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
|
Deutsch R, Kudrina V, Freichel M, Grimm C. Two-pore channel regulators - Who is in control? Front Physiol 2025; 15:1534071. [PMID: 39867224 PMCID: PMC11757267 DOI: 10.3389/fphys.2024.1534071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/30/2024] [Indexed: 01/28/2025] Open
Abstract
Two-pore channels (TPCs) are adenine nucleotide and phosphoinositide regulated cation channels. NAADP activates and ATP blocks TPCs, while the endolysosomal phosphoinositide PI(3,5)P2 activates TPCs. TPCs are ubiquitously expressed including expression in the innate as well as the adaptive immune system. In the immune system TPCs are found, e.g. in macrophages, mast cells and T cells. In cytotoxic T cells, NAADP activates TPCs on cytolytic granules to stimulate exocytosis and killing. TPC inhibition or knockdown increases the number of regulator T cells in a transmembrane TNF/TNFR2 dependent manner, contributing to anti-inflammatory effects in a murine colitis model. TPC1 regulates exocytosis in mast cells in vivo and ex vivo, and TPC1 deficiency in mast cells augments systemic anaphylaxis in mice. In bone marrow derived macrophages NAADP regulates TPCs to control phagocytosis in a calcineurin/dynamin dependent manner, which was recently challenged by data, claiming no effect of TPCs on phagocytosis in macrophages but instead a role in phagosome resolution, a process thought to be mediated by vesiculation and tubulation. In this review we will discuss evidence and recent findings on the different roles of TPCs in immune cell function as well as evidence for adenine nucleotides being involved in these processes. Since the adenine nucleotide effects (NAADP, ATP) are mediated by auxiliary proteins, respectively, another major focus will be on the complex network of TPC regulatory proteins that have been discovered recently.
Collapse
Affiliation(s)
- Rebecca Deutsch
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Veronika Kudrina
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Christian Grimm
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
- Immunology, Infection and Pandemic Research IIP, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Munich/Frankfurt, Germany
| |
Collapse
|
3
|
Abrahamian C, Ouologuem L, Tang R, Fröhlich T, Bartel K, Grimm C. TPC2: From Blond Hair to Melanoma? Cancers (Basel) 2024; 16:4065. [PMID: 39682251 DOI: 10.3390/cancers16234065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/01/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Two-pore channel 2 (TPC2) is expressed in endolysosomes throughout the human body, as well as in melanosomes of melanocytes. Melanocytes produce pigment, i.e., melanin, which determines hair and skin color but also protects from UV light. Extensive exposure to UV light is one of the major risk factors for the development of melanoma, which develops from pigment-producing cells, i.e., melanocytes. In recent years, several human TPC2 single nucleotide polymorphisms have been identified to increase the likelihood of carriers presenting with blond hair and hypopigmentation. These variants were all characterized as gain-of-function versions of TPC2. Vice versa, the loss of function of TPC2 increases melanin production and reduces cancer hallmarks such as proliferation, migration, invasion, tumor growth, and metastasis formation. The activity of TPC2 is controlled in a complex manner, with several endogenous ligands as well as a number of interacting proteins being involved. We will discuss here the role of TPC2 in pigmentation and its potential to impact melanoma development and progression and highlight recent findings on Rab7a as an enhancer of TPC2 activity.
Collapse
Affiliation(s)
- Carla Abrahamian
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig Maximilians University, 80336 Munich, Germany
| | - Lina Ouologuem
- Department of Pharmacy, Ludwig Maximilians University, 80539 Munich, Germany
| | - Rachel Tang
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig Maximilians University, 80336 Munich, Germany
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis LAFUGA, Gene Center, Ludwig Maximilians University, 80539 Munich, Germany
| | - Karin Bartel
- Department of Pharmacy, Ludwig Maximilians University, 80539 Munich, Germany
| | - Christian Grimm
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig Maximilians University, 80336 Munich, Germany
- Immunology, Infection and Pandemic Research IIP, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 80333 Munich, Germany
| |
Collapse
|
4
|
Jin X, Hanbashi AA, Kamli F, Pan X, Goding CR, Parrington J. TPC1 regulates melanoma tumourigenesis via mTORC1 and TFEB. Heliyon 2024; 10:e39752. [PMID: 39524724 PMCID: PMC11550043 DOI: 10.1016/j.heliyon.2024.e39752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/09/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
The major cause of death in cancer patients is a combination of metastatic dissemination combined with therapy resistance. Over recent years, intratumour phenotypic heterogeneity arising from the bi-directional interplay between plastic cancer cells and the microenvironment has been identified as key to disease progression. Most notably metastatic outgrowth and resistance to targeted therapies are frequently associated with activity of mTORC1, a key metabolic hub that promotes protein synthesis and proliferation in the presence of nutrients. Yet while the regulation of mTORC1 by amino acids and glucose availability is well characterized, whether other mechanisms are important in controlling mTORC1 and its downstream signalling is less well understood. Here we show, using the murine B16-F0 melanoma cell line as a model, that mTORC1 activity is decreased following the knockout (KO) of TPC1, a cation channel localised to early and recycling endosomes. Consequently, TPC1 KO melanoma cells exhibit reduced proliferation and invasiveness, as well as increased pigmentation associated with nuclear localisation of the MITF-related transcription factor TFEB. Our results demonstrate that the knockout of TPC1 has induced significant tumour-suppressive effects in melanoma, during which the altered activity of mTORC1 and TFEB play the key roles. The results help us further understand the link between mTORC1 and endolysosomal ion channels, and reveal that TPC1 controls melanoma progression and represents a potential therapeutic target.
Collapse
Affiliation(s)
- Xuhui Jin
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, United Kingdom
| | - Ali A. Hanbashi
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, United Kingdom
- Department of Pharmacology, College of Pharmacy, Jazan University, Jazan, 45142, Saudi Arabia
| | - Faroq Kamli
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, United Kingdom
- Department of Pharmacology, College of Pharmacy, Jazan University, Jazan, 45142, Saudi Arabia
| | - Xiaoqi Pan
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu University of Traditional Chinese Medicine, 611137, Chengdu, China
| | - Colin R. Goding
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, Old Road Campus, University of Oxford, Oxford, OX3 7DQ, United Kingdom
| | - John Parrington
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, United Kingdom
| |
Collapse
|
5
|
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
|
6
|
Cen J, Hu N, Shen J, Gao Y, Lu H. Pathological Functions of Lysosomal Ion Channels in the Central Nervous System. Int J Mol Sci 2024; 25:6565. [PMID: 38928271 PMCID: PMC11203704 DOI: 10.3390/ijms25126565] [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: 04/03/2024] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Lysosomes are highly dynamic organelles that maintain cellular homeostasis and regulate fundamental cellular processes by integrating multiple metabolic pathways. Lysosomal ion channels such as TRPML1-3, TPC1/2, ClC6/7, CLN7, and TMEM175 mediate the flux of Ca2+, Cl-, Na+, H+, and K+ across lysosomal membranes in response to osmotic stimulus, nutrient-dependent signals, and cellular stresses. These ion channels serve as the crucial transducers of cell signals and are essential for the regulation of lysosomal biogenesis, motility, membrane contact site formation, and lysosomal homeostasis. In terms of pathophysiology, genetic variations in these channel genes have been associated with the development of lysosomal storage diseases, neurodegenerative diseases, inflammation, and cancer. This review aims to discuss the current understanding of the role of these ion channels in the central nervous system and to assess their potential as drug targets.
Collapse
Affiliation(s)
| | | | | | - Yongjing Gao
- Institute of Pain Medicine and Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China; (J.C.); (N.H.); (J.S.)
| | - Huanjun Lu
- Institute of Pain Medicine and Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China; (J.C.); (N.H.); (J.S.)
| |
Collapse
|
7
|
Rathore M, Thakur A, Saran S. Deletion of Dictyostelium tpc2 gene forms multi-tipped structures, regulates autophagy and cell-type patterning. Biol Cell 2024; 116:e2300067. [PMID: 38537110 DOI: 10.1111/boc.202300067] [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: 07/17/2023] [Revised: 03/04/2024] [Accepted: 03/08/2024] [Indexed: 05/09/2024]
Abstract
BACKGROUND INFORMATION Two pore channels (TPCs) are voltage-gated ion channel superfamily members that release Ca2+ from acidic intracellular stores and are ubiquitously present in both animals and plants. Starvation initiates multicellular development in Dictyostelium discoideum. Increased intracellular calcium levels bias Dictyostelium cells towards the stalk pathway and thus we decided to analyze the role of TPC2 in development, differentiation, and autophagy. RESULTS We showed TPC2 protein localizes in lysosome-like acidic vesicles and the in situ data showed stalk cell biasness. Deletion of tpc2 showed defective and delayed development with formation of multi-tipped structures attached to a common base, while tpc2OE cells showed faster development with numerous small-sized aggregates and wiry fruiting bodies. The tpc2OE cells showed higher intracellular cAMP levels as compared to the tpc2- cells while pinocytosis was found to be higher in the tpc2- cells. Also, TPC2 regulates cell-substrate adhesion and cellular morphology. Under nutrient starvation, deletion of tpc2 reduced autophagic flux as compared to Ax2. During chimera formation, tpc2- cells showed a bias towards the prestalk/stalk region while tpc2OE cells showed a bias towards the prespore/spore region. tpc2 deficient strain exhibits aberrant cell-type patterning and loss of distinct boundary between the prestalk/prespore regions. CONCLUSION TPC2 is required for effective development and differentiation in Dictyostelium and supports autophagic cell death and cell-type patterning. SIGNIFICANCE Decreased calcium due to deletion of tpc2 inhibit autophagic flux.
Collapse
Affiliation(s)
- Madhubala Rathore
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Ashima Thakur
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Shweta Saran
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
8
|
Barbonari S, D'Amore A, Hanbashi AA, Palombi F, Riccioli A, Parrington J, Filippini A. Endolysosomal two-pore channel 2 plays opposing roles in primary and metastatic malignant melanoma cells. Cell Biol Int 2024; 48:521-540. [PMID: 38263578 DOI: 10.1002/cbin.12129] [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: 07/22/2023] [Revised: 11/10/2023] [Accepted: 01/01/2024] [Indexed: 01/25/2024]
Abstract
The ion channel two-pore channel 2 (TPC2), localised on the membranes of acidic organelles such as endo-lysosomes and melanosomes, has been shown to play a role in pathologies including cancer, and it is differently expressed in primary versus metastatic melanoma cells. Whether TPC2 plays a pro- or anti-oncogenic role in different tumour conditions is a relevant open question which we have explored in melanoma at different stages of tumour progression. The behaviour of primary melanoma cell line B16F0 and its metastatic subline B16F10 were compared in response to TPC2 modulation by silencing (by small interfering RNA), knock-out (by CRISPR/Cas9) and overexpression (by mCherry-TPC2 transfected plasmid). TPC2 silencing increased cell migration, epithelial-to-mesenchymal transition and autophagy in the metastatic samples, but abated them in the silenced primary ones. Interestingly, while TPC2 inactivation failed to affect markers of proliferation in both samples, it strongly enhanced the migratory behaviour of the metastatic cells, again suggesting that in the more aggressive phenotype TPC2 plays a specific antimetastatic role. In line with this, overexpression of TPC2 in B16F10 cells resulted in phenotype rescue, that is, a decrease in migratory ability, thus collectively resuming traits of the B16F0 primary cell line. Our research shows a novel role of TPC2 in melanoma cells that is intriguingly different in initial versus late stages of cancer progression.
Collapse
Affiliation(s)
- Samantha Barbonari
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Unit of Histology and Medical Embryology, Sapienza University, Rome, Italy
| | | | - Ali A Hanbashi
- Department of Pharmacology, University of Oxford, Oxford, UK
- Department of Pharmacology, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Fioretta Palombi
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Unit of Histology and Medical Embryology, Sapienza University, Rome, Italy
| | - Anna Riccioli
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Unit of Histology and Medical Embryology, Sapienza University, Rome, Italy
| | - John Parrington
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Antonio Filippini
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Unit of Histology and Medical Embryology, Sapienza University, Rome, Italy
| |
Collapse
|
9
|
Chadwick SR, Barreda D, Wu JZ, Ye G, Yusuf B, Ren D, Freeman SA. Two-pore channels regulate endomembrane tension to enable remodeling and resolution of phagolysosomes. Proc Natl Acad Sci U S A 2024; 121:e2309465121. [PMID: 38354262 PMCID: PMC10895354 DOI: 10.1073/pnas.2309465121] [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: 06/05/2023] [Accepted: 12/21/2023] [Indexed: 02/16/2024] Open
Abstract
Phagocytes promptly resolve ingested targets to replenish lysosomes and maintain their responsiveness. The resolution process requires that degradative hydrolases, solute transporters, and proteins involved in lipid traffic are delivered and made active in phagolysosomes. It also involves extensive membrane remodeling. We report that cation channels that localize to phagolysosomes were essential for resolution. Specifically, the conductance of Na+ by two-pore channels (TPCs) and the presence of a Na+ gradient between the phagolysosome lumen and the cytosol were critical for the controlled release of membrane tension that permits deformation of the limiting phagolysosome membrane. In turn, membrane deformation was a necessary step to efficiently transport the cholesterol extracted from cellular targets, permeabilizing them to hydrolases. These results place TPCs as regulators of endomembrane remodeling events that precede target degradation in cases when the target is bound by a cholesterol-containing membrane. The findings may help to explain lipid metabolism dysfunction and autophagic flux impairment reported in TPC KO mice and establish stepwise regulation to the resolution process that begins with lysis of the target.
Collapse
Affiliation(s)
- Sarah R. Chadwick
- Program in Cell Biology, Hospital for Sick Children, Toronto, ONM5G 0A4, Canada
| | - Dante Barreda
- Program in Cell Biology, Hospital for Sick Children, Toronto, ONM5G 0A4, Canada
| | - Jing Ze Wu
- Program in Cell Biology, Hospital for Sick Children, Toronto, ONM5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
| | - Gang Ye
- Program in Cell Biology, Hospital for Sick Children, Toronto, ONM5G 0A4, Canada
| | - Bushra Yusuf
- Program in Cell Biology, Hospital for Sick Children, Toronto, ONM5G 0A4, Canada
- Institute of Medical Science, University of Toronto, Toronto, ONM5S 1A8, Canada
| | - Dejian Ren
- Department of Biology, University of Pennsylvania, Philadelphia, PA19104
| | - Spencer A. Freeman
- Program in Cell Biology, Hospital for Sick Children, Toronto, ONM5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A8, Canada
| |
Collapse
|
10
|
Brock VJ, Lory NC, Möckl F, Birus M, Stähler T, Woelk LM, Jaeckstein M, Heeren J, Koch-Nolte F, Rissiek B, Mittrücker HW, Guse AH, Werner R, Diercks BP. Time-resolved role of P2X4 and P2X7 during CD8 + T cell activation. Front Immunol 2024; 15:1258119. [PMID: 38426095 PMCID: PMC10902106 DOI: 10.3389/fimmu.2024.1258119] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 01/11/2024] [Indexed: 03/02/2024] Open
Abstract
CD8+ T cells are a crucial part of the adaptive immune system, responsible for combating intracellular pathogens and tumor cells. The initial activation of T cells involves the formation of highly dynamic Ca2+ microdomains. Recently, purinergic signaling was shown to be involved in the formation of the initial Ca2+ microdomains in CD4+ T cells. In this study, the role of purinergic cation channels, particularly P2X4 and P2X7, in CD8+ T cell signaling from initial events to downstream responses was investigated, focusing on various aspects of T cell activation, including Ca2+ microdomains, global Ca2+ responses, NFAT-1 translocation, cytokine expression, and proliferation. While Ca2+ microdomain formation was significantly reduced in the first milliseconds to seconds in CD8+ T cells lacking P2X4 and P2X7 channels, global Ca2+ responses over minutes were comparable between wild-type (WT) and knockout cells. However, the onset velocity was reduced in P2X4-deficient cells, and P2X4, as well as P2X7-deficient cells, exhibited a delayed response to reach a certain level of free cytosolic Ca2+ concentration ([Ca2+]i). NFAT-1 translocation, a crucial transcription factor in T cell activation, was also impaired in CD8+ T cells lacking P2X4 and P2X7. In addition, the expression of IFN-γ, a major pro-inflammatory cytokine produced by activated CD8+ T cells, and Nur77, a negative regulator of T cell activation, was significantly reduced 18h post-stimulation in the knockout cells. In line, the proliferation of T cells after 3 days was also impaired in the absence of P2X4 and P2X7 channels. In summary, the study demonstrates that purinergic signaling through P2X4 and P2X7 enhances initial Ca2+ events during CD8+ T cell activation and plays a crucial role in regulating downstream responses, including NFAT-1 translocation, cytokine expression, and proliferation on multiple timescales. These findings suggest that targeting purinergic signaling pathways may offer potential therapeutic interventions.
Collapse
Affiliation(s)
- Valerie J. Brock
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Niels Christian Lory
- Department of Immunology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Franziska Möckl
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Melina Birus
- Department of Immunology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Stähler
- Department of Immunology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Lena-Marie Woelk
- Department of Applied Medical Informatics, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department of Computational Neuroscience, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Michelle Jaeckstein
- Department of Biochemistry and Molecular Cell Biology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Department of Immunology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Björn Rissiek
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Willi Mittrücker
- Department of Immunology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas H. Guse
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - René Werner
- Department of Applied Medical Informatics, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department of Computational Neuroscience, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Björn-Philipp Diercks
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
11
|
de Zélicourt A, Fayssoil A, Mansart A, Zarrouki F, Karoui A, Piquereau J, Lefebvre F, Gerbaud P, Mika D, Dakouane-Giudicelli M, Lanchec E, Feng M, Leblais V, Bobe R, Launay JM, Galione A, Gomez AM, de la Porte S, Cancela JM. Two-pore channels (TPCs) acts as a hub for excitation-contraction coupling, metabolism and cardiac hypertrophy signalling. Cell Calcium 2024; 117:102839. [PMID: 38134531 DOI: 10.1016/j.ceca.2023.102839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/01/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023]
Abstract
Ca2+ signaling is essential for cardiac contractility and excitability in heart function and remodeling. Intriguingly, little is known about the role of a new family of ion channels, the endo-lysosomal non-selective cation "two-pore channel" (TPCs) in heart function. Here we have used double TPC knock-out mice for the 1 and 2 isoforms of TPCs (Tpcn1/2-/-) and evaluated their cardiac function. Doppler-echocardiography unveils altered left ventricular (LV) systolic function associated with a LV relaxation impairment. In cardiomyocytes isolated from Tpcn1/2-/- mice, we observed a reduction in the contractile function with a decrease in the sarcoplasmic reticulum Ca2+ content and a reduced expression of various key proteins regulating Ca2+ stores, such as calsequestrin. We also found that two main regulators of the energy metabolism, AMP-activated protein kinase and mTOR, were down regulated. We found an increase in the expression of TPC1 and TPC2 in a model of transverse aortic constriction (TAC) mice and in chronically isoproterenol infused WT mice. In this last model, adaptive cardiac hypertrophy was reduced by Tpcn1/2 deletion. Here, we propose a central role for TPCs and lysosomes that could act as a hub integrating information from the excitation-contraction coupling mechanisms, cellular energy metabolism and hypertrophy signaling.
Collapse
Affiliation(s)
- Antoine de Zélicourt
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, 78000 Versailles, France; Neuroscience Paris-Saclay Institute (Neuro-PSI), UMR 9197, CNRS- Université Paris-Saclay, Saclay, 91400, France
| | - Abdallah Fayssoil
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, 78000 Versailles, France
| | - Arnaud Mansart
- Université Paris-Saclay, UVSQ, Inserm, 2I, 78000 Versailles, France
| | - Faouzi Zarrouki
- Neuroscience Paris-Saclay Institute (Neuro-PSI), UMR 9197, CNRS- Université Paris-Saclay, Saclay, 91400, France
| | - Ahmed Karoui
- UMR-S 1180, INSERM, Signaling and cardiovascular pathophysiology, Université Paris-Saclay, 91400 Orsay, France
| | - Jérome Piquereau
- UMR-S 1180, INSERM, Signaling and cardiovascular pathophysiology, Université Paris-Saclay, 91400 Orsay, France
| | - Florence Lefebvre
- UMR-S 1180, INSERM, Signaling and cardiovascular pathophysiology, Université Paris-Saclay, 91400 Orsay, France
| | - Pascale Gerbaud
- UMR-S 1180, INSERM, Signaling and cardiovascular pathophysiology, Université Paris-Saclay, 91400 Orsay, France
| | - Delphine Mika
- UMR-S 1180, INSERM, Signaling and cardiovascular pathophysiology, Université Paris-Saclay, 91400 Orsay, France
| | | | - Erwan Lanchec
- Neuroscience Paris-Saclay Institute (Neuro-PSI), UMR 9197, CNRS- Université Paris-Saclay, Saclay, 91400, France
| | - Miao Feng
- UMR-S 1176, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | - Véronique Leblais
- UMR-S 1180, INSERM, Signaling and cardiovascular pathophysiology, Université Paris-Saclay, 91400 Orsay, France
| | - Régis Bobe
- UMR-S 1176, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | - Jean-Marie Launay
- Service de Biochimie, INSERM UMR S942, Hôpital Lariboisière, Paris, France
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, United Kingdom
| | - Ana Maria Gomez
- UMR-S 1180, INSERM, Signaling and cardiovascular pathophysiology, Université Paris-Saclay, 91400 Orsay, France
| | - Sabine de la Porte
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, 78000 Versailles, France
| | - José-Manuel Cancela
- Neuroscience Paris-Saclay Institute (Neuro-PSI), UMR 9197, CNRS- Université Paris-Saclay, Saclay, 91400, France.
| |
Collapse
|
12
|
Goretzko J, Pauels I, Heitzig N, Thomas K, Kardell M, Naß J, Krogsaeter EK, Schloer S, Spix B, Linard Matos AL, Leser C, Wegner T, Glorius F, Bracher F, Gerke V, Rossaint J, Grimm C, Rescher U. P-selectin-dependent leukocyte adhesion is governed by endolysosomal two-pore channel 2. Cell Rep 2023; 42:113501. [PMID: 38039128 DOI: 10.1016/j.celrep.2023.113501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/18/2023] [Accepted: 11/10/2023] [Indexed: 12/03/2023] Open
Abstract
Upon proinflammatory challenges, endothelial cell surface presentation of the leukocyte receptor P-selectin, together with the stabilizing co-factor CD63, is needed for leukocyte capture and is mediated via demand-driven exocytosis from the Weibel-Palade bodies that fuse with the plasma membrane. We report that neutrophil recruitment to activated endothelium is significantly reduced in mice deficient for the endolysosomal cation channel TPC2 and in human primary endothelial cells with pharmacological TPC2 block. We observe less CD63 signal in whole-mount stainings of proinflammatory-activated cremaster muscles from TPC2 knockout mice. We find that TPC2 is activated and needed to ensure the transfer of CD63 from endolysosomes via Weibel-Palade bodies to the plasma membrane to retain P-selectin on the cell surface of human primary endothelial cells. Our findings establish TPC2 as a key element to leukocyte interaction with the endothelium and a potential pharmacological target in the control of inflammatory leukocyte recruitment.
Collapse
Affiliation(s)
- Jonas Goretzko
- Research Group Cellular Biochemistry - Regulatory Mechanisms of Inflammation, Institute of Molecular Virology, Center for Molecular Biology of Inflammation, University of Muenster (formerly Research Group Regulatory Mechanisms of Inflammation, Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Muenster), von-Esmarch-Strasse 56, 48149 Muenster, Germany
| | - Inga Pauels
- Research Group Cellular Biochemistry - Regulatory Mechanisms of Inflammation, Institute of Molecular Virology, Center for Molecular Biology of Inflammation, University of Muenster (formerly Research Group Regulatory Mechanisms of Inflammation, Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Muenster), von-Esmarch-Strasse 56, 48149 Muenster, Germany
| | - Nicole Heitzig
- Research Group Cellular Biochemistry - Regulatory Mechanisms of Inflammation, Institute of Molecular Virology, Center for Molecular Biology of Inflammation, University of Muenster (formerly Research Group Regulatory Mechanisms of Inflammation, Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Muenster), von-Esmarch-Strasse 56, 48149 Muenster, Germany
| | - Katharina Thomas
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert Schweitzer Campus 1, A1, 48149 Muenster, Germany
| | - Marina Kardell
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert Schweitzer Campus 1, A1, 48149 Muenster, Germany
| | - Johannes Naß
- Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Muenster, von-Esmarch-Strasse 56, 48149 Muenster, Germany
| | - Einar Kleinhans Krogsaeter
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Nussbaumstrasse 26, 80336 Munich, Germany
| | - Sebastian Schloer
- Research Group Cellular Biochemistry - Regulatory Mechanisms of Inflammation, Institute of Molecular Virology, Center for Molecular Biology of Inflammation, University of Muenster (formerly Research Group Regulatory Mechanisms of Inflammation, Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Muenster), von-Esmarch-Strasse 56, 48149 Muenster, Germany
| | - Barbara Spix
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Nussbaumstrasse 26, 80336 Munich, Germany
| | - Anna Lívia Linard Matos
- Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Muenster, von-Esmarch-Strasse 56, 48149 Muenster, Germany
| | - Charlotte Leser
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Tristan Wegner
- Institute of Organic Chemistry, University of Muenster, Corrensstrasse 40, 48149 Muenster, Germany
| | - Frank Glorius
- Institute of Organic Chemistry, University of Muenster, Corrensstrasse 40, 48149 Muenster, Germany
| | - Franz Bracher
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-University, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Volker Gerke
- Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Muenster, von-Esmarch-Strasse 56, 48149 Muenster, Germany
| | - Jan Rossaint
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Albert Schweitzer Campus 1, A1, 48149 Muenster, Germany
| | - Christian Grimm
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Nussbaumstrasse 26, 80336 Munich, Germany; Immunology, Infection and Pandemic Research IIP, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt am Main, Germany
| | - Ursula Rescher
- Research Group Cellular Biochemistry - Regulatory Mechanisms of Inflammation, Institute of Molecular Virology, Center for Molecular Biology of Inflammation, University of Muenster (formerly Research Group Regulatory Mechanisms of Inflammation, Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Muenster), von-Esmarch-Strasse 56, 48149 Muenster, Germany.
| |
Collapse
|
13
|
Gunaratne GS, Brailoiu E, Kumar S, Yuan Y, Slama JT, Walseth TF, Patel S, Marchant JS. Convergent activation of two-pore channels mediated by the NAADP-binding proteins JPT2 and LSM12. Sci Signal 2023; 16:eadg0485. [PMID: 37607218 PMCID: PMC10639087 DOI: 10.1126/scisignal.adg0485] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 07/27/2023] [Indexed: 08/24/2023]
Abstract
The second messenger nicotinic acid adenine dinucleotide phosphate (NAADP) evokes calcium ion (Ca2+) release from endosomes and lysosomes by activating two-pore channels (TPCs) on these organelles. Rather than directly binding to TPCs, NAADP associates with proteins that indirectly confer NAADP sensitivity to the TPC complex. We investigated whether and how the NAADP-binding proteins Jupiter microtubule-associated homolog 2 (JPT2) and like-Sm protein 12 (LSM12) contributed to NAADP-TPC-Ca2+ signaling in human cells. Biochemical and functional analyses revealed that recombinant JPT2 and LSM12 both bound to NAADP with high affinity and that endogenous JPT2 and LSM12 independently associated with TPC1 and TPC2. On the basis of knockout and rescue analyses, both NAADP-binding proteins were required to support NAADP-evoked Ca2+ signaling and contributed to endolysosomal trafficking of pseudotyped coronavirus particles. These data reveal that the NAADP-binding proteins JPT2 and LSM12 convergently regulate NAADP-evoked Ca2+ release and function through TPCs.
Collapse
Affiliation(s)
- Gihan S. Gunaratne
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Eugen Brailoiu
- Center for Substance Abuse Research and Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Sushil Kumar
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Yu Yuan
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - James T. Slama
- Department of Medicinal and Biological Chemistry, University of Toledo College of Pharmacy and Pharmaceutical Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA
| | - Timothy F. Walseth
- Department of Pharmacology, University of Minnesota Medical School, 312 Church St., Minneapolis, MN 55455, USA
| | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Jonathan S. Marchant
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
14
|
Saito R, Mu Q, Yuan Y, Rubio-Alarcón M, Eznarriaga M, Zhao P, Gunaratne G, Kumar S, Keller M, Bracher F, Grimm C, Brailoiu E, Marchant JS, Rahman T, Patel S. Convergent activation of Ca 2+ permeability in two-pore channel 2 through distinct molecular routes. Sci Signal 2023; 16:eadg0661. [PMID: 37607219 PMCID: PMC10639088 DOI: 10.1126/scisignal.adg0661] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 07/27/2023] [Indexed: 08/24/2023]
Abstract
TPC2 is a pathophysiologically relevant lysosomal ion channel that is activated directly by the phosphoinositide PI(3,5)P2 and indirectly by the calcium ion (Ca2+)-mobilizing molecule NAADP through accessory proteins that associate with the channel. TPC2 toggles between PI(3,5)P2-induced, sodium ion (Na+)-selective and NAADP-induced, Ca2+-permeable states in response to these cues. To address the molecular basis of polymodal gating and ion-selectivity switching, we investigated the mechanism by which NAADP and its synthetic functional agonist, TPC2-A1-N, induced Ca2+ release through TPC2 in human cells. Whereas NAADP required the NAADP-binding proteins JPT2 and LSM12 to evoke endogenous calcium ion signals, TPC2-A1-N did not. Residues in TPC2 that bind to PI(3,5)P2 were required for channel activation by NAADP but not for activation by TPC2-A1-N. The cryptic voltage-sensing region of TPC2 was required for the actions of TPC2-A1-N and PI(3,5)P2 but not for those of NAADP. These data mechanistically distinguish natural and synthetic agonist action at TPC2 despite convergent effects on Ca2+ permeability and delineate a route for pharmacologically correcting impaired NAADP-evoked Ca2+ signals.
Collapse
Affiliation(s)
- Ryo Saito
- Department of Cell and Developmental Biology, University
College London, Gower Street, London WC1E 6BT, UK
- Department of Dermatology, Graduate School of Biomedical
and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Qianru Mu
- Department of Cell and Developmental Biology, University
College London, Gower Street, London WC1E 6BT, UK
| | - Yu Yuan
- Department of Cell and Developmental Biology, University
College London, Gower Street, London WC1E 6BT, UK
| | | | - Maria Eznarriaga
- Department of Pharmacology, University of Cambridge,
Cambridge, UK
| | - Pingwei Zhao
- Center for Substance Abuse Research, Lewis Katz School of
Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Gihan Gunaratne
- Department of Cell Biology, Neurobiology and Anatomy,
Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226,
USA
| | - Sushil Kumar
- Department of Cell Biology, Neurobiology and Anatomy,
Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226,
USA
| | - Marco Keller
- Department of Pharmacy—Center for Drug Research,
Ludwig-Maximilian University, Munich, Germany
| | - Franz Bracher
- Department of Pharmacy—Center for Drug Research,
Ludwig-Maximilian University, Munich, Germany
| | - Christian Grimm
- Walther Straub Institute of Pharmacology and Toxicology,
Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany
| | - Eugen Brailoiu
- Center for Substance Abuse Research, Lewis Katz School of
Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Jonathan S. Marchant
- Department of Cell Biology, Neurobiology and Anatomy,
Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226,
USA
| | - Taufiq Rahman
- Department of Pharmacology, University of Cambridge,
Cambridge, UK
| | - Sandip Patel
- Department of Cell and Developmental Biology, University
College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
15
|
Meng Z, Capel RA, Bose SJ, Bosch E, de Jong S, Planque R, Galione A, Burton RAB, Bueno-Orovio A. Lysosomal calcium loading promotes spontaneous calcium release by potentiating ryanodine receptors. Biophys J 2023; 122:3044-3059. [PMID: 37329137 PMCID: PMC10432190 DOI: 10.1016/j.bpj.2023.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 05/03/2023] [Accepted: 06/12/2023] [Indexed: 06/18/2023] Open
Abstract
Spontaneous calcium release by ryanodine receptors (RyRs) due to intracellular calcium overload results in delayed afterdepolarizations, closely associated with life-threatening arrhythmias. In this regard, inhibiting lysosomal calcium release by two-pore channel 2 (TPC2) knockout has been shown to reduce the incidence of ventricular arrhythmias under β-adrenergic stimulation. However, mechanistic investigations into the role of lysosomal function on RyR spontaneous release remain missing. We investigate the calcium handling mechanisms by which lysosome function modulates RyR spontaneous release, and determine how lysosomes are able to mediate arrhythmias by its influence on calcium loading. Mechanistic studies were conducted using a population of biophysically detailed mouse ventricular models including for the first time modeling of lysosomal function, and calibrated by experimental calcium transients modulated by TPC2. We demonstrate that lysosomal calcium uptake and release can synergistically provide a pathway for fast calcium transport, by which lysosomal calcium release primarily modulates sarcoplasmic reticulum calcium reuptake and RyR release. Enhancement of this lysosomal transport pathway promoted RyR spontaneous release by elevating RyR open probability. In contrast, blocking either lysosomal calcium uptake or release revealed an antiarrhythmic impact. Under conditions of calcium overload, our results indicate that these responses are strongly modulated by intercellular variability in L-type calcium current, RyR release, and sarcoplasmic reticulum calcium-ATPase reuptake. Altogether, our investigations identify that lysosomal calcium handling directly influences RyR spontaneous release by regulating RyR open probability, suggesting antiarrhythmic strategies and identifying key modulators of lysosomal proarrhythmic action.
Collapse
Affiliation(s)
- Zhaozheng Meng
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Rebecca A Capel
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Samuel J Bose
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Erik Bosch
- Department of Mathematics, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Sophia de Jong
- Department of Mathematics, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Robert Planque
- Department of Mathematics, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Rebecca A B Burton
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom.
| | | |
Collapse
|
16
|
Kang Y, Xu L, Dong J, Huang Y, Yuan X, Li R, Chen L, Wang Z, Ji X. Calcium-based nanotechnology for cancer therapy. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
17
|
Takano T, Yule DI. Ca 2+ signals in pancreatic acinar cells in response to physiological stimulation in vivo. J Physiol 2023. [PMID: 36965132 DOI: 10.1113/jp284469] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/08/2023] [Indexed: 03/27/2023] Open
Abstract
The exocrine pancreas secretes fluid and digestive enzymes in response to parasympathetic release of acetylcholine (ACh) via the vagus nerve and the gut hormone cholecystokinin (CCK). Both secretion of fluid and exocytosis of secretory granules containing enzymes and zymogens are dependent on an increase in the cytosolic [Ca2+ ] in acinar cells. It is thought that the specific spatiotemporal characteristics of the Ca2+ signals are fundamental for appropriate secretion and that these properties are disrupted in disease states in the pancreas. While extensive research has been performed to characterize Ca2+ signalling in acinar cells, this has exclusively been achieved in ex vivo preparations of exocrine cells, where it is difficult to mimic physiological conditions. Here we have developed a method to optically observe pancreatic acinar Ca2+ signals in vivo using a genetically expressed Ca2+ indicator and imaged with multi-photon microscopy in live animals. In vivo, acinar cells exhibited baseline activity in fasted animals, which was dependent on CCK1 receptors (CCK1Rs). Both stimulation of intrinsic nervous input and administration of systemic CCK induced oscillatory activity in a proportion of the cells, but the maximum frequencies were vastly different. Upon feeding, oscillatory activity was also observed, which was dependent on CCK1Rs. No evidence of a vago-vagal reflex mediating the effects of CCK was observed. Our in vivo method revealed the spatial and temporal profile of physiologically evoked Ca2+ signals, which will provide new insights into future studies of the mechanisms underlying exocrine physiology and that are disrupted in pathological conditions. KEY POINTS: In the exocrine pancreas, the spatiotemporal properties of Ca2+ signals are fundamentally important for the appropriate stimulation of secretion by the neurotransmitter acetylcholine and gut hormone cholecystokinin. These characteristics were previously defined in ex vivo studies. Here we report the spatiotemporal characteristics of Ca2+ signals in vivo in response to physiological stimulation in a mouse engineered to express a Ca2+ indicator in acinar cells. Specific Ca2+ 'signatures' probably important for stimulating secretion are evoked in vivo in fasted animals, by feeding, neural stimulation and cholecystokinin administration. The Ca2+ signals are probably the result of the direct action of ACh and CCK on acinar cells and not indirectly through a vago-vagal reflex.
Collapse
Affiliation(s)
- Takahiro Takano
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA
| | - David I Yule
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA
| |
Collapse
|
18
|
Wahl-Schott C, Freichel M, Hennis K, Philippaert K, Ottenheijm R, Tsvilovskyy V, Varbanov H. Characterization of Endo-Lysosomal Cation Channels Using Calcium Imaging. Handb Exp Pharmacol 2023; 278:277-304. [PMID: 36894791 DOI: 10.1007/164_2023_637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Endo-lysosomes are membrane-bound acidic organelles that are involved in endocytosis, recycling, and degradation of extracellular and intracellular material. The membranes of endo-lysosomes express several Ca2+-permeable cation ion channels, including two-pore channels (TPC1-3) and transient receptor potential mucolipin channels (TRPML1-3). In this chapter, we will describe four different state-of-the-art Ca2+ imaging approaches, which are well-suited to investigate the function of endo-lysosomal cation channels. These techniques include (1) global cytosolic Ca2+ measurements, (2) peri-endo-lysosomal Ca2+ imaging using genetically encoded Ca2+ sensors that are directed to the cytosolic endo-lysosomal membrane surface, (3) Ca2+ imaging of endo-lysosomal cation channels, which are engineered in order to redirect them to the plasma membrane in combination with approaches 1 and 2, and (4) Ca2+ imaging by directing Ca2+ indicators to the endo-lysosomal lumen. Moreover, we will review useful small molecules, which can be used as valuable tools for endo-lysosomal Ca2+ imaging. Rather than providing complete protocols, we will discuss specific methodological issues related to endo-lysosomal Ca2+ imaging.
Collapse
Affiliation(s)
- Christian Wahl-Schott
- Institut für Kardiovaskuläre Physiologie und Pathophysiologie, Lehrstuhl für Vegetative Physiologie, Biomedizinisches Zentrum, Ludwig-Maximilians-Universität München, München, Germany.
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany. .,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany.
| | - Konstantin Hennis
- Institut für Kardiovaskuläre Physiologie und Pathophysiologie, Lehrstuhl für Vegetative Physiologie, Biomedizinisches Zentrum, Ludwig-Maximilians-Universität München, München, Germany
| | - Koenraad Philippaert
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| | - Roger Ottenheijm
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| | - Volodymyr Tsvilovskyy
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| | - Hristo Varbanov
- Institut für Neurophysiologie, Medizinische Hochschule Hannover(MHH), Hannover, Germany
| |
Collapse
|
19
|
Cai W, Li P, Gu M, Xu H. Lysosomal Ion Channels and Lysosome-Organelle Interactions. Handb Exp Pharmacol 2023; 278:93-108. [PMID: 36882602 DOI: 10.1007/164_2023_640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Intracellular organelles exchange their luminal contents with each other via both vesicular and non-vesicular mechanisms. By forming membrane contact sites (MCSs) with ER and mitochondria, lysosomes mediate bidirectional transport of metabolites and ions between lysosomes and organelles that regulate lysosomal physiology, movement, membrane remodeling, and membrane repair. In this chapter, we will first summarize the current knowledge of lysosomal ion channels and then discuss the molecular and physiological mechanisms that regulate lysosome-organelle MCS formation and dynamics. We will also discuss the roles of lysosome-ER and lysosome-mitochondria MCSs in signal transduction, lipid transport, Ca 2+ transfer, membrane trafficking, and membrane repair, as well as their roles in lysosome-related pathologies.
Collapse
Affiliation(s)
- Weijie Cai
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Ping Li
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Mingxue Gu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dun Neurological Research Institute, Houston, TX, USA
| | - Haoxing Xu
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China. .,Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dun Neurological Research Institute, Houston, TX, USA. .,Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
20
|
Tedeschi V, Vinciguerra A, Sisalli MJ, Pignataro G, Secondo A. Pharmacological inhibition of lysosomal two-pore channel 2 (TPC2) confers neuroprotection in stroke via autophagy regulation. Neurobiol Dis 2023; 178:106020. [PMID: 36708960 DOI: 10.1016/j.nbd.2023.106020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/29/2022] [Accepted: 01/24/2023] [Indexed: 01/27/2023] Open
Abstract
Lysosomal function and organellar Ca2+ homeostasis become dysfunctional in Stroke causing disturbances in autophagy, the major process for the degradation of abnormal protein aggregates and dysfunctional organelles. However, the role of autophagy in Stroke is controversial since excessive or prolonged autophagy activation exacerbates ischemic brain injury. Of note, glutamate evokes NAADP-dependent Ca2+ release via lysosomal TPC2 channels thus controlling basal autophagy. Considering the massive release of excitotoxins in Stroke, autophagic flux becomes uncontrolled with abnormal formation of autophagosomes causing, in turn, disruption of excitotoxins clearance and neurodegeneration. Here, a fine regulation of autophagy via a proper pharmacological modulation of lysosomal TPC2 channel has been tested in preclinical Stroke models. Primary cortical neurons were subjected to oxygen and glucose deprivation+reoxygenation to reproduce in vitro brain ischemia. Focal brain ischemia was induced in rats by transient middle cerebral artery occlusion (tMCAO). Under these conditions, TPC2 protein expression as well as autophagy and endoplasmic reticulum (ER) stress markers were studied by Western blotting, while TPC2 localization and activity were measured by immunocytochemistry and single-cell video-imaging, respectively. TPC2 protein expression and immunosignal were highly modulated in primary cortical neurons exposed to extreme hypoxic conditions causing dysfunction in organellar Ca2+ homeostasis, ER stress and autophagy-induced cell death. TPC2 knocking down and pharmacological inhibition by Ned-19 during hypoxia induced neuroprotection. The effect of Ned-19 was reversed by the permeable form of TPC2 endogenous agonist, NAADP-AM. Of note, Ned-19 prevented ER stress, as measured by GRP78 (78 kDa glucose-regulated protein) protein reduction and caspase 9 downregulation. In this way Ned-19 restored organellar Ca2+ level. Interestingly, Ned-19 reduced the infarct volume and neurological deficits in rats subjected to tMCAO and prevented hypoxia-induced cell death by blocking autophagic flux. Collectively, the pharmacological inhibition of TPC2 lysosomal channel by Ned-19 protects from focal ischemia by hampering a hyperfunctional autophagy.
Collapse
Affiliation(s)
- Valentina Tedeschi
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, Via Sergio Pansini 5, Naples 80131, Italy.
| | - Antonio Vinciguerra
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, Ancona 60126, Italy.
| | - Maria Josè Sisalli
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, Via Sergio Pansini 5, Naples 80131, Italy.
| | - Giuseppe Pignataro
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, Via Sergio Pansini 5, Naples 80131, Italy.
| | - Agnese Secondo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, Via Sergio Pansini 5, Naples 80131, Italy.
| |
Collapse
|
21
|
Just A, Mallmann RT, Grossmann S, Sleman F, Klugbauer N. Two-pore channel protein TPC1 is a determining factor for the adaptation of proximal tubular phosphate handling. Acta Physiol (Oxf) 2023; 237:e13914. [PMID: 36599408 DOI: 10.1111/apha.13914] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 10/27/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023]
Abstract
AIM Two-pore channels (TPCs) constitute a small family of cation channels expressed in endo-lysosomal compartments. TPCs have been characterized as critical elements controlling Ca2+ -mediated vesicular membrane fusion and thereby regulating endo-lysosomal vesicle trafficking. Exo- and endocytotic trafficking and lysosomal degradation are major mechanisms of adaption of epithelial transport. A prime example of highly regulated epithelial transport is the tubular system of the kidney. We therefore studied the localization of TPC protein 1 (TPC1) in the kidney and its functional role in the dynamic regulation of tubular transport. METHODS Immunohistochemistry in combination with tubular markers were used to investigate TPC1 expression in proximal and distal tubules. The excretion of phosphate and ammonium, as well as urine volume and pH were studied in vivo, in response to dynamic challenges induced by bolus injection of parathyroid hormone or acid-base transitions via consecutive infusion of NaCl, Na2 CO3 , and NH4 Cl. RESULTS In TPC1-deficient mice, the PTH-induced rise in phosphate excretion was prolonged and exaggerated, and its recovery delayed in comparison with wildtype littermates. In the acid-base transition experiment, TPC1-deficient mice showed an identical rise in phosphate excretion in response to Na2 CO3 compared with wildtypes, but a delayed NH4Cl-induced recovery. Ammonium-excretion decreased with Na2 CO3 , and increased with NH4 Cl, but without differences between genotypes. CONCLUSIONS We conclude that TPC1 is expressed subapically in the proximal but not distal tubule and plays an important role in the dynamic adaptation of proximal tubular phosphate reabsorption towards enhanced, but not reduced absorption.
Collapse
Affiliation(s)
- Armin Just
- Institut für Physiologie I, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Robert T Mallmann
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Sonja Grossmann
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Faten Sleman
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Norbert Klugbauer
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Medizinische Fakultät, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| |
Collapse
|
22
|
Martucci LL, Launay JM, Kawakami N, Sicard C, Desvignes N, Dakouane-Giudicelli M, Spix B, Têtu M, Gilmaire FO, Paulcan S, Callebert J, Vaillend C, Bracher F, Grimm C, Fossier P, de la Porte S, Sakamoto H, Morris J, Galione A, Granon S, Cancela JM. Endolysosomal TPCs regulate social behavior by controlling oxytocin secretion. Proc Natl Acad Sci U S A 2023; 120:e2213682120. [PMID: 36745816 PMCID: PMC9963339 DOI: 10.1073/pnas.2213682120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 12/14/2022] [Indexed: 02/08/2023] Open
Abstract
Oxytocin (OT) is a prominent regulator of many aspects of mammalian social behavior and stored in large dense-cored vesicles (LDCVs) in hypothalamic neurons. It is released in response to activity-dependent Ca2+ influx, but is also dependent on Ca2+ release from intracellular stores, which primes LDCVs for exocytosis. Despite its importance, critical aspects of the Ca2+-dependent mechanisms of its secretion remain to be identified. Here we show that lysosomes surround dendritic LDCVs, and that the direct activation of endolysosomal two-pore channels (TPCs) provides the critical Ca2+ signals to prime OT release by increasing the releasable LDCV pool without directly stimulating exocytosis. We observed a dramatic reduction in plasma OT levels in TPC knockout mice, and impaired secretion of OT from the hypothalamus demonstrating the importance of priming of neuropeptide vesicles for activity-dependent release. Furthermore, we show that activation of type 1 metabotropic glutamate receptors sustains somatodendritic OT release by recruiting TPCs. The priming effect could be mimicked by a direct application of nicotinic acid adenine dinucleotide phosphate, the endogenous messenger regulating TPCs, or a selective TPC2 agonist, TPC2-A1-N, or blocked by the antagonist Ned-19. Mice lacking TPCs exhibit impaired maternal and social behavior, which is restored by direct OT administration. This study demonstrates an unexpected role for lysosomes and TPCs in controlling neuropeptide secretion, and in regulating social behavior.
Collapse
Affiliation(s)
- Lora L. Martucci
- Neuroscience Paris-Saclay Institute, CNRS UMR 9197, Paris-Sud University, Paris-Saclay University, Saclay91400, France
- Université Paris-Saclay, Université de Versailles Saint-Quentin-en-Yvelines, Inserm, Evolution of Neuromuscular Diseases: Innovative Concepts and Practices, Versailles78000, France
- Department of Pharmacology, University of Oxford, OxfordOX1 3QT, UK
| | | | - Natsuko Kawakami
- Ushimado Marine Institute, Graduate School of Natural Science and Technology, Okayama University, Ushimado, Setouchi, Okayama701-4303, Japan
| | - Cécile Sicard
- Neuroscience Paris-Saclay Institute, CNRS UMR 9197, Paris-Sud University, Paris-Saclay University, Saclay91400, France
| | - Nathalie Desvignes
- Neuroscience Paris-Saclay Institute, CNRS UMR 9197, Paris-Sud University, Paris-Saclay University, Saclay91400, France
| | - Mbarka Dakouane-Giudicelli
- Université Paris-Saclay, Université de Versailles Saint-Quentin-en-Yvelines, Inserm, Evolution of Neuromuscular Diseases: Innovative Concepts and Practices, Versailles78000, France
| | - Barbara Spix
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine Ludwig-Maximilians-University, Munich80336, Germany
| | - Maude Têtu
- Neuroscience Paris-Saclay Institute, CNRS UMR 9197, Paris-Sud University, Paris-Saclay University, Saclay91400, France
| | - Franck-Olivier Gilmaire
- Neuroscience Paris-Saclay Institute, CNRS UMR 9197, Paris-Sud University, Paris-Saclay University, Saclay91400, France
| | - Sloane Paulcan
- Neuroscience Paris-Saclay Institute, CNRS UMR 9197, Paris-Sud University, Paris-Saclay University, Saclay91400, France
| | - Jacques Callebert
- Laboratoire de Biochimie et Biologie Moléculaire, Hôpital Lariboisière, Paris75010, France
- Inserm UMR-S 1144 Universités Paris Descartes-Paris Diderot, Optimisation Thérapeutique en Neuropsychopharmacologie - Faculté des Sciences Pharmaceutiques et Biologiques, Paris Descartes,ParisParis 75006, France
| | - Cyrille Vaillend
- Neuroscience Paris-Saclay Institute, CNRS UMR 9197, Paris-Sud University, Paris-Saclay University, Saclay91400, France
| | - Franz Bracher
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians University, Munich81377, Germany
| | - Christian Grimm
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine Ludwig-Maximilians-University, Munich80336, Germany
| | - Philippe Fossier
- Neuroscience Paris-Saclay Institute, CNRS UMR 9197, Paris-Sud University, Paris-Saclay University, Saclay91400, France
| | - Sabine de la Porte
- Université Paris-Saclay, Université de Versailles Saint-Quentin-en-Yvelines, Inserm, Evolution of Neuromuscular Diseases: Innovative Concepts and Practices, Versailles78000, France
| | - Hirotaka Sakamoto
- Ushimado Marine Institute, Graduate School of Natural Science and Technology, Okayama University, Ushimado, Setouchi, Okayama701-4303, Japan
| | - John Morris
- Department of Physiology, Anatomy & Genetics, University of Oxford, OxfordOX1 3QX, UK
| | - Antony Galione
- Department of Pharmacology, University of Oxford, OxfordOX1 3QT, UK
| | - Sylvie Granon
- Neuroscience Paris-Saclay Institute, CNRS UMR 9197, Paris-Sud University, Paris-Saclay University, Saclay91400, France
| | - José-Manuel Cancela
- Neuroscience Paris-Saclay Institute, CNRS UMR 9197, Paris-Sud University, Paris-Saclay University, Saclay91400, France
| |
Collapse
|
23
|
Rautenberg S, Keller M, Leser C, Chen CC, Bracher F, Grimm C. Expanding the Toolbox: Novel Modulators of Endolysosomal Cation Channels. Handb Exp Pharmacol 2023; 278:249-276. [PMID: 35902436 DOI: 10.1007/164_2022_605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Functional characterization of endolysosomal ion channels is challenging due to their intracellular location. With recent advances in endolysosomal patch clamp technology, it has become possible to directly measure ion channel currents across endolysosomal membranes. Members of the transient receptor potential (TRP) cation channel family, namely the endolysosomal TRPML channels (TRPML1-3), also called mucolipins, as well as the distantly related two-pore channels (TPCs) have recently been characterized in more detail with endolysosomal patch clamp techniques. However, answers to many physiological questions require work in intact cells or animal models. One major obstacle thereby is that the known endogenous ligands of TRPMLs and TPCs are anionic in nature and thus impermeable for cell membranes. Microinjection, on the other hand, is technically demanding. There is also a risk of losing essential co-factors for channel activation or inhibition in isolated preparations. Therefore, lipophilic, membrane-permeable small-molecule activators and inhibitors for TRPMLs and TPCs are urgently needed. Here, we describe and discuss the currently available small-molecule modulators of TRPMLs and TPCs.
Collapse
Affiliation(s)
- Susanne Rautenberg
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-University, Munich, Germany
| | - Marco Keller
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-University, Munich, Germany
| | - Charlotte Leser
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-University, Munich, Germany
| | - Cheng-Chang Chen
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-University, Munich, Germany
| | - Franz Bracher
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-University, Munich, Germany.
| | - Christian Grimm
- Department of Pharmacology and Toxicology, Medical Faculty, Ludwig-Maximilians-University, Munich, Germany.
| |
Collapse
|
24
|
Chen CC. Electrophysiological Techniques on the Study of Endolysosomal Ion Channels. Handb Exp Pharmacol 2023; 278:217-233. [PMID: 36871125 DOI: 10.1007/164_2023_638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Endolysosomal ion channels are a group of ion channel proteins that are functionally expressed on the membrane of endolysosomal vesicles. The electrophysiological properties of these ion channels in the intracellular organelle membrane cannot be observed using conventional electrophysiological techniques. This section compiles the different electrophysiological techniques utilized in recent years to study endolysosomal ion channels and describes their methodological characteristics, emphasizing the most widely used technique for whole endolysosome recordings to date. This includes the use of different pharmacological tools and genetic tools for the application of patch-clamping techniques for specific stages of endolysosomes, allowing the recording of ion channel activity in different organelles, such as recycling endosomes, early endosomes, late endosomes, and lysosomes. These electrophysiological techniques are not only cutting-edge technologies that help to investigate the biophysical properties of known and unknown intracellular ion channels but also help us to investigate the physiopathological role of these ion channels in the distribution of dynamic vesicles and to identify new therapeutic targets for precision medicine and drug screening.
Collapse
Affiliation(s)
- Cheng-Chang Chen
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
25
|
She J, Guo J, Jiang Y. Structure and Function of Plant and Mammalian TPC Channels. Handb Exp Pharmacol 2023; 278:155-180. [PMID: 35879575 DOI: 10.1007/164_2022_599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Two-pore channels (TPCs) belong to the family of voltage-gated tetrameric cation channels and are ubiquitously expressed in organelles of animals and plants. These channels are believed to be evolutionary intermediates between homotetrameric voltage-gated potassium/sodium channels and the four-domain, single subunit, voltage-gated sodium/calcium channels. Each TPC subunit contains 12 transmembrane segments that can be divided into two homologous copies of an S1-S6 Shaker-like 6-TM domain. A functional TPC channel assembles as a dimer - the equivalent of a voltage-gated tetrameric cation channel. The plant TPC channel is localized in the vacuolar membrane and is also called the SV channel for generating the slow vacuolar (SV) current observed long before its molecular identification. Three subfamilies of mammalian TPC channels have been defined - TPC1, 2, and 3 - with the first two being ubiquitously expressed in animals and TPC3 being expressed in some animals but not in humans. Mammalian TPC1 and TPC2 are localized to the endolysosomal membrane and their functions are associated with various physiological processes. TPC3 is localized in the plasma membrane and its physiological function is not well defined.
Collapse
Affiliation(s)
- Ji She
- MOE Key Laboratory for Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jiangtao Guo
- Department of Biophysics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Neurology, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Youxing Jiang
- Howard Hughes Medical Institute, Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
26
|
Abstract
Lysosomes are acidic membrane-bound organelles that use hydrolytic enzymes to break down material through pathways such as endocytosis, phagocytosis, mitophagy, and autophagy. To function properly, intralysosomal environments are strictly controlled by a set of integral membrane proteins such as ion channels and transporters. Potassium ion (K+) channels are a large and diverse family of membrane proteins that control K+ flux across both the plasma membrane and intracellular membranes. In the plasma membrane, they are essential in both excitable and non-excitable cells for the control of membrane potential and cell signaling. However, our understanding of intracellular K+ channels is very limited. In this review, we summarize the recent development in studies of K+ channels in the lysosome. We focus on their characterization, potential roles in maintaining lysosomal membrane potential and lysosomal function, and pathological implications.
Collapse
Affiliation(s)
- Peng Huang
- Collaborative Innovation Center for Biomedicine, School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Mengnan Xu
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, Halifax, NS, Canada
| | - Yi Wu
- Collaborative Innovation Center for Biomedicine, School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Alia Kazim Rizvi Syeda
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, Halifax, NS, Canada
| | - Xian-Ping Dong
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, Halifax, NS, Canada.
| |
Collapse
|
27
|
Wang Q, Zhu MX. NAADP-Dependent TPC Current. Handb Exp Pharmacol 2023; 278:35-56. [PMID: 35902437 DOI: 10.1007/164_2022_606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Two-pore channels, TPC1 and TPC2, are Ca2+- and Na+-permeable cation channels expressed on the membranes of endosomes and lysosomes in nearly all mammalian cells. These channels have been implicated in Ca2+ signaling initiated from the endolysosomes, vesicular trafficking, cellular metabolism, macropinocytosis, and viral infection. Although TPCs have been shown to mediate Ca2+ release from acidic organelles in response to NAADP (nicotinic acid adenine dinucleotide phosphate), the most potent Ca2+ mobilizing messenger, questions remain whether NAADP is a direct ligand of these channels. In whole-endolysosomal patch clamp recordings, it has been difficult to detect NAADP-evoked currents in vacuoles that expressed TPC1 or TPC2, while PI(3,5)P2 (phosphatidylinositol 3,5-bisphosphate) activated a highly Na+-selective current under the same experimental configuration. In this chapter, we summarize recent progress in this area and provide our observations on NAADP-elicited TPC2 currents from enlarged endolysosomes as well as their possible relationships with the currents evoked by PI(3,5)P2. We propose that TPCs are channels dually regulated by PI(3,5)P2 and NAADP in an interdependent manner and the two endogenous ligands may have both distinguished and cooperative roles.
Collapse
Affiliation(s)
- Qiaochu Wang
- Beijing Children's Hospital, Capital Medical University, Beijing, China
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
28
|
Patel S, Zissimopoulos S, Marchant JS. Endo-Lysosomal Two-Pore Channels and Their Protein Partners. Handb Exp Pharmacol 2023; 278:199-214. [PMID: 35902438 DOI: 10.1007/164_2022_601] [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] [Indexed: 04/28/2023]
Abstract
Two-pore channels are ion channels expressed on acidic organelles such as the various vesicles that constitute the endo-lysosomal system. They are permeable to Ca2+ and Na+ and activated by the second messenger NAADP as well as the phosphoinositide, PI(3,5)P2 and/or voltage. Here, we review the proteins that interact with these channels including recently identified NAADP receptors.
Collapse
Affiliation(s)
- Sandip Patel
- Department of Cell and Developmental Biology, University College London, London, UK.
| | | | - Jonathan S Marchant
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
29
|
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
|
30
|
Jaślan D, Ferro IF, Kudrina V, Yuan Y, Patel S, Grimm C. PI(3,5)P 2 and NAADP: Team players or lone warriors? - New insights into TPC activation modes. Cell Calcium 2023; 109:102675. [PMID: 36525777 DOI: 10.1016/j.ceca.2022.102675] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022]
Abstract
NAADP (nicotinic acid adenine dinucleotide phosphate) is a second messenger, releasing Ca2+ from acidic calcium stores such as endosomes and lysosomes. PI(3,5)P2 (phosphatidylinositol 3,5-bisphosphate) is a phospho-inositide, residing on endolysosomal membranes and likewise releasing Ca2+ from endosomes and lysosomes. Both compounds have been shown to activate endolysosomal two-pore channels (TPCs) in mammalian cells. However, their effects on ion permeability as demonstrated specifically for TPC2 differ. While PI(3,5)P2 elicits predominantly Na+-selective currents, NAADP increases the Ca2+ permeability of the channel. What happens when both compounds are applied simultaneously was unclear until recently.
Collapse
Affiliation(s)
- Dawid Jaślan
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Irene Flavia Ferro
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Veronika Kudrina
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Yu Yuan
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, United Kingdom
| | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, United Kingdom
| | - Christian Grimm
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany.
| |
Collapse
|
31
|
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
|
32
|
Prat Castro S, Kudrina V, Jaślan D, Böck J, Scotto Rosato A, Grimm C. Neurodegenerative Lysosomal Storage Disorders: TPC2 Comes to the Rescue! Cells 2022; 11:2807. [PMID: 36139381 PMCID: PMC9496660 DOI: 10.3390/cells11182807] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/01/2022] [Accepted: 09/06/2022] [Indexed: 12/24/2022] Open
Abstract
Lysosomal storage diseases (LSDs) resulting from inherited gene mutations constitute a family of disorders that disturb lysosomal degradative function leading to abnormal storage of macromolecular substrates. In most LSDs, central nervous system (CNS) involvement is common and leads to the progressive appearance of neurodegeneration and early death. A growing amount of evidence suggests that ion channels in the endolysosomal system play a crucial role in the pathology of neurodegenerative LSDs. One of the main basic mechanisms through which the endolysosomal ion channels regulate the function of the endolysosomal system is Ca2+ release, which is thought to be essential for intracellular compartment fusion, fission, trafficking and lysosomal exocytosis. The intracellular TRPML (transient receptor potential mucolipin) and TPC (two-pore channel) ion channel families constitute the main essential Ca2+-permeable channels expressed on endolysosomal membranes, and they are considered potential drug targets for the prevention and treatment of LSDs. Although TRPML1 activation has shown rescue effects on LSD phenotypes, its activity is pH dependent, and it is blocked by sphingomyelin accumulation, which is characteristic of some LSDs. In contrast, TPC2 activation is pH-independent and not blocked by sphingomyelin, potentially representing an advantage over TRPML1. Here, we discuss the rescue of cellular phenotypes associated with LSDs such as cholesterol and lactosylceramide (LacCer) accumulation or ultrastructural changes seen by electron microscopy, mediated by the small molecule agonist of TPC2, TPC2-A1-P, which promotes lysosomal exocytosis and autophagy. In summary, new data suggest that TPC2 is a promising target for the treatment of different types of LSDs such as MLIV, NPC1, and Batten disease, both in vitro and in vivo.
Collapse
Affiliation(s)
| | | | | | | | - Anna Scotto Rosato
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Christian Grimm
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-University, 80336 Munich, Germany
| |
Collapse
|
33
|
Two-pore channels: going with the flows. Biochem Soc Trans 2022; 50:1143-1155. [PMID: 35959977 PMCID: PMC9444070 DOI: 10.1042/bst20220229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 11/26/2022]
Abstract
In recent years, our understanding of the structure, mechanisms and functions of the endo-lysosomal TPC (two-pore channel) family have grown apace. Gated by the second messengers, NAADP and PI(3,5)P2, TPCs are an integral part of fundamental signal-transduction pathways, but their array and plasticity of cation conductances (Na+, Ca2+, H+) allow them to variously signal electrically, osmotically or chemically. Their relative tissue- and organelle-selective distribution, together with agonist-selective ion permeabilities provides a rich palette from which extracellular stimuli can choose. TPCs are emerging as mediators of immunity, cancer, metabolism, viral infectivity and neurodegeneration as this short review attests.
Collapse
|
34
|
Patel S, Yuan Y, Chen CC, Jaślan D, Gunaratne G, Grimm C, Rahman T, Marchant JS. Electrophysiology of Endolysosomal Two-Pore Channels: A Current Account. Cells 2022; 11:2368. [PMID: 35954212 PMCID: PMC9368155 DOI: 10.3390/cells11152368] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/20/2022] [Accepted: 07/20/2022] [Indexed: 12/10/2022] Open
Abstract
Two-pore channels TPC1 and TPC2 are ubiquitously expressed pathophysiologically relevant proteins that reside on endolysosomal vesicles. Here, we review the electrophysiology of these channels. Direct macroscopic recordings of recombinant TPCs expressed in enlarged lysosomes in mammalian cells or vacuoles in plants and yeast demonstrate gating by the Ca2+-mobilizing messenger NAADP and/or the lipid PI(3,5)P2. TPC currents are regulated by H+, Ca2+, and Mg2+ (luminal and/or cytosolic), as well as protein kinases, and they are impacted by single-nucleotide polymorphisms linked to pigmentation. Bisbenzylisoquinoline alkaloids, flavonoids, and several approved drugs demonstrably block channel activity. Endogenous TPC currents have been recorded from a number of primary cell types and cell lines. Many of the properties of endolysosomal TPCs are recapitulated upon rerouting channels to the cell surface, allowing more facile recording through conventional electrophysiological means. Single-channel analyses have provided high-resolution insight into both monovalent and divalent permeability. The discovery of small-molecule activators of TPC2 that toggle the ion selectivity from a Ca2+-permeable (NAADP-like) state to a Na+-selective (PI(3,5)P2-like) state explains discrepancies in the literature relating to the permeability of TPCs. Identification of binding proteins that confer NAADP-sensitive currents confirm that indirect, remote gating likely underpins the inconsistent observations of channel activation by NAADP.
Collapse
Affiliation(s)
- Sandip Patel
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK;
| | - Yu Yuan
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK;
| | - Cheng-Chang Chen
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei 100229, Taiwan;
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Dawid Jaślan
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians University, 80336 Munich, Germany; (D.J.); (C.G.)
| | - Gihan Gunaratne
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA; (G.G.); (J.S.M.)
| | - Christian Grimm
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians University, 80336 Munich, Germany; (D.J.); (C.G.)
| | - Taufiq Rahman
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, UK;
| | - Jonathan S. Marchant
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA; (G.G.); (J.S.M.)
| |
Collapse
|
35
|
He X, Kang Y, Chen L. Identification of ASPDH as a novel NAADP-binding protein. Biochem Biophys Res Commun 2022; 621:168-175. [DOI: 10.1016/j.bbrc.2022.06.100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 06/30/2022] [Indexed: 11/02/2022]
|
36
|
The Three Two-Pore Channel Subtypes from Rabbit Exhibit Distinct Sensitivity to Phosphoinositides, Voltage, and Extracytosolic pH. Cells 2022; 11:cells11132006. [PMID: 35805090 PMCID: PMC9265530 DOI: 10.3390/cells11132006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/17/2022] [Accepted: 06/19/2022] [Indexed: 01/14/2023] Open
Abstract
Two pore channels (TPCs) are implicated in vesicle trafficking, virus infection, and autophagy regulation. As Na+- or Ca2+-permeable channels, TPCs have been reported to be activated by NAADP, PI(3,5)P2, and/or high voltage. However, a comparative study on the function and regulation of the three mammalian TPC subtypes is currently lacking. Here, we used the electrophysiological recording of enlarged endolysosome vacuoles, inside-out and outside-out membrane patches to examine the three TPCs of rabbit (Oryctolagus cuniculus, or Oc) heterologously expressed in HEK293 cells. While PI(3,5)P2 evoked Na+ currents with a potency order of OcTPC1 > OcTPC3 > OcTPC2, only OcTPC2 displayed a strict dependence on PI(3,5)P2. Both OcTPC1 and OcTPC3 were activatable by PI3P and OcTPC3 was also activated by additional phosphoinositide species. While OcTPC2 was voltage-independent, OcTPC1 and OcTPC3 showed voltage dependence with OcTPC3 depending on high positive voltages. Finally, while OcTPC2 preferred a luminal pH of 4.6−6.0 in endolysosomes, OcTPC1 was strongly inhibited by extracytosolic pH 5.0 in both voltage-dependent and -independent manners, and OcTPC3 was inhibited by pH 6.0 but potentiated by pH 8.0. Thus, the three OcTPCs form phosphoinositide-activated Na+ channels with different ligand selectivity, voltage dependence, and extracytosolic pH sensitivity, which likely are optimally tuned for function in specific endolysosomal populations.
Collapse
|
37
|
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
|
38
|
Steiner P, Arlt E, Boekhoff I, Gudermann T, Zierler S. Two-Pore Channels Regulate Inter-Organellar Ca 2+ Homeostasis in Immune Cells. Cells 2022; 11:1465. [PMID: 35563771 PMCID: PMC9103377 DOI: 10.3390/cells11091465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/13/2022] [Accepted: 04/16/2022] [Indexed: 02/01/2023] Open
Abstract
Two-pore channels (TPCs) are ligand-gated cation-selective ion channels that are preserved in plant and animal cells. In the latter, TPCs are located in membranes of acidic organelles, such as endosomes, lysosomes, and endolysosomes. Here, we focus on the function of these unique ion channels in mast cells, which are leukocytes that mature from myeloid hematopoietic stem cells. The cytoplasm of these innate immune cells contains a large number of granules that comprise messenger substances, such as histamine and heparin. Mast cells, along with basophil granulocytes, play an essential role in anaphylaxis and allergic reactions by releasing inflammatory mediators. Signaling in mast cells is mainly regulated via the release of Ca2+ from the endoplasmic reticulum as well as from acidic compartments, such as endolysosomes. For the crosstalk of these organelles TPCs seem essential. Allergic reactions and anaphylaxis were previously shown to be associated with the endolysosomal two-pore channel TPC1. The release of histamine, controlled by intracellular Ca2+ signals, was increased upon genetic or pharmacologic TPC1 inhibition. Conversely, stimulation of TPC channel activity by one of its endogenous ligands, namely nicotinic adenine dinucleotide phosphate (NAADP) or phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2), were found to trigger the release of Ca2+ from the endolysosomes; thereby improving the effect of TPC1 on regulated mast cell degranulation. In this review we discuss the importance of TPC1 for regulating Ca2+ homeostasis in mast cells and the overall potential of TPC1 as a pharmacological target in anti-inflammatory therapy.
Collapse
Affiliation(s)
- Philip Steiner
- Institute of Pharmacology, Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria;
| | - Elisabeth Arlt
- Walther Straub Institute of Pharmacology and Toxicology, Medical Faculty, Ludwig Maximilians University Munich, 80336 Munich, Germany; (E.A.); (I.B.); (T.G.)
| | - Ingrid Boekhoff
- Walther Straub Institute of Pharmacology and Toxicology, Medical Faculty, Ludwig Maximilians University Munich, 80336 Munich, Germany; (E.A.); (I.B.); (T.G.)
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Medical Faculty, Ludwig Maximilians University Munich, 80336 Munich, Germany; (E.A.); (I.B.); (T.G.)
| | - Susanna Zierler
- Institute of Pharmacology, Medical Faculty, Johannes Kepler University Linz, 4020 Linz, Austria;
- Walther Straub Institute of Pharmacology and Toxicology, Medical Faculty, Ludwig Maximilians University Munich, 80336 Munich, Germany; (E.A.); (I.B.); (T.G.)
| |
Collapse
|
39
|
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]
|
40
|
Chen CC, Krogsaeter E, Kuo CY, Huang MC, Chang SY, Biel M. Endolysosomal cation channels point the way towards precision medicine of cancer and infectious diseases. Biomed Pharmacother 2022; 148:112751. [PMID: 35240524 DOI: 10.1016/j.biopha.2022.112751] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 11/02/2022] Open
Abstract
Infectious diseases and cancer are among the key medical challenges that humankind is facing today. A growing amount of evidence suggests that ion channels in the endolysosomal system play a crucial role in the pathology of both groups of diseases. The development of advanced patch-clamp technologies has allowed us to directly characterize ion fluxes through endolysosomal ion channels in their native environments. Endolysosomes are essential organelles for intracellular transport, digestion and metabolism, and maintenance of homeostasis. The endolysosomal ion channels regulate the function of the endolysosomal system through four basic mechanisms: calcium release, control of membrane potential, pH change, and osmolarity regulation. In this review, we put particular emphasis on the endolysosomal cation channels, including TPC2 and TRPML2, which are particularly important in monocyte function. We discuss existing endogenous and synthetic ligands of these channels and summarize current knowledge of their impact on channel activity and function in different cell types. Moreover, we summarize recent findings on the importance of TPC2 and TRPML2 channels as potential drug targets for the prevention and treatment of the emerging infectious diseases and cancer.
Collapse
Affiliation(s)
- Cheng-Chang Chen
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| | | | - Ching-Ying Kuo
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Min-Chuan Huang
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Sui-Yuan Chang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Martin Biel
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
41
|
Regulation of Aging and Longevity by Ion Channels and Transporters. Cells 2022; 11:cells11071180. [PMID: 35406743 PMCID: PMC8997527 DOI: 10.3390/cells11071180] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/22/2022] [Accepted: 03/29/2022] [Indexed: 12/10/2022] Open
Abstract
Despite significant advances in our understanding of the mechanisms that underlie age-related physiological decline, our ability to translate these insights into actionable strategies to extend human healthspan has been limited. One of the major reasons for the existence of this barrier is that with a few important exceptions, many of the proteins that mediate aging have proven to be undruggable. The argument put forth here is that the amenability of ion channels and transporters to pharmacological manipulation could be leveraged to develop novel therapeutic strategies to combat aging. This review delves into the established roles for ion channels and transporters in the regulation of aging and longevity via their influence on membrane excitability, Ca2+ homeostasis, mitochondrial and endolysosomal function, and the transduction of sensory stimuli. The goal is to provide the reader with an understanding of emergent themes, and prompt further investigation into how the activities of ion channels and transporters sculpt the trajectories of cellular and organismal aging.
Collapse
|
42
|
Marchant JS, Gunaratne GS, Cai X, Slama JT, Patel S. NAADP-binding proteins find their identity. Trends Biochem Sci 2022; 47:235-249. [PMID: 34810081 PMCID: PMC8840967 DOI: 10.1016/j.tibs.2021.10.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023]
Abstract
Nicotinic acid adenine dinucleotide phosphate (NAADP) is a second messenger that releases Ca2+ from endosomes and lysosomes by activating ion channels called two-pore channels (TPCs). However, no NAADP-binding site has been identified on TPCs. Rather, NAADP activates TPCs indirectly by engaging NAADP-binding proteins (NAADP-BPs) that form part of the TPC complex. After a decade of searching, two different NAADP-BPs were recently identified: Jupiter microtubule associated homolog 2 (JPT2) and like-Sm protein 12 (LSM12). These discoveries bridge the gap between NAADP generation and NAADP activation of TPCs, providing new opportunity to understand and manipulate the NAADP-signaling pathway. The unmasking of these NAADP-BPs will catalyze future studies to define the molecular choreography of NAADP action.
Collapse
Affiliation(s)
- Jonathan S. Marchant
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA,Correspondence: (J.S. Marchant) and (S. Patel)
| | - Gihan S. Gunaratne
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Xinjiang Cai
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - James T. Slama
- Department of Medicinal and Biological Chemistry, University of Toledo College of Pharmacy and Pharmaceutical Sciences, 3000 Arlington Avenue, Toledo, OH 43614, USA
| | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
43
|
Krogsaeter E, Rosato AS, Grimm C. TRPMLs and TPCs: targets for lysosomal storage and neurodegenerative disease therapy? Cell Calcium 2022; 103:102553. [DOI: 10.1016/j.ceca.2022.102553] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/04/2022] [Accepted: 02/04/2022] [Indexed: 12/25/2022]
|
44
|
Wu Y, Xu M, Wang P, Syeda AKR, Huang P, Dong XP. Lysosomal potassium channels. Cell Calcium 2022; 102:102536. [PMID: 35016151 DOI: 10.1016/j.ceca.2022.102536] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/21/2022]
Abstract
The lysosome is an important membrane-bound acidic organelle that is regarded as the degradative center as well as multifunctional signaling hub. It digests unwanted macromolecules, damaged organelles, microbes, and other materials derived from endocytosis, autophagy, and phagocytosis. To function properly, the ionic homeostasis and membrane potential of the lysosome are strictly regulated by transporters and ion channels. As the most abundant cation inside the cell, potassium ions (K+) are vital for lysosomal membrane potential and lysosomal calcium (Ca2+) signaling. However, our understanding about how lysosomal K+homeostasis is regulated and what are the functions of K+in the lysosome is very limited. Currently, two lysosomal K+channels have been identified: large-conductance Ca2+-activated K+channel (BK) and transmembrane Protein 175 (TMEM175). In this review, we summarize recent development in our understanding of K+ homeostasis and K+channels in the lysosome. We hope to guide the readers into a more in-depth discussion of lysosomal K+ channels in lysosomal physiology and human diseases.
Collapse
Affiliation(s)
- Yi Wu
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Rd, Shanghai 201318, China; School of Pharmacy, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Rd, Shanghai 201318, China
| | - Mengnan Xu
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, Halifax, Nova NS B3H 4R2, Canada
| | - Pingping Wang
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, Halifax, Nova NS B3H 4R2, Canada
| | - Alia Kazim Rizvi Syeda
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, Halifax, Nova NS B3H 4R2, Canada
| | - Peng Huang
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Rd, Shanghai 201318, China; School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Rd, Shanghai 201318, China.
| | - Xian-Ping Dong
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, Halifax, Nova NS B3H 4R2, Canada.
| |
Collapse
|
45
|
Activation of endo-lysosomal two-pore channels by NAADP and PI(3,5)P2. Five things to know. Cell Calcium 2022; 103:102543. [PMID: 35123238 PMCID: PMC9552313 DOI: 10.1016/j.ceca.2022.102543] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 12/31/2022]
Abstract
Two-pore channels are ancient members of the voltage-gated ion channel superfamily that are expressed predominantly on acidic organelles such as endosomes and lysosomes. Here we review recent advances in understanding how TPCs are activated by their ligands and identify five salient features: (1) TPCs are Ca2+-permeable non-selective cation channels gated by NAADP. (2) NAADP activation is indirect through associated NAADP receptors. (3) TPCs are also Na+-selective channels gated by PI(3,5)P2. (4) PI(3,5)P2 activation is direct through a structurally-resolved binding site. (5) TPCs switch their ion selectivity in an agonist-dependent manner.
Collapse
|
46
|
Gan N, Jiang Y. Structural biology of cation channels important for lysosomal calcium release. Cell Calcium 2022; 101:102519. [PMID: 34952412 PMCID: PMC8752501 DOI: 10.1016/j.ceca.2021.102519] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 01/03/2023]
Abstract
Calcium is one of the most important second messengers in cells. The uptake and release of calcium ions are conducted by channels and transporters. Inside a eukaryotic cell, calcium is stored in intracellular organelles including the endoplasmic reticulum (ER), mitochondrion, and lysosome. Lysosomes are acid membrane-bounded organelles serving as the crucial degradation and recycling center of the cell. Lysosomes involve in multiple important signaling events, including nutrient sensing, lipid metabolism, and trafficking. Hitherto, two lysosomal cation channel families have been suggested to function as calcium release channels, namely the Two-pore Channel (TPC) family, and the Transient Receptor Potential Channel Mucolipin (TRPML) family. Additionally, a few plasma membrane calcium channels have also been found in the lysosomal membrane under certain circumstances. In this review, we will discuss the structural mechanism of the cation channels that may be important for lysosomal calcium release, primarily focusing on the TPCs and TRPMLs.
Collapse
Affiliation(s)
- Ninghai Gan
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA,Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390 USA,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390 USA
| | - Youxing Jiang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA,Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390 USA,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390 USA
| |
Collapse
|
47
|
Davis LC, Morgan AJ, Galione A. Acidic Ca 2+ stores and immune-cell function. Cell Calcium 2021; 101:102516. [PMID: 34922066 DOI: 10.1016/j.ceca.2021.102516] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 12/11/2022]
Abstract
Acidic organelles act as intracellular Ca2+ stores; they actively sequester Ca2+ in their lumina and release it to the cytosol upon activation of endo-lysosomal Ca2+ channels. Recent data suggest important roles of endo-lysosomal Ca2+ channels, the Two-Pore Channels (TPCs) and the TRPML channels (mucolipins), in different aspects of immune-cell function, particularly impacting membrane trafficking, vesicle fusion/fission and secretion. Remarkably, different channels on the same acidic vesicles can couple to different downstream physiology. Endo-lysosomal Ca2+ stores can act under different modalities, be they acting alone (via local Ca2+ nanodomains around TPCs/TRPMLs) or in conjunction with the ER Ca2+ store (to either promote or suppress global ER Ca2+ release). These different modalities impinge upon functions as broad as phagocytosis, cell-killing, anaphylaxis, immune memory, thrombostasis, and chemotaxis.
Collapse
Affiliation(s)
- Lianne C Davis
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK.
| | - Anthony J Morgan
- 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.
| |
Collapse
|
48
|
Li ZH, King TP, Ayong L, Asady B, Cai X, Rahman T, Vella SA, Coppens I, Patel S, Moreno SNJ. A plastid two-pore channel essential for inter-organelle communication and growth of Toxoplasma gondii. Nat Commun 2021; 12:5802. [PMID: 34608145 PMCID: PMC8490419 DOI: 10.1038/s41467-021-25987-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/06/2021] [Indexed: 01/04/2023] Open
Abstract
Two-pore channels (TPCs) are a ubiquitous family of cation channels that localize to acidic organelles in animals and plants to regulate numerous Ca2+-dependent events. Little is known about TPCs in unicellular organisms despite their ancient origins. Here, we characterize a TPC from Toxoplasma gondii, the causative agent of toxoplasmosis. TgTPC is a member of a novel clad of TPCs in Apicomplexa, distinct from previously identified TPCs and only present in coccidians. We show that TgTPC localizes not to acidic organelles but to the apicoplast, a non-photosynthetic plastid found in most apicomplexan parasites. Conditional silencing of TgTPC resulted in progressive loss of apicoplast integrity, severely affecting growth and the lytic cycle. Isolation of TPC null mutants revealed a selective role for TPCs in replication independent of apicoplast loss that required conserved residues within the pore-lining region. Using a genetically-encoded Ca2+ indicator targeted to the apicoplast, we show that Ca2+ signals deriving from the ER but not from the extracellular space are selectively transmitted to the lumen. Deletion of the TgTPC gene caused reduced apicoplast Ca2+ uptake and membrane contact site formation between the apicoplast and the ER. Fundamental roles for TPCs in maintaining organelle integrity, inter-organelle communication and growth emerge.
Collapse
Affiliation(s)
- Zhu-Hong Li
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, 30602, USA
| | - Thayer P King
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, 30602, USA
| | - Lawrence Ayong
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, 30602, USA
| | - Beejan Asady
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, MD, 21205, USA
| | - Xinjiang Cai
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Taufiq Rahman
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, USA
| | - Stephen A Vella
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, 30602, USA
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, MD, 21205, USA
| | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK
| | - Silvia N J Moreno
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, 30602, USA. .,Department of Cellular Biology, University of Georgia, Athens, GA, 30602, USA.
| |
Collapse
|
49
|
Abstract
Nicotinic acid adenine dinucleotide 2'-phosphate (NAADP) is a naturally occurring nucleotide that has been shown to be involved in the release of Ca2+ from intracellular stores in a wide variety of cell types, tissues and organisms. Current evidence suggests that NAADP may function as a trigger to initiate a Ca2+ signal that is then amplified by other Ca2+ release mechanisms. A fundamental question that remains unanswered is the identity of the NAADP receptor. Our recent studies have identified HN1L/JPT2 as a high affinity NAADP binding protein that is essential for the modulation of Ca2+ channels.
Collapse
Affiliation(s)
- Timothy F Walseth
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Andreas H Guse
- The Calcium Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
50
|
Jin X, Zhang Y, Alharbi A, Hanbashi A, Alhoshani A, Parrington J. Targeting Two-Pore Channels: Current Progress and Future Challenges. Trends Pharmacol Sci 2021; 41:582-594. [PMID: 32679067 PMCID: PMC7365084 DOI: 10.1016/j.tips.2020.06.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/04/2020] [Accepted: 06/04/2020] [Indexed: 12/19/2022]
Abstract
Two-pore channels (TPCs) are cation-permeable channels located on endolysosomal membranes and important mediators of intracellular Ca2+ signalling. TPCs are involved in various pathophysiological processes, including cell growth and development, metabolism, and cancer progression. Most studies of TPCs have used TPC–/– cell or whole-animal models, or Ned-19, an indirect inhibitor. The TPC activation mechanism remains controversial, which has made it difficult to develop selective modulators. Recent studies of TPC structure and their interactomes are aiding the development of direct pharmacological modulators. This process is still in its infancy, but will facilitate future research and TPC targeting for therapeutical purposes. Here, we review the progress of current research into TPCs, including recent insights into their structures, functional roles, mechanisms of activation, and pharmacological modulators. Two-pore channel (TPC)-mediated endolysosomal Ca2+ signalling regulates a variety of processes, including cell proliferation, differentiation, metabolism, viral infection, and cardiac function. Despite the well-established model that TPCs are Ca2+-selective channels indirectly activated by nicotinic acid adenine dinucleotide phosphate (NAADP), it has also been proposed that TPCs as Na+ channels are activated directly by phosphatidylinositol 3,5-bisphosphate [PI(3,5)P2]. 3D structures of mouse TPC1 and human TPC2 were recently determined, which made it possible for structure-based virtual screening methods to identify pharmacological modulators of TPC. Recent identification by high-throughput screens of pharmacological modulators that target TPCs will help reveal the molecular mechanisms underlying the role of endolysosomal Ca2+ signalling in different pathophysiological processes, and to develop new therapeutics.
Collapse
Affiliation(s)
- Xuhui Jin
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Yuxuan Zhang
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Abeer Alharbi
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Ali Hanbashi
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Ali Alhoshani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, PO Box 2457, Riyadh 11454, Kingdom of Saudi Arabia
| | - John Parrington
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK.
| |
Collapse
|