tethers that are formed by multiple proteins, and local Ca(2+) transfer between SR/ER and mitochondria.
Collapse
Affiliation(s)
- Verónica Eisner
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | | | | |
Collapse
|
26
|
Gene expression profiling of rotenone-mediated cortical neuronal death: Evidence for inhibition of ubiquitin–proteasome system and autophagy-lysosomal pathway, and dysfunction of mitochondrial and calcium signaling. Neurochem Int 2013. [DOI: 10.1016/j.neuint.2012.11.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
27
|
Abstract
Intracellular calcium dynamics is critical for many functions of cerebellar granule cells (GrCs) including membrane excitability, synaptic plasticity, apoptosis, and regulation of gene transcription. Recent measurements of calcium responses in GrCs to depolarization and synaptic stimulation reveal spatial compartmentalization and heterogeneity within dendrites of these cells. However, the main determinants of local calcium dynamics in GrCs are still poorly understood. One reason is that there have been few published studies of calcium dynamics in intact GrCs in their native environment. In the absence of complete information, biophysically realistic models are useful for testing whether specific Ca(2+) handling mechanisms may account for existing experimental observations. Simulation results can be used to identify critical measurements that would discriminate between different models. In this review, we briefly describe experimental studies and phenomenological models of Ca(2+) signaling in GrC, and then discuss a particular biophysical model, with a special emphasis on an approach for obtaining information regarding the distribution of Ca(2+) handling systems under conditions of incomplete experimental data. Use of this approach suggests that Ca(2+) channels and fixed endogenous Ca(2+) buffers are highly heterogeneously distributed in GrCs. Research avenues for investigating calcium dynamics in GrCs by a combination of experimental and modeling studies are proposed.
Collapse
Affiliation(s)
- Elena È Saftenku
- Department of General Physiology of Nervous System, A. A. Bogomoletz Institute of Physiology, 4 Bogomoletz St., Kyiv 01024, Ukraine.
| |
Collapse
|
28
|
Carreras-Sureda A, Cantero-Recasens G, Rubio-Moscardo F, Kiefer K, Peinelt C, Niemeyer BA, Valverde MA, Vicente R. ORMDL3 modulates store-operated calcium entry and lymphocyte activation. Hum Mol Genet 2012; 22:519-30. [PMID: 23100328 DOI: 10.1093/hmg/dds450] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
T lymphocytes rely on a Ca(2+) signal known as store-operated calcium entry (SOCE) for their activation. This Ca(2+) signal is generated by activation of a T-cell receptor, depletion of endoplasmic reticulum (ER) Ca(2+) stores and activation of Ca(2+) release-activated Ca(2+) currents (I(CRAC)). Here, we report that the ER protein orosomucoid like 3 (ORMDL3), the product of the ORMDL3 gene associated with several autoimmune and/or inflammatory diseases, negatively modulates I(CRAC), SOCE, nuclear factor of activated T cells nuclear translocation and interleukin-2 production. ORMDL3 inhibits the Ca(2+) influx mechanism at the outer mitochondrial membrane, resulting in a Ca(2+)-dependent inhibition of I(CRAC) and reduced SOCE. The effect of ORMDL3 could be mimicked by interventions that decreased mitochondrial Ca(2+) influx and reverted by buffering of cytosolic Ca(2+) or activation of mitochondrial Ca(2+) influx. In conclusion, ORMDL3 modifies key steps in the process of T-lymphocyte activation, providing a functional link between the genetic associations of the ORMDL3 gene with autoimmune and/or inflammatory diseases.
Collapse
Affiliation(s)
- Amado Carreras-Sureda
- Laboratory of Molecular Physiology and Channelopathies, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona 08003, Spain
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Mitochondrial regulation of CRAC channel-driven cellular responses. Cell Calcium 2012; 52:52-6. [DOI: 10.1016/j.ceca.2012.02.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 02/08/2012] [Accepted: 02/09/2012] [Indexed: 11/15/2022]
|
30
|
Pizzo P, Drago I, Filadi R, Pozzan T. Mitochondrial Ca²⁺ homeostasis: mechanism, role, and tissue specificities. Pflugers Arch 2012; 464:3-17. [PMID: 22706634 DOI: 10.1007/s00424-012-1122-y] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 05/29/2012] [Indexed: 12/18/2022]
Abstract
Mitochondria from every tissue are quite similar in their capability to accumulate Ca²⁺ in a process that depends on the electrical potential across the inner membrane; it is catalyzed by a gated channel (named mitochondrial Ca²⁺ uniporter), the molecular identity of which has only recently been unraveled. The release of accumulated Ca²⁺ in mitochondria from different tissues is, on the contrary, quite variable, both in terms of speed and mechanism: a Na⁺-dependent efflux in excitable cells (catalyzed by NCLX) and a H⁺/Ca²⁺ exchanger in other cells. The efficacy of mitochondrial Ca²⁺ uptake in living cells is strictly dependent on the topological arrangement of the organelles with respect to the source of Ca²⁺ flowing into the cytoplasm, i.e., plasma membrane or intracellular channels. In turn, the structural and functional relationships between mitochondria and other cellular membranes are dictated by the specific architecture of different cells. Mitochondria not only modulate the amplitude and the kinetics of local and bulk cytoplasmic Ca²⁺ changes but also depend on the Ca²⁺ signal for their own functionality, in particular for their capacity to produce ATP. In this review, we summarize the processes involved in mitochondrial Ca²⁺ handling and its integration in cell physiology, highlighting the main common characteristics as well as key differences, in different tissues.
Collapse
Affiliation(s)
- Paola Pizzo
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | | | | | | |
Collapse
|
31
|
Abstract
Since 1929, when it was discovered that ATP is a substrate for muscle contraction, the knowledge about this purine nucleotide has been greatly expanded. Many aspects of cell metabolism revolve around ATP production and consumption. It is important to understand the concepts of glucose and oxygen consumption in aerobic and anaerobic life and to link bioenergetics with the vast amount of reactions occurring within cells. ATP is universally seen as the energy exchange factor that connects anabolism and catabolism but also fuels processes such as motile contraction, phosphorylations, and active transport. It is also a signalling molecule in the purinergic signalling mechanisms. In this review, we will discuss all the main mechanisms of ATP production linked to ADP phosphorylation as well the regulation of these mechanisms during stress conditions and in connection with calcium signalling events. Recent advances regarding ATP storage and its special significance for purinergic signalling will also be reviewed.
Collapse
|
32
|
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: 68] [Impact Index Per Article: 5.2] [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
|
33
|
De Marchi U, Castelbou C, Demaurex N. Uncoupling protein 3 (UCP3) modulates the activity of Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) by decreasing mitochondrial ATP production. J Biol Chem 2011; 286:32533-41. [PMID: 21775425 PMCID: PMC3173197 DOI: 10.1074/jbc.m110.216044] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Revised: 06/17/2011] [Indexed: 12/17/2022] Open
Abstract
The uncoupling proteins UCP2 and UCP3 have been postulated to catalyze Ca(2+) entry across the inner membrane of mitochondria, but this proposal is disputed, and other, unrelated proteins have since been identified as the mitochondrial Ca(2+) uniporter. To clarify the role of UCPs in mitochondrial Ca(2+) handling, we down-regulated the expression of the only uncoupling protein of HeLa cells, UCP3, and measured Ca(2+) and ATP levels in the cytosol and in organelles with genetically encoded probes. UCP3 silencing did not alter mitochondrial Ca(2+) uptake in permeabilized cells. In intact cells, however, UCP3 depletion increased mitochondrial ATP production and strongly reduced the cytosolic and mitochondrial Ca(2+) elevations evoked by histamine. The reduced Ca(2+) elevations were due to inhibition of store-operated Ca(2+) entry and reduced depletion of endoplasmic reticulum (ER) Ca(2+) stores. UCP3 depletion accelerated the ER Ca(2+) refilling kinetics, indicating that the activity of sarco/endoplasmic reticulum Ca(2+) (SERCA) pumps was increased. Accordingly, SERCA inhibitors reversed the effects of UCP3 depletion on cytosolic, ER, and mitochondrial Ca(2+) responses. Our results indicate that UCP3 is not a mitochondrial Ca(2+) uniporter and that it instead negatively modulates the activity of SERCA by limiting mitochondrial ATP production. The effects of UCP3 on mitochondrial Ca(2+) thus reflect metabolic alterations that impact on cellular Ca(2+) homeostasis. The sensitivity of SERCA to mitochondrial ATP production suggests that mitochondria control the local ATP availability at ER Ca(2+) uptake and release sites.
Collapse
Affiliation(s)
- Umberto De Marchi
- From the Department of Cell Physiology and Metabolism, University of Geneva, rue Michel-Servet, 1, CH-1211 Genève, Switzerland
| | - Cyril Castelbou
- From the Department of Cell Physiology and Metabolism, University of Geneva, rue Michel-Servet, 1, CH-1211 Genève, Switzerland
| | - Nicolas Demaurex
- From the Department of Cell Physiology and Metabolism, University of Geneva, rue Michel-Servet, 1, CH-1211 Genève, Switzerland
| |
Collapse
|
34
|
Van Summeren A, Renes J, Bouwman FG, Noben JP, van Delft JHM, Kleinjans JCS, Mariman ECM. Proteomics Investigations of Drug-Induced Hepatotoxicity in HepG2 Cells. Toxicol Sci 2010; 120:109-22. [DOI: 10.1093/toxsci/kfq380] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
35
|
Abstract
Peripheral taste receptor cells depend on distinct calcium signals to generate appropriate cellular responses that relay taste information to the central nervous system. Some taste cells have conventional chemical synapses and rely on calcium influx through voltage-gated calcium channels. Other taste cells lack these synapses and depend on calcium release from stores to formulate an output signal through a hemichannel. Despite the importance of calcium signaling in taste cells, little is known about how these signals are regulated. This review summarizes recent studies that have identified 2 calcium clearance mechanisms expressed in taste cells, including mitochondrial calcium uptake and sodium/calcium exchangers (NCXs). These studies identified a unique constitutive calcium influx that contributes to maintaining appropriate calcium homeostasis in taste cells and the role of the mitochondria and exchangers in this process. The additional role of NCXs in the regulation of evoked calcium responses is also discussed. Clearly, calcium signaling is a dynamic process in taste cells and appears to be more complex than has previously been appreciated.
Collapse
Affiliation(s)
- Kathryn F Medler
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA.
| |
Collapse
|
36
|
Ghibelli L, Diederich M. Multistep and multitask Bax activation. Mitochondrion 2010; 10:604-13. [PMID: 20709625 DOI: 10.1016/j.mito.2010.08.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Revised: 07/27/2010] [Accepted: 08/04/2010] [Indexed: 12/18/2022]
Abstract
Bax is a pro-apoptotic protein allowing apoptosis to occur through the intrinsic, damage-induced pathway, and amplifying that one occurring via the extrinsic, receptor mediated pathway. Bax is present in viable cells and activated by pro-apoptotic stimuli. Activation implies structural changes, consisting of exposure of the N terminus and hydrophobic domains; changes in localization, consisting in migration from cytosol to mitochondria and endoplasmic reticulum membranes; changes in the aggregation status, from monomer to dimer and multimer. Bax has multiple critical domains, namely the N terminus exposed after activation; two hydrophobic stretches exposed for membrane anchorage; two reactive cysteines allowing multimerization; the BH3 domain for interactions with the Bcl-2 family members; alpha helix 1 for t-Bid interaction. Bax has also multiple functions: it releases different mitochondrial factors such as cytochrome c, SMAC/diablo; it regulates mitochondrial fission, the mitochondrial permeability transition pore; it promotes Ca(2+) leakage through ER membrane. Altogether, Bax activation is a complex multi-step phenomenon. Here, we analyze these events as logically separable or alternative steps, attempting to assess their role, timing and reciprocal relation.
Collapse
Affiliation(s)
- Lina Ghibelli
- Dipartimento di Biologia, Universita' di Roma Tor Vergata, Via della Ricerca Scientifica, I-00133 Rome, Italy.
| | | |
Collapse
|
37
|
Cavanaugh A, McKenna J, Stepanchick A, Breitwieser GE. Calcium-sensing receptor biosynthesis includes a cotranslational conformational checkpoint and endoplasmic reticulum retention. J Biol Chem 2010; 285:19854-64. [PMID: 20421307 DOI: 10.1074/jbc.m110.124792] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Metabolic labeling with [(35)S]cysteine was used to characterize early events in CaSR biosynthesis. [(35)S]CaSR is relatively stable (half-life approximately 8 h), but maturation to the final glycosylated form is slow and incomplete. Incorporation of [(35)S]cysteine is linear over 60 min, and the rate of [(35)S]CaSR biosynthesis is significantly increased by the membrane-permeant allosteric agonist NPS R-568, which acts as a cotranslational pharmacochaperone. The [(35)S]CaSR biosynthetic rate also varies as a function of conformational bias induced by loss- or gain-of-function mutations. In contrast, [(35)S]CaSR maturation to the plasma membrane was not significantly altered by exposure to the pharmacochaperone NPS R-568, the allosteric agonist neomycin, or the orthosteric agonist Ca(2+) (0.5 or 5 mm), suggesting that CaSR does not control its own release from the endoplasmic reticulum. A CaSR chimera containing the mGluR1alpha carboxyl terminus matures completely (half-time of approximately 8 h) and without a lag period, as does the truncation mutant CaSRDelta868 (half-time of approximately 16 h). CaSRDelta898 exhibits maturation comparable with full-length CaSR, suggesting that the CaSR carboxyl terminus between residues Thr(868) and Arg(898) limits maturation. Overall, these results suggest that CaSR is subject to cotranslational quality control, which includes a pharmacochaperone-sensitive conformational checkpoint. The CaSR carboxyl terminus is the chief determinant of intracellular retention of a significant fraction of total CaSR. Intracellular CaSR may reflect a rapidly mobilizable "storage form" of CaSR and/or may subserve distinct intracellular signaling roles that are sensitive to signaling-dependent changes in endoplasmic reticulum Ca(2+) and/or glutathione.
Collapse
Affiliation(s)
- Alice Cavanaugh
- Weis Center for Research, Geisinger Clinic, Danville, Pennsylvania 17822, USA
| | | | | | | |
Collapse
|
38
|
Scullin CS, Partridge LD. Contributions of SERCA pump and ryanodine-sensitive stores to presynaptic residual Ca2+. Cell Calcium 2010; 47:326-38. [PMID: 20153896 DOI: 10.1016/j.ceca.2010.01.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Revised: 12/31/2009] [Accepted: 01/20/2010] [Indexed: 11/24/2022]
Abstract
The presynaptic Ca2+ signal, which triggers vesicle release, disperses to a broadly distributed residual [Ca2+] ([Ca2+](res)) that plays an important role in synaptic plasticity. We have previously reported a slowing in the decay timecourse of [Ca2+](res) during the second of paired pulses. In this study, we investigated the contributions of organelle and plasma membrane Ca2+ flux pathways to the reduction of effectiveness of [Ca2+](res) clearance during short-term plasticity in Schaffer collateral terminals in the CA1 field of the hippocampus. We show that the slowed decay timecourse is mainly the result of a transport-dependent Ca2+ clearance process; that presynaptic caffeine-sensitive Ca2+ stores are not functionally loaded in the unstimulated terminal, but that these stores can effectively take up Ca2+ even during high frequency trains of stimuli; and that a rate limiting step of sarco-endoplasmic reticulum Ca2+-ATPase (SERCA) kinetics following the first pulse is responsible for a large portion of the observed slowing of [Ca2+](res) clearance during the second pulse. We were able to accurately fit our [Ca2+](res) data with a kinetic model based on these observations and this model predicted a reduction in availability of unbound SERCA during paired pulses, but no saturation of Ca2+ buffer in the endoplasmic reticulum.
Collapse
Affiliation(s)
- Chessa S Scullin
- Department of Neurosciences, University of New Mexico, School of Medicine, Albuquerque, NM 87131, USA
| | | |
Collapse
|
39
|
Olson ML, Chalmers S, McCarron JG. Mitochondrial Ca2+ uptake increases Ca2+ release from inositol 1,4,5-trisphosphate receptor clusters in smooth muscle cells. J Biol Chem 2009; 285:2040-50. [PMID: 19889626 DOI: 10.1074/jbc.m109.027094] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Smooth muscle activities are regulated by inositol 1,4,5-trisphosphate (InsP(3))-mediated increases in cytosolic Ca2+ concentration ([Ca2+](c)). Local Ca2+ release from an InsP(3) receptor (InsP(3)R) cluster present on the sarcoplasmic reticulum is termed a Ca2+ puff. Ca2+ released via InsP(3)R may diffuse to adjacent clusters to trigger further release and generate a cell-wide (global) Ca2+ rise. In smooth muscle, mitochondrial Ca2+ uptake maintains global InsP(3)-mediated Ca2+ release by preventing a negative feedback effect of high [Ca2+] on InsP(3)R. Mitochondria may regulate InsP(3)-mediated Ca2+ signals by operating between or within InsP(3)R clusters. In the former mitochondria could regulate only global Ca2+ signals, whereas in the latter both local and global signals would be affected. Here whether mitochondria maintain InsP(3)-mediated Ca2+ release by operating within (local) or between (global) InsP(3)R clusters has been addressed. Ca2+ puffs evoked by localized photolysis of InsP(3) in single voltage-clamped colonic smooth muscle cells had amplitudes of 0.5-4.0 F/F(0), durations of approximately 112 ms at half-maximum amplitude, and were abolished by the InsP(3)R inhibitor 2-aminoethoxydiphenyl borate. The protonophore carbonyl cyanide 3-chloropheylhydrazone and complex I inhibitor rotenone each depolarized DeltaPsi(M) to prevent mitochondrial Ca2+ uptake and attenuated Ca2+ puffs by approximately 66 or approximately 60%, respectively. The mitochondrial uniporter inhibitor, RU360, attenuated Ca2+ puffs by approximately 62%. The "fast" Ca2+ chelator 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid acted like mitochondria to prolong InsP(3)-mediated Ca2+ release suggesting that mitochondrial influence is via their Ca2+ uptake facility. These results indicate Ca2+ uptake occurs quickly enough to influence InsP(3)R communication at the intra-cluster level and that mitochondria regulate both local and global InsP(3)-mediated Ca2+ signals.
Collapse
Affiliation(s)
- Marnie L Olson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, John Arbuthnott Building, Glasgow G40NR, Scotland, United Kingdom
| | | | | |
Collapse
|
40
|
Rizzuto R, Marchi S, Bonora M, Aguiari P, Bononi A, De Stefani D, Giorgi C, Leo S, Rimessi A, Siviero R, Zecchini E, Pinton P. Ca(2+) transfer from the ER to mitochondria: when, how and why. BIOCHIMICA ET BIOPHYSICA ACTA 2009; 1787:1342-51. [PMID: 19341702 PMCID: PMC2730423 DOI: 10.1016/j.bbabio.2009.03.015] [Citation(s) in RCA: 344] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Revised: 03/21/2009] [Accepted: 03/24/2009] [Indexed: 10/25/2022]
Abstract
The heterogenous subcellular distribution of a wide array of channels, pumps and exchangers allows extracellular stimuli to induce increases in cytoplasmic Ca(2+) concentration ([Ca(2+)]c) with highly defined spatial and temporal patterns, that in turn induce specific cellular responses (e.g. contraction, secretion, proliferation or cell death). In this extreme complexity, the role of mitochondria was considered marginal, till the direct measurement with targeted indicators allowed to appreciate that rapid and large increases of the [Ca(2+)] in the mitochondrial matrix ([Ca(2+)]m) invariably follow the cytosolic rises. Given the low affinity of the mitochondrial Ca(2+) transporters, the close proximity to the endoplasmic reticulum (ER) Ca(2+)-releasing channels was shown to be responsible for the prompt responsiveness of mitochondria. In this review, we will summarize the current knowledge of: i) the mitochondrial and ER Ca(2+) channels mediating the ion transfer, ii) the structural and molecular foundations of the signaling contacts between the two organelles, iii) the functional consequences of the [Ca(2+)]m increases, and iv) the effects of oncogene-mediated signals on mitochondrial Ca(2+) homeostasis. Despite the rapid progress carried out in the latest years, a deeper molecular understanding is still needed to unlock the secrets of Ca(2+) signaling machinery.
Collapse
Affiliation(s)
- Rosario Rizzuto
- Dept. Biomedical Sciences, University of Padua, Via Colombo 3, Padua 35121, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
High endoplasmic reticulum activity renders multiple myeloma cells hypersensitive to mitochondrial inhibitors. Cancer Chemother Pharmacol 2009; 66:129-40. [PMID: 19779717 DOI: 10.1007/s00280-009-1143-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2009] [Accepted: 09/12/2009] [Indexed: 10/20/2022]
Abstract
Multiple myeloma (MM) cells continuously secrete large amounts of immunoglobulins that are folded in the endoplasmic reticulum (ER) whose function depend on the Ca(2+) concentration inside its lumen. Recently, it was shown that the ER membrane leaks Ca(2+) that is captured and delivered back by mitochondria in order to prevent its loss. Thus, we hypothesized that the highly active and abundant ER in MM cells results in greater Ca(2+)-regulation by mitochondria which would render them sensitive to mitochondrial inhibitors. Here, we indeed find that Ca(2+) leak is greater in 3 MM, when compared to 2 B-cell leukemia cell lines. Moreover, this greater leak in MM cells is associated with hypersensitivity to various mitochondrial inhibitors, including CCCP. Consistent with our hypothesis, CCCP is more potent in inducing the unfolded protein response marker, CHOP/GADD153 in MM versus B-cell leukemia lines. Additionally, MM cells are found to be significantly more sensitive to clinically used fenofibrate and troglitazone, both of which were recently shown to have inhibitory effects on mitochondrial function. Overall, our results demonstrate that the unusually high ER activity in MM cells may be exploited for therapeutic benefit through the use of mitochondrial inhibitors including troglitazone and fenofibrate.
Collapse
|
42
|
Laskowski AI, Medler KF. Sodium-calcium exchangers contribute to the regulation of cytosolic calcium levels in mouse taste cells. J Physiol 2009; 587:4077-89. [PMID: 19581381 DOI: 10.1113/jphysiol.2009.173567] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Taste cells use multiple signalling mechanisms to generate unique calcium responses to distinct taste stimuli. Some taste stimuli activate G-protein coupled receptors (GPCRs) that cause calcium release from intracellular stores while other stimuli depolarize taste cells to cause calcium influx through voltage-gated calcium channels (VGCCs). We recently demonstrated that a constitutive calcium influx exists in taste cells that is regulated by mitochondrial calcium transport and that the magnitude of this calcium influx correlates with the signalling mechanisms used by the taste cells. In this study, we used calcium imaging to determine that sodium-calcium exchangers (NCXs) also routinely contribute to the regulation of basal cytosolic calcium and that their relative role correlates with the signalling mechanisms used by the taste cells. RT-PCR analysis revealed that multiple NCXs and sodium-calcium-potassium exchangers (NCKXs) are expressed in taste cells. Thus, a dynamic relationship exists between calcium leak channels and calcium regulatory mechanisms in taste cells that functions to keep cytosolic calcium levels in the appropriate range for cell function.
Collapse
Affiliation(s)
- Agnieszka I Laskowski
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | | |
Collapse
|
43
|
Chalmers S, McCarron JG. Inhibition of mitochondrial calcium uptake rather than efflux impedes calcium release by inositol-1,4,5-trisphosphate-sensitive receptors. Cell Calcium 2009; 46:107-13. [PMID: 19577805 DOI: 10.1016/j.ceca.2009.05.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Revised: 05/07/2009] [Accepted: 05/31/2009] [Indexed: 10/20/2022]
Abstract
Mitochondria modulate cellular Ca2+ signals by accumulating the ion via a uniporter and releasing it via Na+- or H+-exchange. In smooth muscle, inhibition of mitochondrial Ca2+ uptake inhibits Ca2+ release from the sarcoplasmic reticulum (SR) via inositol-1,4,5-trisphosphate-sensitive receptors (IP(3)R). At least two mechanisms may explain this effect. First, localised uptake of Ca2+ by mitochondria may prevent negative feedback by cytosolic Ca2+ on IP(3)R activity, or secondly localised provision of Ca2+ by mitochondrial efflux may maintain IP(3)R function or SR Ca2+ content. To distinguish between these possibilities the role of mitochondrial Ca2+ efflux on IP(3)R function was examined. IP(3) was liberated in freshly isolated single colonic smooth muscle cells and mitochondrial Na+-Ca2+ exchanger inhibited with CGP-37157 (10microM). Mitochondria accumulated Ca2+ during IP(3)-evoked [Ca2+](c) rises and released the ion back to the cytosol (within approximately 15s) when mitochondrial Ca2+ efflux was active. When mitochondrial Ca2+ efflux was inhibited by CGP-37157, an extensive and sustained loading of mitochondria with Ca2+ occurred after IP(3)-evoked Ca2+ release. IP(3)-evoked [Ca2+](c) rises were initially unaffected, then only slowly inhibited by CGP-37157. IP(3)R activity was required for inhibition to occur; incubation with CGP-37157 for the same duration without IP(3) release did not inhibit IP(3)R. CGP-37157 directly inhibited voltage-gated Ca2+ channel activity, however SR Ca2+ content was unaltered by the drug. Thus, the gradual decline of IP(3)R function that followed mitochondrial Na+-Ca2+ exchanger inhibition resulted from a gradual overload of mitochondria with Ca2+, leading to a reduced capacity for Ca2+ uptake. Localised uptake of Ca2+ by mitochondria, rather than mitochondrial Ca2+ efflux, appears critical for maintaining IP(3)R activity.
Collapse
Affiliation(s)
- Susan Chalmers
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, John Arbuthnott Building, 27 Taylor Street, Glasgow G4 0NR, UK
| | | |
Collapse
|
44
|
Abstract
Juxtaposition of the endoplasmic reticulum with mitochondria facilitates Ca(2+) fluxes between the organelles. However, the molecular basis of interorganellar tethering remains unknown. A recent report has identified a fundamental role for the dynamin-related mitofusins in the tethering mechanism, thereby ensuring rapid and high fidelity Ca(2+) signalling between the organelles.
Collapse
Affiliation(s)
- Anant Parekh
- Department of Physiology, Anatomy and Genetics, Oxford University, Parks Road, Oxford, UK.
| |
Collapse
|
45
|
Celsi F, Pizzo P, Brini M, Leo S, Fotino C, Pinton P, Rizzuto R. Mitochondria, calcium and cell death: a deadly triad in neurodegeneration. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2009; 1787:335-44. [PMID: 19268425 DOI: 10.1016/j.bbabio.2009.02.021] [Citation(s) in RCA: 223] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Revised: 02/23/2009] [Accepted: 02/24/2009] [Indexed: 12/17/2022]
Abstract
Mitochondrial Ca(2+) accumulation is a tightly controlled process, in turn regulating functions as diverse as aerobic metabolism and induction of cell death. The link between Ca(2+) (dys)regulation, mitochondria and cellular derangement is particularly evident in neurodegenerative disorders, in which genetic models and environmental factors allowed to identify common traits in the pathogenic routes. We will here summarize: i) the current view of mechanisms and functions of mitochondrial Ca(2+) homeostasis, ii) the basic principles of organelle Ca(2+) transport, iii) the role of Ca(2+) in neuronal cell death, and iv) the new information on the pathogenesis of Alzheimer's, Huntington's and Parkinson's diseases, highlighting the role of Ca(2+) and mitochondria.
Collapse
Affiliation(s)
- Fulvio Celsi
- Department of Experimental and Diagnostic Medicine, Interdisciplinary Center for the Study of Inflammation, Italy
| | | | | | | | | | | | | |
Collapse
|
46
|
Laude AJ, Simpson AWM. Compartmentalized signalling: Ca2+ compartments, microdomains and the many facets of Ca2+ signalling. FEBS J 2009; 276:1800-16. [PMID: 19243429 DOI: 10.1111/j.1742-4658.2009.06927.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ca(2+) regulates a multitude of cellular processes and does so by partitioning its actions in space and time. In this review, we discuss how Ca(2+) responses are constructed from small quantal (elementary) events that have the potential to propagate to produce large pan-cellular responses. We review how Ca(2+) is compartmentalized both physically and functionally, and describe how each organelle has its own distinct Ca(2+)-handling properties. We explain how coordination of the movement of Ca(2+) between organelles is used to shape and hone Ca(2+) signals. Finally, we provide a number of specific examples of where compartmentation and localization of Ca(2+) are crucial to cell function.
Collapse
Affiliation(s)
- Alex J Laude
- Department Human Anatomy and Cell Biology, University of Liverpool, Liverpool, UK
| | | |
Collapse
|
47
|
Spät A, Fülöp L, Koncz P, Szanda G. When is high-Ca+ microdomain required for mitochondrial Ca+ uptake? Acta Physiol (Oxf) 2009; 195:139-47. [PMID: 18983456 DOI: 10.1111/j.1748-1716.2008.01928.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ca(2+) release from IP(3)-sensitive stores in the endoplasmic reticulum (ER) induced by Ca(2+)-mobilizing agonists generates high-Ca(2+) microdomains between ER vesicles and neighbouring mitochondria. Here we present a model that describes when such microdomains are required and when submicromolar [Ca(2+)] is sufficient for mitochondrial Ca(2+) uptake. Mitochondrial Ca(2+) uptake rate in angiotensin II-stimulated H295R adrenocortical cells correlates with the proximity between ER vesicles and the mitochondrion, reflecting the uptake promoting effect of high-Ca(2+) peri-mitochondrial microdomains. Silencing or inhibition of p38 mitogen-activated protein kinase (MAPK) or inhibition of the novel isoforms of protein kinase C enhances mitochondrial Ca(2+) uptake and abolishes the positive correlation between Ca(2+) uptake and ER-mitochondrion proximity. Inhibition of protein phosphatases attenuates mitochondrial Ca(2+) uptake and also abolishes its positive correlation with ER-mitochondrion proximity. We postulate that during IP(3)-induced Ca(2+) release, Ca(2+) uptake is confined to ER-close mitochondria, because of the simultaneous activation of the protein kinases. Attenuation of Ca(2+) uptake prevents Ca(2+) overload of mitochondria and thus protects the cell against apoptosis. On the other hand, all the mitochondria accumulate Ca(2+) at a non-inhibited rate during physiological Ca(2+) influx through the plasma membrane. Membrane potential is higher in ER-distant mitochondria, providing a bigger driving force for Ca(2+) uptake. Our model explains why comparable mitochondrial Ca(2+) signals are formed in response to K(+) and angiotensin II (equipotent in respect to global cytosolic Ca(2+) signals), although only the latter generates high-Ca(2+) microdomains.
Collapse
Affiliation(s)
- A Spät
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary.
| | | | | | | |
Collapse
|
48
|
Computational study of non-homogeneous distribution of Ca(2+) handling systems in cerebellar granule cells. J Theor Biol 2008; 257:228-44. [PMID: 19121636 DOI: 10.1016/j.jtbi.2008.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Revised: 10/01/2008] [Accepted: 12/01/2008] [Indexed: 11/21/2022]
Abstract
The spatiotemporal distribution of cytosolic free calcium concentration ([Ca(2+)](i)) in cerebellar granule cells (GrCs) is thought to be critical in defining the occurrence and direction of long-term changes in synaptic strength at cerebellar mossy fiber-GrC synapses. Despite this, the mechanisms responsible for shaping Ca(2+) transients in GrCs are not well understood. To investigate the interplay between Ca(2+) entry, extrusion, buffering and dendritic morphology in shaping Ca(2+) elevations in GrCs, we developed a model of Ca(2+) regulation in these cells and examined the requirements for reproducing fluorescence responses to depolarization and synaptic stimulation previously described in the literature. Two conclusions can be drawn from our simulation results. First, a significant progressive decrease in the amplitudes of depolarization-evoked fluorescence transients from the dendritic endings (digits) toward the soma of GrCs, can be reproduced in the model only if the density of Ca(2+) channels is considerably higher or the concentration of endogenous buffers is much lower in the digits than in the parent dendrites. In contrast, heterogeneities in the distribution of Ca(2+) pumps or in cytosolic fractional volume cannot account for the formation of [Ca(2+)](i) gradients in GrCs. Second, much lower amplitudes of fluorescence transients induced by depolarization and synaptic stimulation than expected from typical measurements of Ca(2+) and NMDA receptor-mediated currents can be reconciled with a pronounced slowing of the decay of fluorescence responses in the digits of GrCs after introducing a high-affinity Ca(2+) indicator if a high-capacity immobile Ca(2+) buffer (presumably plasma membrane-associated) is suggested to be present in the soma and apical part of digits. Mitochondria also are likely to modulate synaptically evoked Ca(2+) responses in GrCs. The alternative hypotheses are thoroughly discussed and research avenues for their testing in future experiments are proposed.
Collapse
|
49
|
Hacker K, Medler KF. Mitochondrial calcium buffering contributes to the maintenance of Basal calcium levels in mouse taste cells. J Neurophysiol 2008; 100:2177-91. [PMID: 18684902 DOI: 10.1152/jn.90534.2008] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Taste stimuli are detected by taste receptor cells present in the oral cavity using diverse signaling pathways. Some taste stimuli are detected by G protein-coupled receptors (GPCRs) that cause calcium release from intracellular stores, whereas other stimuli depolarize taste cells to cause calcium influx through voltage-gated calcium channels (VGCCs). Although taste cells use two distinct mechanisms to transmit taste signals, increases in cytosolic calcium are critical for normal responses in both pathways. This creates a need to tightly control intracellular calcium levels in all transducing taste cells. To date, however, the mechanisms used by taste cells to regulate cytosolic calcium levels have not been identified. Studies in other cell types have shown that mitochondria can be important calcium buffers, even during small changes in calcium loads. In this study, we used calcium imaging to characterize the role of mitochondria in buffering calcium levels in taste cells. We discovered that mitochondria make important contributions to the maintenance of resting calcium levels in taste cells by routinely buffering a constitutive calcium influx across the plasma membrane. This is unusual because in other cell types, mitochondrial calcium buffering primarily affects large evoked calcium responses. We also found that the amount of calcium that is buffered by mitochondria varies with the signaling pathways used by the taste cells. A transient receptor potential (TRP) channel, likely TRPV1 or a taste variant of TRPV1, contributes to the constitutive calcium influx.
Collapse
Affiliation(s)
- Kyle Hacker
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | | |
Collapse
|
50
|
Pacher P, Sharma K, Csordás G, Zhu Y, Hajnóczky G. Uncoupling of ER-mitochondrial calcium communication by transforming growth factor-beta. Am J Physiol Renal Physiol 2008; 295:F1303-12. [PMID: 18653477 DOI: 10.1152/ajprenal.90343.2008] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Transforming growth factor-beta (TGF-beta) has been implicated as a key factor in mediating many cellular processes germane to disease pathogenesis, including diabetic vascular complications. TGF-beta alters cytosolic [Ca2+] ([Ca2+]c) signals, which in some cases may result from the downregulation of the IP3 receptor Ca2+ channels (IP3R). Ca2+ released by IP3Rs is effectively transferred from endoplasmic reticulum (ER) to the mitochondria to stimulate ATP production and to allow feedback control of the Ca2+ mobilization. To assess the effect of TGF-beta on the ER-mitochondrial Ca2+ transfer, we first studied the [Ca2+]c and mitochondrial matrix Ca2+ ([Ca2+]m) signals in single preglomerular afferent arteriolar smooth muscle cells (PGASMC). TGF-beta pretreatment (24 h) decreased both the [Ca2+]c and [Ca2+]m responses evoked by angiotensin II or endothelin. Strikingly, the [Ca2+]m signal was more depressed than the [Ca2+]c signal and was delayed. In permeabilized cells, TGF-beta pretreatment attenuated the rate but not the magnitude of the IP(3)-induced [Ca2+]c rise, yet caused massive depression of the [Ca2+]m responses. ER Ca2+ storage and mitochondrial uptake of added Ca2+ were not affected by TGF-beta. Also, TGF-beta had no effect on mitochondrial distribution and on the ER-mitochondrial contacts assessed by two-photon NAD(P)H imaging and electron microscopy. Downregulation of both IP3R1 and IP3R3 was found in TGF-beta-treated PGASMC. Thus, TGF-beta causes uncoupling of mitochondria from the ER Ca2+ release. The sole source of this would be suppression of the IP3R-mediated Ca2+ efflux, indicating that the ER-mitochondrial Ca2+ transfer depends on the maximal rate of Ca2+ release. The impaired ER-mitochondrial coupling may contribute to the vascular pathophysiology associated with TGF-beta production.
Collapse
Affiliation(s)
- Pál Pacher
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | |
Collapse
|