1
|
Klingl YE, Petrauskas A, Jaślan D, Grimm C. TPCs: FROM PLANT TO HUMAN. Physiol Rev 2025; 105:1695-1732. [PMID: 40197126 DOI: 10.1152/physrev.00044.2024] [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: 10/30/2024] [Revised: 12/15/2024] [Accepted: 03/08/2025] [Indexed: 04/05/2025] Open
Abstract
In 2005, the Arabidopsis thaliana two-pore channel TPC1 channel was identified as a vacuolar Ca2+-release channel. In 2009, three independent groups published studies on mammalian TPCs as nicotinic acid adenine dinucleotide phosphate (NAADP)-activated endolysosomal Ca2+ release channels, results that were eventually challenged by two other groups, claiming mammalian TPCs to be phosphatidylinositol-3,5-bisphosphate [PI(3,5)P2]-activated Na+ channels. By now this dispute seems to have been largely reconciled. Lipophilic small molecule agonists of TPC2, mimicking either the NAADP or the PI(3,5)P2 mode of channel activation, revealed, together with structural evidence, that TPC2 can change its selectivity for Ca2+ versus Na+ in a ligand-dependent fashion (N- vs. P-type activation). Furthermore, the NAADP-binding proteins Jupiter microtubule-associated homolog 2 protein (JPT2) and Lsm12 were discovered, corroborating the hypothesis that NAADP activation of TPCs only works in the presence of these auxiliary NAADP-binding proteins. Pathophysiologically, loss or gain of function of TPCs has effects on autophagy, exocytosis, endocytosis, and intracellular trafficking, e.g., LDL cholesterol trafficking leading to fatty liver disease or viral and bacterial toxin trafficking, corroborating the roles of TPCs in infectious diseases such as Ebola or COVID-19. Defects in the trafficking of epidermal growth factor receptor and β1-integrin suggested roles in cancer. In neurodegenerative lysosomal storage disease models, P-type activation of TPC2 was found to have beneficial effects on both in vitro and in vivo hallmarks of Niemann-Pick disease type C1, Batten disease, and mucolipidosis type IV. Here, we cover the latest on the structure, function, physiology, and pathophysiology of these channels with a focus initially on plants followed by mammalian TPCs, and we discuss their potential as drug targets, including currently available pharmacology.
Collapse
Affiliation(s)
- Yvonne Eileen Klingl
- Walther-Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University Munich, Munich, Germany
- Immunology, Infection and Pandemic Research, Fraunhofer Institute for Translational Medicine and Pharmacology, Munich, Germany
| | - Arnas Petrauskas
- Walther-Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University Munich, Munich, Germany
- Immunology, Infection and Pandemic Research, Fraunhofer Institute for Translational Medicine and Pharmacology, Munich, Germany
| | - Dawid Jaślan
- Walther-Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University Munich, Munich, Germany
| | - Christian Grimm
- Walther-Straub Institute of Pharmacology and Toxicology, Ludwig Maximilian University Munich, Munich, Germany
- Immunology, Infection and Pandemic Research, Fraunhofer Institute for Translational Medicine and Pharmacology, Munich, Germany
- Department of Pharmacology, Faculty of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
2
|
Hu M, Feng X, Liu Q, Liu S, Huang F, Xu H. The ion channels of endomembranes. Physiol Rev 2024; 104:1335-1385. [PMID: 38451235 PMCID: PMC11381013 DOI: 10.1152/physrev.00025.2023] [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: 06/30/2023] [Revised: 02/20/2024] [Accepted: 02/25/2024] [Indexed: 03/08/2024] Open
Abstract
The endomembrane system consists of organellar membranes in the biosynthetic pathway [endoplasmic reticulum (ER), Golgi apparatus, and secretory vesicles] as well as those in the degradative pathway (early endosomes, macropinosomes, phagosomes, autophagosomes, late endosomes, and lysosomes). These endomembrane organelles/vesicles work together to synthesize, modify, package, transport, and degrade proteins, carbohydrates, and lipids, regulating the balance between cellular anabolism and catabolism. Large ion concentration gradients exist across endomembranes: Ca2+ gradients for most endomembrane organelles and H+ gradients for the acidic compartments. Ion (Na+, K+, H+, Ca2+, and Cl-) channels on the organellar membranes control ion flux in response to cellular cues, allowing rapid informational exchange between the cytosol and organelle lumen. Recent advances in organelle proteomics, organellar electrophysiology, and luminal and juxtaorganellar ion imaging have led to molecular identification and functional characterization of about two dozen endomembrane ion channels. For example, whereas IP3R1-3 channels mediate Ca2+ release from the ER in response to neurotransmitter and hormone stimulation, TRPML1-3 and TMEM175 channels mediate lysosomal Ca2+ and H+ release, respectively, in response to nutritional and trafficking cues. This review aims to summarize the current understanding of these endomembrane channels, with a focus on their subcellular localizations, ion permeation properties, gating mechanisms, cell biological functions, and disease relevance.
Collapse
Affiliation(s)
- Meiqin Hu
- Department of Neurology and Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Xinghua Feng
- Department of Neurology and Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Qiang Liu
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Siyu Liu
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Fangqian Huang
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Haoxing Xu
- Department of Neurology and Department of Cardiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- New Cornerstone Science Laboratory, Liangzhu Laboratory and School of Basic Medical Sciences, Zhejiang University, Hangzhou, People's Republic of China
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
3
|
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: 1.0] [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
|
4
|
Riederer E, Cang C, Ren D. Lysosomal Ion Channels: What Are They Good For and Are They Druggable Targets? Annu Rev Pharmacol Toxicol 2023; 63:19-41. [PMID: 36151054 DOI: 10.1146/annurev-pharmtox-051921-013755] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Lysosomes play fundamental roles in material digestion, cellular clearance, recycling, exocytosis, wound repair, Ca2+ signaling, nutrient signaling, and gene expression regulation. The organelle also serves as a hub for important signaling networks involving the mTOR and AKT kinases. Electrophysiological recording and molecular and structural studies in the past decade have uncovered several unique lysosomal ion channels and transporters, including TPCs, TMEM175, TRPMLs, CLN7, and CLC-7. They underlie the organelle's permeability to major ions, including K+, Na+, H+, Ca2+, and Cl-. The channels are regulated by numerous cellular factors, ranging from H+ in the lumen and voltage across the lysosomal membrane to ATP in the cytosol to growth factors outside the cell. Genetic variations in the channel/transporter genes are associated with diseases that include lysosomal storage diseases and neurodegenerative diseases. Recent studies with human genetics and channel activators suggest that lysosomal channels may be attractive targets for the development of therapeutics for the prevention of and intervention in human diseases.
Collapse
Affiliation(s)
- Erika Riederer
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA; ,
| | - Chunlei Cang
- CAS Key Laboratory of Innate Immunity and Chronic Disease, Neurodegenerative Disorder Research Center, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China;
| | - Dejian Ren
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA; ,
| |
Collapse
|
5
|
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
|
6
|
Du C, Guan X, Yan J. Two-pore channel blockade by phosphoinositide kinase inhibitors YM201636 and PI-103 determined by a histidine residue near pore-entrance. Commun Biol 2022; 5:738. [PMID: 35871252 PMCID: PMC9308409 DOI: 10.1038/s42003-022-03701-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 07/11/2022] [Indexed: 12/04/2022] Open
Abstract
Human two-pore channels (TPCs) are endolysosomal cation channels and play an important role in NAADP-evoked Ca2+ release and endomembrane dynamics. We found that YM201636, a PIKfyve inhibitor, potently inhibits PI(3,5)P2-activated human TPC2 with an IC50 of 0.16 μM. YM201636 also effectively inhibits NAADP-activated TPC2 and a constitutively-open TPC2 L690A/L694A mutant channel; whereas it exerts little effect when applied in the channel’s closed state. PI-103, a YM201636 analog and an inhibitor of PI3K and mTOR, also inhibits human TPC2 with an IC50 of 0.64 μM. With mutational, virtual docking, and molecular dynamic simulation analyses, we found that YM201636 and PI-103 directly block the TPC2’s open-state channel pore at the bundle-cross pore-gate region where a nearby H699 residue is a key determinant for channel’s sensitivity to the inhibitors. H699 likely interacts with the blockers around the pore entrance and facilitates their access to the pore. Substitution of a Phe for H699 largely accounts for the TPC1 channel’s insensitivity to YM201636. These findings identify two potent TPC2 channel blockers, reveal a channel pore entrance blockade mechanism, and provide an ion channel target in interpreting the pharmacological effects of two commonly used phosphoinositide kinase inhibitors. YM201636 and PI-103 are potent inhibitors of human two-pore channel 2 that act through a channel pore entrance blockade mechanism.
Collapse
|
7
|
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: 1.0] [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
|
8
|
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: 0.7] [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]
|
9
|
Chen L, Hassani Nia F, Stauber T. Ion Channels and Transporters in Muscle Cell Differentiation. Int J Mol Sci 2021; 22:13615. [PMID: 34948411 PMCID: PMC8703453 DOI: 10.3390/ijms222413615] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/04/2021] [Accepted: 12/14/2021] [Indexed: 01/12/2023] Open
Abstract
Investigations on ion channels in muscle tissues have mainly focused on physiological muscle function and related disorders, but emerging evidence supports a critical role of ion channels and transporters in developmental processes, such as controlling the myogenic commitment of stem cells. In this review, we provide an overview of ion channels and transporters that influence skeletal muscle myoblast differentiation, cardiac differentiation from pluripotent stem cells, as well as vascular smooth muscle cell differentiation. We highlight examples of model organisms or patients with mutations in ion channels. Furthermore, a potential underlying molecular mechanism involving hyperpolarization of the resting membrane potential and a series of calcium signaling is discussed.
Collapse
Affiliation(s)
- Lingye Chen
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany;
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Fatemeh Hassani Nia
- Institute for Molecular Medicine, MSH Medical School Hamburg, 20457 Hamburg, Germany;
| | - Tobias Stauber
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany;
- Institute for Molecular Medicine, MSH Medical School Hamburg, 20457 Hamburg, Germany;
| |
Collapse
|
10
|
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: 54] [Impact Index Per Article: 13.5] [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
|
11
|
Hu W, Zhao F, Chen L, Ni J, Jiang Y. NAADP-induced intracellular calcium ion is mediated by the TPCs (two-pore channels) in hypoxia-induced pulmonary arterial hypertension. J Cell Mol Med 2021; 25:7485-7499. [PMID: 34263977 PMCID: PMC8335677 DOI: 10.1111/jcmm.16783] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 06/21/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a form of obstructive vascular disease. Chronic hypoxic exposure leads to excessive proliferation of pulmonary arterial smooth muscle cells and pulmonary arterial endothelial cells. This condition can potentially be aggravated by [Ca2+] i mobilization. In the present study, hypoxia exposure of rat's model was established. Two‐pore segment channels (TPCs) silencing was achieved in rats' models by injecting Lsh‐TPC1 or Lsh‐TPC2. The effects of TPC1/2 silencing on PAH were evaluated by H&E staining detecting pulmonary artery wall thickness and ELISA assay kit detecting NAADP concentrations in lung tissues. TPC1/2 silencing was achieved in PASMCs and PAECs, and cell proliferation was detected by MTT and BrdU incorporation assays. As the results shown, NAADP‐activated [Ca2+]i shows to be mediated via two‐pore segment channels (TPCs) in PASMCs, with TPC1 being the dominant subtype. NAADP generation and TPC1/2 mRNA and protein levels were elevated in the hypoxia‐induced rat PAH model; NAADP was positively correlated with TPC1 and TPC2 expression, respectively. In vivo, Lsh‐TPC1 or Lsh‐TPC2 infection significantly improved the mean pulmonary artery pressure and PAH morphology. In vitro, TPC1 silencing inhibited NAADP‐AM‐induced PASMC proliferation and [Ca2+]i in PASMCs, whereas TPC2 silencing had minor effects during this process; TPC2 silencing attenuated NAADP‐AM‐ induced [Ca2+]i and ECM in endothelial cells, whereas TPC1 silencing barely ensued any physiological changes. In conclusion, TPC1/2 might provide a unifying mechanism within pulmonary arterial hypertension, which can potentially be regarded as a therapeutic target.
Collapse
Affiliation(s)
- Wen Hu
- Respiratory Medicine, Hunan Provincial People's Hospital, Changsha, China
| | - Fei Zhao
- Respiratory Medicine, Hunan Provincial People's Hospital, Changsha, China
| | - Ling Chen
- Respiratory Medicine, Hunan Provincial People's Hospital, Changsha, China
| | - Jiamin Ni
- Respiratory Medicine, Hunan Provincial People's Hospital, Changsha, China
| | - Yongliang Jiang
- Respiratory Medicine, Hunan Provincial People's Hospital, Changsha, China
| |
Collapse
|
12
|
A comparative analysis of the intrauterine transcriptome in fertile and subfertile mares using cytobrush sampling. BMC Genomics 2021; 22:377. [PMID: 34022808 PMCID: PMC8141133 DOI: 10.1186/s12864-021-07701-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 05/11/2021] [Indexed: 12/13/2022] Open
Abstract
Background Subfertility is a major problem in modern horse breeding. Especially, mares without clinical signs of reproductive diseases, without known uterine pathogens and no evidence of inflammation but not becoming pregnant after several breeding attempts are challenging for veterinarians. To obtain new insights into the cause of these fertility problems and aiming at improving diagnosis of subfertile mares, a comparative analysis of the intrauterine transcriptome in subfertile and fertile mares was performed. Uterine cytobrush samples were collected during estrus from 57 mares without clinical signs of uterine diseases. RNA was extracted from the cytobrush samples and samples from 11 selected subfertile and 11 fertile mares were used for Illumina RNA-sequencing. Results The cytobrush sampling was a suitable technique to isolate enough RNA of high quality for transcriptome analysis. Comparing subfertile and fertile mares, 114 differentially expressed genes (FDR = 10%) were identified. Metascape enrichment analysis revealed that genes with lower mRNA levels in subfertile mares were related to ‘extracellular matrix (ECM)’, ‘ECM-receptor interaction’, ‘focal adhesion’, ‘immune response’ and ‘cytosolic calcium ion concentration’, while DEGs with higher levels in subfertile mares were enriched for ‘monocarboxyl acid transmembrane transport activity’ and ‘protein targeting’. Conclusion Our study revealed significant differences in the uterine transcriptome between fertile and subfertile mares and provides leads for potential uterine molecular biomarkers of subfertility in the mare. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07701-3.
Collapse
|
13
|
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: 4.5] [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
|
14
|
Two-pore and TRPML cation channels: Regulators of phagocytosis, autophagy and lysosomal exocytosis. Pharmacol Ther 2020; 220:107713. [PMID: 33141027 DOI: 10.1016/j.pharmthera.2020.107713] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023]
Abstract
The old Greek saying "Panta Rhei" ("everything flows") is true for all life and all living things in general. It also becomes nicely evident when looking closely into cells. There, material from the extracellular space is taken up by endocytic processes and transported to endosomes where it is sorted either for recycling or degradation. Cargo is also packaged for export through exocytosis involving the Golgi network, lysosomes and other organelles. Everything in this system is in constant motion and many proteins are necessary to coordinate transport along the different intracellular pathways to avoid chaos. Among these proteins are ion channels., in particular TRPML channels (mucolipins) and two-pore channels (TPCs) which reside on endosomal and lysosomal membranes to speed up movement between organelles, e.g. by regulating fusion and fission; they help readjust pH and osmolarity changes due to such processes, or they promote exocytosis of export material. Pathophysiologically, these channels are involved in neurodegenerative, metabolic, retinal and infectious diseases, cancer, pigmentation defects, and immune cell function, and thus have been proposed as novel pharmacological targets, e.g. for the treatment of lysosomal storage disorders, Duchenne muscular dystrophy, or different types of cancer. Here, we discuss the similarities but also differences of TPCs and TRPMLs in regulating phagocytosis, autophagy and lysosomal exocytosis, and we address the contradictions and open questions in the field relating to the roles TPCs and TRPMLs play in these different processes.
Collapse
|
15
|
Wiriyasermkul P, Moriyama S, Nagamori S. Membrane transport proteins in melanosomes: Regulation of ions for pigmentation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183318. [PMID: 32333855 PMCID: PMC7175901 DOI: 10.1016/j.bbamem.2020.183318] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/17/2020] [Accepted: 04/18/2020] [Indexed: 12/12/2022]
Abstract
Melanosomes are unique organelles in melanocytes that produce melanin, the pigment for skin, hair, and eye color. Tyrosinase is the essential and rate-limiting enzyme for melanin production, that strictly requires neutral pH for activity. pH maintenance is a result of the combinational function of multiple ion transport proteins. Thus, ion homeostasis in melanosomes is crucial for melanin synthesis. Defect of the ion transport system causes various pigmentation phenotypes, from mild effect to severe disorders such as albinism. In this review, we summarize the up-to-date knowledge of the ion transport system, such as transport function, structure, and the physiological roles and mechanisms of the ion transport proteins in melanosomes. In addition, we propose a model of melanosomal ion transport system-how the functional coupling of multiple transport proteins modulates and maintains ion homeostasis. We discuss melanin synthesis in terms of the ion transport system.
Collapse
Affiliation(s)
- Pattama Wiriyasermkul
- Department of Collaborative Research for Bio-Molecular Dynamics, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan
| | - Satomi Moriyama
- Department of Collaborative Research for Bio-Molecular Dynamics, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan
| | - Shushi Nagamori
- Department of Collaborative Research for Bio-Molecular Dynamics, Nara Medical University, 840 Shijo-cho, Kashihara, Nara 634-8521, Japan.
| |
Collapse
|
16
|
Nanoparticle-Mediated Therapeutic Application for Modulation of Lysosomal Ion Channels and Functions. Pharmaceutics 2020; 12:pharmaceutics12030217. [PMID: 32131531 PMCID: PMC7150957 DOI: 10.3390/pharmaceutics12030217] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 02/07/2023] Open
Abstract
Applications of nanoparticles in various fields have been addressed. Nanomaterials serve as carriers for transporting conventional drugs or proteins through lysosomes to various cellular targets. The basic function of lysosomes is to trigger degradation of proteins and lipids. Understanding of lysosomal functions is essential for enhancing the efficacy of nanoparticles-mediated therapy and reducing the malfunctions of cellular metabolism. The lysosomal function is modulated by the movement of ions through various ion channels. Thus, in this review, we have focused on the recruited ion channels for lysosomal function, to understand the lysosomal modulation through the nanoparticles and its applications. In the future, lysosomal channels-based targets will expand the therapeutic application of nanoparticles-associated drugs.
Collapse
|
17
|
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: 2.4] [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
|
18
|
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
|
19
|
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: 13] [Impact Index Per Article: 2.2] [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
|
20
|
The Role of Two-Pore Channels in Norepinephrine-Induced [Ca 2+] i Rise in Rat Aortic Smooth Muscle Cells and Aorta Contraction. Cells 2019; 8:cells8101144. [PMID: 31557916 PMCID: PMC6829401 DOI: 10.3390/cells8101144] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/21/2019] [Accepted: 09/23/2019] [Indexed: 12/17/2022] Open
Abstract
Second messenger nicotinic acid adenine dinucleotide phosphate (NAADP) triggers Ca2+ release via two-pore channels (TPCs) localized in endolysosomal vesicles. The aim of the present work is to evaluate the role of TPCs in the action of norepinephrine (NE), angiotensin II (AngII), vasopressin (AVP), and 5-hydroxytriptamine (5-HT) on free cytoplasmic calcium concentration ([Ca2+]i) in smooth muscle cells (SMCs) isolated from rat aorta and on aorta contraction. To address this issue, the NAADP structural analogue and inhibitor of TPCs, NED 19, was applied. We have demonstrated a high degree of colocalization of the fluorescent signals of cis-NED 19 and endolysosmal probe LysoTracker in SMCs. Both cis- or trans-NED 19 inhibited the rise of [Ca2+]i in SMCs induced by 100 μM NE by 50–60%. IC50 for cis- and trans-NED 19 were 2.7 and 8.9 μM, respectively. The inhibition by NED 19 stereoisomers of the effects of AngII, AVP, and 5-HT was much weaker. Both forms of NED 19 caused relaxation of aortic rings preconstricted by NE, with relative potency of cis-NED 19 several times higher than that of trans-NED 19. Inhibition by cis-NED 19 of NE-induced contraction was maintained after intensive washing and slowly reversed within an hour of incubation. Cis- and trans-NED 19 did not cause decrease in the force of aorta contraction in response to Ang II and AVP, and only slightly relaxed aorta preconstricted by 5-HT and by KCl. Suppression of TPC1 in SMCs with siRNA caused a 40% decrease in [Ca2+]i in response to NE, whereas siRNA against TPC2 did not change NE calcium signaling. These data suggest that TPC1 is involved in the NE-stimulated [Ca2+]i rise in SMCs. Inhibition of TPC1 activity by NED 19 could be the reason for partial inhibition of aortic rings contraction in response to NE.
Collapse
|
21
|
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: 1.2] [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
|
22
|
Endolysosomal Ca 2+ Signalling and Cancer Hallmarks: Two-Pore Channels on the Move, TRPML1 Lags Behind! Cancers (Basel) 2018; 11:cancers11010027. [PMID: 30591696 PMCID: PMC6356888 DOI: 10.3390/cancers11010027] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 12/21/2018] [Accepted: 12/21/2018] [Indexed: 12/22/2022] Open
Abstract
The acidic vesicles of the endolysosomal (EL) system are emerging as an intracellular Ca2+ store implicated in the regulation of multiple cellular functions. The EL Ca2+ store releases Ca2+ through a variety of Ca2+-permeable channels, including Transient Receptor Potential (TRP) Mucolipin 1-3 (TRPML1-3) and two-pore channels 1-2 (TPC1-2), whereas EL Ca2+ refilling is sustained by the proton gradient across the EL membrane and/or by the endoplasmic reticulum (ER). EL Ca2+ signals may be either spatially restricted to control vesicle trafficking, autophagy and membrane repair or may be amplified into a global Ca2+ signal through the Ca2+-dependent recruitment of ER-embedded channels. Emerging evidence suggested that nicotinic acid adenine dinucleotide phosphate (NAADP)-gated TPCs sustain multiple cancer hallmarks, such as migration, invasiveness and angiogenesis. Herein, we first survey the EL Ca2+ refilling and release mechanisms and then focus on the oncogenic role of EL Ca2+ signaling. While the evidence in favor of TRPML1 involvement in neoplastic transformation is yet to be clearly provided, TPCs are emerging as an alternative target for anticancer therapies.
Collapse
|
23
|
Sun W, Yue J. TPC2 mediates autophagy progression and extracellular vesicle secretion in cancer cells. Exp Cell Res 2018; 370:478-489. [PMID: 29990474 DOI: 10.1016/j.yexcr.2018.07.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/04/2018] [Accepted: 07/06/2018] [Indexed: 02/06/2023]
Abstract
Autophagy is an evolutionarily conserved lysosomal degradation process, and is involved in various cellular processes. Here we studied the role of two pore channel 2 (TPC2), a lysosomal non-selective Na+/Ca2+ channel, in autophagy progression. We found that TPC overexpression in 4T1 mouse breast cancer cell line or in HeLa human cervical cancer cell line inhibited the fusion between autophagosome and lysosome, resulting in the accumulation of autophagosomes accompanied with increased lysosomal pH and TFEB nuclear localization. Interestingly, we also found that extracellular vesicle (EV) secretion was markedly decreased in TPC2 overexpressing cells but was induced in TPC2 knockdown cells. In addition, migration of TPC2 knockdown cells, not TPC2 overexpressing cells, was inhibited. Taken together, these results support a role of TPC2 in autophagy progression and EV trafficking in cancer cells.
Collapse
Affiliation(s)
- Wei Sun
- City University of Hong Kong ShenZhen Research Institute, ShenZhen, China; Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Jianbo Yue
- City University of Hong Kong ShenZhen Research Institute, ShenZhen, China; Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
24
|
Kelu JJ, Webb SE, Parrington J, Galione A, Miller AL. Ca 2+ release via two-pore channel type 2 (TPC2) is required for slow muscle cell myofibrillogenesis and myotomal patterning in intact zebrafish embryos. Dev Biol 2017; 425:109-129. [PMID: 28390800 DOI: 10.1016/j.ydbio.2017.03.031] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/30/2017] [Accepted: 03/31/2017] [Indexed: 01/14/2023]
Abstract
We recently demonstrated a critical role for two-pore channel type 2 (TPC2)-mediated Ca2+ release during the differentiation of slow (skeletal) muscle cells (SMC) in intact zebrafish embryos, via the introduction of a translational-blocking morpholino antisense oligonucleotide (MO). Here, we extend our study and demonstrate that knockdown of TPC2 with a non-overlapping splice-blocking MO, knockout of TPC2 (via the generation of a tpcn2dhkz1a mutant line of zebrafish using CRISPR/Cas9 gene-editing), or the pharmacological inhibition of TPC2 action with bafilomycin A1 or trans-ned-19, also lead to a significant attenuation of SMC differentiation, characterized by a disruption of SMC myofibrillogenesis and gross morphological changes in the trunk musculature. When the morphants were injected with tpcn2-mRNA or were treated with IP3/BM or caffeine (agonists of the inositol 1,4,5-trisphosphate receptor (IP3R) and ryanodine receptor (RyR), respectively), many aspects of myofibrillogenesis and myotomal patterning (and in the case of the pharmacological treatments, the Ca2+ signals generated in the SMCs), were rescued. STED super-resolution microscopy revealed a close physical relationship between clusters of RyR in the terminal cisternae of the sarcoplasmic reticulum (SR), and TPC2 in lysosomes, with a mean estimated separation of ~52-87nm. Our data therefore add to the increasing body of evidence, which indicate that localized Ca2+ release via TPC2 might trigger the generation of more global Ca2+ release from the SR via Ca2+-induced Ca2+ release.
Collapse
MESH Headings
- Animals
- Base Sequence
- Behavior, Animal/drug effects
- Body Patterning/drug effects
- CRISPR-Cas Systems/genetics
- Caffeine/pharmacology
- Calcium/metabolism
- Calcium Channels/metabolism
- Calcium Signaling/drug effects
- Cell Death/drug effects
- Cells, Cultured
- Embryo, Nonmammalian/drug effects
- Embryo, Nonmammalian/metabolism
- Gene Knockdown Techniques
- Gene Knockout Techniques
- Inositol 1,4,5-Trisphosphate Receptors/metabolism
- Kinesins/metabolism
- Macrolides/pharmacology
- Models, Biological
- Morpholinos/pharmacology
- Motor Activity/drug effects
- Muscle Cells/cytology
- Muscle Cells/drug effects
- Muscle Cells/metabolism
- Muscle Development/drug effects
- Muscle Fibers, Slow-Twitch/cytology
- Muscle Fibers, Slow-Twitch/drug effects
- Muscle Fibers, Slow-Twitch/metabolism
- Phenotype
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Ryanodine Receptor Calcium Release Channel/metabolism
- Sarcomeres/drug effects
- Sarcomeres/metabolism
- Zebrafish/embryology
- Zebrafish/metabolism
- Zebrafish Proteins/metabolism
Collapse
Affiliation(s)
- Jeffrey J Kelu
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, HKUST, Clear Water Bay, Hong Kong, PR China
| | - Sarah E Webb
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, HKUST, Clear Water Bay, Hong Kong, PR China
| | - John Parrington
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, UK
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, UK
| | - Andrew L Miller
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, HKUST, Clear Water Bay, Hong Kong, PR China; Marine Biological Laboratory, Woods Hole, MA, USA.
| |
Collapse
|
25
|
García-Rúa V, Feijóo-Bandín S, García-Vence M, Aragón-Herrera A, Bravo SB, Rodríguez-Penas D, Mosquera-Leal A, Lear PV, Parrington J, Alonso J, Roselló-Lletí E, Portolés M, Rivera M, González-Juanatey JR, Lago F. Metabolic alterations derived from absence of Two-Pore Channel 1 at cardiac level. J Biosci 2016; 41:643-658. [DOI: 10.1007/s12038-016-9647-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
26
|
Kelu JJ, Chan HLH, Webb SE, Cheng AHH, Ruas M, Parrington J, Galione A, Miller AL. Two-Pore Channel 2 activity is required for slow muscle cell-generated Ca(2+) signaling during myogenesis in intact zebrafish. THE INTERNATIONAL JOURNAL OF DEVELOPMENTAL BIOLOGY 2016; 59:313-25. [PMID: 26679948 DOI: 10.1387/ijdb.150206am] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
We have recently characterized essential inositol 1,4,5-trisphosphate receptor (IP 3R) and ryanodine receptor (RyR)-mediated Ca(2+) signals generated during the differentiation of slow muscle cells (SMCs) in intact zebrafish embryos. Here, we show that the lysosomal two-pore channel 2 (TPC2) also plays a crucial role in generating, and perhaps triggering, these essential Ca(2+) signals, and thus contributes to the regulation of skeletal muscle myogenesis. We used a transgenic line of zebrafish that expresses the bioluminescent Ca(2+) reporter, aequorin, specifically in skeletal muscle, in conjunction with morpholino (MO)-based and pharmacological inhibition of TPC2, in both intact embryos and isolated SMCs. MO-based knock-down of TPC2 resulted in a dramatic attenuation of the Ca(2+) signals, whereas the introduction of TPCN2-MO and TPCN2 mRNA together partially rescued the Ca(2+) signaling signature. Embryos treated with trans-ned-19 or bafilomycin A1, a specific NAADP receptor inhibitor and vacuolar-type H(+)ATPase inhibitor, respectively, also displayed a similar disruption of SMC Ca(2+) signaling. TPC2 and lysosomes were shown via immunohistochemistry and confocal laser scanning microscopy to be localized in perinuclear and striated cytoplasmic domains of SMCs, coincident with patterns of IP 3R and RyR expression. These data together imply that TPC2-mediated Ca(2+) release from lysosomes acts upstream from RyR- and IP 3R-mediated Ca(2+) release, suggesting that the former might act as a sensitive trigger to initiate the SR-mediated Ca(2+)-induced-Ca(2+)-release essential for SMC myogenesis and function.
Collapse
Affiliation(s)
- Jeffrey J Kelu
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, HKUST, Clear Water Bay, Hong Kong, PRC
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Brailoiu GC, Brailoiu E. Modulation of Calcium Entry by the Endo-lysosomal System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:423-47. [PMID: 27161239 DOI: 10.1007/978-3-319-26974-0_18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Endo-lysosomes are acidic organelles that besides the role in macromolecules degradation, act as intracellular Ca(2+) stores. Nicotinic acid adenine dinucleotide phosphate (NAADP), the most potent Ca(2+)-mobilizing second messenger, produced in response to agonist stimulation, activates Ca(2+)-releasing channels on endo-lysosomes and modulates a variety of cellular functions. NAADP-evoked signals are amplified by Ca(2+) release from endoplasmic reticulum, via the recruitment of inositol 1,4,5-trisphosphate and/or ryanodine receptors through a Ca(2+)-induced Ca(2+)- release (CICR) mechanism. The endo-lysosomal Ca(2+) channels activated by NAADP were recently identified as the two-pore channels (TPCs). In addition to TPCs, endo-lysosomes express another distinct family of Ca(2+)- permeable channels, namely the transient receptor potential mucolipin (TRPML) channels, functionally distinct from TPCs. TPCs belong to the voltage-gated channels, resembling voltage-gated Na(+) and Ca(2+) channels. TPCs have important roles in vesicular fusion and trafficking, in triggering a global Ca(2+) signal and in modulation of the membrane excitability. Depletion of acidic Ca(2+) stores has been shown to activate store-operated Ca(2+) entry in human platelets and mouse pancreatic β-cells. In human platelets, Ca(2+) influx in response to acidic stores depletion is facilitated by the tubulin-cytoskeleton and occurs through non-selective cation channels and transient receptor potential canonical (TRPC) channels. Emerging evidence indicates that activation of intracellular receptors, situated on endo-lysosomes, elicits canonical and non-canonical signaling mechanisms that involve CICR and activation of non-selective cation channels in plasma membrane. The ability of endo-lysosomal Ca(2+) stores to modulate the Ca(2+) release from other organelles and the Ca(2+) entry increases the diversity and complexity of cellular signaling mechanisms.
Collapse
Affiliation(s)
- G Cristina Brailoiu
- Department of Pharmaceutical Sciences, Jefferson School of Pharmacy, Thomas Jefferson University, 901 Walnut St, Rm 916, Philadelphia, PA, 19107, USA.
| | - Eugen Brailoiu
- Center for Substance Abuse Research, Temple University School of Medicine, 3500 N. Broad Street, Room 848, Philadelphia, PA, 19140, USA
| |
Collapse
|
28
|
Feijóo-Bandín S, García-Vence M, García-Rúa V, Roselló-Lletí E, Portolés M, Rivera M, González-Juanatey JR, Lago F. Two-pore channels (TPCs): Novel voltage-gated ion channels with pleiotropic functions. Channels (Austin) 2016; 11:20-33. [PMID: 27440385 DOI: 10.1080/19336950.2016.1213929] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Two-pore channels (TPC1-3) comprise a subfamily of the eukaryotic voltage-gated ion channels (VGICs) superfamily that are mainly expressed in acidic stores in plants and animals. TPCS are widespread across the animal kingdom, with primates, mice and rats lacking TPC3, and mainly act as Ca+ and Na+ channels, although it was also suggested that they could be permeable to other ions. Nowadays, TPCs have been related to the development of different diseases, including Parkinson´s disease, obesity or myocardial ischemia. Due to this, their study has raised the interest of the scientific community to try to understand their mechanism of action in order to be able to develop an efficient drug that could regulate TPCs activity. In this review, we will provide an updated view regarding TPCs structure, function and activation, as well as their role in different pathophysiological processes.
Collapse
Affiliation(s)
- Sandra Feijóo-Bandín
- a Cellular and Molecular Cardiology Research Unit and Department of Cardiology , Institute of Biomedical Research and University Clinical Hospital , Santiago de Compostela , Spain
| | - María García-Vence
- a Cellular and Molecular Cardiology Research Unit and Department of Cardiology , Institute of Biomedical Research and University Clinical Hospital , Santiago de Compostela , Spain
| | - Vanessa García-Rúa
- a Cellular and Molecular Cardiology Research Unit and Department of Cardiology , Institute of Biomedical Research and University Clinical Hospital , Santiago de Compostela , Spain
| | - Esther Roselló-Lletí
- b Cardiocirculatory Unit, Health Institute of La Fe University Hospital , Valencia , Spain
| | - Manuel Portolés
- b Cardiocirculatory Unit, Health Institute of La Fe University Hospital , Valencia , Spain
| | - Miguel Rivera
- b Cardiocirculatory Unit, Health Institute of La Fe University Hospital , Valencia , Spain
| | - José Ramón González-Juanatey
- a Cellular and Molecular Cardiology Research Unit and Department of Cardiology , Institute of Biomedical Research and University Clinical Hospital , Santiago de Compostela , Spain
| | - Francisca Lago
- a Cellular and Molecular Cardiology Research Unit and Department of Cardiology , Institute of Biomedical Research and University Clinical Hospital , Santiago de Compostela , Spain
| |
Collapse
|
29
|
Horton JS, Wakano CT, Speck M, Stokes AJ. Two-pore channel 1 interacts with citron kinase, regulating completion of cytokinesis. Channels (Austin) 2015; 9:21-9. [PMID: 25665131 PMCID: PMC4594595 DOI: 10.4161/19336950.2014.978676] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Two-pore channels (TPC1, 2, and 3) are recently identified endolysosmal ion channels, but remain poorly characterized. In this study, we show for the first time a role for TPC1 in cytokinesis, the final step in cell division. HEK 293 T-REx cells inducibly overexpressing TPC1 demonstrated a lack of proliferation accompanied by multinucleation and an increase in G2/M cycling cells. Increased TPC1 was associated with a concomitant accumulation of active RhoGTP and a decrease in phosphorylated myosin light chain (MLC). Finally, we demonstrated a novel interaction between TPC1 and citron kinase (CIT). These results identify TPC1 as a central component of cytokinetic control, specifically during abscission, and introduce a means by which the endolysosomal system may play an active role in this process.
Collapse
Affiliation(s)
- Jaime S Horton
- a Laboratory of Experimental Medicine; John A. Burns School of Medicine ; University of Hawaii ; Honolulu , HI USA
| | | | | | | |
Collapse
|
30
|
Höglinger D, Haberkant P, Aguilera-Romero A, Riezman H, Porter FD, Platt FM, Galione A, Schultz C. Intracellular sphingosine releases calcium from lysosomes. eLife 2015; 4:e10616. [PMID: 26613410 PMCID: PMC4744193 DOI: 10.7554/elife.10616] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 11/27/2015] [Indexed: 12/15/2022] Open
Abstract
To elucidate new functions of sphingosine (Sph), we demonstrate that the spontaneous elevation of intracellular Sph levels via caged Sph leads to a significant and transient calcium release from acidic stores that is independent of sphingosine 1-phosphate, extracellular and ER calcium levels. This photo-induced Sph-driven calcium release requires the two-pore channel 1 (TPC1) residing on endosomes and lysosomes. Further, uncaging of Sph leads to the translocation of the autophagy-relevant transcription factor EB (TFEB) to the nucleus specifically after lysosomal calcium release. We confirm that Sph accumulates in late endosomes and lysosomes of cells derived from Niemann-Pick disease type C (NPC) patients and demonstrate a greatly reduced calcium release upon Sph uncaging. We conclude that sphingosine is a positive regulator of calcium release from acidic stores and that understanding the interplay between Sph homeostasis, calcium signaling and autophagy will be crucial in developing new therapies for lipid storage disorders such as NPC.
Collapse
Affiliation(s)
- Doris Höglinger
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Per Haberkant
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Howard Riezman
- Department of Biochemistry, University of Geneva, Geneva, Switzerland
| | - Forbes D Porter
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Frances M Platt
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Carsten Schultz
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| |
Collapse
|
31
|
Abstract
Lysosomes are acidic compartments filled with more than 60 different types of hydrolases. They mediate the degradation of extracellular particles from endocytosis and of intracellular components from autophagy. The digested products are transported out of the lysosome via specific catabolite exporters or via vesicular membrane trafficking. Lysosomes also contain more than 50 membrane proteins and are equipped with the machinery to sense nutrient availability, which determines the distribution, number, size, and activity of lysosomes to control the specificity of cargo flux and timing (the initiation and termination) of degradation. Defects in degradation, export, or trafficking result in lysosomal dysfunction and lysosomal storage diseases (LSDs). Lysosomal channels and transporters mediate ion flux across perimeter membranes to regulate lysosomal ion homeostasis, membrane potential, catabolite export, membrane trafficking, and nutrient sensing. Dysregulation of lysosomal channels underlies the pathogenesis of many LSDs and possibly that of metabolic and common neurodegenerative diseases.
Collapse
Affiliation(s)
- Haoxing Xu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109;
| | | |
Collapse
|
32
|
Calcium signaling in membrane repair. Semin Cell Dev Biol 2015; 45:24-31. [PMID: 26519113 DOI: 10.1016/j.semcdb.2015.10.031] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 10/20/2015] [Accepted: 10/20/2015] [Indexed: 11/21/2022]
Abstract
Resealing allows cells to mend damaged membranes rapidly when plasma membrane (PM) disruptions occur. Models of PM repair mechanisms include the "lipid-patch", "endocytic removal", and "macro-vesicle shedding" models, all of which postulate a dependence on local increases in intracellular Ca(2+) at injury sites. Multiple calcium sensors, including synaptotagmin (Syt) VII, dysferlin, and apoptosis-linked gene-2 (ALG-2), are involved in PM resealing, suggesting that Ca(2+) may regulate multiple steps of the repair process. Although earlier studies focused exclusively on external Ca(2+), recent studies suggest that Ca(2+) release from intracellular stores may also be important for PM resealing. Hence, depending on injury size and the type of injury, multiple sources of Ca(2+) may be recruited to trigger and orchestrate repair processes. In this review, we discuss the mechanisms by which the resealing process is promoted by vesicular Ca(2+) channels and Ca(2+) sensors that accumulate at damage sites.
Collapse
|
33
|
Capel RA, Bolton EL, Lin WK, Aston D, Wang Y, Liu W, Wang X, Burton RAB, Bloor-Young D, Shade KT, Ruas M, Parrington J, Churchill GC, Lei M, Galione A, Terrar DA. Two-pore Channels (TPC2s) and Nicotinic Acid Adenine Dinucleotide Phosphate (NAADP) at Lysosomal-Sarcoplasmic Reticular Junctions Contribute to Acute and Chronic β-Adrenoceptor Signaling in the Heart. J Biol Chem 2015; 290:30087-98. [PMID: 26438825 PMCID: PMC4705968 DOI: 10.1074/jbc.m115.684076] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Indexed: 12/12/2022] Open
Abstract
Ca2+-permeable type 2 two-pore channels (TPC2) are lysosomal proteins required for nicotinic acid adenine dinucleotide phosphate (NAADP)-evoked Ca2+ release in many diverse cell types. Here, we investigate the importance of TPC2 proteins for the physiology and pathophysiology of the heart. NAADP-AM failed to enhance Ca2+ responses in cardiac myocytes from Tpcn2−/− mice, unlike myocytes from wild-type (WT) mice. Ca2+/calmodulin-dependent protein kinase II inhibitors suppressed actions of NAADP in myocytes. Ca2+ transients and contractions accompanying action potentials were increased by isoproterenol in myocytes from WT mice, but these effects of β-adrenoreceptor stimulation were reduced in myocytes from Tpcn2−/− mice. Increases in amplitude of L-type Ca2+ currents evoked by isoproterenol remained unchanged in myocytes from Tpcn2−/− mice showing no loss of β-adrenoceptors or coupling mechanisms. Whole hearts from Tpcn2−/− mice also showed reduced inotropic effects of isoproterenol and a reduced tendency for arrhythmias following acute β-adrenoreceptor stimulation. Hearts from Tpcn2−/− mice chronically exposed to isoproterenol showed less cardiac hypertrophy and increased threshold for arrhythmogenesis compared with WT controls. Electron microscopy showed that lysosomes form close contacts with the sarcoplasmic reticulum (separation ∼25 nm). We propose that Ca2+-signaling nanodomains between lysosomes and sarcoplasmic reticulum dependent on NAADP and TPC2 comprise an important element in β-adrenoreceptor signal transduction in cardiac myocytes. In summary, our observations define a role for NAADP and TPC2 at lysosomal/sarcoplasmic reticulum junctions as unexpected but major contributors in the acute actions of β-adrenergic signaling in the heart and also in stress pathways linking chronic stimulation of β-adrenoceptors to hypertrophy and associated arrhythmias.
Collapse
Affiliation(s)
- Rebecca A Capel
- From the Department of Pharmacology, BHF Centre of Research Excellence, University of Oxford, Mansfield Road, Oxford OX1 3QT
| | - Emma L Bolton
- From the Department of Pharmacology, BHF Centre of Research Excellence, University of Oxford, Mansfield Road, Oxford OX1 3QT
| | - Wee K Lin
- From the Department of Pharmacology, BHF Centre of Research Excellence, University of Oxford, Mansfield Road, Oxford OX1 3QT
| | - Daniel Aston
- From the Department of Pharmacology, BHF Centre of Research Excellence, University of Oxford, Mansfield Road, Oxford OX1 3QT
| | - Yanwen Wang
- From the Department of Pharmacology, BHF Centre of Research Excellence, University of Oxford, Mansfield Road, Oxford OX1 3QT
| | - Wei Liu
- the Faculty of Life Science, University of Manchester, Manchester M13 9NT, and
| | - Xin Wang
- the Faculty of Life Science, University of Manchester, Manchester M13 9NT, and
| | - Rebecca-Ann B Burton
- the Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Sherrington Road, Oxford OX1 3PT, United Kingdom
| | - Duncan Bloor-Young
- From the Department of Pharmacology, BHF Centre of Research Excellence, University of Oxford, Mansfield Road, Oxford OX1 3QT
| | - Kai-Ting Shade
- From the Department of Pharmacology, BHF Centre of Research Excellence, University of Oxford, Mansfield Road, Oxford OX1 3QT
| | - Margarida Ruas
- From the Department of Pharmacology, BHF Centre of Research Excellence, University of Oxford, Mansfield Road, Oxford OX1 3QT
| | - John Parrington
- From the Department of Pharmacology, BHF Centre of Research Excellence, University of Oxford, Mansfield Road, Oxford OX1 3QT
| | - Grant C Churchill
- From the Department of Pharmacology, BHF Centre of Research Excellence, University of Oxford, Mansfield Road, Oxford OX1 3QT
| | - Ming Lei
- From the Department of Pharmacology, BHF Centre of Research Excellence, University of Oxford, Mansfield Road, Oxford OX1 3QT
| | - Antony Galione
- From the Department of Pharmacology, BHF Centre of Research Excellence, University of Oxford, Mansfield Road, Oxford OX1 3QT
| | - Derek A Terrar
- From the Department of Pharmacology, BHF Centre of Research Excellence, University of Oxford, Mansfield Road, Oxford OX1 3QT,
| |
Collapse
|
34
|
Arredouani A, Ruas M, Collins SC, Parkesh R, Clough F, Pillinger T, Coltart G, Rietdorf K, Royle A, Johnson P, Braun M, Zhang Q, Sones W, Shimomura K, Morgan AJ, Lewis AM, Chuang KT, Tunn R, Gadea J, Teboul L, Heister PM, Tynan PW, Bellomo EA, Rutter GA, Rorsman P, Churchill GC, Parrington J, Galione A. Nicotinic Acid Adenine Dinucleotide Phosphate (NAADP) and Endolysosomal Two-pore Channels Modulate Membrane Excitability and Stimulus-Secretion Coupling in Mouse Pancreatic β Cells. J Biol Chem 2015; 290:21376-21392. [PMID: 26152717 PMCID: PMC4571866 DOI: 10.1074/jbc.m115.671248] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 07/03/2015] [Indexed: 12/02/2022] Open
Abstract
Pancreatic β cells are electrically excitable and respond to elevated glucose concentrations with bursts of Ca(2+) action potentials due to the activation of voltage-dependent Ca(2+) channels (VDCCs), which leads to the exocytosis of insulin granules. We have examined the possible role of nicotinic acid adenine dinucleotide phosphate (NAADP)-mediated Ca(2+) release from intracellular stores during stimulus-secretion coupling in primary mouse pancreatic β cells. NAADP-regulated Ca(2+) release channels, likely two-pore channels (TPCs), have recently been shown to be a major mechanism for mobilizing Ca(2+) from the endolysosomal system, resulting in localized Ca(2+) signals. We show here that NAADP-mediated Ca(2+) release from endolysosomal Ca(2+) stores activates inward membrane currents and depolarizes the β cell to the threshold for VDCC activation and thereby contributes to glucose-evoked depolarization of the membrane potential during stimulus-response coupling. Selective pharmacological inhibition of NAADP-evoked Ca(2+) release or genetic ablation of endolysosomal TPC1 or TPC2 channels attenuates glucose- and sulfonylurea-induced membrane currents, depolarization, cytoplasmic Ca(2+) signals, and insulin secretion. Our findings implicate NAADP-evoked Ca(2+) release from acidic Ca(2+) storage organelles in stimulus-secretion coupling in β cells.
Collapse
Affiliation(s)
- Abdelilah Arredouani
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom,
| | - Margarida Ruas
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Stephan C Collins
- the Centre des Sciences du Gout et de l'Alimentation, Equipe 5, 9E Boulevard Jeanne d'Arc 21000 Dijon, France
| | - Raman Parkesh
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Frederick Clough
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Toby Pillinger
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - George Coltart
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Katja Rietdorf
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Andrew Royle
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Paul Johnson
- the Nuffield Department of Surgery, John Radcliffe Hospital, Headley Way, Headington, Oxford OX3 9DU, United Kingdom
| | - Matthias Braun
- the The Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford OX3 7LJ, United Kingdom
| | - Quan Zhang
- the The Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford OX3 7LJ, United Kingdom
| | - William Sones
- the The Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford OX3 7LJ, United Kingdom
| | - Kenju Shimomura
- the Henry Wellcome Centre for Gene Function, Department of Physiology, Anatomy, and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, United Kingdom
| | - Anthony J Morgan
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Alexander M Lewis
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Kai-Ting Chuang
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Ruth Tunn
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Joaquin Gadea
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Lydia Teboul
- The Mary Lyon Centre, Medical Research Council Harwell, Oxfordshire OX11 0RD, United Kingdom
| | - Paula M Heister
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Patricia W Tynan
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Elisa A Bellomo
- the Centre des Sciences du Gout et de l'Alimentation, Equipe 5, 9E Boulevard Jeanne d'Arc 21000 Dijon, France
| | - Guy A Rutter
- the Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Medicine, Imperial College London, Hammersmith Hospital, du Cane Road, London W12 0NN, United Kingdom, and
| | - Patrik Rorsman
- the The Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford OX3 7LJ, United Kingdom
| | - Grant C Churchill
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - John Parrington
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom,
| | - Antony Galione
- From the Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom,
| |
Collapse
|
35
|
Abstract
Two-pore channels (TPCs) are evolutionarily important members of the voltage-gated ion channel superfamily. TPCs localize to acidic Ca(2+) stores within the endolysosomal system. Most evidence indicate that TPCs mediate Ca(2+) signals through the Ca(2+)-mobilizing messenger nicotinic acid adenine dinucleotide phosphate (NAADP) to control a range of Ca(2+)-dependent events. Recent studies clarify the mechanism of TPC activation and identify roles for TPCs in disease, highlighting the regulation of endolysosomal membrane traffic by local Ca(2+) fluxes. Chemical targeting of TPCs to maintain endolysosomal "well-being" may be beneficial in disorders as diverse as Parkinson's disease, fatty liver disease, and Ebola virus infection.
Collapse
Affiliation(s)
- Sandip Patel
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK. E-mail:
| |
Collapse
|
36
|
Ruas M, Galione A, Parrington J. Two-Pore Channels: Lessons from Mutant Mouse Models. ACTA ACUST UNITED AC 2015; 4:4-22. [PMID: 27330869 DOI: 10.1166/msr.2015.1041] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Recent interest in two-pore channels (TPCs) has resulted in a variety of studies dealing with the functional role and mechanism of action of these endo-lysosomal proteins in diverse physiological processes. With the availability of mouse lines harbouring mutant alleles for Tpcnl and/or Tpcn2 genes, several studies have made use of them to validate, consolidate and discover new roles for these channels not only at the cellular level but, importantly, also at the level of the whole organism. The different mutant mouse lines that have been used were derived from distinct genetic manipulation strategies, with the aim of knocking out expression of TPC proteins. However, the expression of different residual TPC sequences predicted to occur in these mutant mouse lines, together with the varied degree to which the effects on Tpcn expression have been studied, makes it important to assess the true knockout status of some of the lines. In this review we summarize these Tpcn mutant mouse lines with regard to their predicted effect on Tpcn expression and the extent to which they have been characterized. Additionally, we discuss how results derived from studies using these Tpcn mutant mouse lines have consolidated previously proposed roles for TPCs, such as mediators of NAADP signalling, endo-lysosomal functions, and pancreatic β cell physiology. We will also review how they have been instrumental in the assignment of new physiological roles for these cation channels in processes such as membrane electrical excitability, neoangiogenesis, viral infection and brown adipose tissue and heart function, revealing, in some cases, a specific contribution of a particular TPC isoform.
Collapse
Affiliation(s)
- Margarida Ruas
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - John Parrington
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| |
Collapse
|
37
|
Ruas M, Davis LC, Chen CC, Morgan AJ, Chuang KT, Walseth TF, Grimm C, Garnham C, Powell T, Platt N, Platt FM, Biel M, Wahl-Schott C, Parrington J, Galione A. Expression of Ca²⁺-permeable two-pore channels rescues NAADP signalling in TPC-deficient cells. EMBO J 2015; 34:1743-58. [PMID: 25872774 PMCID: PMC4516428 DOI: 10.15252/embj.201490009] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 03/11/2015] [Indexed: 01/26/2023] Open
Abstract
The second messenger NAADP triggers Ca2+ release from endo-lysosomes. Although two-pore channels (TPCs) have been proposed to be regulated by NAADP, recent studies have challenged this. By generating the first mouse line with demonstrable absence of both Tpcn1 and Tpcn2 expression (Tpcn1/2−/−), we show that the loss of endogenous TPCs abolished NAADP-dependent Ca2+ responses as assessed by single-cell Ca2+ imaging or patch-clamp of single endo-lysosomes. In contrast, currents stimulated by PI(3,5)P2 were only partially dependent on TPCs. In Tpcn1/2−/− cells, NAADP sensitivity was restored by re-expressing wild-type TPCs, but not by mutant versions with impaired Ca2+-permeability, nor by TRPML1. Another mouse line formerly reported as TPC-null likely expresses truncated TPCs, but we now show that these truncated proteins still support NAADP-induced Ca2+ release. High-affinity [32P]NAADP binding still occurs in Tpcn1/2−/− tissue, suggesting that NAADP regulation is conferred by an accessory protein. Altogether, our data establish TPCs as Ca2+-permeable channels indispensable for NAADP signalling.
Collapse
Affiliation(s)
- Margarida Ruas
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Lianne C Davis
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Cheng-Chang Chen
- Center for Integrated Protein Science CIPS-M and Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, München, Germany
| | | | - Kai-Ting Chuang
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Timothy F Walseth
- Pharmacology Department, University of Minnesota, Minneapolis, MN, USA
| | - Christian Grimm
- Center for Integrated Protein Science CIPS-M and Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, München, Germany
| | - Clive Garnham
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Trevor Powell
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Nick Platt
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Frances M Platt
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Martin Biel
- Center for Integrated Protein Science CIPS-M and Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, München, Germany
| | - Christian Wahl-Schott
- Center for Integrated Protein Science CIPS-M and Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, München, Germany
| | - John Parrington
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Oxford, UK
| |
Collapse
|
38
|
Penny CJ, Kilpatrick BS, Eden ER, Patel S. Coupling acidic organelles with the ER through Ca²⁺ microdomains at membrane contact sites. Cell Calcium 2015; 58:387-96. [PMID: 25866010 DOI: 10.1016/j.ceca.2015.03.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 03/13/2015] [Accepted: 03/14/2015] [Indexed: 10/23/2022]
Abstract
Acidic organelles such as lysosomes serve as non-canonical Ca(2+) stores. The Ca(2+) mobilising messenger NAADP is thought to trigger local Ca(2+) release from such stores. These events are then amplified by Ca(2+) channels on canonical ER Ca(2+) stores to generate physiologically relevant global Ca(2+) signals. Coupling likely occurs at microdomains formed at membrane contact sites between acidic organelles and the ER. Molecular analyses and computational modelling suggest heterogeneity in the composition of these contacts and predicted Ca(2+) microdomain behaviour. Conversely, acidic organelles might also locally amplify and temper ER-evoked Ca(2+) signals. Ca(2+) microdomains between distinct Ca(2+) stores are thus likely to be integral to the genesis of complex Ca(2+) signals.
Collapse
Affiliation(s)
- Christopher J Penny
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Bethan S Kilpatrick
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Emily R Eden
- Department of Cell Biology, Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
39
|
Rahman T, Cai X, Brailoiu GC, Abood ME, Brailoiu E, Patel S. Two-pore channels provide insight into the evolution of voltage-gated Ca2+ and Na+ channels. Sci Signal 2014; 7:ra109. [PMID: 25406377 PMCID: PMC4327855 DOI: 10.1126/scisignal.2005450] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Four-domain voltage-gated Ca(2+) and Na(+) channels (CaV, NaV) underpin nervous system function and likely emerged upon intragenic duplication of a primordial two-domain precursor. To investigate if two-pore channels (TPCs) may represent an intermediate in this evolutionary transition, we performed molecular docking simulations with a homology model of TPC1, which suggested that the pore region could bind antagonists of CaV or NaV. CaV or NaV antagonists blocked NAADP (nicotinic acid adenine dinucleotide phosphate)-evoked Ca(2+) signals in sea urchin egg preparations and in intact cells that overexpressed TPC1. By sequence analysis and inspection of the model, we predicted a noncanonical selectivity filter in animal TPCs in which the carbonyl groups of conserved asparagine residues are positioned to coordinate cations. In contrast, a distinct clade of TPCs [TPCR (for TPC-related)] in several unicellular species had ion selectivity filters with acidic residues more akin to CaV. TPCRs were predicted to interact strongly with CaV antagonists. Our data suggest that acquisition of a "blueprint" pharmacological profile and changes in ion selectivity within four-domain voltage-gated ion channels may have predated intragenic duplication of an ancient two-domain ancestor.
Collapse
Affiliation(s)
- Taufiq Rahman
- Department of Pharmacology, Cambridge University, Cambridge CB2 1PD, UK.
| | - Xinjiang Cai
- Department of Cell Developmental Biology, University College London, London WC1E 6BT, UK
| | - G Cristina Brailoiu
- Department of Pharmaceutical Sciences, Thomas Jefferson University School of Pharmacy, Philadelphia, PA 19107, USA
| | - Mary E Abood
- Department of Anatomy and Cell Biology and Center for Substance Abuse Research, Temple University, Philadelphia, PA 19140, USA
| | - Eugen Brailoiu
- Department of Pharmacology and Center for Substance Abuse Research, Temple University, Philadelphia, PA 19140, USA
| | - Sandip Patel
- Department of Cell Developmental Biology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
40
|
Favia A, Desideri M, Gambara G, D'Alessio A, Ruas M, Esposito B, Del Bufalo D, Parrington J, Ziparo E, Palombi F, Galione A, Filippini A. VEGF-induced neoangiogenesis is mediated by NAADP and two-pore channel-2-dependent Ca2+ signaling. Proc Natl Acad Sci U S A 2014; 111:E4706-15. [PMID: 25331892 PMCID: PMC4226099 DOI: 10.1073/pnas.1406029111] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) and its receptors VEGFR1/VEGFR2 play major roles in controlling angiogenesis, including vascularization of solid tumors. Here we describe a specific Ca(2+) signaling pathway linked to the VEGFR2 receptor subtype, controlling the critical angiogenic responses of endothelial cells (ECs) to VEGF. Key steps of this pathway are the involvement of the potent Ca(2+) mobilizing messenger, nicotinic acid adenine-dinucleotide phosphate (NAADP), and the specific engagement of the two-pore channel TPC2 subtype on acidic intracellular Ca(2+) stores, resulting in Ca(2+) release and angiogenic responses. Targeting this intracellular pathway pharmacologically using the NAADP antagonist Ned-19 or genetically using Tpcn2(-/-) mice was found to inhibit angiogenic responses to VEGF in vitro and in vivo. In human umbilical vein endothelial cells (HUVECs) Ned-19 abolished VEGF-induced Ca(2+) release, impairing phosphorylation of ERK1/2, Akt, eNOS, JNK, cell proliferation, cell migration, and capillary-like tube formation. Interestingly, Tpcn2 shRNA treatment abolished VEGF-induced Ca(2+) release and capillary-like tube formation. Importantly, in vivo VEGF-induced vessel formation in matrigel plugs in mice was abolished by Ned-19 and, most notably, failed to occur in Tpcn2(-/-) mice, but was unaffected in Tpcn1(-/-) animals. These results demonstrate that a VEGFR2/NAADP/TPC2/Ca(2+) signaling pathway is critical for VEGF-induced angiogenesis in vitro and in vivo. Given that VEGF can elicit both pro- and antiangiogenic responses depending upon the balance of signal transduction pathways activated, targeting specific VEGFR2 downstream signaling pathways could modify this balance, potentially leading to more finely tailored therapeutic strategies.
Collapse
Affiliation(s)
- Annarita Favia
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Marianna Desideri
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, 00128 Rome, Italy
| | - Guido Gambara
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Alessio D'Alessio
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy; Institute of Histology and Embryology, Catholic University of the Sacred Heart, 00168 Rome, Italy; and
| | - Margarida Ruas
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, United Kingdom
| | - Bianca Esposito
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Donatella Del Bufalo
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, 00128 Rome, Italy
| | - John Parrington
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, United Kingdom
| | - Elio Ziparo
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Fioretta Palombi
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, United Kingdom
| | - Antonio Filippini
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy;
| |
Collapse
|
41
|
Pantazaka E, Taylor EJA, Bernard WG, Taylor CW. Ca(2+) signals evoked by histamine H1 receptors are attenuated by activation of prostaglandin EP2 and EP4 receptors in human aortic smooth muscle cells. Br J Pharmacol 2014; 169:1624-34. [PMID: 23638853 PMCID: PMC3724117 DOI: 10.1111/bph.12239] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 04/19/2013] [Accepted: 04/25/2013] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE Histamine and prostaglandin E2 (PGE2 ), directly and via their effects on other cells, regulate the behaviour of vascular smooth muscle (VSM), but their effects on human VSM are incompletely resolved. EXPERIMENTAL APPROACH The effects of PGE2 on histamine-evoked changes in intracellular free Ca(2+) concentration ([Ca(2+) ]i ) and adenylyl cyclase activity were measured in populations of cultured human aortic smooth muscle cells (ASMCs). Selective ligands of histamine and EP receptors were used to identify the receptors that mediate the responses. KEY RESULTS Histamine, via H1 receptors, stimulates an increase in [Ca(2+) ]i that is entirely mediated by activation of inositol 1,4,5-trisphosphate receptors. Selective stimulation of EP2 or EP4 receptors attenuates histamine-evoked Ca(2+) signals, but the effects of PGE2 on both Ca(2+) signals and AC activity are largely mediated by EP2 receptors. CONCLUSIONS AND IMPLICATIONS Two important inflammatory mediators, histamine via H1 receptors and PGE2 acting largely via EP2 receptors, exert opposing effects on [Ca(2+) ]i in human ASMCs.
Collapse
|
42
|
Galione A. A primer of NAADP-mediated Ca(2+) signalling: From sea urchin eggs to mammalian cells. Cell Calcium 2014; 58:27-47. [PMID: 25449298 DOI: 10.1016/j.ceca.2014.09.010] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 09/28/2014] [Accepted: 09/29/2014] [Indexed: 02/04/2023]
Abstract
Since the discovery of the Ca(2+) mobilizing effects of the pyridine nucleotide metabolite, nicotinic acid adenine dinucleotide phosphate (NAADP), this molecule has been demonstrated to function as a Ca(2+) mobilizing intracellular messenger in a wide range of cell types. In this review, I will briefly summarize the distinct principles behind NAADP-mediated Ca(2+) signalling before going on to outline the role of this messenger in the physiology of specific cell types. Central to the discussion here is the finding that NAADP principally mobilizes Ca(2+) from acidic organelles such as lysosomes and it is this property that allows NAADP to play a unique role in intracellular Ca(2+) signalling. Lysosomes and related organelles are small Ca(2+) stores but importantly may also initiate a two-way dialogue with other Ca(2+) storage organelles to amplify Ca(2+) release, and may be strategically localized to influence localized Ca(2+) signalling microdomains. The study of NAADP signalling has created a new and fruitful focus on the lysosome and endolysosomal system as major players in calcium signalling and pathophysiology.
Collapse
Affiliation(s)
- Antony Galione
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.
| |
Collapse
|
43
|
TPC1 has two variant isoforms, and their removal has different effects on endo-lysosomal functions compared to loss of TPC2. Mol Cell Biol 2014; 34:3981-92. [PMID: 25135478 PMCID: PMC4386455 DOI: 10.1128/mcb.00113-14] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Organelle ion homeostasis within the endo-lysosomal system is critical for physiological functions. Two-pore channels (TPCs) are cation channels that reside in endo-lysosomal organelles, and overexpression results in endo-lysosomal trafficking defects. However, the impact of a lack of TPC expression on endo-lysosomal trafficking is unknown. Here, we characterize Tpcn1 expression in two transgenic mouse lines (Tpcn1XG716 and Tpcn1T159) and show expression of a novel evolutionarily conserved Tpcn1B transcript from an alternative promoter, raising important questions regarding the status of Tpcn1 expression in mice recently described to be Tpcn1 knockouts. We show that the transgenic Tpcn1T159 line lacks expression of both Tpcn1 isoforms in all tissues analyzed. Using mouse embryonic fibroblasts (MEFs) from Tpcn1−/− and Tpcn2−/− animals, we show that a lack of Tpcn1 or Tpcn2 expression has no significant impact on resting endo-lysosomal pH or morphology. However, differential effects in endo-lysosomal function were observed upon the loss of Tpcn1 or Tpcn2 expression; thus, while Tpcn1−/− MEFs have impaired trafficking of cholera toxin from the plasma membrane to the Golgi apparatus, Tpcn2−/− MEFs show slower kinetics of ligand-induced platelet-derived growth factor receptor β (PDGFRβ) degradation, which is dependent on trafficking to lysosomes. Our findings indicate that TPC1 and TPC2 have important but distinct roles in the endo-lysosomal pathway.
Collapse
|
44
|
Pitt SJ, Lam AKM, Rietdorf K, Galione A, Sitsapesan R. Reconstituted human TPC1 is a proton-permeable ion channel and is activated by NAADP or Ca2+. Sci Signal 2014; 7:ra46. [PMID: 24847115 DOI: 10.1126/scisignal.2004854] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
NAADP potently triggers Ca2+ release from acidic lysosomal and endolysosomal Ca2+ stores. Human two-pore channels (TPC1 and TPC2), which are located on these stores, are involved in this process, but there is controversy over whether TPC1 and TPC2 constitute the Ca2+ release channels. We therefore examined the single-channel properties of human TPC1 after reconstitution into bilayers of controlled composition. We found that TPC1 was permeable not only to Ca2+ but also to monovalent cations and that permeability to protons was the highest (relative permeability sequence: H+ >> K+ > Na(+) ≥ Ca2+). NAADP or Ca2+ activated TPC1, and the presence of one of these ligands was required for channel activation. The endolysosome-located lipid phosphatidylinositol 3,5-bisphosphate [PI(3,5)P2] had no effect on TPC1 open probability but significantly increased the relative permeability of Na+ to Ca2+ and of H+ to Ca2+. Furthermore, our data showed that, although both TPC1 and TPC2 are stimulated by NAADP, these channels differ in ion selectivity and modulation by Ca2+ and pH. We propose that NAADP triggers H+ release from lysosomes and endolysomes through activation of TPC1, but that the Ca2+ -releasing ability of TPC1 will depend on the ionic composition of the acidic stores and may be influenced by other regulators that affect TPC1 ion permeation.
Collapse
Affiliation(s)
- Samantha J Pitt
- School of Medicine, University of St Andrews, St Andrews KY16 9TF, UK
| | - Andy K M Lam
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Katja Rietdorf
- 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
| | - Rebecca Sitsapesan
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.
| |
Collapse
|
45
|
Pereira GJS, Hirata H, do Carmo LG, Stilhano RS, Ureshino RP, Medaglia NC, Han SW, Churchill G, Bincoletto C, Patel S, Smaili SS. NAADP-sensitive two-pore channels are present and functional in gastric smooth muscle cells. Cell Calcium 2014; 56:51-8. [PMID: 24882212 DOI: 10.1016/j.ceca.2014.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 04/11/2014] [Accepted: 04/21/2014] [Indexed: 01/06/2023]
Abstract
Nicotinic acid adenine dinucleotide phosphate (NAADP) has been identified as an important modulator of Ca(2+) release from the endo-lysosomal system in a variety of cells by a new and ubiquitous class of endo-lysosomal ion channels known as the two-pore channels (TPCs). However, the role of TPCs in NAADP action in smooth muscle is not known. In the present work, we investigated the effects of NAADP in gastric smooth muscle cells and its ability to release Ca(2+) by TPCs. We show that Ca(2+) signals mediated by NAADP were inhibited by disrupting Ca(2+) handling by either acidic organelles (using bafilomycin A1) or the Endoplasmic Reticulum (using thapsigargin, ryanodine or 2-APB). Transcripts for endogenous TPC1 and TPC2 were readily detected and recombinant TPCs localized to the endosomes and/or lysosomes. Overexpression of wild-type TPCs but not pore mutants enhanced NAADP-mediated cytosolic Ca(2+) signals. Desensitizing the NAADP pathway inhibited Ca(2+)-responses to extracellular stimulation with carbachol but not ATP. Taken together, these results indicate that NAADP likely induces Ca(2+) release from the endolysosomal system through TPCs which is subsequently amplified via the ER in an agonist-specific manner. Thus, we suggest a second messenger role for NAADP in smooth muscle cells.
Collapse
Affiliation(s)
- Gustavo J S Pereira
- Department of Pharmacology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Hanako Hirata
- Department of Pharmacology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Lúcia G do Carmo
- Department of Pharmacology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Roberta S Stilhano
- Interdisciplinary Center for Gene Therapy, Federal University of São Paulo, São Paulo, Brazil
| | - Rodrigo P Ureshino
- Department of Pharmacology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Natalia C Medaglia
- Department of Pharmacology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Sang W Han
- Interdisciplinary Center for Gene Therapy, Federal University of São Paulo, São Paulo, Brazil
| | - Grant Churchill
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Claudia Bincoletto
- Department of Pharmacology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Soraya S Smaili
- Department of Pharmacology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil.
| |
Collapse
|
46
|
Jha A, Ahuja M, Patel S, Brailoiu E, Muallem S. Convergent regulation of the lysosomal two-pore channel-2 by Mg²⁺, NAADP, PI(3,5)P₂ and multiple protein kinases. EMBO J 2014; 33:501-11. [PMID: 24502975 DOI: 10.1002/embj.201387035] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Lysosomal Ca(2+) homeostasis is implicated in disease and controls many lysosomal functions. A key in understanding lysosomal Ca(2+) signaling was the discovery of the two-pore channels (TPCs) and their potential activation by NAADP. Recent work concluded that the TPCs function as a PI(3,5)P2 activated channels regulated by mTORC1, but not by NAADP. Here, we identified Mg(2+) and the MAPKs, JNK and P38 as novel regulators of TPC2. Cytoplasmic Mg(2+) specifically inhibited TPC2 outward current, whereas lysosomal Mg(2+) partially inhibited both outward and inward currents in a lysosomal lumen pH-dependent manner. Under controlled Mg(2+), TPC2 is readily activated by NAADP with channel properties identical to those in response to PI(3,5)P2. Moreover, TPC2 is robustly regulated by P38 and JNK. Notably, NAADP-mediated Ca(2+) release in intact cells is regulated by Mg(2+), PI(3,5)P2, and P38/JNK kinases, thus paralleling regulation of TPC2 currents. Our data affirm a key role for TPC2 in NAADP-mediated Ca(2+) signaling and link this pathway to Mg(2+) homeostasis and MAP kinases, pointing to roles for lysosomal Ca(2+) in cell growth, inflammation and cancer.
Collapse
Affiliation(s)
- Archana Jha
- Epithelial Signaling and Transport Section, Molecular Physiology and Therapeutics Branch, NIDCR NIH, Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
47
|
Arndt L, Castonguay J, Arlt E, Meyer D, Hassan S, Borth H, Zierler S, Wennemuth G, Breit A, Biel M, Wahl-Schott C, Gudermann T, Klugbauer N, Boekhoff I. NAADP and the two-pore channel protein 1 participate in the acrosome reaction in mammalian spermatozoa. Mol Biol Cell 2014; 25:948-64. [PMID: 24451262 PMCID: PMC3952862 DOI: 10.1091/mbc.e13-09-0523] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
A TPCN1 gene–deficient mouse strain is used to show that two convergent working NAADP-dependent pathways with nonoverlapping activation and self-inactivation profiles for distinct NAADP concentrations drive acrosomal exocytosis, by which TPC1 is central for the pathway activated by low-micromolar NAADP concentrations. The functional relationship between the formation of hundreds of fusion pores during the acrosome reaction in spermatozoa and the mobilization of calcium from the acrosome has been determined only partially. Hence, the second messenger NAADP, promoting efflux of calcium from lysosome-like compartments and one of its potential molecular targets, the two-pore channel 1 (TPC1), were analyzed for its involvement in triggering the acrosome reaction using a TPCN1 gene–deficient mouse strain. The present study documents that TPC1 and NAADP-binding sites showed a colocalization at the acrosomal region and that treatment of spermatozoa with NAADP resulted in a loss of the acrosomal vesicle that showed typical properties described for TPCs: Registered responses were not detectable for its chemical analogue NADP and were blocked by the NAADP antagonist trans-Ned-19. In addition, two narrow bell-shaped dose-response curves were identified with maxima in either the nanomolar or low micromolar NAADP concentration range, where TPC1 was found to be responsible for activating the low affinity pathway. Our finding that two convergent NAADP-dependent pathways are operative in driving acrosomal exocytosis supports the concept that both NAADP-gated cascades match local NAADP concentrations with the efflux of acrosomal calcium, thereby ensuring complete fusion of the large acrosomal vesicle.
Collapse
Affiliation(s)
- Lilli Arndt
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians University, 81377 München, Germany Department of Pharmacy, Ludwig-Maximilians University, 81377 München, Germany Institute for Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-University, 79104 Freiburg, Germany Institute for Anatomy, University of Duisburg-Essen, 45141 Essen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Much excitement surrounded the proposal that a family of endo-lysosomal channels, the two-pore channels (TPCs) were the long sought after targets of the Ca(2+) -mobilising messenger, nicotinic acid adenine dinucleotide phosphate (NAADP). However, the role of TPCs in NAADP signalling may be more complex than originally envisaged. First, NAADP may not bind directly to TPCs but via an accessory protein. Second, two papers recently challenged the notion that TPCs are NAADP-regulated Ca(2+) channels by suggesting that they are highly selective Na(+) channels regulated by the lipid phosphatidylinositol 3,5-bisphosphate and by ATP. This paper aims critically to evaluate the evidence for TPCs as NAADP targets and to discuss how the new findings fit in with what we know about endo-lysosomal Ca(2+) stores.
Collapse
|
49
|
The N-terminal region of two-pore channel 1 regulates trafficking and activation by NAADP. Biochem J 2013; 453:147-51. [PMID: 23634879 DOI: 10.1042/bj20130474] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
TPCs (two-pore channels) are NAADP (nicotinic acid-adenine dinucleotide phosphate)-sensitive Ca2+-permeable ion channels expressed on acidic organelles. In the present study we show that deletion of the N-terminal region redirects TPC1 to the ER (endoplasmic reticulum). The introduction of fluorophores at the N-terminus of TPC1 does not affect its subcellular location, but does reversibly abolish NAADP sensitivity. Our results reveal a dual role for the N-terminus in localization and function of TPC1.
Collapse
|
50
|
Lu Y, Hao BX, Graeff R, Wong CWM, Wu WT, Yue J. Two pore channel 2 (TPC2) inhibits autophagosomal-lysosomal fusion by alkalinizing lysosomal pH. J Biol Chem 2013; 288:24247-24263. [PMID: 23836916 PMCID: PMC3745369 DOI: 10.1074/jbc.m113.484253] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 07/01/2013] [Indexed: 12/31/2022] Open
Abstract
Autophagy is an evolutionarily conserved lysosomal degradation pathway, yet the underlying mechanisms remain poorly understood. Nicotinic acid adenine dinucleotide phosphate (NAADP), one of the most potent Ca(2+) mobilizing messengers, elicits Ca(2+) release from lysosomes via the two pore channel 2 (TPC2) in many cell types. Here we found that overexpression of TPC2 in HeLa or mouse embryonic stem cells inhibited autophagosomal-lysosomal fusion, thereby resulting in the accumulation of autophagosomes. Treatment of TPC2 expressing cells with a cell permeant-NAADP agonist, NAADP-AM, further induced autophagosome accumulation. On the other hand, TPC2 knockdown or treatment of cells with Ned-19, a NAADP antagonist, markedly decreased the accumulation of autophagosomes. TPC2-induced accumulation of autophagosomes was also markedly blocked by ATG5 knockdown. Interestingly, inhibiting mTOR activity failed to increase TPC2-induced autophagosome accumulation. Instead, we found that overexpression of TPC2 alkalinized lysosomal pH, and lysosomal re-acidification abolished TPC2-induced autophagosome accumulation. In addition, TPC2 overexpression had no effect on general endosomal-lysosomal degradation but prevented the recruitment of Rab-7 to autophagosomes. Taken together, our data demonstrate that TPC2/NAADP/Ca(2+) signaling alkalinizes lysosomal pH to specifically inhibit the later stage of basal autophagy progression.
Collapse
Affiliation(s)
- Yingying Lu
- From the Department of Physiology, University of Hong Kong, Hong Kong, China
| | - Bai-Xia Hao
- From the Department of Physiology, University of Hong Kong, Hong Kong, China
| | - Richard Graeff
- From the Department of Physiology, University of Hong Kong, Hong Kong, China
| | - Connie W. M. Wong
- Department of Anatomy and State Key Laboratory of Brain and Cognitive Sciences, University of Hong Kong, Hong Kong, China, and
| | - Wu-Tian Wu
- Department of Anatomy and State Key Laboratory of Brain and Cognitive Sciences, University of Hong Kong, Hong Kong, China, and
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Jianbo Yue
- From the Department of Physiology, University of Hong Kong, Hong Kong, China
| |
Collapse
|