1
|
Moccia F, Fiorio Pla A, Lim D, Lodola F, Gerbino A. Intracellular Ca 2+ signalling: unexpected new roles for the usual suspect. Front Physiol 2023; 14:1210085. [PMID: 37576340 PMCID: PMC10413985 DOI: 10.3389/fphys.2023.1210085] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/18/2023] [Indexed: 08/15/2023] Open
Abstract
Cytosolic Ca2+ signals are organized in complex spatial and temporal patterns that underlie their unique ability to regulate multiple cellular functions. Changes in intracellular Ca2+ concentration ([Ca2+]i) are finely tuned by the concerted interaction of membrane receptors and ion channels that introduce Ca2+ into the cytosol, Ca2+-dependent sensors and effectors that translate the elevation in [Ca2+]i into a biological output, and Ca2+-clearing mechanisms that return the [Ca2+]i to pre-stimulation levels and prevent cytotoxic Ca2+ overload. The assortment of the Ca2+ handling machinery varies among different cell types to generate intracellular Ca2+ signals that are selectively tailored to subserve specific functions. The advent of novel high-speed, 2D and 3D time-lapse imaging techniques, single-wavelength and genetic Ca2+ indicators, as well as the development of novel genetic engineering tools to manipulate single cells and whole animals, has shed novel light on the regulation of cellular activity by the Ca2+ handling machinery. A symposium organized within the framework of the 72nd Annual Meeting of the Italian Society of Physiology, held in Bari on 14-16th September 2022, has recently addressed many of the unexpected mechanisms whereby intracellular Ca2+ signalling regulates cellular fate in healthy and disease states. Herein, we present a report of this symposium, in which the following emerging topics were discussed: 1) Regulation of water reabsorption in the kidney by lysosomal Ca2+ release through Transient Receptor Potential Mucolipin 1 (TRPML1); 2) Endoplasmic reticulum-to-mitochondria Ca2+ transfer in Alzheimer's disease-related astroglial dysfunction; 3) The non-canonical role of TRP Melastatin 8 (TRPM8) as a Rap1A inhibitor in the definition of some cancer hallmarks; and 4) Non-genetic optical stimulation of Ca2+ signals in the cardiovascular system.
Collapse
Affiliation(s)
- Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | | | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, Novara, Italy
| | - Francesco Lodola
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan, Italy
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, Milan, Italy
| | - Andrea Gerbino
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
2
|
Scorza SI, Milano S, Saponara I, Certini M, De Zio R, Mola MG, Procino G, Carmosino M, Moccia F, Svelto M, Gerbino A. TRPML1-Induced Lysosomal Ca 2+ Signals Activate AQP2 Translocation and Water Flux in Renal Collecting Duct Cells. Int J Mol Sci 2023; 24:ijms24021647. [PMID: 36675161 PMCID: PMC9861594 DOI: 10.3390/ijms24021647] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/04/2023] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
Lysosomes are acidic Ca2+ storage organelles that actively generate local Ca2+ signaling events to regulate a plethora of cell functions. Here, we characterized lysosomal Ca2+ signals in mouse renal collecting duct (CD) cells and we assessed their putative role in aquaporin 2 (AQP2)-dependent water reabsorption. Bafilomycin A1 and ML-SA1 triggered similar Ca2+ oscillations, in the absence of extracellular Ca2+, by alkalizing the acidic lysosomal pH or activating the lysosomal cation channel mucolipin 1 (TRPML1), respectively. TRPML1-dependent Ca2+ signals were blocked either pharmacologically or by lysosomes' osmotic permeabilization, thus indicating these organelles as primary sources of Ca2+ release. Lysosome-induced Ca2+ oscillations were sustained by endoplasmic reticulum (ER) Ca2+ content, while bafilomycin A1 and ML-SA1 did not directly interfere with ER Ca2+ homeostasis per se. TRPML1 activation strongly increased AQP2 apical expression and depolymerized the actin cytoskeleton, thereby boosting water flux in response to an hypoosmotic stimulus. These effects were strictly dependent on the activation of the Ca2+/calcineurin pathway. Conversely, bafilomycin A1 led to perinuclear accumulation of AQP2 vesicles without affecting water permeability. Overall, lysosomal Ca2+ signaling events can be differently decoded to modulate Ca2+-dependent cellular functions related to the dock/fusion of AQP2-transporting vesicles in principal cells of the CD.
Collapse
Affiliation(s)
- Simona Ida Scorza
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Serena Milano
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Ilenia Saponara
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Maira Certini
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Roberta De Zio
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Maria Grazia Mola
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Giuseppe Procino
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Monica Carmosino
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy
| | - Francesco Moccia
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, 27100 Pavia, Italy
| | - Maria Svelto
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Andrea Gerbino
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
- Correspondence: ; Tel.: +39-0805443334
| |
Collapse
|
3
|
Steiner P, Arlt E, Boekhoff I, Gudermann T, Zierler S. TPC Functions in the Immune System. Handb Exp Pharmacol 2023; 278:71-92. [PMID: 36639434 DOI: 10.1007/164_2022_634] [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) are novel intracellular cation channels, which play a key role in numerous (patho-)physiological and immunological processes. In this chapter, we focus on their function in immune cells and immune reactions. Therefore, we first give an overview of the cellular immune response and the partaking immune cells. Second, we concentrate on ion channels which in the past have been shown to play an important role in the regulation of immune cells. The main focus is then directed to TPCs, which are primarily located in the membranes of acidic organelles, such as lysosomes or endolysosomes but also certain other vesicles. They regulate Ca2+ homeostasis and thus Ca2+ signaling in immune cells. Due to this important functional role, TPCs are enjoying increasing attention within the field of immunology in the last few decades but are also becoming more pertinent as pharmacological targets for the treatment of pro-inflammatory diseases such as allergic hypersensitivity. However, to uncover the precise molecular mechanism of TPCs in immune cell responses, further molecular, genetic, and ultrastructural investigations on TPCs are necessary, which then may pave the way to develop novel therapeutic strategies to treat diseases such as anaphylaxis more specifically.
Collapse
Affiliation(s)
- Philip Steiner
- Institute of Pharmacology, Faculty of Medicine, Johannes Kepler University Linz, Linz, Austria
| | - Elisabeth Arlt
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ingrid Boekhoff
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Susanna Zierler
- Institute of Pharmacology, Faculty of Medicine, Johannes Kepler University Linz, Linz, Austria.
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
4
|
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
|
5
|
Zajac M, Modi S, Krishnan Y. The evolution of organellar calcium mapping technologies. Cell Calcium 2022; 108:102658. [PMID: 36274564 PMCID: PMC10224794 DOI: 10.1016/j.ceca.2022.102658] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/05/2022] [Accepted: 10/08/2022] [Indexed: 01/25/2023]
Abstract
Intracellular Ca2+ fluxes are dynamically controlled by the co-involvement of multiple organellar pools of stored Ca2+. Endolysosomes are emerging as physiologically critical, yet underexplored, sources and sinks of intracellular Ca2+. Delineating the role of organelles in Ca2+ signaling has relied on chemical fluorescent probes and electrophysiological strategies. However, the acidic endolysosomal environment presents unique issues, which preclude the use of traditional chemical reporter strategies to map lumenal Ca2+. Here, we broadly address the current state of knowledge about organellar Ca2+ pools. We then outline the application of traditional probes, and their sensing paradigms. We then discuss how a new generation of probes overcomes the limitations of traditional Ca2+probes, emphasizing their ability to offer critical insights into endolysosomal Ca2+, and its feedback with other organellar pools.
Collapse
Affiliation(s)
- Matthew Zajac
- Department of Chemistry, The University of Chicago, Chicago, Illinois, 60637, USA; Neuroscience Institute, The University of Chicago, Chicago, IL, 60637, USA
| | - Souvik Modi
- Esya Labs, Translation and Innovation Hub, Imperial College White City Campus, 84 Wood Lane, London, W12 0BZ, UK
| | - Yamuna Krishnan
- Department of Chemistry, The University of Chicago, Chicago, Illinois, 60637, USA; Neuroscience Institute, The University of Chicago, Chicago, IL, 60637, USA; Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois, 60637, USA.
| |
Collapse
|
6
|
Scotto Rosato A, Krogsaeter EK, Jaślan D, Abrahamian C, Montefusco S, Soldati C, Spix B, Pizzo MT, Grieco G, Böck J, Wyatt A, Wünkhaus D, Passon M, Stieglitz M, Keller M, Hermey G, Markmann S, Gruber-Schoffnegger D, Cotman S, Johannes L, Crusius D, Boehm U, Wahl-Schott C, Biel M, Bracher F, De Leonibus E, Polishchuk E, Medina DL, Paquet D, Grimm C. TPC2 rescues lysosomal storage in mucolipidosis type IV, Niemann-Pick type C1, and Batten disease. EMBO Mol Med 2022; 14:e15377. [PMID: 35929194 PMCID: PMC9449600 DOI: 10.15252/emmm.202115377] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 01/05/2023] Open
Abstract
Lysosomes are cell organelles that degrade macromolecules to recycle their components. If lysosomal degradative function is impaired, e.g., due to mutations in lysosomal enzymes or membrane proteins, lysosomal storage diseases (LSDs) can develop. LSDs manifest often with neurodegenerative symptoms, typically starting in early childhood, and going along with a strongly reduced life expectancy and quality of life. We show here that small molecule activation of the Ca2+‐permeable endolysosomal two‐pore channel 2 (TPC2) results in an amelioration of cellular phenotypes associated with LSDs such as cholesterol or lipofuscin accumulation, or the formation of abnormal vacuoles seen by electron microscopy. Rescue effects by TPC2 activation, which promotes lysosomal exocytosis and autophagy, were assessed in mucolipidosis type IV (MLIV), Niemann–Pick type C1, and Batten disease patient fibroblasts, and in neurons derived from newly generated isogenic human iPSC models for MLIV and Batten disease. For in vivo proof of concept, we tested TPC2 activation in the MLIV mouse model. In sum, our data suggest that TPC2 is a promising target for the treatment of different types of LSDs, both in vitro and in‐vivo.
Collapse
Affiliation(s)
- Anna Scotto Rosato
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Einar K Krogsaeter
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Dawid Jaślan
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Carla Abrahamian
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | | | - Chiara Soldati
- Telethon Institute of Genetics and Medicine, Naples, Italy
| | - Barbara Spix
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | | | | | - Julia Böck
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Amanda Wyatt
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | | | - Marcel Passon
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Marc Stieglitz
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität, Munich, Germany
| | - Marco Keller
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität, Munich, Germany
| | - Guido Hermey
- Center for Molecular Neurobiology Hamburg (ZMNH), Institute of Molecular and Cellular Cognition, UKE, Hamburg, Germany
| | | | | | - Susan Cotman
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ludger Johannes
- Cellular and Chemical Biology Department, Institut Curie, U1143 INSERM, UMR3666 CNRS, PSL Research University, Paris, France
| | - Dennis Crusius
- Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-University (LMU) Hospital, Munich, Germany
| | - Ulrich Boehm
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | | | - Martin Biel
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität, Munich, Germany
| | - Franz Bracher
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität, Munich, Germany
| | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine, Naples, Italy.,Institute of Biochemistry and Cell Biology (IBBC), CNR, Rome, Italy
| | | | - Diego L Medina
- Telethon Institute of Genetics and Medicine, Naples, Italy.,Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Naples, Italy
| | - Dominik Paquet
- Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-University (LMU) Hospital, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Christian Grimm
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| |
Collapse
|
7
|
Zhu F, Miao Y, Cheng M, Ye X, Chen A, Zheng G, Tian X. The CACNA1A Mutant Disrupts Lysosome Calcium Homeostasis in Cerebellar Neurons and the Resulting Endo-Lysosomal Fusion Defect Can be Improved by Calcium Modulation. Neurochem Res 2021; 47:249-263. [PMID: 34476720 DOI: 10.1007/s11064-021-03438-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 11/24/2022]
Abstract
Mutations in P/Q type voltage gated calcium channel (VGCC) lead severe human neurological diseases such as episodic ataxia 2, familial hemiplegic migraine 1, absence epilepsy, progressive ataxia and spinocerebellar ataxia 6. The pathogenesis of these diseases remains unclear. Mice with spontaneous mutation in the Cacna1a gene encoding the pore-forming subunit of P/Q type VGCC also exhibit ataxia, epilepsy and neurodegeneration. Based on the previous work showing that the P/Q type VGCC in neurons regulates lysosomal fusion through its calcium channel activity on lysosomes, we utilized CACNA1A mutant mice to further investigate the mechanism by which P/Q-type VGCCs regulate lysosomal function and neuronal homeostasis. We found CACNA1A mutant neurons have reduced lysosomal calcium storage without changing the resting calcium concentration in cytoplasm and the acidification of lysosomes. Immunohistochemistry and transmission electron microscopy reveal axonal degeneration due to lysosome dysfunction in the CACNA1A mutant cerebella. The calcium modulating drug thapsigargin, by depleting the ER calcium store, which locally increases the calcium concentration can alleviate the defective lysosomal fusion in mutant neurons. We propose a model that in cerebellar neurons, P/Q-type VGCC maintains the integrity of the nervous system by regulating lysosomal calcium homeostasis to affect lysosomal fusion, which in turn regulates multiple important cellular processes such as autophagy and endocytosis. This study helps us to better understand the pathogenesis of P/Q-type VGCC related neurodegenerative diseases and provides a feasible direction for future pharmacological treatment.
Collapse
Affiliation(s)
- Feng Zhu
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, 310013, China.,Department of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Yunping Miao
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, 310013, China.,Department of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Min Cheng
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, 310013, China.,Department of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Xiaodi Ye
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, 310013, China.,Department of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Aiying Chen
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, 310013, China.,Department of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Gaoli Zheng
- National Zhejiang Center for Safety Evaluation of New Drugs, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, 310013, China
| | - Xuejun Tian
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, 310013, China. .,Department of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China. .,Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
8
|
Boley N, Patil S, Garnett EO, Li H, Chugani DC, Chang SE, Chow HM. Association Between Gray Matter Volume Variations and Energy Utilization in the Brain: Implications for Developmental Stuttering. JOURNAL OF SPEECH, LANGUAGE, AND HEARING RESEARCH : JSLHR 2021; 64:2317-2324. [PMID: 33719533 PMCID: PMC8740693 DOI: 10.1044/2020_jslhr-20-00325] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/23/2020] [Accepted: 10/02/2020] [Indexed: 06/12/2023]
Abstract
Purpose The biological mechanisms underlying developmental stuttering remain unclear. In a previous investigation, we showed that there is significant spatial correspondence between regional gray matter structural anomalies and the expression of genes linked to energy metabolism. In the current study, we sought to further examine the relationship between structural anomalies in the brain in children with persistent stuttering and brain regional energy metabolism. Method High-resolution structural MRI scans were acquired from 26 persistent stuttering and 44 typically developing children. Voxel-based morphometry was used to quantify the between-group gray matter volume (GMV) differences across the whole brain. Group differences in GMV were then compared with published values for the pattern of glucose metabolism measured via F18 fluorodeoxyglucose uptake in the brains of 29 healthy volunteers using positron emission tomography. Results A significant positive correlation between GMV differences and F18 fluorodeoxyglucose uptake was found in the left hemisphere (ρ = .36, p < .01), where speech-motor and language processing are typically localized. No such correlation was observed in the right hemisphere (ρ = .05, p = .70). Conclusions Corroborating our previous gene expression studies, the results of the current study suggest a potential connection between energy metabolism and stuttering. Brain regions with high energy utilization may be particularly vulnerable to anatomical changes associated with stuttering. Such changes may be further exacerbated when there are sharp increases in brain energy utilization, which coincides with the developmental period of rapid speech/language acquisition and the onset of stuttering during childhood. Supplemental Material https://doi.org/10.23641/asha.14110454.
Collapse
Affiliation(s)
- Nathaniel Boley
- The Institute for Biomedical Sciences, School of Medicine and Health Sciences, The George Washington University, Washington, DC
- Katzin Diagnostic & Research PET/MRI Center, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE
| | - Sanath Patil
- Katzin Diagnostic & Research PET/MRI Center, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE
- Premedical-Medical Program, Eberly College of Science, The Pennsylvania State University, University Park
| | - Emily O. Garnett
- Department of Psychiatry, University of Michigan Medical School, Ann Arbor
| | - Hua Li
- Katzin Diagnostic & Research PET/MRI Center, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE
| | - Diane C. Chugani
- Department of Communication Sciences and Disorders, College of Health Sciences, University of Delaware, Newark
| | - Soo-Eun Chang
- Department of Psychiatry, University of Michigan Medical School, Ann Arbor
- Cognitive Imaging Research Center, Department of Radiology, College of Osteopathic Medicine, Michigan State University, East Lansing
- Department of Communicative Sciences and Disorders, College of Communication Arts and Sciences, Michigan State University, East Lansing
| | - Ho Ming Chow
- Katzin Diagnostic & Research PET/MRI Center, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE
- Department of Communication Sciences and Disorders, College of Health Sciences, University of Delaware, Newark
| |
Collapse
|
9
|
Ordway B, Gillies RJ, Damaghi M. Extracellular Acidification Induces Lysosomal Dysregulation. Cells 2021; 10:1188. [PMID: 34067971 PMCID: PMC8152284 DOI: 10.3390/cells10051188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/05/2021] [Accepted: 05/09/2021] [Indexed: 02/05/2023] Open
Abstract
Many invasive cancers emerge through a years-long process of somatic evolution, characterized by an accumulation of heritable genetic and epigenetic changes and the emergence of increasingly aggressive clonal populations. In solid tumors, such as breast ductal carcinoma, the extracellular environment for cells within the nascent tumor is harsh and imposes different types of stress on cells, such as hypoxia, nutrient deprivation, and cytokine inflammation. Acidosis is a constant stressor of most cancer cells due to its production through fermentation of glucose to lactic acid in hypoxic or normoxic regions (Warburg effect). Over a short period of time, acid stress can have a profound effect on the function of lysosomes within the cells exposed to this environment, and after long term exposure, lysosomal function of the cancer cells can become completely dysregulated. Whether this dysregulation is due to an epigenetic change or evolutionary selection has yet to be determined, but understanding the mechanisms behind this dysregulation could identify therapeutic opportunities.
Collapse
Affiliation(s)
- Bryce Ordway
- Department of Cancer Physiology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (B.O.); (R.J.G.)
| | - Robert J. Gillies
- Department of Cancer Physiology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (B.O.); (R.J.G.)
| | - Mehdi Damaghi
- Department of Cancer Physiology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (B.O.); (R.J.G.)
- Department of Oncological Sciences, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
10
|
Cui W, Sathyanarayan A, Lopresti M, Aghajan M, Chen C, Mashek DG. Lipophagy-derived fatty acids undergo extracellular efflux via lysosomal exocytosis. Autophagy 2021; 17:690-705. [PMID: 32070194 PMCID: PMC8032247 DOI: 10.1080/15548627.2020.1728097] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 01/31/2020] [Accepted: 02/06/2020] [Indexed: 12/16/2022] Open
Abstract
The autophagic degradation of lipid droplets (LDs), termed lipophagy, is a major mechanism that contributes to lipid turnover in numerous cell types. While numerous factors, including nutrient deprivation or overexpression of PNPLA2/ATGL (patatin-like phospholipase domain containing 2) drive lipophagy, the trafficking of fatty acids (FAs) produced from this pathway is largely unknown. Herein, we show that PNPLA2 and nutrient deprivation promoted the extracellular efflux of FAs. Inhibition of autophagy or lysosomal lipid degradation attenuated FA efflux highlighting a critical role for lipophagy in this process. Rather than direct transport of FAs across the lysosomal membrane, lipophagy-derived FA efflux requires lysosomal fusion to the plasma membrane. The lysosomal Ca2+ channel protein MCOLN1/TRPML1 (mucolipin 1) regulates lysosomal-plasma membrane fusion and its overexpression increased, while inhibition blocked FA efflux. In addition, inhibition of autophagy/lipophagy or MCOLN1, or sequestration of extracellular FAs with BSA attenuated the oxidation and re-esterification of lipophagy-derived FAs. Overall, these studies show that the well-established pathway of lysosomal fusion to the plasma membrane is the primary route for the disposal of FAs derived from lipophagy. Moreover, the efflux of FAs and their reuptake or subsequent extracellular trafficking to adjacent cells may play an important role in cell-to-cell lipid exchange and signaling.Abbreviations: ACTB: beta actin; ADRA1A: adrenergic receptor alpha, 1a; ALB: albumin; ATG5: autophagy related 5; ATG7: autophagy related 7; BafA1: bafilomycin A1; BECN1: beclin 1; BHBA: beta-hydroxybutyrate; BSA: bovine serum albumin; CDH1: e-cadherin; CQ: chloroquine; CTSB: cathepsin B; DGAT: diacylglycerol O-acyltransferase; FA: fatty acid; HFD: high-fat diet; LAMP1: lysosomal-associated membrane protein 1; LD: lipid droplet; LIPA/LAL: lysosomal acid lipase A; LLME: Leu-Leu methyl ester hydrobromide; MAP1LC3B/LC3: microtubule associated protein 1 light chain 3 beta; MCOLN1/TRPML1: mucolipin 1; MEF: mouse embryo fibroblast; PBS: phosphate-buffered saline; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; PLIN: perilipin; PNPLA2/ATGL patatin-like phospholipase domain containing 2; RUBCN (rubicon autophagy regulator); SM: sphingomyelin; TAG: triacylglycerol; TMEM192: transmembrane protein 192; VLDL: very low density lipoprotein.
Collapse
Affiliation(s)
- Wenqi Cui
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Aishwarya Sathyanarayan
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Michael Lopresti
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | | | - Chi Chen
- Department of Food Science and Nutrition, University of Minnesota, Minneapolis, MN, USA
| | - Douglas G. Mashek
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
11
|
Childers WE, Elokely KM, Abou-Gharbia M. The Resurrection of Phenotypic Drug Discovery. ACS Med Chem Lett 2020; 11:1820-1828. [PMID: 33062159 DOI: 10.1021/acsmedchemlett.0c00006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 03/06/2020] [Indexed: 12/12/2022] Open
Abstract
Prior to genetic mapping, the majority of drug discovery efforts involved phenotypic screening, wherein compounds were screened in either in vitro or in vivo models thought to mimic the disease state of interest. While never completely abandoning phenotypic approaches, the labor intensive nature of such tests encouraged the pharmaceutical industry to move away from them in favor of target-based drug discovery, which facilitated throughput and allowed for the efficient screening of large numbers of compounds. However, a consequence of reliance on target-based screening was an increased number of failures in clinical trials due to poor correlation between novel mechanistic targets and the actual disease state. As a result, the field has seen a recent resurrection in phenotypic drug discovery approaches. In this work, we highlight some recent phenotypic projects from our industrial past and in our current academic drug discovery environment that have provided encouraging results.
Collapse
Affiliation(s)
- Wayne E. Childers
- Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, 3307 N. Broad Street, Philadelphia, Pennsylvania 19140, United States
| | - Khaled M. Elokely
- Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, 3307 N. Broad Street, Philadelphia, Pennsylvania 19140, United States
- Department of Chemistry, College of Science and Technology, Temple University, 1925 N. 12th Street, Philadelphia, Pennsylvania 19122, United States
- Department of Pharmaceutical Chemistry, Tanta University, Tanta 31527, Egypt
| | - Magid Abou-Gharbia
- Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, 3307 N. Broad Street, Philadelphia, Pennsylvania 19140, United States
| |
Collapse
|
12
|
Childers W, Fan R, Martinez R, Colussi DJ, Melenski E, Liu Y, Gordon J, Abou-Gharbia M, Jacobson MA. Novel compounds that reverse the disease phenotype in Type 2 Gaucher disease patient-derived cells. Bioorg Med Chem Lett 2020; 30:126806. [PMID: 31757667 PMCID: PMC7569734 DOI: 10.1016/j.bmcl.2019.126806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/30/2019] [Accepted: 11/01/2019] [Indexed: 11/30/2022]
Abstract
Gaucher disease (GD) results from inherited mutations in the lysosomal enzyme β-glucocerobrosidase (GCase). Currently available treatment options for Type 1 GD are not efficacious for treating neuronopathic Type 2 and 3 GD due to their inability to cross the blood-brain barrier. In an effort to identify small molecules which could be optimized for CNS penetration we identified tamoxifen from a high throughput phenotypic screen on Type 2 GD patient-derived fibroblasts which reversed the disease phenotype. Structure activity studies around this scaffold led to novel molecules that displayed improved potency, efficacy and reduced estrogenic/antiestrogenic activity compared to the original hits. Here we present the design, synthesis and structure activity relationships that led to the lead molecule Compound 31.
Collapse
Affiliation(s)
- Wayne Childers
- Moulder Center for Drug Discovery Research, Department of Pharmaceutical Sciences, Temple University School of Pharmacy, 3307 N. Broad Street, Philadelphia, PA 19140, USA.
| | - Rong Fan
- Moulder Center for Drug Discovery Research, Department of Pharmaceutical Sciences, Temple University School of Pharmacy, 3307 N. Broad Street, Philadelphia, PA 19140, USA
| | - Rogelio Martinez
- Moulder Center for Drug Discovery Research, Department of Pharmaceutical Sciences, Temple University School of Pharmacy, 3307 N. Broad Street, Philadelphia, PA 19140, USA
| | - Dennis J Colussi
- Moulder Center for Drug Discovery Research, Department of Pharmaceutical Sciences, Temple University School of Pharmacy, 3307 N. Broad Street, Philadelphia, PA 19140, USA
| | - Edward Melenski
- Moulder Center for Drug Discovery Research, Department of Pharmaceutical Sciences, Temple University School of Pharmacy, 3307 N. Broad Street, Philadelphia, PA 19140, USA
| | - Yuxiao Liu
- Moulder Center for Drug Discovery Research, Department of Pharmaceutical Sciences, Temple University School of Pharmacy, 3307 N. Broad Street, Philadelphia, PA 19140, USA
| | - John Gordon
- Moulder Center for Drug Discovery Research, Department of Pharmaceutical Sciences, Temple University School of Pharmacy, 3307 N. Broad Street, Philadelphia, PA 19140, USA
| | - Magid Abou-Gharbia
- Moulder Center for Drug Discovery Research, Department of Pharmaceutical Sciences, Temple University School of Pharmacy, 3307 N. Broad Street, Philadelphia, PA 19140, USA
| | - Marlene A Jacobson
- Moulder Center for Drug Discovery Research, Department of Pharmaceutical Sciences, Temple University School of Pharmacy, 3307 N. Broad Street, Philadelphia, PA 19140, USA.
| |
Collapse
|
13
|
Kobolák J, Molnár K, Varga E, Bock I, Jezsó B, Téglási A, Zhou S, Lo Giudice M, Hoogeveen-Westerveld M, Pijnappel WP, Phanthong P, Varga N, Kitiyanant N, Freude K, Nakanishi H, László L, Hyttel P, Dinnyés A. Modelling the neuropathology of lysosomal storage disorders through disease-specific human induced pluripotent stem cells. Exp Cell Res 2019; 380:216-233. [PMID: 31039347 DOI: 10.1016/j.yexcr.2019.04.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/12/2019] [Accepted: 04/17/2019] [Indexed: 12/15/2022]
Abstract
Mucopolysaccharidosis II (MPS II) is a lysosomal storage disorder (LSD), caused by iduronate 2-sulphatase (IDS) enzyme dysfunction. The neuropathology of the disease is not well understood, although the neural symptoms are currently incurable. MPS II-patient derived iPSC lines were established and differentiated to neuronal lineage. The disease phenotype was confirmed by IDS enzyme and glycosaminoglycan assay. MPS II neuronal precursor cells (NPCs) showed significantly decreased self-renewal capacity, while their cortical neuronal differentiation potential was not affected. Major structural alterations in the ER and Golgi complex, accumulation of storage vacuoles, and increased apoptosis were observed both at protein expression and ultrastructural level in the MPS II neuronal cells, which was more pronounced in GFAP + astrocytes, with increased LAMP2 expression but unchanged in their RAB7 compartment. Based on these finding we hypothesize that lysosomal membrane protein (LMP) carrier vesicles have an initiating role in the formation of storage vacuoles leading to impaired lysosomal function. In conclusion, a novel human MPS II disease model was established for the first time which recapitulates the in vitro neuropathology of the disorder, providing novel information on the disease mechanism which allows better understanding of further lysosomal storage disorders and facilitates drug testing and gene therapy approaches.
Collapse
Affiliation(s)
| | - Kinga Molnár
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, 1117, Hungary
| | | | | | - Bálint Jezsó
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, 1117, Hungary
| | | | - Shuling Zhou
- BioTalentum Ltd., Gödöllő, 2100, Hungary; Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Copenhagen, Denmark
| | | | | | - Wwm Pim Pijnappel
- Department of Clinical Genetics, Erasmus MC Rotterdam, 3015 CN, Rotterdam, the Netherlands
| | - Phetcharat Phanthong
- BioTalentum Ltd., Gödöllő, 2100, Hungary; Institute of Molecular Biosciences, Mahidol University, Bangkok, 73170, Thailand
| | - Norbert Varga
- Department of Metabolic Diseases, Heim Pál Children's Hospital, Budapest, 1089, Hungary
| | - Narisorn Kitiyanant
- Institute of Molecular Biosciences, Mahidol University, Bangkok, 73170, Thailand
| | - Kristine Freude
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Copenhagen, Denmark
| | - Hideyuki Nakanishi
- Department of Macromolecular Science and Engineering, Graduate School of Science and Technology, Kyoto Institute of Technology, Matsugasaki, Kyoto, 606-8585, Japan
| | - Lajos László
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, 1117, Hungary
| | - Poul Hyttel
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Copenhagen, Denmark
| | - András Dinnyés
- BioTalentum Ltd., Gödöllő, 2100, Hungary; Molecular Animal Biotechnology Laboratory, Szent István University, Gödöllő, 2101, Hungary.
| |
Collapse
|
14
|
Colussi DJ, Jacobson MA. Patient-Derived Phenotypic High-Throughput Assay to Identify Small Molecules Restoring Lysosomal Function in Tay-Sachs Disease. SLAS DISCOVERY 2019; 24:295-303. [PMID: 30616450 DOI: 10.1177/2472555218814538] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tay-Sachs disease is an inherited lysosomal storage disease resulting from mutations in the lysosomal enzyme, β-hexosaminidase A, and leads to excessive accumulation of GM2 ganglioside. Tay-Sachs patients with the infantile form do not live beyond 2-4 years of age due to rapid, progressive neurodegeneration. Enzyme replacement therapy is not a therapeutic option due to its inability to cross the blood-brain barrier. As an alternative, small molecules identified from high-throughput screening could provide leads suitable for chemical optimization to target the central nervous system. We developed a new high-throughput phenotypic assay utilizing infantile Tay-Sachs patient cells based on disrupted lysosomal calcium signaling as a monitor of diseased phenotype. The assay was validated in a pilot screen on a collection of Food and Drug Administration-approved drugs to identify compounds that could reverse or attenuate the disease. Pyrimethamine, a known pharmacological chaperone of β-hexosaminidase A, was identified from the primary screen. The mechanism of action of pyrimethamine in reversing the defective lysosomal phenotype was by improving autophagy. This new high-throughput screening assay in patient cells will enable the screening of larger chemical compound collections. Importantly, this approach could lead to identification of new molecular targets previously unknown to impact the disease and accelerate the discovery of new treatments for Tay-Sachs disease.
Collapse
Affiliation(s)
- Dennis J Colussi
- 1 Moulder Center for Drug Discovery Research and Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA, USA
| | - Marlene A Jacobson
- 1 Moulder Center for Drug Discovery Research and Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA, USA
| |
Collapse
|
15
|
Niemann-Pick type C disease: The atypical sphingolipidosis. Adv Biol Regul 2018; 70:82-88. [PMID: 30205942 DOI: 10.1016/j.jbior.2018.08.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 08/27/2018] [Indexed: 12/29/2022]
Abstract
Niemann-Pick type C (NPC) disease is a lysosomal storage disorder resulting from mutations in either the NPC1 (95%) or NPC2 (5%) genes. NPC typically presents in childhood with visceral lipid accumulation and complex progressive neurodegeneration characterized by cerebellar ataxia, dysphagia, and dementia, resulting in a shortened lifespan. While cholesterol is widely acknowledged as the principal storage lipid in NPC, multiple species of sphingolipids accumulate as well. This accumulation of sphingolipids led to the initial assumption that NPC disease was caused by a deficiency in a sphingolipid catabolism enzyme, similar to sphingomyelinase deficiencies with which it shares a family name. It took about half a century to determine that NPC was in fact caused by a cholesterol trafficking defect, and still as we approach a century after the initial identification of the disease, the mechanisms by which sphingolipids accumulate remain poorly understood. Here we focus on the defects of sphingolipid catabolism in the endolysosomal compartment and how they contribute to the biology and pathology observed in NPC disease. This review highlights the need for further work on understanding and possibly developing treatments to correct the accumulation of sphingolipids in addition to cholesterol in this currently untreatable disease.
Collapse
|
16
|
Abstract
Cells utilize calcium ions (Ca2+) to signal almost all aspects of cellular life, ranging from cell proliferation to cell death, in a spatially and temporally regulated manner. A key aspect of this regulation is the compartmentalization of Ca2+ in various cytoplasmic organelles that act as intracellular Ca2+ stores. Whereas Ca2+ release from the large-volume Ca2+ stores, such as the endoplasmic reticulum (ER) and Golgi apparatus, are preferred for signal transduction, Ca2+ release from the small-volume individual vesicular stores that are dispersed throughout the cell, such as lysosomes, may be more useful in local regulation, such as membrane fusion and individualized vesicular movements. Conceivably, these two types of Ca2+ stores may be established, maintained or refilled via distinct mechanisms. ER stores are refilled through sustained Ca2+ influx at ER-plasma membrane (PM) membrane contact sites (MCSs). In this review, we discuss the release and refilling mechanisms of intracellular small vesicular Ca2+ stores, with a special focus on lysosomes. Recent imaging studies of Ca2+ release and organelle MCSs suggest that Ca2+ exchange may occur between two types of stores, such that the small stores acquire Ca2+ from the large stores via ER-vesicle MCSs. Hence vesicular stores like lysosomes may be viewed as secondary Ca2+ stores in the cell.
Collapse
|
17
|
Dysregulation of autophagy as a common mechanism in lysosomal storage diseases. Essays Biochem 2017; 61:733-749. [PMID: 29233882 PMCID: PMC5869865 DOI: 10.1042/ebc20170055] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 10/08/2017] [Accepted: 10/12/2017] [Indexed: 12/19/2022]
Abstract
The lysosome plays a pivotal role between catabolic and anabolic processes as the nexus for signalling pathways responsive to a variety of factors, such as growth, nutrient availability, energetic status and cellular stressors. Lysosomes are also the terminal degradative organelles for autophagy through which macromolecules and damaged cellular components and organelles are degraded. Autophagy acts as a cellular homeostatic pathway that is essential for organismal physiology. Decline in autophagy during ageing or in many diseases, including late-onset forms of neurodegeneration is considered a major contributing factor to the pathology. Multiple lines of evidence indicate that impairment in autophagy is also a central mechanism underlying several lysosomal storage disorders (LSDs). LSDs are a class of rare, inherited disorders whose histopathological hallmark is the accumulation of undegraded materials in the lysosomes due to abnormal lysosomal function. Inefficient degradative capability of the lysosomes has negative impact on the flux through the autophagic pathway, and therefore dysregulated autophagy in LSDs is emerging as a relevant disease mechanism. Pathology in the LSDs is generally early-onset, severe and life-limiting but current therapies are limited or absent; recognizing common autophagy defects in the LSDs raises new possibilities for therapy. In this review, we describe the mechanisms by which LSDs occur, focusing on perturbations in the autophagy pathway and present the latest data supporting the development of novel therapeutic approaches related to the modulation of autophagy.
Collapse
|
18
|
Liou B, Peng Y, Li R, Inskeep V, Zhang W, Quinn B, Dasgupta N, Blackwood R, Setchell KDR, Fleming S, Grabowski GA, Marshall J, Sun Y. Modulating ryanodine receptors with dantrolene attenuates neuronopathic phenotype in Gaucher disease mice. Hum Mol Genet 2017; 25:5126-5141. [PMID: 27655403 DOI: 10.1093/hmg/ddw322] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 09/14/2016] [Indexed: 12/12/2022] Open
Abstract
Neuronopathic Gaucher disease (nGD) manifests as severe neurological symptoms in patients with no effective treatment available. Ryanodine receptors (Ryrs) are a family of calcium release channels on intracellular stores. The goal of this study is to determine if Ryrs are potential targets for nGD treatment. A nGD cell model (CBE-N2a) was created by inhibiting acid β-glucosidase (GCase) in N2a cells with conduritol B epoxide (CBE). Enhanced cytosolic calcium in CBE-N2a cells was blocked by either ryanodine or dantrolene, antagonists of Ryrs and by Genz-161, a glucosylceramide synthase inhibitor, suggesting substrate-mediated ER-calcium efflux occurs through ryanodine receptors. In the brain of a nGD (4L;C*) mouse model, expression of Ryrs was normal at 13 days of age, but significantly decreased below the wild type level in end-stage 4L;C* brains at 40 days. Treatment with dantrolene in 4L;C* mice starting at postnatal day 5 delayed neurological pathology and prolonged survival. Compared to untreated 4L;C* mice, dantrolene treatment significantly improved gait, reduced LC3-II levels, improved mitochondrial ATP production and reduced inflammation in the brain. Dantrolene treatment partially normalized Ryr expression and its potential regulators, CAMK IV and calmodulin. Furthermore, dantrolene treatment increased residual mutant GCase activity in 4L;C* brains. These data demonstrate that modulating Ryrs has neuroprotective effects in nGD through mechanisms that protect the mitochondria, autophagy, Ryr expression and enhance GCase activity. This study suggests that calcium signalling stabilization, e.g. with dantrolene, could be a potential disease modifying therapy for nGD.
Collapse
Affiliation(s)
- Benjamin Liou
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Yanyan Peng
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Ronghua Li
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Venette Inskeep
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Wujuan Zhang
- Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Brian Quinn
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Nupur Dasgupta
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Rachel Blackwood
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kenneth D R Setchell
- Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Sheila Fleming
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Gregory A Grabowski
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | - Ying Sun
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
19
|
Methods for monitoring Ca 2+ and ion channels in the lysosome. Cell Calcium 2017; 64:20-28. [DOI: 10.1016/j.ceca.2016.12.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 12/07/2016] [Accepted: 12/07/2016] [Indexed: 12/22/2022]
|
20
|
Wang W, Zhang X, Gao Q, Lawas M, Yu L, Cheng X, Gu M, Sahoo N, Li X, Li P, Ireland S, Meredith A, Xu H. A voltage-dependent K + channel in the lysosome is required for refilling lysosomal Ca 2+ stores. J Cell Biol 2017; 216:1715-1730. [PMID: 28468834 PMCID: PMC5461029 DOI: 10.1083/jcb.201612123] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/05/2017] [Accepted: 04/10/2017] [Indexed: 01/04/2023] Open
Abstract
Ion-dependent channels and transporters have been identified in lysosomes, including the V-ATPase H+ pump and transient receptor potential mucolipin channels (TRPMLs), the principle Ca2+ release channels in the lysosome, but much less is understood about the roles of Na+ and K+ in lysosomal physiology. Wang et al. describe a voltage-sensitive, Ca2+-activated K+ current in the lysosome (LysoKVCa) and show that LysoKVCa regulates lysosomal membrane potential and refilling of lysosomal Ca2+ stores. The resting membrane potential (Δψ) of the cell is negative on the cytosolic side and determined primarily by the plasma membrane’s selective permeability to K+. We show that lysosomal Δψ is set by lysosomal membrane permeabilities to Na+ and H+, but not K+, and is positive on the cytosolic side. An increase in juxta-lysosomal Ca2+ rapidly reversed lysosomal Δψ by activating a large voltage-dependent and K+-selective conductance (LysoKVCa). LysoKVCa is encoded molecularly by SLO1 proteins known for forming plasma membrane BK channels. Opening of single LysoKVCa channels is sufficient to cause the rapid, striking changes in lysosomal Δψ. Lysosomal Ca2+ stores may be refilled from endoplasmic reticulum (ER) Ca2+ via ER–lysosome membrane contact sites. We propose that LysoKVCa serves as the perilysosomal Ca2+ effector to prime lysosomes for the refilling process. Consistently, genetic ablation or pharmacological inhibition of LysoKVCa, or abolition of its Ca2+ sensitivity, blocks refilling and maintenance of lysosomal Ca2+ stores, resulting in lysosomal cholesterol accumulation and a lysosome storage phenotype.
Collapse
Affiliation(s)
- Wuyang Wang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Xiaoli Zhang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Qiong Gao
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Maria Lawas
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Lu Yu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Xiping Cheng
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Mingxue Gu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Nirakar Sahoo
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Xinran Li
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Ping Li
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109.,Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Stephen Ireland
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Andrea Meredith
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Haoxing Xu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
21
|
Viana GM, do Nascimento CC, Paredes-Gamero EJ, D'Almeida V. Altered Cellular Homeostasis in Murine MPS I Fibroblasts: Evidence of Cell-Specific Physiopathology. JIMD Rep 2017; 36:109-116. [PMID: 28220405 DOI: 10.1007/8904_2017_5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/11/2017] [Accepted: 01/13/2017] [Indexed: 12/12/2022] Open
Abstract
Mucopolysaccharidosis type I (MPS I), a rare autosomal recessive disease, is caused by a deficiency of the lysosomal enzyme alfa-L-iduronidase. Impaired enzyme activity promotes glycosaminoglycans accumulation in several tissues and organs, leading to complex multisystemic complications. Several studies using animal models indicated different intracellular pathways involving MPS I physiopathology; however, the exact mechanisms underlying this syndrome are still not understood. Previous results from our group showed alterations in ionic homeostasis and cell viability of splenocytes and macrophages in Idua-/- mice. In the present study, we found altered intracellular ionic homeostasis in a different cell type (fibroblasts) from the same murine model. Idua-/- fibroblasts from 3-month-old mice presented higher cytoplasmatic and endoplasmic reticulum Ca2+ concentration, lower levels of mitochondrial Ca2+ and mitochondrial membrane potential and higher cytoplasmatic pH when compared to Idua+/+ animals. Also, Idua-/- fibroblasts were more resistant to the apoptotic induction with staurosporine, indicating a possible resistance to apoptotic induction in those cells. In addition, despite the intracellular ionic imbalance, no significant alterations were found in apoptosis and autophagy in Idua-/- fibroblasts, which implies that the ionic alterations did not activate those pathways. The investigation of mechanisms underlying the cellular physiopathology of lysosomal diseases is crucial for a better understanding about the progression of these diseases. Since splenocytes, macrophages, and fibroblasts have different embryonic origins and distinct structural and functional features, potentially altered signaling pathways found in a cell-specific manner in an alfa-L-iduronidase-deficient environment provide additional understanding of the clinical multisystemic presentation of this disease and provide new basis for improved therapeutic approaches.
Collapse
Affiliation(s)
- Gustavo Monteiro Viana
- Department of Pediatrics, Universidade Federal de São Paulo, Rua Napoleão de Barros, 325, 3rd floor, São Paulo, 04024-002, Brazil. .,Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, Brazil.
| | | | - Edgar Julian Paredes-Gamero
- Centro Interdisciplinar de Investigação Bioquímica, Universidade de Mogi das Cruzes, Mogi das Cruzes, Brazil
| | - Vânia D'Almeida
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
22
|
Sbano L, Bonora M, Marchi S, Baldassari F, Medina DL, Ballabio A, Giorgi C, Pinton P. TFEB-mediated increase in peripheral lysosomes regulates store-operated calcium entry. Sci Rep 2017; 7:40797. [PMID: 28084445 PMCID: PMC5233950 DOI: 10.1038/srep40797] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/09/2016] [Indexed: 01/04/2023] Open
Abstract
Lysosomes are membrane-bound organelles mainly involved in catabolic processes. In addition, lysosomes can expel their contents outside of the cell via lysosomal exocytosis. Some of the key steps involved in these important cellular processes, such as vesicular fusion and trafficking, require calcium (Ca2+) signaling. Recent data show that lysosomal functions are transcriptionally regulated by transcription factor EB (TFEB) through the induction of genes involved in lysosomal biogenesis and exocytosis. Given these observations, we investigated the roles of TFEB and lysosomes in intracellular Ca2+ homeostasis. We studied the effect of transient modulation of TFEB expression in HeLa cells by measuring the cytosolic Ca2+ response after capacitative Ca2+ entry activation and Ca2+ dynamics in the endoplasmic reticulum (ER) and directly in lysosomes. Our observations show that transient TFEB overexpression significantly reduces cytosolic Ca2+ levels under a capacitative influx model and ER re-uptake of calcium, increasing the lysosomal Ca2+ buffering capacity. Moreover, lysosomal destruction or damage abolishes these TFEB-dependent effects in both the cytosol and ER. These results suggest a possible Ca2+ buffering role for lysosomes and shed new light on lysosomal functions during intracellular Ca2+ homeostasis.
Collapse
Affiliation(s)
- Luigi Sbano
- Dept. of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, 44121, Italy
| | - Massimo Bonora
- Dept. of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, 44121, Italy
| | - Saverio Marchi
- Dept. of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, 44121, Italy
| | - Federica Baldassari
- Dept. of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, 44121, Italy
| | - Diego L Medina
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Naples, Italy
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Naples, Italy.,Dept. of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA.,Medical Genetics, Dept. of Translational Medicine, Federico II University, 80131 Naples, Italy
| | - Carlotta Giorgi
- Dept. of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, 44121, Italy
| | - Paolo Pinton
- Dept. of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, 44121, Italy
| |
Collapse
|
23
|
Zhong XZ, Sun X, Cao Q, Dong G, Schiffmann R, Dong XP. BK channel agonist represents a potential therapeutic approach for lysosomal storage diseases. Sci Rep 2016; 6:33684. [PMID: 27670435 PMCID: PMC5037385 DOI: 10.1038/srep33684] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 08/31/2016] [Indexed: 12/21/2022] Open
Abstract
Efficient lysosomal Ca2+ release plays an essential role in lysosomal trafficking. We have recently shown that lysosomal big conductance Ca2+-activated potassium (BK) channel forms a physical and functional coupling with the lysosomal Ca2+ release channel Transient Receptor Potential Mucolipin-1 (TRPML1). BK and TRPML1 forms a positive feedback loop to facilitate lysosomal Ca2+ release and subsequent lysosome membrane trafficking. However, it is unclear whether the positive feedback mechanism is common for other lysosomal storage diseases (LSDs) and whether BK channel agonists rescue abnormal lysosomal storage in LSDs. In this study, we assessed the effect of BK agonist, NS1619 and NS11021 in a number of LSDs including NPC1, mild cases of mucolipidosis type IV (ML4) (TRPML1-F408∆), Niemann-Pick type A (NPA) and Fabry disease. We found that TRPML1-mediated Ca2+ release was compromised in these LSDs. BK activation corrected the impaired Ca2+ release in these LSDs and successfully rescued the abnormal lysosomal storage of these diseases by promoting TRPML1-mediated lysosomal exocytosis. Our study suggests that BK channel activation stimulates the TRPML1-BK positive reinforcing loop to correct abnormal lysosomal storage in LSDs. Drugs targeting BK channel represent a potential therapeutic approach for LSDs.
Collapse
Affiliation(s)
- Xi Zoë Zhong
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, Halifax, B3H 4R2, Nova Scotia, Canada
| | - Xue Sun
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, Halifax, B3H 4R2, Nova Scotia, Canada
| | - Qi Cao
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, Halifax, B3H 4R2, Nova Scotia, Canada
| | - Gaofeng Dong
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, Halifax, B3H 4R2, Nova Scotia, Canada
| | - Raphael Schiffmann
- Institute of Metabolic Disease, Baylor Research Institute, 3812 Elm Street, Dallas, TX, 75226, USA
| | - Xian-Ping Dong
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, Halifax, B3H 4R2, Nova Scotia, Canada
| |
Collapse
|
24
|
Brailoiu GC, Brailoiu E. Modulation of Calcium Entry by the Endo-lysosomal System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:423-47. [PMID: 27161239 DOI: 10.1007/978-3-319-26974-0_18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Endo-lysosomes are acidic organelles that besides the role in macromolecules degradation, act as intracellular Ca(2+) stores. Nicotinic acid adenine dinucleotide phosphate (NAADP), the most potent Ca(2+)-mobilizing second messenger, produced in response to agonist stimulation, activates Ca(2+)-releasing channels on endo-lysosomes and modulates a variety of cellular functions. NAADP-evoked signals are amplified by Ca(2+) release from endoplasmic reticulum, via the recruitment of inositol 1,4,5-trisphosphate and/or ryanodine receptors through a Ca(2+)-induced Ca(2+)- release (CICR) mechanism. The endo-lysosomal Ca(2+) channels activated by NAADP were recently identified as the two-pore channels (TPCs). In addition to TPCs, endo-lysosomes express another distinct family of Ca(2+)- permeable channels, namely the transient receptor potential mucolipin (TRPML) channels, functionally distinct from TPCs. TPCs belong to the voltage-gated channels, resembling voltage-gated Na(+) and Ca(2+) channels. TPCs have important roles in vesicular fusion and trafficking, in triggering a global Ca(2+) signal and in modulation of the membrane excitability. Depletion of acidic Ca(2+) stores has been shown to activate store-operated Ca(2+) entry in human platelets and mouse pancreatic β-cells. In human platelets, Ca(2+) influx in response to acidic stores depletion is facilitated by the tubulin-cytoskeleton and occurs through non-selective cation channels and transient receptor potential canonical (TRPC) channels. Emerging evidence indicates that activation of intracellular receptors, situated on endo-lysosomes, elicits canonical and non-canonical signaling mechanisms that involve CICR and activation of non-selective cation channels in plasma membrane. The ability of endo-lysosomal Ca(2+) stores to modulate the Ca(2+) release from other organelles and the Ca(2+) entry increases the diversity and complexity of cellular signaling mechanisms.
Collapse
Affiliation(s)
- G Cristina Brailoiu
- Department of Pharmaceutical Sciences, Jefferson School of Pharmacy, Thomas Jefferson University, 901 Walnut St, Rm 916, Philadelphia, PA, 19107, USA.
| | - Eugen Brailoiu
- Center for Substance Abuse Research, Temple University School of Medicine, 3500 N. Broad Street, Room 848, Philadelphia, PA, 19140, USA
| |
Collapse
|
25
|
Garrity AG, Wang W, Collier CM, Levey SA, Gao Q, Xu H. The endoplasmic reticulum, not the pH gradient, drives calcium refilling of lysosomes. eLife 2016; 5. [PMID: 27213518 PMCID: PMC4909396 DOI: 10.7554/elife.15887] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/20/2016] [Indexed: 12/15/2022] Open
Abstract
Impaired homeostasis of lysosomal Ca2+ causes lysosome dysfunction and lysosomal storage diseases (LSDs), but the mechanisms by which lysosomes acquire and refill Ca2+ are not known. We developed a physiological assay to monitor lysosomal Ca2+ store refilling using specific activators of lysosomal Ca2+ channels to repeatedly induce lysosomal Ca2+ release. In contrast to the prevailing view that lysosomal acidification drives Ca2+ into the lysosome, inhibiting the V-ATPase H+ pump did not prevent Ca2+ refilling. Instead, pharmacological depletion or chelation of Endoplasmic Reticulum (ER) Ca2+ prevented lysosomal Ca2+ stores from refilling. More specifically, antagonists of ER IP3 receptors (IP3Rs) rapidly and completely blocked Ca2+ refilling of lysosomes, but not in cells lacking IP3Rs. Furthermore, reducing ER Ca2+ or blocking IP3Rs caused a dramatic LSD-like lysosome storage phenotype. By closely apposing each other, the ER may serve as a direct and primary source of Ca2+for the lysosome. DOI:http://dx.doi.org/10.7554/eLife.15887.001
Collapse
Affiliation(s)
- Abigail G Garrity
- Neuroscience Program, University of Michigan, Ann Arbor, United States
| | - Wuyang Wang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Crystal Md Collier
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Sara A Levey
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Qiong Gao
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Haoxing Xu
- Neuroscience Program, University of Michigan, Ann Arbor, United States.,Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, United States
| |
Collapse
|
26
|
Onyenwoke RU, Brenman JE. Lysosomal Storage Diseases-Regulating Neurodegeneration. J Exp Neurosci 2016; 9:81-91. [PMID: 27081317 PMCID: PMC4822725 DOI: 10.4137/jen.s25475] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/11/2015] [Accepted: 11/16/2015] [Indexed: 12/19/2022] Open
Abstract
Autophagy is a complex pathway regulated by numerous signaling events that recycles macromolecules and can be perturbed in lysosomal storage diseases (LSDs). The concept of LSDs, which are characterized by aberrant, excessive storage of cellular material in lysosomes, developed following the discovery of an enzyme deficiency as the cause of Pompe disease in 1963. Great strides have since been made in better understanding the biology of LSDs. Defective lysosomal storage typically occurs in many cell types, but the nervous system, including the central nervous system and peripheral nervous system, is particularly vulnerable to LSDs, being affected in two-thirds of LSDs. This review provides a summary of some of the better characterized LSDs and the pathways affected in these disorders.
Collapse
Affiliation(s)
- Rob U Onyenwoke
- Department of Pharmaceutical Science, Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC, USA
| | - Jay E Brenman
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.; Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
27
|
Abstract
Lysosomes are acidic compartments filled with more than 60 different types of hydrolases. They mediate the degradation of extracellular particles from endocytosis and of intracellular components from autophagy. The digested products are transported out of the lysosome via specific catabolite exporters or via vesicular membrane trafficking. Lysosomes also contain more than 50 membrane proteins and are equipped with the machinery to sense nutrient availability, which determines the distribution, number, size, and activity of lysosomes to control the specificity of cargo flux and timing (the initiation and termination) of degradation. Defects in degradation, export, or trafficking result in lysosomal dysfunction and lysosomal storage diseases (LSDs). Lysosomal channels and transporters mediate ion flux across perimeter membranes to regulate lysosomal ion homeostasis, membrane potential, catabolite export, membrane trafficking, and nutrient sensing. Dysregulation of lysosomal channels underlies the pathogenesis of many LSDs and possibly that of metabolic and common neurodegenerative diseases.
Collapse
Affiliation(s)
- Haoxing Xu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109;
| | | |
Collapse
|
28
|
Cao Q, Zhong XZ, Zou Y, Zhang Z, Toro L, Dong XP. BK Channels Alleviate Lysosomal Storage Diseases by Providing Positive Feedback Regulation of Lysosomal Ca2+ Release. Dev Cell 2015; 33:427-41. [PMID: 25982675 DOI: 10.1016/j.devcel.2015.04.010] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 03/25/2015] [Accepted: 04/10/2015] [Indexed: 11/25/2022]
Abstract
Promoting lysosomal trafficking represents a promising therapeutic approach for lysosome storage diseases. Efficient Ca(2+) mobilization from lysosomes is important for lysosomal trafficking. Ca(2+) release from lysosomes could generate a negative potential in the lumen to disturb subsequent Ca(2+) release in the absence of counter ion flux. Here we report that lysosomes express big-conductance Ca(2+)-activated potassium (BK) channels that form physical and functional coupling with the lysosomal Ca(2+) release channel, TRPML1. Ca(2+) release via TRPML1 causes BK activation, which in turn facilitates further lysosomal Ca(2+) release and membrane trafficking. Importantly, BK overexpression rescues the impaired TRPML1-mediated Ca(2+) release and abnormal lysosomal storage in cells from Niemann-Pick C1 patients. Therefore, we have identified a lysosomal K(+) channel that provides a positive feedback mechanism to facilitate TRPML1-mediated Ca(2+) release and membrane trafficking. Our findings suggest that upregulating BK may be a potential therapeutic strategy for certain lysosomal storage diseases and common neurodegenerative disorders.
Collapse
Affiliation(s)
- Qi Cao
- Department of Physiology and Biophysics, Sir Charles Tupper Medical Building, Dalhousie University, 5850 College Street, Halifax, NS B3H 4R2, Canada
| | - Xi Zoë Zhong
- Department of Physiology and Biophysics, Sir Charles Tupper Medical Building, Dalhousie University, 5850 College Street, Halifax, NS B3H 4R2, Canada
| | - Yuanjie Zou
- Department of Physiology and Biophysics, Sir Charles Tupper Medical Building, Dalhousie University, 5850 College Street, Halifax, NS B3H 4R2, Canada
| | - Zhu Zhang
- Division of Molecular Medicine, Department of Anesthesiology and Department of Molecular & Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-7115, USA
| | - Ligia Toro
- Division of Molecular Medicine, Department of Anesthesiology and Department of Molecular & Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-7115, USA
| | - Xian-Ping Dong
- Department of Physiology and Biophysics, Sir Charles Tupper Medical Building, Dalhousie University, 5850 College Street, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
29
|
López Sanjurjo CI, Tovey SC, Taylor CW. Rapid recycling of Ca2+ between IP3-sensitive stores and lysosomes. PLoS One 2014; 9:e111275. [PMID: 25337829 PMCID: PMC4206489 DOI: 10.1371/journal.pone.0111275] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 09/28/2014] [Indexed: 11/26/2022] Open
Abstract
Inositol 1,4,5-trisphosphate (IP3) evokes release of Ca2+ from the endoplasmic reticulum (ER), but the resulting Ca2+ signals are shaped by interactions with additional intracellular organelles. Bafilomycin A1, which prevents lysosomal Ca2+ uptake by inhibiting H+ pumping into lysosomes, increased the amplitude of the initial Ca2+ signals evoked by carbachol in human embryonic kidney (HEK) cells. Carbachol alone and carbachol in combination with parathyroid hormone (PTH) evoke Ca2+ release from distinct IP3-sensitive Ca2+ stores in HEK cells stably expressing human type 1 PTH receptors. Bafilomycin A1 similarly exaggerated the Ca2+ signals evoked by carbachol or carbachol with PTH, indicating that Ca2+ released from distinct IP3-sensitive Ca2+ stores is sequestered by lysosomes. The Ca2+ signals resulting from store-operated Ca2+ entry, whether evoked by thapsigargin or carbachol, were unaffected by bafilomycin A1. Using Gd3+ (1 mM) to inhibit both Ca2+ entry and Ca2+ extrusion, HEK cells were repetitively stimulated with carbachol to assess the effectiveness of Ca2+ recycling to the ER after IP3-evoked Ca2+ release. Blocking lysosomal Ca2+ uptake with bafilomycin A1 increased the amplitude of each carbachol-evoked Ca2+ signal without affecting the rate of Ca2+ recycling to the ER. This suggests that Ca2+ accumulated by lysosomes is rapidly returned to the ER. We conclude that lysosomes rapidly, reversibly and selectively accumulate the Ca2+ released by IP3 receptors residing within distinct Ca2+ stores, but not the Ca2+ entering cells via receptor-regulated, store-operated Ca2+ entry pathways.
Collapse
Affiliation(s)
| | - Stephen C. Tovey
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Colin W. Taylor
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
30
|
Alroy J, Garganta C, Wiederschain G. Secondary biochemical and morphological consequences in lysosomal storage diseases. BIOCHEMISTRY (MOSCOW) 2014; 79:619-36. [DOI: 10.1134/s0006297914070049] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
31
|
Rasmussen JE, Sheridan JT, Polk W, Davies CM, Tarran R. Cigarette smoke-induced Ca2+ release leads to cystic fibrosis transmembrane conductance regulator (CFTR) dysfunction. J Biol Chem 2014; 289:7671-81. [PMID: 24448802 DOI: 10.1074/jbc.m113.545137] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Chronic obstructive pulmonary disease affects 64 million people and is currently the fourth leading cause of death worldwide. Chronic obstructive pulmonary disease includes both emphysema and chronic bronchitis, and in the case of chronic bronchitis represents an inflammatory response of the airways that is associated with mucus hypersecretion and obstruction of small airways. Recently, it has emerged that exposure to cigarette smoke (CS) leads to an inhibition of the cystic fibrosis transmembrane conductance regulator (CFTR) Cl(-) channel, causing airway surface liquid dehydration, which may play a role in the development of chronic bronchitis. CS rapidly clears CFTR from the plasma membrane and causes it to be deposited into aggresome-like compartments. However, little is known about the mechanism(s) responsible for the internalization of CFTR following CS exposure. Our studies revealed that CS triggered a rise in cytoplasmic Ca(2+) that may have emanated from lysosomes. Furthermore, chelation of cytoplasmic Ca(2+), but not inhibition of protein kinases/phosphatases, prevented CS-induced CFTR internalization. The macrolide antibiotic bafilomycin A1 inhibited CS-induced Ca(2+) release and prevented CFTR clearance from the plasma membrane, further linking cytoplasmic Ca(2+) and CFTR internalization. We hypothesize that CS-induced Ca(2+) release prevents normal sorting/degradation of CFTR and causes internalized CFTR to reroute to aggresomes. Our data provide mechanistic insight into the potentially deleterious effects of CS on airway epithelia and outline a hitherto unrecognized signaling event triggered by CS that may affect the long term transition of the lung into a hyper-inflammatory/dehydrated environment.
Collapse
|
32
|
Pfaender S, Grabrucker AM. Characterization of biometal profiles in neurological disorders. Metallomics 2014; 6:960-77. [DOI: 10.1039/c4mt00008k] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This review summarizes the findings on dysregulation of metal ions in neurological diseases and tries to develop and predict specific biometal profiles.
Collapse
Affiliation(s)
| | - Andreas M. Grabrucker
- Institute for Anatomy and Cell Biology
- Ulm University
- Ulm, Germany
- WG Molecular Analysis of Synaptopathies
- Neurology Dept
| |
Collapse
|
33
|
Martyn C, Li J. Fig4 deficiency: a newly emerged lysosomal storage disorder? Prog Neurobiol 2012; 101-102:35-45. [PMID: 23165282 DOI: 10.1016/j.pneurobio.2012.11.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 09/07/2012] [Accepted: 11/09/2012] [Indexed: 12/31/2022]
Abstract
FIG4 (Sac3 in mammals) is a 5'-phosphoinositide phosphatase that coordinates the turnover of phosphatidylinositol-3,5-bisphosphate (PI(3,5)P(2)), a very low abundance phosphoinositide. Deficiency of FIG4 severely affects the human and mouse nervous systems by causing two distinct forms of abnormal lysosomal storage. The first form occurs in spinal sensory neurons, where vacuolated endolysosomes accumulate in perinuclear regions. A second form occurs in cortical/spinal motor neurons and glia, in which enlarged endolysosomes become filled with electron dense materials in a manner indistinguishable from other lysosomal storage disorders. Humans with a deficiency of FIG4 (known as Charcot-Marie-Tooth disease type 4J or CMT4J) present with clinical and pathophysiological phenotypes indicative of spinal motor neuron degeneration and segmental demyelination. These findings reveal a signaling pathway involving FIG4 that appears to be important for lysosomal function. In this review, we discuss the biology of FIG4 and describe how the deficiency of FIG4 results in lysosomal phenotypes. We also discuss the implications of FIG4/PI(3,5)P(2) signaling in understanding other lysosomal storage diseases, neuropathies, and acquired demyelinating diseases.
Collapse
Affiliation(s)
- Colin Martyn
- VA Tennessee Valley Healthcare System, Nashville, TN, USA
| | | |
Collapse
|
34
|
Exocytosis is impaired in mucopolysaccharidosis IIIA mouse chromaffin cells. Neuroscience 2012; 227:110-8. [PMID: 23022219 DOI: 10.1016/j.neuroscience.2012.09.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 08/28/2012] [Accepted: 09/13/2012] [Indexed: 11/22/2022]
Abstract
Mucopolysaccharidosis IIIA (MPS IIIA) is a lysosomal storage disorder caused by a deficiency in the activity of the lysosomal hydrolase, sulphamidase, an enzyme involved in the degradation of heparan sulphate. MPS IIIA patients exhibit progressive mental retardation and behavioural disturbance. While neuropathology is the major clinical problem in MPS IIIA patients, there is little understanding of how lysosomal storage generates this phenotype. As reduced neuronal communication can underlie cognitive deficiencies, we investigated whether the secretion of neurotransmitters is altered in MPS IIIA mice; utilising adrenal chromaffin cells, a classical model for studying secretion via exocytosis. MPS IIIA chromaffin cells displayed heparan sulphate storage and electron microscopy revealed large electron-lucent storage compartments. There were also increased numbers of large/elongated chromaffin granules, with a morphology that was similar to immature secretory granules. Carbon fibre amperometry illustrated a significant decrease in the number of exocytotic events for MPS IIIA, when compared to control chromaffin cells. However, there were no changes in the kinetics of release, the amount of catecholamine released per exocytotic event, or the amount of Ca(2+) entry upon stimulation. The increased number of large/elongated granules and reduced number of exocytotic events suggests that either the biogenesis and/or the cell surface docking and fusion potential of these vesicles is impaired in MPS IIIA. If this also occurs in central nervous system neurons, the reduction in neurotransmitter release could help to explain the development of neuropathology in MPS IIIA.
Collapse
|
35
|
Kiselyov KK, Ahuja M, Rybalchenko V, Patel S, Muallem S. The intracellular Ca²⁺ channels of membrane traffic. Channels (Austin) 2012; 6:344-51. [PMID: 22907062 DOI: 10.4161/chan.21723] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Regulation of organellar fusion and fission by Ca ( 2+) has emerged as a central paradigm in intracellular membrane traffic. Originally formulated for Ca ( 2+) -driven SNARE-mediated exocytosis in the presynaptic terminals, it was later expanded to explain membrane traffic in other exocytic events within the endo-lysosomal system. The list of processes and conditions that depend on the intracellular membrane traffic includes aging, antigen and lipid processing, growth factor signaling and enzyme secretion. Characterization of the ion channels that regulate intracellular membrane fusion and fission promises novel pharmacological approaches in these processes when their function becomes aberrant. The recent identification of Ca ( 2+) permeability through the intracellular ion channels comprising the mucolipin (TRPMLs) and the two-pore channels (TPCs) families pinpoints the candidates for the Ca ( 2+) channel that drive intracellular membrane traffic. The present review summarizes the recent developments and the current questions relevant to this topic.
Collapse
Affiliation(s)
- Kirill K Kiselyov
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | |
Collapse
|
36
|
Taylor CW, Dale P. Intracellular Ca(2+) channels - a growing community. Mol Cell Endocrinol 2012; 353:21-8. [PMID: 21889573 DOI: 10.1016/j.mce.2011.08.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 08/16/2011] [Accepted: 08/18/2011] [Indexed: 12/24/2022]
Abstract
The Ca(2+) signals that control almost every cellular activity are generated by regulating Ca(2+) transport, usually via Ca(2+)-permeable channels, across the plasma membrane or the membranes of intracellular organelles. The most widespread and best understood of the intracellular Ca(2+) channels are inositol trisphosphate receptors (IP(3)R) and ryanodine receptors, most of which are expressed in the endoplasmic or sarcoplasmic reticulum. However, accumulating evidence suggests physiological roles for many additional Ca(2+) channels in both ER and other intracellular organelles. Interactions between these channels, whether mediated by Ca(2+) itself or interactions between proteins, is a recurrent feature of the Ca(2+) signals evoked by physiological stimuli. We focus on two specific examples, clustering of IP(3)Rs and NAADP (nicotinic acid dinucleotide phosphate)-evoked Ca(2+) release from endo-lysosomes, to illustrate the diversity of Ca(2+) channels and the interplay between them.
Collapse
|
37
|
Rybalchenko V, Ahuja M, Coblentz J, Churamani D, Patel S, Kiselyov K, Muallem S. Membrane potential regulates nicotinic acid adenine dinucleotide phosphate (NAADP) dependence of the pH- and Ca2+-sensitive organellar two-pore channel TPC1. J Biol Chem 2012; 287:20407-16. [PMID: 22500018 DOI: 10.1074/jbc.m112.359612] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Nicotinic acid adenine dinucleotide phosphate (NAADP) is a potent second messenger that mobilizes Ca(2+) from the acidic endolysosomes by activation of the two-pore channels TPC1 and TPC2. The channel properties of human TPC1 have not been studied before, and its cellular function is not known. In the present study, we characterized TPC1 incorporated into lipid bilayers. The native and recombinant TPC1 channels are activated by NAADP. TPC1 activity requires acidic luminal pH and high luminal Ca(2+). With Ba(2+) as the permeable ion, luminal Ca(2+) activates TPC1 with an apparent K(m) of 180 μm. TPC1 operates in two tightly coupled conductance states of 47 ± 8 and 200 ± 9 picosiemens. Importantly, opening of the large conductance markedly increases the small conductance mean open time. Changes in membrane potential from 0 to -60 mV increased linearly both the small and the large conductances and NP(o), indicating that TPC1 is regulated by voltage. Intriguingly, the apparent affinity for activation of TPC1 by its ligand NAADP is not constant. Rather, hyperpolarization increases the apparent affinity of TPC1 for NAADP by 10 nm/mV. The concerted regulation of TPC1 activity by luminal Ca(2+) and by membrane potential thus provides a potential mechanism to explain NAADP-induced Ca(2+) oscillations. These findings reveal unique properties of TPC1 to explain its role in Ca(2+) oscillations and cell function.
Collapse
Affiliation(s)
- Volodymyr Rybalchenko
- Epithelial Signaling and Transport Section, Molecular Physiology and Therapeutics Branch, NIDCR, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Shen D, Wang X, Li X, Zhang X, Yao Z, Dibble S, Dong XP, Yu T, Lieberman AP, Showalter HD, Xu H. Lipid storage disorders block lysosomal trafficking by inhibiting a TRP channel and lysosomal calcium release. Nat Commun 2012; 3:731. [PMID: 22415822 DOI: 10.1038/ncomms1735] [Citation(s) in RCA: 378] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 02/07/2012] [Indexed: 11/09/2022] Open
Abstract
Lysosomal lipid accumulation, defects in membrane trafficking and altered Ca(2+) homoeostasis are common features in many lysosomal storage diseases. Mucolipin transient receptor potential channel 1 (TRPML1) is the principle Ca(2+) channel in the lysosome. Here we show that TRPML1-mediated lysosomal Ca(2+) release, measured using a genetically encoded Ca(2+) indicator (GCaMP3) attached directly to TRPML1 and elicited by a potent membrane-permeable synthetic agonist, is dramatically reduced in Niemann-Pick (NP) disease cells. Sphingomyelins (SMs) are plasma membrane lipids that undergo sphingomyelinase (SMase)-mediated hydrolysis in the lysosomes of normal cells, but accumulate distinctively in lysosomes of NP cells. Patch-clamp analyses revealed that TRPML1 channel activity is inhibited by SMs, but potentiated by SMases. In NP-type C cells, increasing TRPML1's expression or activity was sufficient to correct the trafficking defects and reduce lysosome storage and cholesterol accumulation. We propose that abnormal accumulation of luminal lipids causes secondary lysosome storage by blocking TRPML1- and Ca(2+)-dependent lysosomal trafficking.
Collapse
Affiliation(s)
- Dongbiao Shen
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, 3089 Natural Science Building (Kraus), 830 North University, Ann Arbor 48109, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Ion channels and G-protein-coupled receptors (GPCRs) play a fundamental role in cancer progression by influencing Ca(2+) influx and signaling pathways in transformed cells. Transformed cells thrive in a hostile environment that is characterized by extracellular acidosis that promotes the pathological phenotype. The pathway(s) by which extracellular protons achieve this remain unclear. Here, a role for proton-sensing ion channels and GPCRs as mediators of the effects of extracellular protons in cancer cells is discussed.
Collapse
Affiliation(s)
- Maike Glitsch
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford, United Kingdom.
| |
Collapse
|
40
|
Abstract
The Transient receptor potential (TRP) family of cation channels is a large protein family, which is mainly structurally uniform. Proteins consist typically of six transmembrane domains and mostly four subunits are necessary to form a functional channel. Apart from this, TRP channels display a wide variety of activation mechanisms (ligand binding, G-protein coupled receptor dependent, physical stimuli such as temperature, pressure, etc.) and ion selectivity profiles (from highly Ca(2+) selective to non-selective for cations). They have been described now in almost every tissue of the body, including peripheral and central neurons. Especially in the sensory nervous system the role of several TRP channels is already described on a detailed level. This review summarizes data that is currently available on their role in the central nervous system. TRP channels are involved in neurogenesis and brain development, synaptic transmission and they play a key role in the development of several neurological diseases.
Collapse
|
41
|
Yamaguchi S, Jha A, Li Q, Soyombo AA, Dickinson GD, Churamani D, Brailoiu E, Patel S, Muallem S. Transient receptor potential mucolipin 1 (TRPML1) and two-pore channels are functionally independent organellar ion channels. J Biol Chem 2011; 286:22934-42. [PMID: 21540176 DOI: 10.1074/jbc.m110.210930] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
NAADP is a potent second messenger that mobilizes Ca(2+) from acidic organelles such as endosomes and lysosomes. The molecular basis for Ca(2+) release by NAADP, however, is uncertain. TRP mucolipins (TRPMLs) and two-pore channels (TPCs) are Ca(2+)-permeable ion channels present within the endolysosomal system. Both have been proposed as targets for NAADP. In the present study, we probed possible physical and functional association of these ion channels. Exogenously expressed TRPML1 showed near complete colocalization with TPC2 and partial colocalization with TPC1. TRPML3 overlap with TPC2 was more modest. TRPML1 and to some extent TRPML3 co-immunoprecipitated with TPC2 but less so with TPC1. Current recording, however, showed that TPC1 and TPC2 did not affect the activity of wild-type TRPML1 or constitutively active TRPML1(V432P). N-terminally truncated TPC2 (TPC2delN), which is targeted to the plasma membrane, also failed to affect TRPML1 and TRPML1(V432P) channel function or TRPML1(V432P)-mediated Ca(2+) influx. Whereas overexpression of TPCs enhanced NAADP-mediated Ca(2+) signals, overexpression of TRPML1 did not, and the dominant negative TRPML1(D471K) was without affect on endogenous NAADP-mediated Ca(2+) signals. Furthermore, the single channel properties of NAADP-activated TPC2delN were not affected by TRPML1. Finally, NAADP-evoked Ca(2+) oscillations in pancreatic acinar cells were identical in wild-type and TRPML1(-/-) cells. We conclude that although TRPML1 and TPCs are present in the same complex, they function as two independent organellar ion channels and that TPCs, not TRPMLs, are the targets for NAADP.
Collapse
Affiliation(s)
- Soichiro Yamaguchi
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75235, USA
| | | | | | | | | | | | | | | | | |
Collapse
|