1
|
Calle-Ciborro B, Santos FJ, Espin-Jaime T, Gomez-Martin A, Camello PJ, Camello-Almaraz C. Pharmacological inhibition reveals participation of the endocytic compartment in positive feedback IL-6 secretion in human skeletal myotubes. Eur J Pharmacol 2024; 984:177055. [PMID: 39395584 DOI: 10.1016/j.ejphar.2024.177055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 10/14/2024]
Abstract
IL-6 is an important cytokine involved in metabolic, immunological, and cell-fate responses. It is released upon stimulation by skeletal muscle cells through partially characterized mechanisms. In some cell types, IL-6 has been reported to activate a positive feedback loop involving endocytic vesicles, but evidence is mostly based on transcription and signal transduction mechanisms and is very scarce in muscle cells. Our aim was to directly demonstrate the presence of positive feedback in the ATP-induced release of IL-6 into the supernatant of human skeletal muscle cultures. The total release (production) of IL-6 was reduced for higher volumes of supernatant, when the secreted IL-6 molecules are more diluted, and enhanced when the supernatant volume was lower. In addition, secretion was impaired both by tocilizumab, a blocker of human IL-6 receptors, and by the soluble form of the receptor. The secretion in response to ATP was also inhibited by treatment with the endocytosis inhibitor dynasore, and by disruption of the acidic gradient of the endocytic compartment using different methods (chloroquine, NH4Cl or monensin). IL-6 secretion was also impaired by NED-19, a specific inhibitor of the two pore channels receptor mediating Ca2+ release from the endolysosomal compartment. IL-6 and ATP increased IL-6 mRNA levels, an effect blocked by tocilizumab. Altogether, our results demonstrate that ATP-secreted IL-6 activates a positive loop based on IL-6 receptors, endocytosis, two pore channels and IL-6 transcription. Given the importance of muscle IL-6 as a systemic regulator and as an inflammatory mediator, our study can help to understand muscle pathophysiology.
Collapse
Affiliation(s)
- Blanca Calle-Ciborro
- Department of Physiology, Instituto de Biomarcadores Patológicos Moleculares y Metabólicos, Universidad de Extremadura, Cáceres, Spain
| | | | | | - Ana Gomez-Martin
- Dept. of Nursing, Facultad de Enfermeria y Terapia Ocupacional, Cáceres, Spain
| | - Pedro J Camello
- Department of Physiology, Instituto de Biomarcadores Patológicos Moleculares y Metabólicos, Universidad de Extremadura, Cáceres, Spain.
| | - Cristina Camello-Almaraz
- Department of Physiology, Instituto de Biomarcadores Patológicos Moleculares y Metabólicos, Universidad de Extremadura, Cáceres, Spain
| |
Collapse
|
2
|
Patel S, Yule DI. Stretching the role of TMEM63a to gatekeeping Ca 2+ release in pancreatic acinar cells. Cell Calcium 2024; 121:102890. [PMID: 38759307 DOI: 10.1016/j.ceca.2024.102890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 05/19/2024]
Affiliation(s)
- Sandip Patel
- Department of Cell and Developmental Biology, University College London, Gower Street, WC1E 6BT, London, UK
| | - David I Yule
- Department of Pharmacology and Physiology, University of Rochester, 601 Elmwood Avenue, Rochester, NY, 14642, USA.
| |
Collapse
|
3
|
Tsvilovskyy V, Ottenheijm R, Kriebs U, Schütz A, Diakopoulos KN, Jha A, Bildl W, Wirth A, Böck J, Jaślan D, Ferro I, Taberner FJ, Kalinina O, Hildebrand S, Wissenbach U, Weissgerber P, Vogt D, Eberhagen C, Mannebach S, Berlin M, Kuryshev V, Schumacher D, Philippaert K, Camacho-Londoño JE, Mathar I, Dieterich C, Klugbauer N, Biel M, Wahl-Schott C, Lipp P, Flockerzi V, Zischka H, Algül H, Lechner SG, Lesina M, Grimm C, Fakler B, Schulte U, Muallem S, Freichel M. OCaR1 endows exocytic vesicles with autoregulatory competence by preventing uncontrolled Ca2+ release, exocytosis, and pancreatic tissue damage. J Clin Invest 2024; 134:e169428. [PMID: 38557489 PMCID: PMC10977991 DOI: 10.1172/jci169428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 02/13/2024] [Indexed: 04/04/2024] Open
Abstract
Regulated exocytosis is initiated by increased Ca2+ concentrations in close spatial proximity to secretory granules, which is effectively prevented when the cell is at rest. Here we showed that exocytosis of zymogen granules in acinar cells was driven by Ca2+ directly released from acidic Ca2+ stores including secretory granules through NAADP-activated two-pore channels (TPCs). We identified OCaR1 (encoded by Tmem63a) as an organellar Ca2+ regulator protein integral to the membrane of secretory granules that controlled Ca2+ release via inhibition of TPC1 and TPC2 currents. Deletion of OCaR1 led to extensive Ca2+ release from NAADP-responsive granules under basal conditions as well as upon stimulation of GPCR receptors. Moreover, OCaR1 deletion exacerbated the disease phenotype in murine models of severe and chronic pancreatitis. Our findings showed OCaR1 as a gatekeeper of Ca2+ release that endows NAADP-sensitive secretory granules with an autoregulatory mechanism preventing uncontrolled exocytosis and pancreatic tissue damage.
Collapse
Affiliation(s)
- Volodymyr Tsvilovskyy
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Roger Ottenheijm
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Ulrich Kriebs
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Aline Schütz
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Kalliope Nina Diakopoulos
- Comprehensive Cancer Center München, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Archana Jha
- Epithelial Signaling and Transport Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, USA
| | - Wolfgang Bildl
- Institute for Physiology, University of Freiburg, Freiburg, Germany
| | - Angela Wirth
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Julia Böck
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Dawid Jaślan
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Irene Ferro
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Francisco J. Taberner
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández–Consejo Superior de Investigaciones Científicas, Sant Joan d’Alacant, Spain
| | - Olga Kalinina
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany
| | - Staffan Hildebrand
- Institut für Pharmakologie und Toxikologie, Universität Bonn, Bonn, Germany
| | - Ulrich Wissenbach
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Homburg, Germany
| | - Petra Weissgerber
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Homburg, Germany
| | - Dominik Vogt
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Carola Eberhagen
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Stefanie Mannebach
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Homburg, Germany
| | - Michael Berlin
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Vladimir Kuryshev
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Dagmar Schumacher
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Koenraad Philippaert
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | | | - Ilka Mathar
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Christoph Dieterich
- University Hospital Heidelberg, Department of Medicine III: Cardiology, Angiology and Pneumology, Heidelberg, Germany
| | - Norbert Klugbauer
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Fakultät für Medizin, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Martin Biel
- Center for Integrated Protein Science Munich (CIPS-M) and Center for Drug Research, Department of Pharmacy, Ludwig-Maximilians-Universität München, and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Christian Wahl-Schott
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Medical Faculty, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Peter Lipp
- Institute for Molecular Cell Biology, Center for Molecular Signaling (PZMS), Universität des Saarlandes, Homburg, Germany
| | - Veit Flockerzi
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Homburg, Germany
| | - Hans Zischka
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Toxicology and Environmental Hygiene, Technical University Munich, School of Medicine, Munich, Germany
| | - Hana Algül
- Comprehensive Cancer Center München, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Stefan G. Lechner
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Marina Lesina
- Comprehensive Cancer Center München, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Christian Grimm
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, Munich, Germany
- Immunology, Infection and Pandemic Research (IIP), Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Munich, Germany
| | - Bernd Fakler
- Institute for Physiology, University of Freiburg, Freiburg, Germany
| | - Uwe Schulte
- Institute for Physiology, University of Freiburg, Freiburg, Germany
| | - Shmuel Muallem
- Epithelial Signaling and Transport Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, USA
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
4
|
Chakraborty A, Dissanayake R, Wall KA. Nicotinic Acid Adenine Dinucleotide Phosphate (NAADP)-Mediated Calcium Signaling Is Active in Memory CD4 + T Cells. Molecules 2024; 29:907. [PMID: 38398657 PMCID: PMC10892544 DOI: 10.3390/molecules29040907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Nicotinic acid adenine dinucleotide phosphate (NAADP), identified as one of the most potent calcium-mobilizing second messengers, has been studied in different eukaryotic cell types, including lymphocytes. Although aspects of NAADP-mediated calcium release in lymphocytes are still under debate, the organelles pertaining to NAADP-mediated calcium release are often characterized as acidic and related to lysosomes. Although NAADP-mediated calcium release in different subsets of T cells, including naïve, effector and natural regulatory T cells, has been studied, it has not been widely studied in memory CD4+ T cells, which show a different calcium flux profile. Using a pharmacological approach, the effect of Ned-19, an NAADP pathway antagonist, on the involvement of NAADP in TCR activation in murine memory CD4+ T cells and their downstream effector functions, such as proliferation and cytokine production, was studied. According to this study, Ned-19 inhibited TCR-mediated calcium flux and its downstream effector functions in primary memory CD4+ T cells. The study also revealed that both extracellular and intracellular calcium stores, including endoplasmic reticulum and lysosome-like acidic calcium stores, contribute to the TCR-mediated calcium flux in memory CD4+ T cells. NAADP-AM, a cell permeable analogue of NAADP, was shown to release calcium in memory CD4+ T cells and calcium flux was inhibited by Ned-19.
Collapse
Affiliation(s)
| | | | - Katherine A. Wall
- Department of Medicinal and Biological Chemistry, University of Toledo, Toledo, OH 43614, USA; (A.C.); (R.D.)
| |
Collapse
|
5
|
Sun S, Zhao G, Jia M, Jiang Q, Li S, Wang H, Li W, Wang Y, Bian X, Zhao YG, Huang X, Yang G, Cai H, Pastor-Pareja JC, Ge L, Zhang C, Hu J. Stay in touch with the endoplasmic reticulum. SCIENCE CHINA. LIFE SCIENCES 2024; 67:230-257. [PMID: 38212460 DOI: 10.1007/s11427-023-2443-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 08/28/2023] [Indexed: 01/13/2024]
Abstract
The endoplasmic reticulum (ER), which is composed of a continuous network of tubules and sheets, forms the most widely distributed membrane system in eukaryotic cells. As a result, it engages a variety of organelles by establishing membrane contact sites (MCSs). These contacts regulate organelle positioning and remodeling, including fusion and fission, facilitate precise lipid exchange, and couple vital signaling events. Here, we systematically review recent advances and converging themes on ER-involved organellar contact. The molecular basis, cellular influence, and potential physiological functions for ER/nuclear envelope contacts with mitochondria, Golgi, endosomes, lysosomes, lipid droplets, autophagosomes, and plasma membrane are summarized.
Collapse
Affiliation(s)
- Sha Sun
- National Laboratory of Biomacromolecules, Institute of Biophysics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Gan Zhao
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Mingkang Jia
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Qing Jiang
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Shulin Li
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Haibin Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wenjing Li
- Laboratory of Computational Biology & Machine Intelligence, School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yunyun Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xin Bian
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Yan G Zhao
- Brain Research Center, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Xun Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Ge Yang
- Laboratory of Computational Biology & Machine Intelligence, School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Huaqing Cai
- National Laboratory of Biomacromolecules, Institute of Biophysics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jose C Pastor-Pareja
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Institute of Neurosciences, Consejo Superior de Investigaciones Cientfflcas-Universidad Miguel Hernandez, San Juan de Alicante, 03550, Spain.
| | - Liang Ge
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| | - Chuanmao Zhang
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, China.
| | - Junjie Hu
- National Laboratory of Biomacromolecules, Institute of Biophysics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
6
|
Yuan Y, Arige V, Saito R, Mu Q, Brailoiu GC, Pereira GJS, Bolsover SR, Keller M, Bracher F, Grimm C, Brailoiu E, Marchant JS, Yule DI, Patel S. Two-pore channel-2 and inositol trisphosphate receptors coordinate Ca 2+ signals between lysosomes and the endoplasmic reticulum. Cell Rep 2024; 43:113628. [PMID: 38160394 PMCID: PMC10931537 DOI: 10.1016/j.celrep.2023.113628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/13/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024] Open
Abstract
Lysosomes and the endoplasmic reticulum (ER) are Ca2+ stores mobilized by the second messengers NAADP and IP3, respectively. Here, we establish Ca2+ signals between the two sources as fundamental building blocks that couple local release to global changes in Ca2+. Cell-wide Ca2+ signals evoked by activation of endogenous NAADP-sensitive channels on lysosomes comprise both local and global components and exhibit a major dependence on ER Ca2+ despite their lysosomal origin. Knockout of ER IP3 receptor channels delays these signals, whereas expression of lysosomal TPC2 channels accelerates them. High-resolution Ca2+ imaging reveals elementary events upon TPC2 opening and signals coupled to IP3 receptors. Biasing TPC2 activation to a Ca2+-permeable state sensitizes local Ca2+ signals to IP3. This increases the potency of a physiological agonist to evoke global Ca2+ signals and activate a downstream target. Our data provide a conceptual framework to understand how Ca2+ release from physically separated stores is coordinated.
Collapse
Affiliation(s)
- Yu Yuan
- Department of Cell and Developmental Biology, University College London, Gower Street, WC1E 6BT London, UK
| | - Vikas Arige
- Department of Pharmacology and Physiology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Ryo Saito
- Department of Cell and Developmental Biology, University College London, Gower Street, WC1E 6BT London, UK; Department of Dermatology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Qianru Mu
- Department of Cell and Developmental Biology, University College London, Gower Street, WC1E 6BT London, UK
| | - Gabriela C Brailoiu
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, 901 Walnut Street, Philadelphia, PA 19107, USA
| | - Gustavo J S Pereira
- Department of Cell and Developmental Biology, University College London, Gower Street, WC1E 6BT London, UK; Department of Pharmacology, Federal University of São Paulo (UNIFESP), São Paulo 04044-020, Brazil
| | - Stephen R Bolsover
- Department of Cell and Developmental Biology, University College London, Gower Street, WC1E 6BT London, UK
| | - Marco Keller
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilian University, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Franz Bracher
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilian University, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Christian Grimm
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilian University, Nussbaumstrasse 26, 80336 Munich, Germany; Immunology, Infection and Pandemic Research IIP, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt, Germany
| | - Eugen Brailoiu
- Department of Neural Sciences and Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Jonathan S Marchant
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - David I Yule
- Department of Pharmacology and Physiology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA.
| | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, Gower Street, WC1E 6BT London, UK.
| |
Collapse
|
7
|
Mushtaq M, Mahmood M, Jabbar U, Kim UH. Essential role of CD38 in platelet aggregation through the PKC-mediated internalization and activation. BIOIMPACTS : BI 2023; 14:27780. [PMID: 38505670 PMCID: PMC10945299 DOI: 10.34172/bi.2023.27780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 07/20/2023] [Accepted: 07/26/2023] [Indexed: 03/21/2024]
Abstract
Introduction CD38 is a multifunctional enzyme with a potent Ca2+ mobilizing effect, cyclic ADP-ribose (cADPR), and nicotinic acid adenine dinucleotide phosphate (NAADP). Here, we aimed to demonstrate the role of CD38 in platelets via protein kinase C (PKC)-mediated internalization and activation. Methods Mouse platelets were used in this study. Thrombin, an agonist of platelet function, provoked a prompt and long-lasting increase in intracellular Ca2+ concentration ([Ca2+]i), resulting from an interplay of multifold Ca2+ mobilizing messengers.The signaling pathway was delineated using different inhibitors and techniques such as platelet aggregation assay, intracellular calcium measurements, immunoprecipitation, immunoblotting, and flow cytometry. Results We observed a sequential formation of cADPR and NAADP through CD38 activation by PKC of non-muscle myosin heavy chain IIA (MHCIIA), resulting in phospholipase C (PLC) activation in the thrombin-stimulated platelets. These findings reveal that PKC is fundamental in activating CD38 and elicits a physiological response in the murine platelets. Conclusion PKC is involved in many signaling pathways. Specifically, PKC is involved in the internalization of CD38 via MHCIIA in CD38+/+ wild-type (WT) and CD38-/- knockout mice (KO). CD38 generates calcium-mobilizing agents that act on specific receptors of the calcium stores. Calcium triggered platelet aggregation while serving as a secondary messenger.
Collapse
Affiliation(s)
- Mazhar Mushtaq
- Basic Medical Sciences, Sulaiman Al Rajhi University, Al-Qaseem, Kingdom of Saudi Arabia
| | - Maira Mahmood
- Department of Biochemistry, FMH College of Medicine and Dentistry, Lahore, Pakistan
| | - Uzma Jabbar
- Department of Biochemistry, Gujranwala Medical College, Gujranwala, Pakistan
| | - Uh-Hyun Kim
- Department of Biochemistry, Chonbuk National University, Chonbuk, South Korea
| |
Collapse
|
8
|
Terrar DA. Timing mechanisms to control heart rhythm and initiate arrhythmias: roles for intracellular organelles, signalling pathways and subsarcolemmal Ca 2. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220170. [PMID: 37122228 PMCID: PMC10150226 DOI: 10.1098/rstb.2022.0170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Rhythms of electrical activity in all regions of the heart can be influenced by a variety of intracellular membrane bound organelles. This is true both for normal pacemaker activity and for abnormal rhythms including those caused by early and delayed afterdepolarizations under pathological conditions. The influence of the sarcoplasmic reticulum (SR) on cardiac electrical activity is widely recognized, but other intracellular organelles including lysosomes and mitochondria also contribute. Intracellular organelles can provide a timing mechanism (such as an SR clock driven by cyclic uptake and release of Ca2+, with an important influence of intraluminal Ca2+), and/or can act as a Ca2+ store involved in signalling mechanisms. Ca2+ plays many diverse roles including carrying electric current, driving electrogenic sodium-calcium exchange (NCX) particularly when Ca2+ is extruded across the surface membrane causing depolarization, and activation of enzymes which target organelles and surface membrane proteins. Heart function is also influenced by Ca2+ mobilizing agents (cADP-ribose, nicotinic acid adenine dinucleotide phosphate and inositol trisphosphate) acting on intracellular organelles. Lysosomal Ca2+ release exerts its effects via calcium/calmodulin-dependent protein kinase II to promote SR Ca2+ uptake, and contributes to arrhythmias resulting from excessive beta-adrenoceptor stimulation. A separate arrhythmogenic mechanism involves lysosomes, mitochondria and SR. Interacting intracellular organelles, therefore, have profound effects on heart rhythms and NCX plays a central role. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.
Collapse
Affiliation(s)
- Derek A Terrar
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| |
Collapse
|
9
|
Merve D, Irfan A, Tugba DKN, Inci SE. Determination of the roles of cADPR and NAADP as intracellular calcium mobilizing messengers in S1P-induced contractions in rat bladders having IC/PBS. Life Sci 2023; 322:121651. [PMID: 37023954 DOI: 10.1016/j.lfs.2023.121651] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 04/08/2023]
Abstract
AIMS Interstitial cystitis/painful bladder syndrome (IC/PBS) is characterized by lower abdominal pain and increased frequency and urgency of urine. Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid that plays role in calcium homeostasis in smooth muscle. The intracellular calcium mobilizing secondary messengers are also involved in smooth muscle contraction. The role of intracellular calcium storing depots in S1P-induced contraction was investigated in permeabilized detrusor smooth muscle having cystitis. MAIN METHODS IC/PBS was induced by cyclophosphamide injection. The detrusor smooth muscle strips isolated from rats were permeabilized with β-escin. KEY FINDINGS S1P-induced contraction was increased in cystitis. S1P-induced enhanced contraction was inhibited by cyclopiazonic acid, ryanodine and heparin showing involvement of sarcoplasmic reticulum (SR) calcium stores. Inhibition of S1P-induced contraction by bafilomycin and NAADP suggested the participation of lysosome-related organelles. SIGNIFICANCE IC/PBS triggers S1P-induced increase in intracellular calcium from SR and lysosome-related organelles in permeabilized detrusor smooth muscle.
Collapse
Affiliation(s)
- Denizalti Merve
- Faculty of Pharmacy, Department of Pharmacology, Hacettepe University, Ankara, Turkey
| | - Anjum Irfan
- Faculty of Pharmacy, Department of Pharmacology, Hacettepe University, Ankara, Turkey
| | | | - Sahin-Erdemli Inci
- Faculty of Pharmacy, Department of Pharmacology, Hacettepe University, Ankara, Turkey
| |
Collapse
|
10
|
Takano T, Yule DI. Ca 2+ signals in pancreatic acinar cells in response to physiological stimulation in vivo. J Physiol 2023. [PMID: 36965132 DOI: 10.1113/jp284469] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/08/2023] [Indexed: 03/27/2023] Open
Abstract
The exocrine pancreas secretes fluid and digestive enzymes in response to parasympathetic release of acetylcholine (ACh) via the vagus nerve and the gut hormone cholecystokinin (CCK). Both secretion of fluid and exocytosis of secretory granules containing enzymes and zymogens are dependent on an increase in the cytosolic [Ca2+ ] in acinar cells. It is thought that the specific spatiotemporal characteristics of the Ca2+ signals are fundamental for appropriate secretion and that these properties are disrupted in disease states in the pancreas. While extensive research has been performed to characterize Ca2+ signalling in acinar cells, this has exclusively been achieved in ex vivo preparations of exocrine cells, where it is difficult to mimic physiological conditions. Here we have developed a method to optically observe pancreatic acinar Ca2+ signals in vivo using a genetically expressed Ca2+ indicator and imaged with multi-photon microscopy in live animals. In vivo, acinar cells exhibited baseline activity in fasted animals, which was dependent on CCK1 receptors (CCK1Rs). Both stimulation of intrinsic nervous input and administration of systemic CCK induced oscillatory activity in a proportion of the cells, but the maximum frequencies were vastly different. Upon feeding, oscillatory activity was also observed, which was dependent on CCK1Rs. No evidence of a vago-vagal reflex mediating the effects of CCK was observed. Our in vivo method revealed the spatial and temporal profile of physiologically evoked Ca2+ signals, which will provide new insights into future studies of the mechanisms underlying exocrine physiology and that are disrupted in pathological conditions. KEY POINTS: In the exocrine pancreas, the spatiotemporal properties of Ca2+ signals are fundamentally important for the appropriate stimulation of secretion by the neurotransmitter acetylcholine and gut hormone cholecystokinin. These characteristics were previously defined in ex vivo studies. Here we report the spatiotemporal characteristics of Ca2+ signals in vivo in response to physiological stimulation in a mouse engineered to express a Ca2+ indicator in acinar cells. Specific Ca2+ 'signatures' probably important for stimulating secretion are evoked in vivo in fasted animals, by feeding, neural stimulation and cholecystokinin administration. The Ca2+ signals are probably the result of the direct action of ACh and CCK on acinar cells and not indirectly through a vago-vagal reflex.
Collapse
Affiliation(s)
- Takahiro Takano
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA
| | - David I Yule
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA
| |
Collapse
|
11
|
Wahl-Schott C, Freichel M, Hennis K, Philippaert K, Ottenheijm R, Tsvilovskyy V, Varbanov H. Characterization of Endo-Lysosomal Cation Channels Using Calcium Imaging. Handb Exp Pharmacol 2023; 278:277-304. [PMID: 36894791 DOI: 10.1007/164_2023_637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Endo-lysosomes are membrane-bound acidic organelles that are involved in endocytosis, recycling, and degradation of extracellular and intracellular material. The membranes of endo-lysosomes express several Ca2+-permeable cation ion channels, including two-pore channels (TPC1-3) and transient receptor potential mucolipin channels (TRPML1-3). In this chapter, we will describe four different state-of-the-art Ca2+ imaging approaches, which are well-suited to investigate the function of endo-lysosomal cation channels. These techniques include (1) global cytosolic Ca2+ measurements, (2) peri-endo-lysosomal Ca2+ imaging using genetically encoded Ca2+ sensors that are directed to the cytosolic endo-lysosomal membrane surface, (3) Ca2+ imaging of endo-lysosomal cation channels, which are engineered in order to redirect them to the plasma membrane in combination with approaches 1 and 2, and (4) Ca2+ imaging by directing Ca2+ indicators to the endo-lysosomal lumen. Moreover, we will review useful small molecules, which can be used as valuable tools for endo-lysosomal Ca2+ imaging. Rather than providing complete protocols, we will discuss specific methodological issues related to endo-lysosomal Ca2+ imaging.
Collapse
Affiliation(s)
- Christian Wahl-Schott
- Institut für Kardiovaskuläre Physiologie und Pathophysiologie, Lehrstuhl für Vegetative Physiologie, Biomedizinisches Zentrum, Ludwig-Maximilians-Universität München, München, Germany.
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany. .,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany.
| | - Konstantin Hennis
- Institut für Kardiovaskuläre Physiologie und Pathophysiologie, Lehrstuhl für Vegetative Physiologie, Biomedizinisches Zentrum, Ludwig-Maximilians-Universität München, München, Germany
| | - Koenraad Philippaert
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| | - Roger Ottenheijm
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| | - Volodymyr Tsvilovskyy
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| | - Hristo Varbanov
- Institut für Neurophysiologie, Medizinische Hochschule Hannover(MHH), Hannover, Germany
| |
Collapse
|
12
|
Wang Q, Zhu MX. NAADP-Dependent TPC Current. Handb Exp Pharmacol 2023; 278:35-56. [PMID: 35902437 DOI: 10.1007/164_2022_606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Two-pore channels, TPC1 and TPC2, are Ca2+- and Na+-permeable cation channels expressed on the membranes of endosomes and lysosomes in nearly all mammalian cells. These channels have been implicated in Ca2+ signaling initiated from the endolysosomes, vesicular trafficking, cellular metabolism, macropinocytosis, and viral infection. Although TPCs have been shown to mediate Ca2+ release from acidic organelles in response to NAADP (nicotinic acid adenine dinucleotide phosphate), the most potent Ca2+ mobilizing messenger, questions remain whether NAADP is a direct ligand of these channels. In whole-endolysosomal patch clamp recordings, it has been difficult to detect NAADP-evoked currents in vacuoles that expressed TPC1 or TPC2, while PI(3,5)P2 (phosphatidylinositol 3,5-bisphosphate) activated a highly Na+-selective current under the same experimental configuration. In this chapter, we summarize recent progress in this area and provide our observations on NAADP-elicited TPC2 currents from enlarged endolysosomes as well as their possible relationships with the currents evoked by PI(3,5)P2. We propose that TPCs are channels dually regulated by PI(3,5)P2 and NAADP in an interdependent manner and the two endogenous ligands may have both distinguished and cooperative roles.
Collapse
Affiliation(s)
- Qiaochu Wang
- Beijing Children's Hospital, Capital Medical University, Beijing, China
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
13
|
Abstract
The discovery of NAADP-evoked Ca2+ release in sea urchin eggs and then as a ubiquitous Ca2+ mobilizing messenger has introduced several novel paradigms to our understanding of Ca2+ signalling, not least in providing a link between cell stimulation and Ca2+ release from lysosomes and other acidic Ca2+ storage organelles. In addition, the hallmark concentration-response relationship of NAADP-mediated Ca2+ release, shaped by striking activation/desensitization mechanisms, influences its actions as an intracellular messenger. There has been recent progress in our understanding of the molecular mechanisms underlying NAADP-evoked Ca2+ release, such as the identification of the endo-lysosomal two-pore channel family of cation channels (TPCs) as their principal target and the identity of NAADP-binding proteins that complex with them. The NAADP/TPC signalling axis has gained recent prominence in pathophysiology for their roles in such disease processes as neurodegeneration, tumorigenesis and cellular viral entry.
Collapse
Affiliation(s)
- Antony Galione
- Department of Pharmacology, University of Oxford, Oxford, UK.
| | - Lianne C Davis
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Lora L Martucci
- Department of Pharmacology, University of Oxford, Oxford, UK
| | | |
Collapse
|
14
|
Guse AH. NAADP-Evoked Ca 2+ Signaling: The DUOX2-HN1L/JPT2-Ryanodine Receptor 1 Axis. Handb Exp Pharmacol 2023; 278:57-70. [PMID: 36443544 DOI: 10.1007/164_2022_623] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Nicotinic acid adenine dinucleotide phosphate (NAADP) is the most potent Ca2+ mobilizing second messenger known to date. Major steps elucidating metabolism and Ca2+ mobilizing activity of NAADP are reviewed, with emphasis on a novel redox cycle between the inactive reduced form, NAADPH, and the active oxidized form, NAADP. Oxidation from NAADPH to NAADP is catalyzed in cell free system by (dual) NADPH oxidases NOX5, DUOX1, and DUOX2, whereas reduction from NAADP to NAADPH is catalyzed by glucose 6-phosphate dehydrogenase. Using different knockout models for NOX and DUOX isozymes, DUOX2 was identified as NAADP forming enzyme in early T-cell activation.Recently, receptors or binding proteins for NAADP were identified: hematological and neurological expressed 1-like protein (HN1L)/Jupiter microtubule associated homolog 2 (JPT2) and Lsm12 are small cytosolic proteins that bind NAADP. In addition, they interact with NAADP-sensitive Ca2+ channels, such as ryanodine receptor type 1 (RYR1) or two-pore channels (TPC).Due to its role as Ca2+ mobilizing second messenger in T cells, NAADP's involvement in inflammation is also reviewed. In the central nervous system (CNS), NAADP regulates autoimmunity because NAADP antagonism affects a couple of T-cell migration and re-activation events, e.g. secretion of the pro-inflammatory cytokine interleukin-17. Further, the role of NAADP in transdifferentiation of IL-17-producing Th17 cells into T regulatory type 1 cells in vitro and in vivo is discussed.
Collapse
Affiliation(s)
- Andreas H Guse
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
15
|
Jaślan D, Ferro IF, Kudrina V, Yuan Y, Patel S, Grimm C. PI(3,5)P 2 and NAADP: Team players or lone warriors? - New insights into TPC activation modes. Cell Calcium 2023; 109:102675. [PMID: 36525777 DOI: 10.1016/j.ceca.2022.102675] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022]
Abstract
NAADP (nicotinic acid adenine dinucleotide phosphate) is a second messenger, releasing Ca2+ from acidic calcium stores such as endosomes and lysosomes. PI(3,5)P2 (phosphatidylinositol 3,5-bisphosphate) is a phospho-inositide, residing on endolysosomal membranes and likewise releasing Ca2+ from endosomes and lysosomes. Both compounds have been shown to activate endolysosomal two-pore channels (TPCs) in mammalian cells. However, their effects on ion permeability as demonstrated specifically for TPC2 differ. While PI(3,5)P2 elicits predominantly Na+-selective currents, NAADP increases the Ca2+ permeability of the channel. What happens when both compounds are applied simultaneously was unclear until recently.
Collapse
Affiliation(s)
- Dawid Jaślan
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Irene Flavia Ferro
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Veronika Kudrina
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Yu Yuan
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, United Kingdom
| | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, United Kingdom
| | - Christian Grimm
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany.
| |
Collapse
|
16
|
Patel S, Yuan Y, Chen CC, Jaślan D, Gunaratne G, Grimm C, Rahman T, Marchant JS. Electrophysiology of Endolysosomal Two-Pore Channels: A Current Account. Cells 2022; 11:2368. [PMID: 35954212 PMCID: PMC9368155 DOI: 10.3390/cells11152368] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/20/2022] [Accepted: 07/20/2022] [Indexed: 12/10/2022] Open
Abstract
Two-pore channels TPC1 and TPC2 are ubiquitously expressed pathophysiologically relevant proteins that reside on endolysosomal vesicles. Here, we review the electrophysiology of these channels. Direct macroscopic recordings of recombinant TPCs expressed in enlarged lysosomes in mammalian cells or vacuoles in plants and yeast demonstrate gating by the Ca2+-mobilizing messenger NAADP and/or the lipid PI(3,5)P2. TPC currents are regulated by H+, Ca2+, and Mg2+ (luminal and/or cytosolic), as well as protein kinases, and they are impacted by single-nucleotide polymorphisms linked to pigmentation. Bisbenzylisoquinoline alkaloids, flavonoids, and several approved drugs demonstrably block channel activity. Endogenous TPC currents have been recorded from a number of primary cell types and cell lines. Many of the properties of endolysosomal TPCs are recapitulated upon rerouting channels to the cell surface, allowing more facile recording through conventional electrophysiological means. Single-channel analyses have provided high-resolution insight into both monovalent and divalent permeability. The discovery of small-molecule activators of TPC2 that toggle the ion selectivity from a Ca2+-permeable (NAADP-like) state to a Na+-selective (PI(3,5)P2-like) state explains discrepancies in the literature relating to the permeability of TPCs. Identification of binding proteins that confer NAADP-sensitive currents confirm that indirect, remote gating likely underpins the inconsistent observations of channel activation by NAADP.
Collapse
Affiliation(s)
- Sandip Patel
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK;
| | - Yu Yuan
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK;
| | - Cheng-Chang Chen
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei 100229, Taiwan;
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Dawid Jaślan
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians University, 80336 Munich, Germany; (D.J.); (C.G.)
| | - Gihan Gunaratne
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA; (G.G.); (J.S.M.)
| | - Christian Grimm
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians University, 80336 Munich, Germany; (D.J.); (C.G.)
| | - Taufiq Rahman
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, UK;
| | - Jonathan S. Marchant
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA; (G.G.); (J.S.M.)
| |
Collapse
|
17
|
Segregated cation flux by TPC2 biases Ca 2+ signaling through lysosomes. Nat Commun 2022; 13:4481. [PMID: 35918320 PMCID: PMC9346130 DOI: 10.1038/s41467-022-31959-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 07/12/2022] [Indexed: 12/19/2022] Open
Abstract
Two-pore channels are endo-lysosomal cation channels with malleable selectivity filters that drive endocytic ion flux and membrane traffic. Here we show that TPC2 can differentially regulate its cation permeability when co-activated by its endogenous ligands, NAADP and PI(3,5)P2. Whereas NAADP rendered the channel Ca2+-permeable and PI(3,5)P2 rendered the channel Na+-selective, a combination of the two increased Ca2+ but not Na+ flux. Mechanistically, this was due to an increase in Ca2+ permeability independent of changes in ion selectivity. Functionally, we show that cell permeable NAADP and PI(3,5)P2 mimetics synergistically activate native TPC2 channels in live cells, globalizing cytosolic Ca2+ signals and regulating lysosomal pH and motility. Our data reveal that flux of different ions through the same pore can be independently controlled and identify TPC2 as a likely coincidence detector that optimizes lysosomal Ca2+ signaling. TPC2 is a lysosomal ion channel permeable to both calcium and sodium ions. Here, the authors show that TPC2 can selectively increase its calcium permeability when simultaneously challenged by both its natural activators- NAADP and PI(3,5)P2.
Collapse
|
18
|
He X, Kang Y, Chen L. Identification of ASPDH as a novel NAADP-binding protein. Biochem Biophys Res Commun 2022; 621:168-175. [DOI: 10.1016/j.bbrc.2022.06.100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 06/30/2022] [Indexed: 11/02/2022]
|
19
|
Shah KR, Guan X, Yan J. Diversity of two-pore channels and the accessory NAADP receptors in intracellular Ca 2+ signaling. Cell Calcium 2022; 104:102594. [PMID: 35561646 PMCID: PMC9645597 DOI: 10.1016/j.ceca.2022.102594] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/10/2022] [Accepted: 05/04/2022] [Indexed: 02/03/2023]
Abstract
Intracellular Ca2+ signaling via changes or oscillation in cytosolic Ca2+ concentration controls almost every aspect of cellular function and physiological processes, such as gene transcription, cell motility and proliferation, muscle contraction, and learning and memory. Two-pore channels (TPCs) are a class of eukaryotic cation channels involved in intracellular Ca2+ signaling, likely present in a multitude of organisms from unicellular organisms to mammals. Accumulated evidence indicates that TPCs play a critical role in Ca2+ mobilization from intracellular stores mediated by the second messenger molecule, nicotinic acid adenine dinucleotide phosphate (NAADP). In recent years, significant progress has been made regarding our understanding of the structures and function of TPCs, including Cryo-EM structure determination of mammalian TPCs and characterization of a plastid TPC in a single-celled parasite.. The recent identification of Lsm12 and JPT2 as NAADP-binding proteins provides a new molecular basis for understanding NAADP-evoked Ca2+ signaling. In this review, we summarize basic structural and functional aspects of TPCs and highlight the most recent studies on the newly discovered TPC in a parasitic protozoan and the NAADP-binding proteins LSM12 and JPT2 as new key players in NAADP signaling.
Collapse
Affiliation(s)
- Kunal R. Shah
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xin Guan
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jiusheng Yan
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,Neuroscience and Biochemistry and Cell Biology Programs, The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX, USA,Corresponding author at: Department of Anesthesiology & Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA., (J. Yan)
| |
Collapse
|
20
|
Martucci LL, Cancela JM. Neurophysiological functions and pharmacological tools of acidic and non-acidic Ca2+ stores. Cell Calcium 2022; 104:102582. [DOI: 10.1016/j.ceca.2022.102582] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/07/2022] [Accepted: 03/23/2022] [Indexed: 02/08/2023]
|
21
|
NAADP Signaling: New Kids on the Block. Cells 2022; 11:cells11061054. [PMID: 35326505 PMCID: PMC8947471 DOI: 10.3390/cells11061054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 01/20/2023] Open
Abstract
Nicotinic acid adenine dinucleotide phosphate (NAADP) is a universal Ca2+ mobilizing second messenger essential for initiation of Ca2+ signaling. Recently, novel molecular mechanisms of both its rapid formation upon receptor stimulation and its mode of action were discovered. Dual NADPH oxidase 2 (DUOX2) and hematological and neurological expressed 1-like protein (HN1L)/Jupiter microtubule-associated homolog 2 (JPT2) were discovered as NAADP-forming enzyme and NAADP receptor/binding protein—the new kids on the block. These novel aspects are reviewed and integrated into the previous view of NAADP signaling.
Collapse
|
22
|
Terrar DA. Endolysosomal Calcium Release and Cardiac Physiology. Cell Calcium 2022; 104:102565. [DOI: 10.1016/j.ceca.2022.102565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 11/25/2022]
|
23
|
Barbonari S, D'Amore A, Palombi F, De Cesaris P, Parrington J, Riccioli A, Filippini A. RELEVANCE OF LYSOSOMAL Ca2+ SIGNALLING MACHINERY IN CANCER. Cell Calcium 2022; 102:102539. [DOI: 10.1016/j.ceca.2022.102539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 12/23/2022]
|
24
|
Abstract
Nicotinic acid adenine dinucleotide 2'-phosphate (NAADP) is a naturally occurring nucleotide that has been shown to be involved in the release of Ca2+ from intracellular stores in a wide variety of cell types, tissues and organisms. Current evidence suggests that NAADP may function as a trigger to initiate a Ca2+ signal that is then amplified by other Ca2+ release mechanisms. A fundamental question that remains unanswered is the identity of the NAADP receptor. Our recent studies have identified HN1L/JPT2 as a high affinity NAADP binding protein that is essential for the modulation of Ca2+ channels.
Collapse
Affiliation(s)
- Timothy F Walseth
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Andreas H Guse
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
25
|
Jin X, Zhang Y, Alharbi A, Hanbashi A, Alhoshani A, Parrington J. Targeting Two-Pore Channels: Current Progress and Future Challenges. Trends Pharmacol Sci 2021; 41:582-594. [PMID: 32679067 PMCID: PMC7365084 DOI: 10.1016/j.tips.2020.06.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/04/2020] [Accepted: 06/04/2020] [Indexed: 12/19/2022]
Abstract
Two-pore channels (TPCs) are cation-permeable channels located on endolysosomal membranes and important mediators of intracellular Ca2+ signalling. TPCs are involved in various pathophysiological processes, including cell growth and development, metabolism, and cancer progression. Most studies of TPCs have used TPC–/– cell or whole-animal models, or Ned-19, an indirect inhibitor. The TPC activation mechanism remains controversial, which has made it difficult to develop selective modulators. Recent studies of TPC structure and their interactomes are aiding the development of direct pharmacological modulators. This process is still in its infancy, but will facilitate future research and TPC targeting for therapeutical purposes. Here, we review the progress of current research into TPCs, including recent insights into their structures, functional roles, mechanisms of activation, and pharmacological modulators. Two-pore channel (TPC)-mediated endolysosomal Ca2+ signalling regulates a variety of processes, including cell proliferation, differentiation, metabolism, viral infection, and cardiac function. Despite the well-established model that TPCs are Ca2+-selective channels indirectly activated by nicotinic acid adenine dinucleotide phosphate (NAADP), it has also been proposed that TPCs as Na+ channels are activated directly by phosphatidylinositol 3,5-bisphosphate [PI(3,5)P2]. 3D structures of mouse TPC1 and human TPC2 were recently determined, which made it possible for structure-based virtual screening methods to identify pharmacological modulators of TPC. Recent identification by high-throughput screens of pharmacological modulators that target TPCs will help reveal the molecular mechanisms underlying the role of endolysosomal Ca2+ signalling in different pathophysiological processes, and to develop new therapeutics.
Collapse
Affiliation(s)
- Xuhui Jin
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Yuxuan Zhang
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Abeer Alharbi
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Ali Hanbashi
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Ali Alhoshani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, PO Box 2457, Riyadh 11454, Kingdom of Saudi Arabia
| | - John Parrington
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK.
| |
Collapse
|
26
|
Lsm12 is an NAADP receptor and a two-pore channel regulatory protein required for calcium mobilization from acidic organelles. Nat Commun 2021; 12:4739. [PMID: 34362892 PMCID: PMC8346516 DOI: 10.1038/s41467-021-24735-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/01/2021] [Indexed: 01/19/2023] Open
Abstract
Nicotinic acid adenine dinucleotide phosphate (NAADP) is a potent Ca2+-mobilizing second messenger which uniquely mobilizes Ca2+ from acidic endolysosomal organelles. However, the molecular identity of the NAADP receptor remains unknown. Given the necessity of the endolysosomal two-pore channel (TPC1 or TPC2) in NAADP signaling, we performed affinity purification and quantitative proteomic analysis of the interacting proteins of NAADP and TPCs. We identified a Sm-like protein Lsm12 complexed with NAADP, TPC1, and TPC2. Lsm12 directly binds to NAADP via its Lsm domain, colocalizes with TPC2, and mediates the apparent association of NAADP to isolated TPC2 or TPC2-containing membranes. Lsm12 is essential and immediately participates in NAADP-evoked TPC activation and Ca2+ mobilization from acidic stores. These findings reveal a putative RNA-binding protein to function as an NAADP receptor and a TPC regulatory protein and provides a molecular basis for understanding the mechanisms of NAADP signaling. Nicotinic acid adenine dinucleotide phosphate (NAADP) potent Ca2+ mobilizing second messenger which uniquely triggers Ca2+ release from acidic endolysosomal organelles. Here the authors identify Lsm12 as an NAADP receptor essential for NAADP-evoked Ca2+ release from lysosomes via NAADP binding on its Lsm domain.
Collapse
|
27
|
Glucose and NAADP trigger elementary intracellular β-cell Ca 2+ signals. Sci Rep 2021; 11:10714. [PMID: 34021189 PMCID: PMC8140081 DOI: 10.1038/s41598-021-88906-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/15/2021] [Indexed: 11/12/2022] Open
Abstract
Pancreatic β-cells release insulin upon a rise in blood glucose. The precise mechanisms of stimulus-secretion coupling, and its failure in Diabetes Mellitus Type 2, remain to be elucidated. The consensus model, as well as a class of currently prescribed anti-diabetic drugs, are based around the observation that glucose-evoked ATP production in β-cells leads to closure of cell membrane ATP-gated potassium (KATP) channels, plasma membrane depolarisation, Ca2+ influx, and finally the exocytosis of insulin granules. However, it has been demonstrated by the inactivation of this pathway using genetic and pharmacological means that closure of the KATP channel alone may not be sufficient to explain all β-cell responses to glucose elevation. We have previously proposed that NAADP-evoked Ca2+ release is an important step in stimulus-secretion coupling in pancreatic β-cells. Here we show using total internal reflection fluorescence (TIRF) microscopy that glucose as well as the Ca2+ mobilising messenger nicotinic acid adenine dinucleotide phosphate (NAADP), known to operate in β-cells, lead to highly localised elementary intracellular Ca2+ signals. These were found to be obscured by measurements of global Ca2+ signals and the action of powerful SERCA-based sequestration mechanisms at the endoplasmic reticulum (ER). Building on our previous work demonstrating that NAADP-evoked Ca2+ release is an important step in stimulus-secretion coupling in pancreatic β-cells, we provide here the first demonstration of elementary Ca2+ signals in response to NAADP, whose occurrence was previously suspected. Optical quantal analysis of these events reveals a unitary event amplitude equivalent to that of known elementary Ca2+ signalling events, inositol trisphosphate (IP3) receptor mediated blips, and ryanodine receptor mediated quarks. We propose that a mechanism based on these highly localised intracellular Ca2+ signalling events mediated by NAADP may initially operate in β-cells when they respond to elevations in blood glucose.
Collapse
|
28
|
Petersen OH, Gerasimenko JV, Gerasimenko OV, Gryshchenko O, Peng S. The roles of calcium and ATP in the physiology and pathology of the exocrine pancreas. Physiol Rev 2021; 101:1691-1744. [PMID: 33949875 DOI: 10.1152/physrev.00003.2021] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
This review deals with the roles of calcium ions and ATP in the control of the normal functions of the different cell types in the exocrine pancreas as well as the roles of these molecules in the pathophysiology of acute pancreatitis. Repetitive rises in the local cytosolic calcium ion concentration in the apical part of the acinar cells not only activate exocytosis but also, via an increase in the intramitochondrial calcium ion concentration, stimulate the ATP formation that is needed to fuel the energy-requiring secretion process. However, intracellular calcium overload, resulting in a global sustained elevation of the cytosolic calcium ion concentration, has the opposite effect of decreasing mitochondrial ATP production, and this initiates processes that lead to necrosis. In the last few years it has become possible to image calcium signaling events simultaneously in acinar, stellate, and immune cells in intact lobules of the exocrine pancreas. This has disclosed processes by which these cells interact with each other, particularly in relation to the initiation and development of acute pancreatitis. By unraveling the molecular mechanisms underlying this disease, several promising therapeutic intervention sites have been identified. This provides hope that we may soon be able to effectively treat this often fatal disease.
Collapse
Affiliation(s)
- Ole H Petersen
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | | | | | | | - Shuang Peng
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
29
|
Negri S, Faris P, Moccia F. Endolysosomal Ca 2+ signaling in cardiovascular health and disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 363:203-269. [PMID: 34392930 DOI: 10.1016/bs.ircmb.2021.03.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
An increase in intracellular Ca2+ concentration ([Ca2+]i) regulates a plethora of functions in the cardiovascular (CV) system, including contraction in cardiomyocytes and vascular smooth muscle cells (VSMCs), and angiogenesis in vascular endothelial cells and endothelial colony forming cells. The sarco/endoplasmic reticulum (SR/ER) represents the largest endogenous Ca2+ store, which releases Ca2+ through ryanodine receptors (RyRs) and/or inositol-1,4,5-trisphosphate receptors (InsP3Rs) upon extracellular stimulation. The acidic vesicles of the endolysosomal (EL) compartment represent an additional endogenous Ca2+ store, which is targeted by several second messengers, including nicotinic acid adenine dinucleotide phosphate (NAADP) and phosphatidylinositol 3,5-bisphosphate [PI(3,5)P2], and may release intraluminal Ca2+ through multiple Ca2+ permeable channels, including two-pore channels 1 and 2 (TPC1-2) and Transient Receptor Potential Mucolipin 1 (TRPML1). Herein, we discuss the emerging, pathophysiological role of EL Ca2+ signaling in the CV system. We describe the role of cardiac TPCs in β-adrenoceptor stimulation, arrhythmia, hypertrophy, and ischemia-reperfusion injury. We then illustrate the role of EL Ca2+ signaling in VSMCs, where TPCs promote vasoconstriction and contribute to pulmonary artery hypertension and atherosclerosis, whereas TRPML1 sustains vasodilation and is also involved in atherosclerosis. Subsequently, we describe the mechanisms whereby endothelial TPCs promote vasodilation, contribute to neurovascular coupling in the brain and stimulate angiogenesis and vasculogenesis. Finally, we discuss about the possibility to target TPCs, which are likely to mediate CV cell infection by the Severe Acute Respiratory Disease-Coronavirus-2, with Food and Drug Administration-approved drugs to alleviate the detrimental effects of Coronavirus Disease-19 on the CV system.
Collapse
Affiliation(s)
- Sharon Negri
- Laboratory of Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Pawan Faris
- Laboratory of Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Francesco Moccia
- Laboratory of Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy.
| |
Collapse
|
30
|
Morgan AJ, Davis LC, Galione A. Choreographing endo-lysosomal Ca 2+ throughout the life of a phagosome. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119040. [PMID: 33872669 DOI: 10.1016/j.bbamcr.2021.119040] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 12/20/2022]
Abstract
The emergence of endo-lysosomes as ubiquitous Ca2+ stores with their unique cohort of channels has resulted in their being implicated in a growing number of processes in an ever-increasing number of cell types. The architectural and regulatory constraints of these acidic Ca2+ stores distinguishes them from other larger Ca2+ sources such as the ER and influx across the plasma membrane. In view of recent advances in the understanding of the modes of operation, we discuss phagocytosis as a template for how endo-lysosomal Ca2+ signals (generated via TPC and TRPML channels) can be integrated in multiple sophisticated ways into biological processes. Phagocytosis illustrates how different endo-lysosomal Ca2+ signals drive different phases of a process, and how these can be altered by disease or infection.
Collapse
Affiliation(s)
- Anthony J Morgan
- Department of Pharmacology, University of Oxford, Mansfield Park, Oxford OX1 3QT, UK.
| | - Lianne C Davis
- Department of Pharmacology, University of Oxford, Mansfield Park, Oxford OX1 3QT, UK
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Mansfield Park, Oxford OX1 3QT, UK.
| |
Collapse
|
31
|
Ibata K, Yuzaki M. Destroy the old to build the new: Activity-dependent lysosomal exocytosis in neurons. Neurosci Res 2021; 167:38-46. [PMID: 33845090 DOI: 10.1016/j.neures.2021.03.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 12/14/2022]
Abstract
Lysosomes are organelles that support diverse cellular functions such as terminal degradation of macromolecules and nutrient recycling. Additionally, lysosomes can fuse with the plasma membrane, a phenomenon referred to as lysosomal exocytosis, to release their contents, including hydrolytic enzymes and cargo proteins. Recently, neuronal activity has been shown to induce lysosomal exocytosis in dendrites and axons. Secreted lysosomal enzyme cathepsin B induces and stabilizes synaptic structural changes by degrading the local extracellular matrix. Extracellular matrix reorganization could also enhance the lateral diffusion of the co-released synaptic organizer Cbln1 along the surface of axons to facilitate new synapse formation. Similarly, lateral diffusion of dendritic AMPA-type glutamate receptors could be facilitated to enhance functional synaptic plasticity. Therefore, lysosomal exocytosis is a powerful way of building new cellular structures through the coordinated destruction of the old environment. Understanding the mechanisms by which lysosomal exocytosis is regulated in neurons is expected to lead to the development of new therapeutics for neuronal plasticity following spinal cord injury or neurodegenerative disease.
Collapse
Affiliation(s)
- Keiji Ibata
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan; Department of Physiology, St. Marianna University School of Medicine, 216-8511, Kanagawa, Japan
| | - Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan.
| |
Collapse
|
32
|
Roggenkamp HG, Khansahib I, Hernandez C LC, Zhang Y, Lodygin D, Krüger A, Gu F, Möckl F, Löhndorf A, Wolters V, Woike D, Rosche A, Bauche A, Schetelig D, Werner R, Schlüter H, Failla AV, Meier C, Fliegert R, Walseth TF, Flügel A, Diercks BP, Guse AH. HN1L/JPT2: A signaling protein that connects NAADP generation to Ca 2+ microdomain formation. Sci Signal 2021; 14:14/675/eabd5647. [PMID: 33758062 DOI: 10.1126/scisignal.abd5647] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
NAADP-evoked Ca2+ release through type 1 ryanodine receptors (RYR1) is a major mechanism underlying the earliest signals in T cell activation, which are the formation of Ca2+ microdomains. In our characterization of the molecular machinery underlying NAADP action, we identified an NAADP-binding protein, called hematological and neurological expressed 1-like protein (HN1L) [also known as Jupiter microtubule-associated homolog 2 (JPT2)]. Gene deletion of Hn1l/Jpt2 in human Jurkat and primary rat T cells resulted in decreased numbers of initial Ca2+ microdomains and delayed the onset and decreased the amplitude of global Ca2+ signaling. Photoaffinity labeling demonstrated direct binding of NAADP to recombinant HN1L/JPT2. T cell receptor/CD3-dependent coprecipitation of HN1L/JPT2 with RYRs and colocalization of these proteins suggest that HN1L/JPT2 connects NAADP formation with the activation of RYR channels within the first seconds of T cell activation. Thus, HN1L/JPT2 enables NAADP to activate Ca2+ release from the endoplasmic reticulum through RYR.
Collapse
Affiliation(s)
- Hannes G Roggenkamp
- The Ca Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Imrankhan Khansahib
- The Ca Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lola C Hernandez C
- The Ca Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Yunpeng Zhang
- The Ca Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Dmitri Lodygin
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre Göttingen, 37075 Göttingen, Germany
| | - Aileen Krüger
- The Ca Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Feng Gu
- The Ca Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Franziska Möckl
- The Ca Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Anke Löhndorf
- The Ca Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Valerie Wolters
- The Ca Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Daniel Woike
- The Ca Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Anette Rosche
- The Ca Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Andreas Bauche
- The Ca Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Daniel Schetelig
- Department of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - René Werner
- Department of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Hartmut Schlüter
- Mass Spectrometric Proteomics Group, Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Antonio V Failla
- Microscopy Imaging Facility (UMIF), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Chris Meier
- Organic Chemistry, University of Hamburg, 20146 Hamburg, Germany
| | - Ralf Fliegert
- The Ca Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Timothy F Walseth
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455-0217, USA
| | - Alexander Flügel
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre Göttingen, 37075 Göttingen, Germany
| | - Björn-Philipp Diercks
- The Ca Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Andreas H Guse
- The Ca Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| |
Collapse
|
33
|
Cytosolic and intra-organellar Ca2+ oscillations: mechanisms and function. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
34
|
The role of Ca2+ signalling in the physiology and pathophysiology of exocrine pancreas. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.07.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
35
|
Nam TS, Park DR, Rah SY, Woo TG, Chung HT, Brenner C, Kim UH. Interleukin-8 drives CD38 to form NAADP from NADP + and NAAD in the endolysosomes to mobilize Ca 2+ and effect cell migration. FASEB J 2020; 34:12565-12576. [PMID: 32717131 DOI: 10.1096/fj.202001249r] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/28/2020] [Accepted: 07/08/2020] [Indexed: 01/22/2023]
Abstract
Nicotinic acid adenine dinucleotide phosphate (NAADP) is the most potent Ca2+ mobilizing second messenger whose formation has remained elusive. In vitro, CD38-mediated NAADP synthesis requires an acidic pH and a nonphysiological concentration of nicotinic acid (NA). We discovered that CD38 catalyzes synthesis of NAADP by exchanging the nicotinamide moiety of nicotinamide adenine dinucleotide phosphate (NADP+ ) for the NA group of nicotinic acid adenine dinucleotide (NAAD) inside endolysosomes of interleukin 8 (IL8)-treated lymphokine-activated killer (LAK) cells. Upon IL8 stimulation, cytosolic NADP+ is transported to acidified endolysosomes via connexin 43 (Cx43) and gated by cAMP-EPAC-RAP1-PP2A signaling. CD38 then performs a base-exchange reaction with the donor NA group deriving from NAAD, produced by newly described endolysosomal activities of NA phosphoribosyltransferase (NAPRT) and NMN adenyltransferase (NMNAT) 3. Thus, the membrane organization of endolysosomal CD38, a signal-mediated transport system for NADP+ and luminal NAD+ biosynthetic enzymes integrate signals from a chemokine and cAMP to specify the spatiotemporal mobilization of Ca2+ to drive cell migration.
Collapse
Affiliation(s)
- Tae-Sik Nam
- Department of Biochemistry & National Creative Research Laboratory for Ca2+ Signaling, Chonbuk National University Medical School, Jeonju, Korea
| | - Dae-Ryoung Park
- Department of Biochemistry & National Creative Research Laboratory for Ca2+ Signaling, Chonbuk National University Medical School, Jeonju, Korea
| | - So-Young Rah
- Department of Biochemistry & National Creative Research Laboratory for Ca2+ Signaling, Chonbuk National University Medical School, Jeonju, Korea
| | - Tae-Gyu Woo
- Department of Biochemistry & National Creative Research Laboratory for Ca2+ Signaling, Chonbuk National University Medical School, Jeonju, Korea
| | - Hun Taeg Chung
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea
| | - Charles Brenner
- Department of Biochemistry, University of Iowa, Iowa City, IA, USA
| | - Uh-Hyun Kim
- Department of Biochemistry & National Creative Research Laboratory for Ca2+ Signaling, Chonbuk National University Medical School, Jeonju, Korea
| |
Collapse
|
36
|
Masoli S, Tognolina M, Laforenza U, Moccia F, D'Angelo E. Parameter tuning differentiates granule cell subtypes enriching transmission properties at the cerebellum input stage. Commun Biol 2020; 3:222. [PMID: 32385389 PMCID: PMC7210112 DOI: 10.1038/s42003-020-0953-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 04/13/2020] [Indexed: 02/06/2023] Open
Abstract
The cerebellar granule cells (GrCs) are classically described as a homogeneous neuronal population discharging regularly without adaptation. We show that GrCs in fact generate diverse response patterns to current injection and synaptic activation, ranging from adaptation to acceleration of firing. Adaptation was predicted by parameter optimization in detailed computational models based on available knowledge on GrC ionic channels. The models also predicted that acceleration required additional mechanisms. We found that yet unrecognized TRPM4 currents specifically accounted for firing acceleration and that adapting GrCs outperformed accelerating GrCs in transmitting high-frequency mossy fiber (MF) bursts over a background discharge. This implied that GrC subtypes identified by their electroresponsiveness corresponded to specific neurotransmitter release probability values. Simulations showed that fine-tuning of pre- and post-synaptic parameters generated effective MF-GrC transmission channels, which could enrich the processing of input spike patterns and enhance spatio-temporal recoding at the cerebellar input stage.
Collapse
Affiliation(s)
- Stefano Masoli
- Department of Brain and Behavioral Sciences, University of Pavia, Via Forlanini 6, 27100, Pavia, Italy
| | - Marialuisa Tognolina
- Department of Brain and Behavioral Sciences, University of Pavia, Via Forlanini 6, 27100, Pavia, Italy
| | - Umberto Laforenza
- Department of Molecular Medicine, University of Pavia, Via Forlanini 6, 27100, Pavia, Italy
| | - Francesco Moccia
- Department of Biology and Biotechnology, University of Pavia, Via Forlanini 6, 27100, Pavia, Italy
| | - Egidio D'Angelo
- Department of Brain and Behavioral Sciences, University of Pavia, Via Forlanini 6, 27100, Pavia, Italy. .,Brain Connectivity Center, IRCCS Mondino Foundation, Via Mondino 2, 27100, Pavia, Italy.
| |
Collapse
|
37
|
Webb SE, Kelu JJ, Miller AL. Role of Two-Pore Channels in Embryonic Development and Cellular Differentiation. Cold Spring Harb Perspect Biol 2020; 12:a035170. [PMID: 31358517 PMCID: PMC6942120 DOI: 10.1101/cshperspect.a035170] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Since the identification of nicotinic acid adenine dinucleotide phosphate (NAADP) and its putative target, the two-pore channel (TPC), the NAADP/TPC/Ca2+ signaling pathway has been reported to play a role in a diverse range of functions in a variety of different cell types. TPCs have also been associated with a number of diseases, which arise when their activity is perturbed. In addition, TPCs have been shown to play key roles in various embryological processes and during the differentiation of a variety of cell types. Here, we review the role of NAADP/TPC/Ca2+ signaling during early embryonic development and cellular differentiation. We pay particular attention to the role of TPC2 in the development and maturation of early neuromuscular activity in zebrafish, and during the differentiation of isolated osteoclasts, endothelial cells, and keratinocytes. Our aim is to emphasize the conserved features of TPC-mediated Ca2+ signaling in a number of selected examples.
Collapse
Affiliation(s)
- Sarah E Webb
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology (HKUST), Clearwater Bay, Hong Kong, PRC
| | - Jeffrey J Kelu
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology (HKUST), Clearwater Bay, Hong Kong, PRC
| | - Andrew L Miller
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology (HKUST), Clearwater Bay, Hong Kong, PRC
| |
Collapse
|
38
|
Abstract
Of the established Ca2+-mobilizing messengers, NAADP is arguably the most tantalizing. It is the most potent, often efficacious at low nanomolar concentrations, and its receptors undergo dramatic desensitization. Recent studies have identified a new class of calcium-release channel, the two-pore channels (TPCs), as the likely targets for NAADP regulation, even though the effect may be indirect. These channels localized at endolysosomes, where they mediate local Ca2+ release, and have highlighted a new role of acidic organelles as targets for messenger-evoked Ca2+ mobilization. Three distinct roles of TPCs have been identified. The first is to effect local Ca2+ release that may play a role in endolysosomal function including vesicular fusion and trafficking. The second is to trigger global calcium release by recruiting Ca2+-induced Ca2+-release (CICR) channels at lysosomal-endoplasmic reticulum (ER) junctions. The third is to regulate plasma membrane excitability by the targeting of Ca2+ release from appropriately positioned subplasma membrane stores to regulate plasma membrane Ca2+-activated channels. In this review, I discuss the role of nicotinic acid adenine nucleotide diphosphate (NAADP)-mediated Ca2+ release from endolysosomal stores as a widespread trigger for intracellular calcium signaling mechanisms, and how studies of TPCs are beginning to enhance our understanding of the central role of lysosomes in Ca2+ signaling.
Collapse
Affiliation(s)
- Antony Galione
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, United Kingdom
| |
Collapse
|
39
|
Galione A, Chuang KT. Pyridine Nucleotide Metabolites and Calcium Release from Intracellular Stores. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1131:371-394. [PMID: 31646518 DOI: 10.1007/978-3-030-12457-1_15] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Ca2+ signals are probably the most common intracellular signaling cellular events, controlling an extensive range of responses in virtually all cells. Many cellular stimuli, often acting at cell surface receptors, evoke Ca2+ signals by mobilizing Ca2+ from intracellular stores. Inositol trisphosphate (IP3) was the first messenger shown to link events at the plasma membrane to release Ca2+ from the endoplasmic reticulum (ER), through the activation of IP3-gated Ca2+ release channels (IP3 receptors). Subsequently, two additional Ca2+ mobilizing messengers were discovered, cADPR and NAADP. Both are metabolites of pyridine nucleotides, and may be produced by the same class of enzymes, ADP-ribosyl cyclases, such as CD38. Whilst cADPR mobilizes Ca2+ from the ER by activation of ryanodine receptors (RyRs), NAADP releases Ca2+ from acidic stores by a mechanism involving the activation of two pore channels (TPCs). In addition, other pyridine nucleotides have emerged as intracellular messengers. ADP-ribose and 2'-deoxy-ADPR both activate TRPM2 channels which are expressed at the plasma membrane and in lysosomes.
Collapse
Affiliation(s)
- Antony Galione
- Department of Pharmacology, University of Oxford, Oxford, UK.
| | - Kai-Ting Chuang
- Department of Pharmacology, University of Oxford, Oxford, UK
| |
Collapse
|
40
|
Abstract
The aim of this chapter is to discuss evidence concerning the many roles of calcium ions, Ca2+, in cell signaling pathways that control heart function. Before considering details of these signaling pathways, the control of contraction in ventricular muscle by Ca2+ transients accompanying cardiac action potentials is first summarized, together with a discussion of how myocytes from the atrial and pacemaker regions of the heart diverge from this basic scheme. Cell signaling pathways regulate the size and timing of the Ca2+ transients in the different heart regions to influence function. The simplest Ca2+ signaling elements involve enzymes that are regulated by cytosolic Ca2+. Particularly important examples to be discussed are those that are stimulated by Ca2+, including Ca2+-calmodulin-dependent kinase (CaMKII), Ca2+ stimulated adenylyl cyclases, Ca2+ stimulated phosphatase and NO synthases. Another major aspect of Ca2+ signaling in the heart concerns actions of the Ca2+ mobilizing agents, inositol trisphosphate (IP3), cADP-ribose (cADPR) and nicotinic acid adenine dinucleotide phosphate, (NAADP). Evidence concerning roles of these Ca2+ mobilizing agents in different regions of the heart is discussed in detail. The focus of the review will be on short term regulation of Ca2+ transients and contractile function, although it is recognized that Ca2+ regulation of gene expression has important long term functional consequences which will also be briefly discussed.
Collapse
|
41
|
Asfaha TY, Gunaratne GS, Johns ME, Marchant JS, Walseth TF, Slama JT. The synthesis and characterization of a clickable-photoactive NAADP analog active in human cells. Cell Calcium 2019; 83:102060. [PMID: 31442840 DOI: 10.1016/j.ceca.2019.102060] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/11/2019] [Accepted: 07/15/2019] [Indexed: 12/11/2022]
Abstract
Nicotinic acid adenine dinucleotide phosphate (NAADP) is a potent Ca2+ mobilizing second messenger which triggers Ca2+ release in both sea urchin egg homogenates and in mammalian cells. The NAADP binding protein has not been identified and the regulation of NAADP mediated Ca2+ release remains controversial. To address this issue, we have synthesized an NAADP analog in which 3-azido-5-azidomethylbenzoic acid is attached to the amino group of 5-(3-aminopropyl)-NAADP to produce an NAADP analog which is both a photoaffinity label and clickable. This 'all-in-one-clickable' NAADP (AIOC-NAADP) elicited Ca2+ release when microinjected into cultured human SKBR3 cells at low concentrations. In contrast, it displayed little activity in sea urchin egg homogenates where very high concentrations were required to elicit Ca2+ release. In mammalian cell homogenates, incubation with low concentrations of [32P]AIOC-NAADP followed by irradiation with UV light resulted in labeling 23 kDa protein(s). Competition between [32P]AIOC-NAADP and increasing concentrations of NAADP demonstrated that the labeling was selective. We show that this label recognizes and selectively photodervatizes the 23 kDa NAADP binding protein(s) in cultured human cells identified in previous studies using [32P]5-N3-NAADP.
Collapse
Affiliation(s)
- Timnit Yosef Asfaha
- Department of Medicinal and Biological Chemistry, University of Toledo College of Pharmacy and Pharmaceutical Sciences, 3000 Arlington Avenue, Toledo, OH, 43614, United States
| | - Gihan S Gunaratne
- Department of Pharmacology, University of Minnesota Medical School, 312 Church St., Minneapolis, MN, 55455-0217, United States
| | - Malcolm E Johns
- Department of Pharmacology, University of Minnesota Medical School, 312 Church St., Minneapolis, MN, 55455-0217, United States
| | - Jonathan S Marchant
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226-0509, United States
| | - Timothy F Walseth
- Department of Pharmacology, University of Minnesota Medical School, 312 Church St., Minneapolis, MN, 55455-0217, United States.
| | - James T Slama
- Department of Medicinal and Biological Chemistry, University of Toledo College of Pharmacy and Pharmaceutical Sciences, 3000 Arlington Avenue, Toledo, OH, 43614, United States.
| |
Collapse
|
42
|
Tai L, Li BB, Nie XM, Zhang PP, Hu CH, Zhang L, Liu WT, Li WQ, Chen KM. Calmodulin Is the Fundamental Regulator of NADK-Mediated NAD Signaling in Plants. FRONTIERS IN PLANT SCIENCE 2019; 10:681. [PMID: 31275331 PMCID: PMC6593290 DOI: 10.3389/fpls.2019.00681] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/06/2019] [Indexed: 05/02/2023]
Abstract
Calcium (Ca2+) signaling and nicotinamide adenine dinucleotide (NAD) signaling are two basic signal regulation pathways in organisms, playing crucial roles in signal transduction, energy metabolism, stress tolerance, and various developmental processes. Notably, calmodulins (CaMs) and NAD kinases (NADKs) are important hubs for connecting these two types of signaling networks, where CaMs are the unique activators of NADKs. NADK is a key enzyme for NADP (including NADP+ and NADPH) biosynthesis by phosphorylating NAD (including NAD+ and NADH) and therefore, maintains the balance between NAD pool and NADP pool through an allosteric regulation mode. In addition, the two respective derivatives from NAD+ (substrate of NADK) and NADP+ (product of NADK), cyclic ADP-ribose (cADPR) and nicotinic acid adenine dinucleotide phosphate (NAADP), have been considered to be the important messengers for intracellular Ca2+ homeostasis which could finally influence the combination between CaM and NADK, forming a feedback regulation mechanism. In this review article, we briefly summarized the major research advances related to the feedback regulation pathway, which is activated by the interaction of CaM and NADK during plant development and signaling. The theories and fact will lay a solid foundation for further studies related to CaM and NADK and their regulatory mechanisms as well as the NADK-mediated NAD signaling behavior in plant development and response to stress.
Collapse
Affiliation(s)
- Li Tai
- State Key Laboratory of Crop Stress Biology in Arid Area/College of Life Sciences, Northwest A&F University, Yangling, China
| | - Bin-Bin Li
- State Key Laboratory of Crop Stress Biology in Arid Area/College of Life Sciences, Northwest A&F University, Yangling, China
| | - Xiu-Min Nie
- State Key Laboratory of Crop Stress Biology in Arid Area/College of Life Sciences, Northwest A&F University, Yangling, China
| | - Peng-Peng Zhang
- State Key Laboratory of Crop Stress Biology in Arid Area/College of Life Sciences, Northwest A&F University, Yangling, China
| | - Chun-Hong Hu
- State Key Laboratory of Crop Stress Biology in Arid Area/College of Life Sciences, Northwest A&F University, Yangling, China
- Department of General Biology, College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou, China
| | - Lu Zhang
- State Key Laboratory of Crop Stress Biology in Arid Area/College of Life Sciences, Northwest A&F University, Yangling, China
| | - Wen-Ting Liu
- State Key Laboratory of Crop Stress Biology in Arid Area/College of Life Sciences, Northwest A&F University, Yangling, China
| | - Wen-Qiang Li
- State Key Laboratory of Crop Stress Biology in Arid Area/College of Life Sciences, Northwest A&F University, Yangling, China
| | - Kun-Ming Chen
- State Key Laboratory of Crop Stress Biology in Arid Area/College of Life Sciences, Northwest A&F University, Yangling, China
| |
Collapse
|
43
|
Westman J, Grinstein S, Maxson ME. Revisiting the role of calcium in phagosome formation and maturation. J Leukoc Biol 2019; 106:837-851. [DOI: 10.1002/jlb.mr1118-444r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/24/2019] [Accepted: 04/25/2019] [Indexed: 12/19/2022] Open
Affiliation(s)
- Johannes Westman
- Program in Cell BiologyHospital for Sick Children Toronto Ontario Canada
| | - Sergio Grinstein
- Program in Cell BiologyHospital for Sick Children Toronto Ontario Canada
- Department of BiochemistryUniversity of Toronto Toronto Ontario Canada
- Keenan Research Centre of the Li Ka Shing Knowledge InstituteSt. Michael's Hospital Toronto Ontario Canada
| | - Michelle E. Maxson
- Program in Cell BiologyHospital for Sick Children Toronto Ontario Canada
| |
Collapse
|
44
|
Martucci LL, Amar M, Chaussenot R, Benet G, Bauer O, de Zélicourt A, Nosjean A, Launay JM, Callebert J, Sebrié C, Galione A, Edeline JM, de la Porte S, Fossier P, Granon S, Vaillend C, Cancela JM. A multiscale analysis in CD38 -/- mice unveils major prefrontal cortex dysfunctions. FASEB J 2019; 33:5823-5835. [PMID: 30844310 DOI: 10.1096/fj.201800489r] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Autism spectrum disorder (ASD) is characterized by early onset of behavioral and cognitive alterations. Low plasma levels of oxytocin (OT) have also been found in ASD patients; recently, a critical role for the enzyme CD38 in the regulation of OT release was demonstrated. CD38 is important in regulating several Ca2+-dependent pathways, but beyond its role in regulating OT secretion, it is not known whether a deficit in CD38 expression leads to functional modifications of the prefrontal cortex (PFC), a structure involved in social behavior. Here, we report that CD38-/- male mice show an abnormal cortex development, an excitation-inhibition balance shifted toward a higher excitation, and impaired synaptic plasticity in the PFC such as those observed in various mouse models of ASD. We also show that a lack of CD38 alters social behavior and emotional responses. Finally, examining neuromodulators known to control behavioral flexibility, we found elevated monoamine levels in the PFC of CD38-/- adult mice. Overall, our study unveiled major changes in PFC physiologic mechanisms and provides new evidence that the CD38-/- mouse could be a relevant model to study pathophysiological brain mechanisms of mental disorders such as ASD.-Martucci, L. L., Amar, M., Chaussenot, R., Benet, G., Bauer, O., de Zélicourt, A., Nosjean, A., Launay, J.-M., Callebert, J., Sebrié, C., Galione, A., Edeline, J.-M., de la Porte, S., Fossier, P., Granon, S., Vaillend, C., Cancela, J.-M., A multiscale analysis in CD38-/- mice unveils major prefrontal cortex dysfunctions.
Collapse
Affiliation(s)
- Lora L Martucci
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France.,INSERM Unité 1179, Handicap Neuromusculaire: Physiologie, Biothérapie et Pharmacologie Appliquées, Unité de Formation et de Recherche (UFR) des Sciences de la Santé Simone Veil, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Montigny-le-Bretonneux, France
| | - Muriel Amar
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France
| | - Remi Chaussenot
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France
| | - Gabriel Benet
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France
| | - Oscar Bauer
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France.,Génétique Humaine et Fonctions Cognitives, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 3571, Gènes, Synapses et Cognition, CNRS, Institut Pasteur, Paris, France
| | - Antoine de Zélicourt
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France.,INSERM Unité 1179, Handicap Neuromusculaire: Physiologie, Biothérapie et Pharmacologie Appliquées, Unité de Formation et de Recherche (UFR) des Sciences de la Santé Simone Veil, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Montigny-le-Bretonneux, France
| | - Anne Nosjean
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France
| | | | | | - Catherine Sebrié
- Imagerie par Résonance Magnétique Médicale et Multimodalité (IR4M) Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8081, Paris-Sud University, Paris-Saclay University, CNRS, Orsay, France
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Jean-Marc Edeline
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France
| | - Sabine de la Porte
- INSERM Unité 1179, Handicap Neuromusculaire: Physiologie, Biothérapie et Pharmacologie Appliquées, Unité de Formation et de Recherche (UFR) des Sciences de la Santé Simone Veil, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Montigny-le-Bretonneux, France
| | - Philippe Fossier
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France
| | - Sylvie Granon
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France
| | - Cyrille Vaillend
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France
| | - José-Manuel Cancela
- Neuroscience Paris-Saclay Institute (Neuro-PSI), Unité Mixte de Recherche (UMR) 9197, Paris-Sud University, Paris-Saclay University, Orsay, France
| |
Collapse
|
45
|
Imbery JF, Iqbal AK, Desai T, Giovannucci DR. Role of NAADP for calcium signaling in the salivary gland. Cell Calcium 2019; 80:29-37. [PMID: 30947088 DOI: 10.1016/j.ceca.2019.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 03/02/2019] [Accepted: 03/03/2019] [Indexed: 11/26/2022]
Abstract
Coordination of intracellular Ca2+ signaling in parotid acini is crucial for controlling the secretion of primary saliva. Previous work from our lab has demonstrated acidic-organelle Ca2+ release as a participant in agonist-evoked signaling dynamics of the parotid acinar cell. Furthermore, results implicated a potential role for the potent Ca2+ releasing second messenger NAADP in these events. The current study interrogated a direct role of NAADP for Ca2+ signaling in the parotid salivary gland acinar cell. Use of live-cell Ca2+ imaging, patch-clamp methods, and confocal microscopy revealed for the first time NAADP can evoke or enhance Ca2+ dynamics in parotid acini. These results were compared with pancreatic acini, a morphologically similar cell type previously shown to display NAADP-dependent Ca2+ signals. Findings presented here may be relevant in establishing new therapeutic targets for those suffering from xerostomia produced by hypofunctioning salivary glands.
Collapse
Affiliation(s)
- John F Imbery
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, 3000 Arlington Ave., Toledo, OH, 43614, United States
| | - Azwar K Iqbal
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, 3000 Arlington Ave., Toledo, OH, 43614, United States
| | - Tanvi Desai
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, 3000 Arlington Ave., Toledo, OH, 43614, United States
| | - David R Giovannucci
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, 3000 Arlington Ave., Toledo, OH, 43614, United States.
| |
Collapse
|
46
|
Yuan Y, Gunaratne GS, Marchant JS, Patel S. Probing Ca 2+ release mechanisms using sea urchin egg homogenates. Methods Cell Biol 2019; 151:445-458. [PMID: 30948025 DOI: 10.1016/bs.mcb.2018.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Sea urchin eggs have been extensively used to study Ca2+ release through intracellular Ca2+-permeable channels. Their amenability to homogenization yields a robust, cell-free preparation that was central to establishing the Ca2+ mobilizing actions of cyclic ADP-ribose and NAADP. Egg homogenates have continued to provide insight into the basic properties and pharmacology of intracellular Ca2+ release channels. In this chapter, we describe methods for the preparation of egg homogenates and monitoring Ca2+ release using fluorimetry and radiotracer flux.
Collapse
Affiliation(s)
- Yu Yuan
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Gihan S Gunaratne
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States
| | - Jonathan S Marchant
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, London, United Kingdom.
| |
Collapse
|
47
|
Foster WJ, Taylor HBC, Padamsey Z, Jeans AF, Galione A, Emptage NJ. Hippocampal mGluR1-dependent long-term potentiation requires NAADP-mediated acidic store Ca 2+ signaling. Sci Signal 2018; 11:11/558/eaat9093. [PMID: 30482851 DOI: 10.1126/scisignal.aat9093] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Acidic organelles, such as endosomes and lysosomes, store Ca2+ that is released in response to intracellular increases in the second messenger nicotinic acid adenine dinucleotide phosphate (NAADP). In neurons, NAADP and Ca2+ signaling contribute to synaptic plasticity, a process of activity-dependent long-term potentiation (LTP) [or, alternatively, long-term depression (LTD)] of synaptic strength and neuronal transmission that is critical for neuronal function and memory formation. We explored the function of and mechanisms regulating acidic Ca2+ store signaling in murine hippocampal neurons. We found that metabotropic glutamate receptor 1 (mGluR1) was coupled to NAADP signaling that elicited Ca2+ release from acidic stores. In turn, this released Ca2+-mediated mGluR1-dependent LTP by transiently inhibiting SK-type K+ channels, possibly through the activation of protein phosphatase 2A. Genetically removing two-pore channels (TPCs), which are endolysosomal-specific ion channels, switched the polarity of plasticity from LTP to LTD, indicating the importance of specific receptor store coupling and providing mechanistic insight into how mGluR1 can produce both synaptic potentiation and synaptic depression.
Collapse
Affiliation(s)
- William J Foster
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.
| | - Henry B C Taylor
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Zahid Padamsey
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Alexander F Jeans
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.
| | - Nigel J Emptage
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.
| |
Collapse
|
48
|
Zuccolo E, Kheder DA, Lim D, Perna A, Nezza FD, Botta L, Scarpellino G, Negri S, Martinotti S, Soda T, Forcaia G, Riboni L, Ranzato E, Sancini G, Ambrosone L, D'Angelo E, Guerra G, Moccia F. Glutamate triggers intracellular Ca 2+ oscillations and nitric oxide release by inducing NAADP- and InsP 3 -dependent Ca 2+ release in mouse brain endothelial cells. J Cell Physiol 2018; 234:3538-3554. [PMID: 30451297 DOI: 10.1002/jcp.26953] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/13/2018] [Indexed: 12/13/2022]
Abstract
The neurotransmitter glutamate increases cerebral blood flow by activating postsynaptic neurons and presynaptic glial cells within the neurovascular unit. Glutamate does so by causing an increase in intracellular Ca2+ concentration ([Ca2+ ]i ) in the target cells, which activates the Ca2+ /Calmodulin-dependent nitric oxide (NO) synthase to release NO. It is unclear whether brain endothelial cells also sense glutamate through an elevation in [Ca2+ ]i and NO production. The current study assessed whether and how glutamate drives Ca2+ -dependent NO release in bEND5 cells, an established model of brain endothelial cells. We found that glutamate induced a dose-dependent oscillatory increase in [Ca2+ ]i , which was maximally activated at 200 μM and inhibited by α-methyl-4-carboxyphenylglycine, a selective blocker of Group 1 metabotropic glutamate receptors. Glutamate-induced intracellular Ca2+ oscillations were triggered by rhythmic endogenous Ca2+ mobilization and maintained over time by extracellular Ca2+ entry. Pharmacological manipulation revealed that glutamate-induced endogenous Ca2+ release was mediated by InsP3 -sensitive receptors and nicotinic acid adenine dinucleotide phosphate (NAADP) gated two-pore channel 1. Constitutive store-operated Ca2+ entry mediated Ca2+ entry during ongoing Ca2+ oscillations. Finally, glutamate evoked a robust, although delayed increase in NO levels, which was blocked by pharmacologically inhibition of the accompanying intracellular Ca2+ signals. Of note, glutamate induced Ca2+ -dependent NO release also in hCMEC/D3 cells, an established model of human brain microvascular endothelial cells. This investigation demonstrates for the first time that metabotropic glutamate-induced intracellular Ca2+ oscillations and NO release have the potential to impact on neurovascular coupling in the brain.
Collapse
Affiliation(s)
- Estella Zuccolo
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani," University of Pavia, Pavia, Italy
| | - Dlzar A Kheder
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani," University of Pavia, Pavia, Italy.,Department of Biology, University of Zakho, Duhok, Kurdistan-Region of Iraq
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, University of Eastern Piedmont "Amedeo Avogadro,", Novara, Italy
| | - Angelica Perna
- Department of Medicine and Health Sciences "Vincenzo Tiberio,", University of Molise, Campobasso, Italy
| | - Francesca Di Nezza
- Department of Bioscience and Territory (DIBT), University of Molise, Contrada Lappone Pesche, Isernia, Italy
| | - Laura Botta
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani," University of Pavia, Pavia, Italy
| | - Giorgia Scarpellino
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani," University of Pavia, Pavia, Italy
| | - Sharon Negri
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani," University of Pavia, Pavia, Italy
| | - Simona Martinotti
- Dipartimento di Scienze e Innovazione Tecnologica (DiSIT), University of Piemonte Orientale, Alessandria, Italy
| | - Teresa Soda
- Museo Storico della Fisica e Centro Studi e Ricerche Enrico Fermi, Rome, Italy.,Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Greta Forcaia
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Laura Riboni
- Department of Medical Biotechnology and Translational Medicine, LITA-Segrate, University of Milan, Segrate, Milan, Italy
| | - Elia Ranzato
- Dipartimento di Scienze e Innovazione Tecnologica (DiSIT), University of Piemonte Orientale, Alessandria, Italy
| | - Giulio Sancini
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Luigi Ambrosone
- Department of Medicine and Health Sciences "Vincenzo Tiberio,", Centre of Nanomedicine, University of Molise, Campobasso, Italy
| | - Egidio D'Angelo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,Brain Connectivity Center, C. Mondino National Neurological Institute, Pavia, Italy
| | - Germano Guerra
- Department of Medicine and Health Sciences "Vincenzo Tiberio,", University of Molise, Campobasso, Italy
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology "Lazzaro Spallanzani," University of Pavia, Pavia, Italy
| |
Collapse
|
49
|
Liu B, Cao W, Li J, Liu J. Lysosomal exocytosis of ATP is coupled to P2Y 2 receptor in marginal cells in the stria vascular in neonatal rats. Cell Calcium 2018; 76:62-71. [PMID: 30273839 DOI: 10.1016/j.ceca.2018.09.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/09/2018] [Accepted: 09/20/2018] [Indexed: 01/06/2023]
Abstract
Adenosine triphosphate (ATP) is stored as lysosomal vesicles in marginal cells of the stria vascular in neonatal rats, but the mechanisms of ATP release are unclear. Primary cultures of marginal cells from 1-day-old Sprague-Dawley rats were established. P2Y2 receptor and inositol 1,4,5-trisphosphate (IP3) receptor were immunolabelled in marginal cells of the stria vascular. We found that 30 μM ATP and 30 μM uridine triphosphate (UTP) evoked comparable significant increases in the intracellular Ca2+ concentration ([Ca2+]i) in the absence of extracellular Ca2+, whereas the response was suppressed by 100 μM suramin, 10 μM 1-(6-(17β-3-methoxyester-1,3,5(10)-trien-17-yl)amino)-hexyl)-1H-pyrrole-2,5-dione(U-73122), 100 μM 2-aminoethoxydiphenyl borate (2-APB) and 5 μM thapsigargin (TG), thus indicating that ATP coupled with the P2Y2R-PLC-IP3 pathway to evoke Ca2+ release from the endoplasmic reticulum (ER). Incubation with 200 μM Gly-Phe-β-naphthylamide (GPN) selectively disrupted lysosomes and caused significant increases in [Ca2+]I; this effect was partly inhibited by P2Y2R-PLC-IP3 pathway antagonists. After pre-treatment with 5 μM TG, [Ca2+]i was significantly lower than that after treatment with P2Y2R-PLC-IP3 pathway antagonists under the same conditions, thus indicating that lysosomal Ca2+ triggers Ca2+ release from ER Ca2+ stores. Baseline [Ca2+]i declined after treatment with the Ca2+ chelator 50 μM bis-(aminophenolxy) ethane-N,N,N',N'-tetra-acetic acid acetoxyme-thyl ester (BAPTA-AM) and 4 IU/ml apyrase. 30 μM ATP decrease of the number of quinacrine-positive vesicles via lysosome exocytosis, whereas the number of lysosomes did not change. However, lysosome exocytosis was significantly suppressed by pre-treatment with 5 μM vacuolin-1. Release of ATP and β-hexosaminidase both increased after treatment with 200 μM GPN and 5 μM TG, but decreased after incubation with 50 μM BAPTA-AM, 4 IU/ml apyrase and 5 μM vacuolin-1. We suggest that ATP triggers Ca2+ release from the ER, thereby contributing to secretion of lysosomal ATP via lysosomal exocytosis. Lysosomal stored Ca2+ triggers Ca2+ release from the ER directly though the IP3 receptors, and lysosomal ATP evokes Ca2+ signals indirectly via the P2Y2R-PLC-IP3 pathway.
Collapse
Affiliation(s)
- Bin Liu
- Department of Otorhinolaryngology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wanxin Cao
- Department of Otorhinolaryngology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jiping Li
- Department of Otorhinolaryngology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Jun Liu
- Department of Otorhinolaryngology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
50
|
Roest G, La Rovere RM, Bultynck G, Parys JB. IP 3 Receptor Properties and Function at Membrane Contact Sites. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 981:149-178. [PMID: 29594861 DOI: 10.1007/978-3-319-55858-5_7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) is a ubiquitously expressed Ca2+-release channel localized in the endoplasmic reticulum (ER). The intracellular Ca2+ signals originating from the activation of the IP3R regulate multiple cellular processes including the control of cell death versus cell survival via their action on apoptosis and autophagy. The exact role of the IP3Rs in these two processes does not only depend on their activity, which is modulated by the cytosolic composition (Ca2+, ATP, redox status, …) and by various types of regulatory proteins, including kinases and phosphatases as well as by a number of oncogenes and tumor suppressors, but also on their intracellular localization, especially at the ER-mitochondrial and ER-lysosomal interfaces. At these interfaces, Ca2+ microdomains are formed, in which the Ca2+ concentration is finely regulated by the different ER, mitochondrial and lysosomal Ca2+-transport systems and also depends on the functional and structural interactions existing between them. In this review, we therefore discuss the most recent insights in the role of Ca2+ signaling in general, and of the IP3R in particular, in the control of basal mitochondrial bioenergetics, apoptosis, and autophagy at the level of inter-organellar contact sites.
Collapse
Affiliation(s)
- Gemma Roest
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Leuven, Belgium
| | - Rita M La Rovere
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Leuven, Belgium
| | - Geert Bultynck
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Leuven, Belgium.
| | - Jan B Parys
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Leuven, Belgium.
| |
Collapse
|