201
|
Kasitinon SY, Eskiocak U, Martin M, Bezwada D, Khivansara V, Tasdogan A, Zhao Z, Mathews T, Aurora AB, Morrison SJ. TRPML1 Promotes Protein Homeostasis in Melanoma Cells by Negatively Regulating MAPK and mTORC1 Signaling. Cell Rep 2019; 28:2293-2305.e9. [PMID: 31461647 PMCID: PMC6813770 DOI: 10.1016/j.celrep.2019.07.086] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/01/2019] [Accepted: 07/23/2019] [Indexed: 12/14/2022] Open
Abstract
We screen ion channels and transporters throughout the genome to identify those required by human melanoma cells but not by normal human melanocytes. We discover that Mucolipin-1 (MCOLN1), which encodes the lysosomal cation channel TRPML1, is preferentially required for the survival and proliferation of melanoma cells. Loss of MCOLN1/TRPML1 function impairs the growth of patient-derived melanomas in culture and in xenografts but does not affect the growth of human melanocytes. TRPML1 expression and macropinocytosis are elevated in melanoma cells relative to melanocytes. TRPML1 is required in melanoma cells to negatively regulate MAPK pathway and mTORC1 signaling. TRPML1-deficient melanoma cells exhibit decreased survival, proliferation, tumor growth, and macropinocytosis, as well as serine depletion and proteotoxic stress. All of these phenotypes are partially or completely rescued by mTORC1 inhibition. Melanoma cells thus increase TRPML1 expression relative to melanocytes to attenuate MAPK and mTORC1 signaling, to sustain macropinocytosis, and to avoid proteotoxic stress.
Collapse
Affiliation(s)
- Stacy Y Kasitinon
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ugur Eskiocak
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Misty Martin
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Divya Bezwada
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Vishal Khivansara
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Alpaslan Tasdogan
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zhiyu Zhao
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Thomas Mathews
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Arin B Aurora
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sean J Morrison
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
202
|
BK channels regulate extracellular Tat-mediated HIV-1 LTR transactivation. Sci Rep 2019; 9:12285. [PMID: 31439883 PMCID: PMC6706582 DOI: 10.1038/s41598-019-48777-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 08/05/2019] [Indexed: 01/22/2023] Open
Abstract
HIV-1 Tat is essential for HIV-1 replication and plays an important role in latent HIV-1 infection, HIV-1 associated neurological complication, and other HIV-1 comorbidities. Secreted from HIV-1 infected or transfected cells, Tat can be up-taken into cells by receptor-mediated endocytosis and internalized into endolysosomes. To reach nucleus where it can facilitate HIV-1 viral replication, exogenous Tat has to escape the degradation by endolysosomes. Because of findings that endolysosome de-acidification with, for example, the weak-base anti-malarial drug chloroquine prevents exogenous Tat degradation and enhances the amount of Tat available to activate HIV-1 LTR, we hypothesize that acidifying endolysosomes may enhance Tat degradation in endolysosomes and restrict LTR transactivation. Here, we determined the involvement of endolysosome-resident transient receptor potential mucolipin 1 channel (TRPML1) and the big conductance Ca2+-activated potassium (BK) channel in regulating endolysosome pH, as well as Tat-mediated HIV-1 LTR transactivation in U87MG cells stably integrated with HIV-1 LTR luciferase reporter. Activating TRPML1 channels with ML-SA1 acidified endolysosomes and restricted Tat-mediated HIV-1 LTR transactivation. These effects of ML-SA1 appeared to be mediated through activation of BK channels, because the effects of ML-SA1 on Tat-mediated HIV-1 LTR transactivation were blocked using pharmacological inhibitors or shRNA knock-down of BK channels. On the other hand, activating TRPML1 and BK channels enhanced cellular degradation of exogenous Tat. These results suggest that acidifying endolysosomes by activating TRPML1 or BK channels may provide therapeutic benefit against latent HIV-1 infection, HIV-1 associated neurocognitive disorders, and other HIV-1 comorbidities.
Collapse
|
203
|
Khan N, Haughey NJ, Nath A, Geiger JD. Involvement of organelles and inter-organellar signaling in the pathogenesis of HIV-1 associated neurocognitive disorder and Alzheimer's disease. Brain Res 2019; 1722:146389. [PMID: 31425679 DOI: 10.1016/j.brainres.2019.146389] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/26/2019] [Accepted: 08/13/2019] [Indexed: 12/30/2022]
Abstract
Endolysosomes, mitochondria, peroxisomes, endoplasmic reticulum, and plasma membranes are now known to physically and functionally interact with each other. Such findings of inter-organellar signaling and communication has led to a resurgent interest in cell biology and an increased appreciation for the physiological actions and pathological consequences of the dynamic physical and chemical communications occurring between intracellular organelles. Others and we have shown that HIV-1 proteins implicated in the pathogenesis of neuroHIV and that Alzheimer's disease both affects the structure and function of intracellular organelles. Intracellular organelles are highly mobile, and their intracellular distribution almost certainly affects their ability to interact with other organelles and to regulate such important physiological functions as endolysosome acidification, cell motility, and nutrient homeostasis. Indeed, compounds that acidify endolysosomes cause endolysosomes to exhibit a mainly perinuclear pattern while compounds that de-acidify endolysosomes cause these organelles to exhibit a larger profile as well as movement towards plasma membranes. Endolysosome pH might be an early event in the pathogenesis of neuroHIV and Alzheimer's disease and in terms of organellar biology endolysosome changes might be upstream of HIV-1 protein-induced changes to other organelles. Thus, inter-organellar signaling mechanisms might be involved in the pathogenesis of neuroHIV and other neurological disorders, and a better understanding of inter-organellar signaling might lead to improved therapeutic strategies.
Collapse
Affiliation(s)
- Nabab Khan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, United States
| | - Norman J Haughey
- Department of Neurology, Johns Hopkins University, Baltimore, MD, United States
| | - Avindra Nath
- National Institute of Neurological Diseases and Stroke, Bethesda, MD, United States
| | - Jonathan D Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, United States.
| |
Collapse
|
204
|
Nash B, Tarn K, Irollo E, Luchetta J, Festa L, Halcrow P, Datta G, Geiger JD, Meucci O. Morphine-Induced Modulation of Endolysosomal Iron Mediates Upregulation of Ferritin Heavy Chain in Cortical Neurons. eNeuro 2019; 6:ENEURO.0237-19.2019. [PMID: 31300544 PMCID: PMC6675873 DOI: 10.1523/eneuro.0237-19.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 07/01/2019] [Indexed: 01/01/2023] Open
Abstract
HIV-associated neurocognitive disorders (HAND) remain prevalent and are aggravated by µ-opioid use. We have previously shown that morphine and other µ-opioids may contribute to HAND by inhibiting the homeostatic and neuroprotective chemokine receptor CXCR4 in cortical neurons, and this novel mechanism depends on upregulation of the protein ferritin heavy chain (FHC). Here, we examined the cellular events and potential mechanisms involved in morphine-mediated FHC upregulation using rat cortical neurons of either sex in vitro and in vivo. Morphine dose dependently increased FHC protein levels in primary neurons through µ-opioid receptor (µOR) and Gαi-protein signaling. Cytoplasmic FHC levels were significantly elevated, but nuclear FHC levels and FHC gene expression were unchanged. Morphine-treated rats also displayed increased FHC levels in layer 2/3 neurons of the prefrontal cortex. Importantly, both in vitro and in vivo FHC upregulation was accompanied by loss of mature dendritic spines, which was also dependent on µOR and Gαi-protein signaling. Moreover, morphine upregulated ferritin light chain (FLC), a component of the ferritin iron storage complex, suggesting that morphine altered neuronal iron metabolism. Indeed, prior to FHC upregulation, morphine increased cytoplasmic labile iron levels as a function of decreased endolysosomal iron. In line with this, chelation of endolysosomal iron (but not extracellular iron) blocked morphine-induced FHC upregulation and dendritic spine reduction, whereas iron overloading mimicked the effect of morphine on FHC and dendritic spines. Overall, these data demonstrate that iron mediates morphine-induced FHC upregulation and consequent dendritic spine deficits and implicate endolysosomal iron efflux to the cytoplasm in these effects.
Collapse
Affiliation(s)
- Bradley Nash
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Kevin Tarn
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Elena Irollo
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Jared Luchetta
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Lindsay Festa
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
| | - Peter Halcrow
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203
| | - Gaurav Datta
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203
| | - Jonathan D Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203
| | - Olimpia Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19102
| |
Collapse
|
205
|
The activation of Mucolipin TRP channel 1 (TRPML1) protects motor neurons from L-BMAA neurotoxicity by promoting autophagic clearance. Sci Rep 2019; 9:10743. [PMID: 31341250 PMCID: PMC6656764 DOI: 10.1038/s41598-019-46708-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 07/04/2019] [Indexed: 12/14/2022] Open
Abstract
Cellular clearance mechanisms including the autophagy-lysosome pathway are impaired in amyotrophic lateral sclerosis (ALS). One of the most important proteins involved in the regulation of autophagy is the lysosomal Ca2+ channel Mucolipin TRP channel 1 (TRPML1). Therefore, we investigated the role of TRPML1 in a neuronal model of ALS/Parkinson-dementia complex reproduced by the exposure of motor neurons to the cyanobacterial neurotoxin beta-methylamino-L-alanine (L-BMAA). Under these conditions, L-BMAA induces a dysfunction of the endoplasmic reticulum (ER) leading to ER stress and cell death. Therefore we hypothesized a dysfunctional coupling between lysosomes and ER in L-BMAA-treated motor neurons. Here, we showed that in motor neuronal cells TRPML1 as well as the lysosomal protein LAMP1 co-localized with ER. In addition, TRPML1 co-immunoprecipitated with the ER Ca2+ sensor STIM1. Functionally, the TRPML1 agonist ML-SA1 induced lysosomal Ca2+ release in a dose-dependent way in motor neuronal cells. The SERCA inhibitor thapsigargin increased the fluorescent signal associated with lysosomal Ca2+ efflux in the cells transfected with the genetically encoded Ca2+ indicator GCaMP3-ML1, thus suggesting an interplay between the two organelles. Moreover, chronic exposure to L-BMAA reduced TRPML1 protein expression and produced an impairment of both lysosomal and ER Ca2+ homeostasis in primary motor neurons. Interestingly, the preincubation of ML-SA1, by an early activation of AMPK and beclin 1, rescued motor neurons from L-BMAA-induced cell death and reduced the expression of the ER stress marker GRP78. Finally, ML-SA1 reduced the accumulation of the autophagy-related proteins p62/SQSTM1 and LC3-II in L-BMAA-treated motor neurons. Collectively, we propose that the pharmacological stimulation of TRPML1 can rescue motor neurons from L-BMAA-induced toxicity by boosting autophagy and reducing ER stress.
Collapse
|
206
|
Wheeler S, Haberkant P, Bhardwaj M, Tongue P, Ferraz MJ, Halter D, Sprong H, Schmid R, Aerts JM, Sullo N, Sillence DJ. Cytosolic glucosylceramide regulates endolysosomal function in Niemann-Pick type C disease. Neurobiol Dis 2019; 127:242-252. [DOI: 10.1016/j.nbd.2019.03.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 03/06/2019] [Accepted: 03/09/2019] [Indexed: 12/22/2022] Open
|
207
|
Krogsaeter EK, Biel M, Wahl-Schott C, Grimm C. The protein interaction networks of mucolipins and two-pore channels. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2019; 1866:1111-1123. [PMID: 30395881 PMCID: PMC7111325 DOI: 10.1016/j.bbamcr.2018.10.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/24/2018] [Accepted: 10/26/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND The endolysosomal, non-selective cation channels, two-pore channels (TPCs) and mucolipins (TRPMLs), regulate intracellular membrane dynamics and autophagy. While partially compensatory for each other, isoform-specific intracellular distribution, cell-type expression patterns, and regulatory mechanisms suggest different channel isoforms confer distinct properties to the cell. SCOPE OF REVIEW Briefly, established TPC/TRPML functions and interaction partners ('interactomes') are discussed. Novel TRPML3 interactors are shown, and a meta-analysis of experimentally obtained channel interactomes conducted. Accordingly, interactomes are compared and contrasted, and subsequently described in detail for TPC1, TPC2, TRPML1, and TRPML3. MAJOR CONCLUSIONS TPC interactomes are well-defined, encompassing intracellular membrane organisation proteins. TRPML interactomes are varied, encompassing cardiac contractility- and chaperone-mediated autophagy proteins, alongside regulators of intercellular signalling. GENERAL SIGNIFICANCE Comprising recently proposed targets to treat cancers, infections, metabolic disease and neurodegeneration, the advancement of TPC/TRPML understanding is of considerable importance. This review proposes novel directions elucidating TPC/TRPML relevance in health and disease. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.
Collapse
Affiliation(s)
- Einar K Krogsaeter
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Munich (LMU) Nussbaumstrasse 26, 80336 Munich
| | - Martin Biel
- Department of Pharmacy - Center for Drug Research and Center for Integrated Protein Science Munich (CIPSM), Ludwig-Maximilians-Universität München, Germany
| | - Christian Wahl-Schott
- Hannover Medical School, Institute for Neurophysiology, OE 4230, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Christian Grimm
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Munich (LMU) Nussbaumstrasse 26, 80336 Munich.
| |
Collapse
|
208
|
Fiorenza MT, Moro E, Erickson RP. The pathogenesis of lysosomal storage disorders: beyond the engorgement of lysosomes to abnormal development and neuroinflammation. Hum Mol Genet 2019; 27:R119-R129. [PMID: 29718288 DOI: 10.1093/hmg/ddy155] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 04/24/2018] [Indexed: 01/03/2023] Open
Abstract
There is growing evidence that the complex clinical manifestations of lysosomal storage diseases (LSDs) are not fully explained by the engorgement of the endosomal-autophagic-lysosomal system. In this review, we explore current knowledge of common pathogenetic mechanisms responsible for the early onset of tissue abnormalities of two LSDs, Mucopolysaccharidosis type II (MPSII) and Niemann-Pick type C (NPC) diseases. In particular, perturbations of the homeostasis of glycosaminoglycans (GAGs) and cholesterol (Chol) in MPSII and NPC diseases, respectively, affect key biological processes, including morphogen signaling. Both GAGs and Chol finely regulate the release, reception and tissue distribution of Shh. Hence, not surprisingly, developmental processes depending on correct Shh signaling have been found altered in both diseases. Besides abnormal signaling, exaggerated activation of microglia and impairment of autophagy and mitophagy occur in both diseases, largely before the appearance of typical pathological signs.
Collapse
Affiliation(s)
- Maria Teresa Fiorenza
- Division of Neuroscience, Department of Psychology and "Daniel Bovet" Neurobiology Research Center, Sapienza University of Rome, Rome, Italy.,IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Enrico Moro
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | |
Collapse
|
209
|
Zhang X, Chen W, Gao Q, Yang J, Yan X, Zhao H, Su L, Yang M, Gao C, Yao Y, Inoki K, Li D, Shao R, Wang S, Sahoo N, Kudo F, Eguchi T, Ruan B, Xu H. Rapamycin directly activates lysosomal mucolipin TRP channels independent of mTOR. PLoS Biol 2019; 17:e3000252. [PMID: 31112550 PMCID: PMC6528971 DOI: 10.1371/journal.pbio.3000252] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 04/18/2019] [Indexed: 02/06/2023] Open
Abstract
Rapamycin (Rap) and its derivatives, called rapalogs, are being explored in clinical trials targeting cancer and neurodegeneration. The underlying mechanisms of Rap actions, however, are not well understood. Mechanistic target of rapamycin (mTOR), a lysosome-localized protein kinase that acts as a critical regulator of cellular growth, is believed to mediate most Rap actions. Here, we identified mucolipin 1 (transient receptor potential channel mucolipin 1 [TRPML1], also known as MCOLN1), the principle Ca2+ release channel in the lysosome, as another direct target of Rap. Patch-clamping of isolated lysosomal membranes showed that micromolar concentrations of Rap and some rapalogs activated lysosomal TRPML1 directly and specifically. Pharmacological inhibition or genetic inactivation of mTOR failed to mimic the Rap effect. In vitro binding assays revealed that Rap bound directly to purified TRPML1 proteins with a micromolar affinity. In both healthy and disease human fibroblasts, Rap and rapalogs induced autophagic flux via nuclear translocation of transcription factor EB (TFEB). However, such effects were abolished in TRPML1-deficient cells or by TRPML1 inhibitors. Hence, Rap and rapalogs promote autophagy via a TRPML1-dependent mechanism. Given the demonstrated roles of TRPML1 and TFEB in cellular clearance, we propose that lysosomal TRPML1 may contribute a significant portion to the in vivo neuroprotective and anti-aging effects of Rap via an augmentation of autophagy and lysosomal biogenesis.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Wei Chen
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Qiong Gao
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Junsheng Yang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Xueni Yan
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Han Zhao
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Lin Su
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Meimei Yang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Neurology, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Chenlang Gao
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Yao Yao
- Department of Integrative and Molecular Physiology and Internal Medicine, Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ken Inoki
- Department of Integrative and Molecular Physiology and Internal Medicine, Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Dan Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Rong Shao
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Shiyi Wang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Nirakar Sahoo
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Fumitaka Kudo
- Department of Chemistry, Tokyo Institute of Technology, Ookayama, Meguro-ku, Tokyo, Japan
| | - Tadashi Eguchi
- Department of Chemistry, Tokyo Institute of Technology, Ookayama, Meguro-ku, Tokyo, Japan
| | - Benfang Ruan
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
- * E-mail: (HX); (BR)
| | - Haoxing Xu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail: (HX); (BR)
| |
Collapse
|
210
|
Capurro MI, Greenfield LK, Prashar A, Xia S, Abdullah M, Wong H, Zhong XZ, Bertaux-Skeirik N, Chakrabarti J, Siddiqui I, O'Brien C, Dong X, Robinson L, Peek RM, Philpott DJ, Zavros Y, Helmrath M, Jones NL. VacA generates a protective intracellular reservoir for Helicobacter pylori that is eliminated by activation of the lysosomal calcium channel TRPML1. Nat Microbiol 2019; 4:1411-1423. [PMID: 31110360 DOI: 10.1038/s41564-019-0441-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 03/28/2019] [Indexed: 01/15/2023]
Abstract
Helicobacter pylori infection is a proven carcinogen for gastric cancer. Its virulence factor vacuolating cytotoxin A (VacA) promotes more severe disease and gastric colonization. VacA, by an unknown mechanism, usurps lysosomal and autophagy pathways to generate a protected reservoir for H. pylori that confers bacterial survival in vitro. Here, we show the existence of a VacA-generated intracellular niche in vivo that protects the bacteria from antibiotic treatment and leads to infection recrudescence after therapy. Furthermore, we report that VacA targets the lysosomal calcium channel TRPML1 to disrupt endolysosomal trafficking and mediate these effects. Remarkably, H. pylori that lack toxigenic VacA colonize enlarged dysfunctional lysosomes in the gastric epithelium of trpml1-null mice, where they are protected from eradication therapy. Furthermore, a small molecule agonist directed against TRPML1 reversed the toxic effects of VacA on endolysosomal trafficking, culminating in the clearance of intracellular bacteria. These results suggest that TRPML1 may represent a therapeutic target for chronic H. pylori infection.
Collapse
Affiliation(s)
- Mariana I Capurro
- Department of Paediatrics and Physiology, University of Toronto; Division of Gastroenterology, Hepatology and Nutrition, and Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Laura K Greenfield
- Department of Paediatrics and Physiology, University of Toronto; Division of Gastroenterology, Hepatology and Nutrition, and Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Akriti Prashar
- Department of Paediatrics and Physiology, University of Toronto; Division of Gastroenterology, Hepatology and Nutrition, and Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sunny Xia
- Department of Paediatrics and Physiology, University of Toronto; Division of Gastroenterology, Hepatology and Nutrition, and Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Majd Abdullah
- Department of Paediatrics and Physiology, University of Toronto; Division of Gastroenterology, Hepatology and Nutrition, and Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Harikesh Wong
- Department of Paediatrics, University of Toronto; Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Xi Zoe Zhong
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Nina Bertaux-Skeirik
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Jayati Chakrabarti
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Iram Siddiqui
- Department of Pathology, University of Toronto; The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Catherine O'Brien
- University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Xianping Dong
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Lisa Robinson
- Department of Paediatrics, University of Toronto; Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Richard M Peek
- Division of Gastroenterology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Yana Zavros
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Michael Helmrath
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Nicola L Jones
- Department of Paediatrics and Physiology, University of Toronto; Division of Gastroenterology, Hepatology and Nutrition, and Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada.
| |
Collapse
|
211
|
Increased Lysosomal Exocytosis Induced by Lysosomal Ca 2+ Channel Agonists Protects Human Dopaminergic Neurons from α-Synuclein Toxicity. J Neurosci 2019; 39:5760-5772. [PMID: 31097622 DOI: 10.1523/jneurosci.3085-18.2019] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 04/06/2019] [Accepted: 05/04/2019] [Indexed: 01/08/2023] Open
Abstract
The accumulation of misfolded proteins is a common pathological feature of many neurodegenerative disorders, including synucleinopathies such as Parkinson's disease (PD), which is characterized by the presence of α-synuclein (α-syn)-containing Lewy bodies. However, although recent studies have investigated α-syn accumulation and propagation in neurons, the molecular mechanisms underlying α-syn transmission have been largely unexplored. Here, we examined a monogenic form of synucleinopathy caused by loss-of-function mutations in lysosomal ATP13A2/PARK9. These studies revealed that lysosomal exocytosis regulates intracellular levels of α-syn in human neurons. Loss of PARK9 function in patient-derived dopaminergic neurons disrupted lysosomal Ca2+ homeostasis, reduced lysosomal Ca2+ storage, increased cytosolic Ca2+, and impaired lysosomal exocytosis. Importantly, this dysfunction in lysosomal exocytosis impaired α-syn secretion from both axons and soma, promoting α-syn accumulation. However, activation of the lysosomal Ca2+ channel transient receptor potential mucolipin 1 (TRPML1) was sufficient to upregulate lysosomal exocytosis, rescue defective α-syn secretion, and prevent α-syn accumulation. Together, these results suggest that intracellular α-syn levels are regulated by lysosomal exocytosis in human dopaminergic neurons and may represent a potential therapeutic target for PD and other synucleinopathies.SIGNIFICANCE STATEMENT Parkinson's disease (PD) is the second most common neurodegenerative disease linked to the accumulation of α-synuclein (α-syn) in patient neurons. However, it is unclear what the mechanism might be. Here, we demonstrate a novel role for lysosomal exocytosis in clearing intracellular α-syn and show that impairment of this pathway by mutations in the PD-linked gene ATP13A2/PARK9 contributes to α-syn accumulation in human dopaminergic neurons. Importantly, upregulating lysosomal exocytosis by increasing lysosomal Ca2+ levels was sufficient to rescue defective α-syn secretion and accumulation in patient neurons. These studies identify lysosomal exocytosis as a potential therapeutic target in diseases characterized by the accumulation of α-syn, including PD.
Collapse
|
212
|
Westman J, Grinstein S, Maxson ME. Revisiting the role of calcium in phagosome formation and maturation. J Leukoc Biol 2019; 106:837-851. [DOI: 10.1002/jlb.mr1118-444r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/24/2019] [Accepted: 04/25/2019] [Indexed: 12/19/2022] Open
Affiliation(s)
- Johannes Westman
- Program in Cell BiologyHospital for Sick Children Toronto Ontario Canada
| | - Sergio Grinstein
- Program in Cell BiologyHospital for Sick Children Toronto Ontario Canada
- Department of BiochemistryUniversity of Toronto Toronto Ontario Canada
- Keenan Research Centre of the Li Ka Shing Knowledge InstituteSt. Michael's Hospital Toronto Ontario Canada
| | - Michelle E. Maxson
- Program in Cell BiologyHospital for Sick Children Toronto Ontario Canada
| |
Collapse
|
213
|
Vrieling F, Wilson L, Rensen PCN, Walzl G, Ottenhoff THM, Joosten SA. Oxidized low-density lipoprotein (oxLDL) supports Mycobacterium tuberculosis survival in macrophages by inducing lysosomal dysfunction. PLoS Pathog 2019; 15:e1007724. [PMID: 30998773 PMCID: PMC6490946 DOI: 10.1371/journal.ppat.1007724] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 04/30/2019] [Accepted: 03/21/2019] [Indexed: 02/07/2023] Open
Abstract
Type 2 diabetes mellitus (DM) is a major risk factor for developing tuberculosis (TB). TB-DM comorbidity is expected to pose a serious future health problem due to the alarming rise in global DM incidence. At present, the causal underlying mechanisms linking DM and TB remain unclear. DM is associated with elevated levels of oxidized low-density lipoprotein (oxLDL), a pathologically modified lipoprotein which plays a key role during atherosclerosis development through the formation of lipid-loaded foamy macrophages, an event which also occurs during progression of the TB granuloma. We therefore hypothesized that oxLDL could be a common factor connecting DM to TB. To study this, we measured oxLDL levels in plasma samples of healthy controls, TB, DM and TB-DM patients, and subsequently investigated the effect of oxLDL treatment on human macrophage infection with Mycobacterium tuberculosis (Mtb). Plasma oxLDL levels were significantly elevated in DM patients and associated with high triglyceride levels in TB-DM. Strikingly, incubation with oxLDL strongly increased macrophage Mtb load compared to native or acetylated LDL (acLDL). Mechanistically, oxLDL -but not acLDL- treatment induced macrophage lysosomal cholesterol accumulation and increased protein levels of lysosomal and autophagy markers, while reducing Mtb colocalization with lysosomes. Importantly, combined treatment of acLDL and intracellular cholesterol transport inhibitor (U18666A) mimicked the oxLDL-induced lysosomal phenotype and impaired macrophage Mtb control, illustrating that the localization of lipid accumulation is critical. Collectively, these results demonstrate that oxLDL could be an important DM-associated TB-risk factor by causing lysosomal dysfunction and impaired control of Mtb infection in human macrophages. Tuberculosis (TB) is an infectious disease of the lungs caused by a bacterium, Mycobacterium tuberculosis (Mtb), and is responsible for over a million deaths per year worldwide. Population studies have demonstrated that type 2 diabetes mellitus (DM) is a risk factor for TB as it triples the risk of developing the disease. DM is a metabolic disorder which is generally associated with obesity, and is characterized by resistance to the pancreatic hormone insulin and high blood glucose and lipid levels. As the global incidence of DM is rising at an alarming rate, especially in regions where TB is common, it is important to understand precisely how DM increases the risk of developing TB. Both TB and DM are associated with the development of foamy macrophages, lipid-loaded white blood cells, which can be the result of a specific lipoprotein particle called oxidized low-density lipoprotein (oxLDL). Here, we demonstrated that DM patients have high blood levels of oxLDL, and generating foamy macrophages with oxLDL supported Mtb survival after infection as a result of faulty intracellular cholesterol accumulation. Our results propose a proof of concept for oxLDL as a risk factor for TB development, encouraging future studies on lipid-lowering therapies for TB-DM.
Collapse
Affiliation(s)
- Frank Vrieling
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, ZA Leiden, The Netherlands
| | - Louis Wilson
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, ZA Leiden, The Netherlands
| | - Patrick C. N. Rensen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Albinusdreef 2, ZA Leiden, The Netherlands
| | - Gerhard Walzl
- DST/NRF Center of Excellence for Biomedical Tuberculosis Research, SA MRC Center for TB Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences Stellenbosch University, Francie van Zijl Drive, Tygerberg, Cape Town, South Africa
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, ZA Leiden, The Netherlands
| | - Simone A. Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, ZA Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
214
|
Transient Receptor Potential Mucolipin-1 Channels in Glioblastoma: Role in Patient's Survival. Cancers (Basel) 2019; 11:cancers11040525. [PMID: 31013784 PMCID: PMC6521337 DOI: 10.3390/cancers11040525] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 04/09/2019] [Indexed: 02/06/2023] Open
Abstract
A link between mucolipin channels and tumors has been recently suggested. Herein, we aim to investigate the transient receptor potential mucolipin (TRPML)-1 relevance in glioblastoma. The expression of this channel was evaluated via qRT-PCR and immunohistochemistry in biopsies from 66 glioblastoma patients and two human glioblastoma cell lines and compared to normal human brain, astrocytes, and epileptic tissues. The subcellular distribution of TRPML-1 was examined via confocal microscopy in the glioma cell lines. Then, to assess the role of TRPML-1, cell viability assays have been conducted in T98 and U251 cell lines treated with the specific TRPML-1 agonist, MK6-83. We found that MK6-83 reduced cell viability and induced caspase-3-dependent apoptosis. Indeed, the TRPML-1 silencing or the blockage of TRPML-1 dependent [Ca2+]i release abrogated these effects. In addition, exposure of glioma cells to the reactive oxygen species (ROS) inducer, carbonyl cyanide m-chlorophenylhydrazone (CCCP), stimulated a TRPML-1-dependent autophagic cell death, as demonstrated by the ability of the autophagic inhibitor bafilomycin A, the TRPML-1 inhibitor sphingomyelin, and the TRPML-1 silencing to completely inhibit the CCCP-mediated effects. To test a possible correlation with patient’s survival, Kaplan–Meier, univariate, and multivariate analysis have been performed. Data showed that the loss/reduction of TRPML-1 mRNA expression strongly correlates with short survival in glioblastoma (GBM) patients, suggesting that the reduction of TRPML-1 expression represents a negative prognostic factor in GBM patients.
Collapse
|
215
|
Jung J, Cho KJ, Naji AK, Clemons KN, Wong CO, Villanueva M, Gregory S, Karagas NE, Tan L, Liang H, Rousseau MA, Tomasevich KM, Sikora AG, Levental I, van der Hoeven D, Zhou Y, Hancock JF, Venkatachalam K. HRAS-driven cancer cells are vulnerable to TRPML1 inhibition. EMBO Rep 2019; 20:e46685. [PMID: 30787043 PMCID: PMC6446245 DOI: 10.15252/embr.201846685] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 01/28/2019] [Accepted: 02/01/2019] [Indexed: 12/28/2022] Open
Abstract
By serving as intermediaries between cellular metabolism and the bioenergetic demands of proliferation, endolysosomes allow cancer cells to thrive under normally detrimental conditions. Here, we show that an endolysosomal TRP channel, TRPML1, is necessary for the proliferation of cancer cells that bear activating mutations in HRAS Expression of MCOLN1, which encodes TRPML1, is significantly elevated in HRAS-positive tumors and inversely correlated with patient prognosis. Concordantly, MCOLN1 knockdown or TRPML1 inhibition selectively reduces the proliferation of cancer cells that express oncogenic, but not wild-type, HRAS Mechanistically, TRPML1 maintains oncogenic HRAS in signaling-competent nanoclusters at the plasma membrane by mediating cholesterol de-esterification and transport. TRPML1 inhibition disrupts the distribution and levels of cholesterol and thereby attenuates HRAS nanoclustering and plasma membrane abundance, ERK phosphorylation, and cell proliferation. These findings reveal a selective vulnerability of HRAS-driven cancers to TRPML1 inhibition, which may be leveraged as an actionable therapeutic strategy.
Collapse
Affiliation(s)
- Jewon Jung
- Department of Integrative Biology and Pharmacology, McGovern Medical School, the University of Texas Health Sciences Center (UTHealth), Houston, TX, USA
| | - Kwang-Jin Cho
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - Ali K Naji
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center, Houston, TX, USA
| | - Kristen N Clemons
- Department of Integrative Biology and Pharmacology, McGovern Medical School, the University of Texas Health Sciences Center (UTHealth), Houston, TX, USA
- Graduate Program in Biochemistry and Cell Biology, MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Ching On Wong
- Department of Integrative Biology and Pharmacology, McGovern Medical School, the University of Texas Health Sciences Center (UTHealth), Houston, TX, USA
| | - Mariana Villanueva
- Bobby R. Alford Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
- Patient Derived Xenografts and Advanced in vivo Models Core Facility, Baylor College of Medicine, Houston, TX, USA
| | - Steven Gregory
- Department of Integrative Biology and Pharmacology, McGovern Medical School, the University of Texas Health Sciences Center (UTHealth), Houston, TX, USA
- Graduate Program in Biochemistry and Cell Biology, MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Nicholas E Karagas
- Department of Integrative Biology and Pharmacology, McGovern Medical School, the University of Texas Health Sciences Center (UTHealth), Houston, TX, USA
- Graduate Program in Biochemistry and Cell Biology, MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Lingxiao Tan
- Department of Integrative Biology and Pharmacology, McGovern Medical School, the University of Texas Health Sciences Center (UTHealth), Houston, TX, USA
- Graduate Program in Biochemistry and Cell Biology, MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Hong Liang
- Department of Integrative Biology and Pharmacology, McGovern Medical School, the University of Texas Health Sciences Center (UTHealth), Houston, TX, USA
| | - Morgan A Rousseau
- Department of Integrative Biology and Pharmacology, McGovern Medical School, the University of Texas Health Sciences Center (UTHealth), Houston, TX, USA
| | - Kelly M Tomasevich
- Department of Integrative Biology and Pharmacology, McGovern Medical School, the University of Texas Health Sciences Center (UTHealth), Houston, TX, USA
| | - Andrew G Sikora
- Bobby R. Alford Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
- Patient Derived Xenografts and Advanced in vivo Models Core Facility, Baylor College of Medicine, Houston, TX, USA
| | - Ilya Levental
- Department of Integrative Biology and Pharmacology, McGovern Medical School, the University of Texas Health Sciences Center (UTHealth), Houston, TX, USA
- Graduate Program in Biochemistry and Cell Biology, MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Dharini van der Hoeven
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center, Houston, TX, USA
| | - Yong Zhou
- Department of Integrative Biology and Pharmacology, McGovern Medical School, the University of Texas Health Sciences Center (UTHealth), Houston, TX, USA
- Graduate Program in Biochemistry and Cell Biology, MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - John F Hancock
- Department of Integrative Biology and Pharmacology, McGovern Medical School, the University of Texas Health Sciences Center (UTHealth), Houston, TX, USA
- Graduate Program in Biochemistry and Cell Biology, MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Kartik Venkatachalam
- Department of Integrative Biology and Pharmacology, McGovern Medical School, the University of Texas Health Sciences Center (UTHealth), Houston, TX, USA
- Graduate Program in Biochemistry and Cell Biology, MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
216
|
Guan Y, Li X, Umetani M, Boini KM, Li PL, Zhang Y. Tricyclic antidepressant amitriptyline inhibits autophagic flux and prevents tube formation in vascular endothelial cells. Basic Clin Pharmacol Toxicol 2019; 124:370-384. [PMID: 30311396 PMCID: PMC6226027 DOI: 10.1111/bcpt.13146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 10/04/2018] [Indexed: 02/06/2023]
Abstract
Amitriptyline is a tricyclic antidepressant and an inhibitor of lysosomal acid sphingomyelinase (ASM). Amitriptyline is well known for its cardiovascular side effects and toxicity in psychiatric patients. However, the mechanisms underlying the cardiovascular side effects of amitriptyline remain largely undefined. This study aimed to determine the effects of amitriptyline on angiogenic capability of vascular endothelial cells in physiological settings and identify its mechanism of action. The ex vivo aortic ring angiogenesis and in vitro-cultured endothelial cell tube formation assay were used to assess the effects of amitriptyline on endothelial angiogenic capability. It was demonstrated that amitriptyline impaired the angiogenesis of aortic rings, which was similar to that found in aortic rings with haploinsufficiency of the ASM gene. In cultured mouse microvascular endothelial cells (MVECs), amitriptyline impaired the proliferation and tube formation under basal condition, which were accompanied by attenuated angiogenic signalling pathways such as endothelial nitric oxide synthase, Akt and Erk1/2 pathways. Mechanistically, amitriptyline inhibited autophagic flux without affecting autophagosome biogenesis at basal condition. ASM gene silencing or autophagy inhibition mimics the inhibitory effects of amitriptyline on endothelial cell proliferation and tube formation. Collectively, our data suggest that amitriptyline inhibits endothelial cell proliferation and angiogenesis via blockade of ASM-autophagic flux axis. It is implicated that the cardiovascular side effects of amitriptyline may be associated with its inhibitory action on physiological angiogenesis.
Collapse
Affiliation(s)
- Yinglu Guan
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Xiang Li
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Michihisa Umetani
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Krishna M. Boini
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Yang Zhang
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| |
Collapse
|
217
|
Liu EA, Lieberman AP. The intersection of lysosomal and endoplasmic reticulum calcium with autophagy defects in lysosomal diseases. Neurosci Lett 2019; 697:10-16. [PMID: 29704574 PMCID: PMC6202281 DOI: 10.1016/j.neulet.2018.04.049] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 04/13/2018] [Accepted: 04/24/2018] [Indexed: 01/01/2023]
Abstract
The lysosomal storage disorders (LSDs) encompass a group of more than 50 inherited diseases characterized by the accumulation of lysosomal substrates. Two-thirds of patients experience significant neurological symptoms, but the mechanisms of neurodegeneration are not well understood. Interestingly, a wide range of LSDs show defects in both autophagy and Ca2+ homeostasis, which is notable as Ca2+ is a key regulator of autophagy. The crosstalk between these pathways in the context of LSD pathogenesis is not well characterized, but further understanding of this relationship could open up promising therapeutic targets. This review discusses the role of endoplasmic reticulum and lysosomal Ca2+ in autophagy regulation and highlights what is known about defects in autophagy and Ca2+ homeostasis in two LSDs, Niemann-Pick type C disease and Gaucher disease.
Collapse
Affiliation(s)
- Elaine A Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, United States; Medical Scientist Training Program, University of Michigan Medical School, Ann Arbor, MI 48109, United States; Cellular and Molecular Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Andrew P Lieberman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, United States.
| |
Collapse
|
218
|
Fernandez-Mosquera L, Yambire KF, Couto R, Pereyra L, Pabis K, Ponsford AH, Diogo CV, Stagi M, Milosevic I, Raimundo N. Mitochondrial respiratory chain deficiency inhibits lysosomal hydrolysis. Autophagy 2019; 15:1572-1591. [PMID: 30917721 PMCID: PMC6693470 DOI: 10.1080/15548627.2019.1586256] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Mitochondria are key organelles for cellular metabolism, and regulate several processes including cell death and macroautophagy/autophagy. Here, we show that mitochondrial respiratory chain (RC) deficiency deactivates AMP-activated protein kinase (AMPK, a key regulator of energy homeostasis) signaling in tissue and in cultured cells. The deactivation of AMPK in RC-deficiency is due to increased expression of the AMPK-inhibiting protein FLCN (folliculin). AMPK is found to be necessary for basal lysosomal function, and AMPK deactivation in RC-deficiency inhibits lysosomal function by decreasing the activity of the lysosomal Ca2+ channel MCOLN1 (mucolipin 1). MCOLN1 is regulated by phosphoinositide kinase PIKFYVE and its product PtdIns(3,5)P2, which is also decreased in RC-deficiency. Notably, reactivation of AMPK, in a PIKFYVE-dependent manner, or of MCOLN1 in RC-deficient cells, restores lysosomal hydrolytic capacity. Building on these data and the literature, we propose that downregulation of the AMPK-PIKFYVE-PtdIns(3,5)P2-MCOLN1 pathway causes lysosomal Ca2+ accumulation and impaired lysosomal catabolism. Besides unveiling a novel role of AMPK in lysosomal function, this study points to the mechanism that links mitochondrial malfunction to impaired lysosomal catabolism, underscoring the importance of AMPK and the complexity of organelle cross-talk in the regulation of cellular homeostasis. Abbreviation: ΔΨm: mitochondrial transmembrane potential; AMP: adenosine monophosphate; AMPK: AMP-activated protein kinase; ATG5: autophagy related 5; ATP: adenosine triphosphate; ATP6V0A1: ATPase, H+ transporting, lysosomal, V0 subbunit A1; ATP6V1A: ATPase, H+ transporting, lysosomal, V0 subbunit A; BSA: bovine serum albumin; CCCP: carbonyl cyanide-m-chlorophenylhydrazone; CREB1: cAMP response element binding protein 1; CTSD: cathepsin D; CTSF: cathepsin F; DMEM: Dulbecco’s modified Eagle’s medium; DMSO: dimethyl sulfoxide; EBSS: Earl’s balanced salt solution; ER: endoplasmic reticulum; FBS: fetal bovine serum; FCCP: carbonyl cyanide-p-trifluoromethoxyphenolhydrazone; GFP: green fluorescent protein; GPN: glycyl-L-phenylalanine 2-naphthylamide; LAMP1: lysosomal associated membrane protein 1; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MCOLN1/TRPML1: mucolipin 1; MEF: mouse embryonic fibroblast; MITF: melanocyte inducing transcription factor; ML1N*2-GFP: probe used to detect PtdIns(3,5)P2 based on the transmembrane domain of MCOLN1; MTORC1: mechanistic target of rapamycin kinase complex 1; NDUFS4: NADH:ubiquinone oxidoreductase subunit S4; OCR: oxygen consumption rate; PBS: phosphate-buffered saline; pcDNA: plasmid cytomegalovirus promoter DNA; PCR: polymerase chain reaction; PtdIns3P: phosphatidylinositol-3-phosphate; PtdIns(3,5)P2: phosphatidylinositol-3,5-bisphosphate; PIKFYVE: phosphoinositide kinase, FYVE-type zinc finger containing; P/S: penicillin-streptomycin; PVDF: polyvinylidene fluoride; qPCR: quantitative real time polymerase chain reaction; RFP: red fluorescent protein; RNA: ribonucleic acid; SDS-PAGE: sodium dodecyl sulfate polyacrylamide gel electrophoresis; shRNA: short hairpin RNA; siRNA: small interfering RNA; TFEB: transcription factor EB; TFE3: transcription factor binding to IGHM enhancer 3; TMRM: tetramethylrhodamine, methyl ester, perchlorate; ULK1: unc-51 like autophagy activating kinase 1; ULK2: unc-51 like autophagy activating kinase 2; UQCRC1: ubiquinol-cytochrome c reductase core protein 1; v-ATPase: vacuolar-type H+-translocating ATPase; WT: wild-type
Collapse
Affiliation(s)
- Lorena Fernandez-Mosquera
- a Institute of Cellular Biochemistry, University Medical Center Goettingen , Goettingen , Germany.,b Doctoral Program in Molecular Medicine, Georg August University Goettingen , Goettingen , Germany
| | - King Faisal Yambire
- a Institute of Cellular Biochemistry, University Medical Center Goettingen , Goettingen , Germany.,c International Max-Planck Research School in Neuroscience , Goettingen , Germany.,d European Neuroscience Institute Goettingen, University Medical Center Goettingen and Max-Planck Society , Goettingen , Germany
| | - Renata Couto
- a Institute of Cellular Biochemistry, University Medical Center Goettingen , Goettingen , Germany.,e Doctoral Program in Molecular Biology of Cells, Göttingen Graduate School for Neurosciences, Biophysics, and Molecular Biosciences, University of Goettingen , Goettingen , Germany
| | - Leonardo Pereyra
- a Institute of Cellular Biochemistry, University Medical Center Goettingen , Goettingen , Germany.,e Doctoral Program in Molecular Biology of Cells, Göttingen Graduate School for Neurosciences, Biophysics, and Molecular Biosciences, University of Goettingen , Goettingen , Germany
| | - Kamil Pabis
- a Institute of Cellular Biochemistry, University Medical Center Goettingen , Goettingen , Germany
| | - Amy H Ponsford
- f Institute of Translational Medicine, University of Liverpool , Liverpool , UK
| | - Cátia V Diogo
- a Institute of Cellular Biochemistry, University Medical Center Goettingen , Goettingen , Germany
| | - Massimiliano Stagi
- f Institute of Translational Medicine, University of Liverpool , Liverpool , UK
| | - Ira Milosevic
- d European Neuroscience Institute Goettingen, University Medical Center Goettingen and Max-Planck Society , Goettingen , Germany
| | - Nuno Raimundo
- a Institute of Cellular Biochemistry, University Medical Center Goettingen , Goettingen , Germany
| |
Collapse
|
219
|
Exosomal release through TRPML1-mediated lysosomal exocytosis is required for adipogenesis. Biochem Biophys Res Commun 2019; 510:409-415. [PMID: 30711251 PMCID: PMC9883805 DOI: 10.1016/j.bbrc.2019.01.115] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 01/26/2019] [Indexed: 01/31/2023]
Abstract
The lysosomal Ca2+ permeable channel TRPML1 (MCOLN1) plays key roles in lysosomal membrane trafficking, including the fusion of late endosomes to lysosomes and lysosomal exocytosis, both of which are essential for release of exosomes into the extracellular milieu. Multiple lines of evidence indicate that the contents of adipocyte-derived exosomes mediate diverse cellular responses, including adipogenic differentiation. In this study, we aimed to define the potential roles of TRPML1 in lysosomal membrane trafficking during adipogenesis and in exosomal release. In response to adipogenic stimuli, the endogenous TRPML1 expression in OP9 pre-adipocytes was increased in a time-dependent manner, and the acute deletion of TRPML1 reduced lipid synthesis and expression of differentiation-related marker genes. Notably, mature adipocyte-derived exosomes were found to be necessary for adipogenesis and were dependent on TRPML1-mediated lysosomal exocytosis. Taken together, our findings indicate that TRPML1 mediates diverse roles in adipocyte differentiation and exosomal release. Further, we propose that TRPML1 should be considered as a regulator of obesity-related diseases.
Collapse
|
220
|
Boudewyn LC, Walkley SU. Current concepts in the neuropathogenesis of mucolipidosis type IV. J Neurochem 2019; 148:669-689. [PMID: 29770442 PMCID: PMC6239999 DOI: 10.1111/jnc.14462] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/29/2018] [Accepted: 05/02/2018] [Indexed: 12/11/2022]
Abstract
Mucolipidosis type IV (MLIV) is an autosomal recessive, lysosomal storage disorder causing progressively severe intellectual disability, motor and speech deficits, retinal degeneration often culminating in blindness, and systemic disease causing a shortened lifespan. MLIV results from mutations in the gene MCOLN1 encoding the transient receptor potential channel mucolipin-1. It is an ultra-rare disease and is currently known to affect just over 100 diagnosed individuals. The last decade has provided a wealth of research focused on understanding the role of the enigmatic mucolipin-1 protein in cell and brain function and how its absence causes disease. This review explores our current understanding of the mucolipin-1 protein in relation to neuropathogenesis in MLIV and describes recent findings implicating mucolipin-1's important role in mechanistic target of rapamycin and TFEB (transcription factor EB) signaling feedback loops as well as in the function of the greater endosomal/lysosomal system. In addition to addressing the vital role of mucolipin-1 in the brain, we also report new data on the question of whether haploinsufficiency as would be anticipated in MCOLN1 heterozygotes is associated with any evidence of neuron dysfunction or disease. Greater insights into the role of mucolipin-1 in the nervous system can be expected to shed light not only on MLIV disease but also on numerous processes governing normal brain function. This article is part of the Special Issue "Lysosomal Storage Disorders".
Collapse
Affiliation(s)
- Lauren C. Boudewyn
- Dominick P. Purpura Department of Neuroscience, Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, New York
| | - Steven U. Walkley
- Dominick P. Purpura Department of Neuroscience, Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
221
|
Wheeler S, Schmid R, Sillence DJ. Lipid⁻Protein Interactions in Niemann⁻Pick Type C Disease: Insights from Molecular Modeling. Int J Mol Sci 2019; 20:E717. [PMID: 30736449 PMCID: PMC6387118 DOI: 10.3390/ijms20030717] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 01/31/2019] [Accepted: 02/03/2019] [Indexed: 12/19/2022] Open
Abstract
The accumulation of lipids in the late endosomes and lysosomes of Niemann⁻Pick type C disease (NPCD) cells is a consequence of the dysfunction of one protein (usually NPC1) but induces dysfunction in many proteins. We used molecular docking to propose (a) that NPC1 exports not just cholesterol, but also sphingosine, (b) that the cholesterol sensitivity of big potassium channel (BK) can be traced to a previously unappreciated site on the channel's voltage sensor, (c) that transient receptor potential mucolipin 1 (TRPML1) inhibition by sphingomyelin is likely an indirect effect, and (d) that phosphoinositides are responsible for both the mislocalization of annexin A2 (AnxA2) and a soluble NSF (N-ethylmaleimide Sensitive Fusion) protein attachment receptor (SNARE) recycling defect. These results are set in the context of existing knowledge of NPCD to sketch an account of the endolysosomal pathology key to this disease.
Collapse
Affiliation(s)
- Simon Wheeler
- School of Pharmacy, De Montfort University, The Gateway, Leicester LE1 9BH, UK.
| | - Ralf Schmid
- Leicester Institute of Structural and Chemical Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 7RH, UK.
| | - Dan J Sillence
- School of Pharmacy, De Montfort University, The Gateway, Leicester LE1 9BH, UK.
| |
Collapse
|
222
|
Li P, Gu M, Xu H. Lysosomal Ion Channels as Decoders of Cellular Signals. Trends Biochem Sci 2019; 44:110-124. [PMID: 30424907 PMCID: PMC6340733 DOI: 10.1016/j.tibs.2018.10.006] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/09/2018] [Accepted: 10/15/2018] [Indexed: 02/08/2023]
Abstract
Lysosomes, the degradation center of the cell, are filled with acidic hydrolases. Lysosomes generate nutrient-sensitive signals to regulate the import of H+, hydrolases, and endocytic and autophagic cargos, as well as the export of their degradation products (catabolites). In response to environmental and cellular signals, lysosomes change their positioning, number, morphology, size, composition, and activity within minutes to hours to meet the changing cellular needs. Ion channels in the lysosome are essential transducers that mediate signal-initiated Ca2+/Fe2+/Zn2+ release and H+/Na+/K+-dependent changes of membrane potential across the perimeter membrane. Dysregulation of lysosomal ion flux impairs lysosome movement, membrane trafficking, nutrient sensing, membrane repair, organelle membrane contact, and lysosome biogenesis and adaptation. Hence, activation and inhibition of lysosomal channels by synthetic modulators may tune lysosome function to maintain cellular health and promote cellular clearance in lysosome storage disorders.
Collapse
Affiliation(s)
- Ping Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China; Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA; These authors contributed equally to this work
| | - Mingxue Gu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA; These authors contributed equally to this work
| | - Haoxing Xu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
223
|
Plesch E, Chen CC, Butz E, Scotto Rosato A, Krogsaeter EK, Yinan H, Bartel K, Keller M, Robaa D, Teupser D, Holdt LM, Vollmar AM, Sippl W, Puertollano R, Medina D, Biel M, Wahl-Schott C, Bracher F, Grimm C. Selective agonist of TRPML2 reveals direct role in chemokine release from innate immune cells. eLife 2018; 7:39720. [PMID: 30479274 PMCID: PMC6257821 DOI: 10.7554/elife.39720] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 10/26/2018] [Indexed: 12/21/2022] Open
Abstract
Cytokines and chemokines are produced and secreted by a broad range of immune cells including macrophages. Remarkably, little is known about how these inflammatory mediators are released from the various immune cells. Here, the endolysosomal cation channel TRPML2 is shown to play a direct role in chemokine trafficking and secretion from murine macrophages. To demonstrate acute and direct involvement of TRPML2 in these processes, the first isoform-selective TRPML2 channel agonist was generated, ML2-SA1. ML2-SA1 was not only found to directly stimulate release of the chemokine CCL2 from macrophages but also to stimulate macrophage migration, thus mimicking CCL2 function. Endogenous TRPML2 is expressed in early/recycling endosomes as demonstrated by endolysosomal patch-clamp experimentation and ML2-SA1 promotes trafficking through early/recycling endosomes, suggesting CCL2 being transported and secreted via this pathway. These data provide a direct link between TRPML2 activation, CCL2 release and stimulation of macrophage migration in the innate immune response.
Collapse
Affiliation(s)
- Eva Plesch
- Department of Pharmacy, Center for Drug Research and Center for Integrated Protein Science Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Cheng-Chang Chen
- Department of Pharmacy, Center for Drug Research and Center for Integrated Protein Science Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Elisabeth Butz
- Department of Pharmacy, Center for Drug Research and Center for Integrated Protein Science Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | | | - Einar K Krogsaeter
- Department of Pharmacology and Toxicology, Medical Faculty, Ludwig Maximilian University of Munich, Munich, Germany
| | - Hua Yinan
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - Karin Bartel
- Department of Pharmacy, Center for Drug Research and Center for Integrated Protein Science Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Marco Keller
- Department of Pharmacy, Center for Drug Research and Center for Integrated Protein Science Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Dina Robaa
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Daniel Teupser
- Institute of Laboratory Medicine, University Hospital Munich, Munich, Germany
| | - Lesca M Holdt
- Institute of Laboratory Medicine, University Hospital Munich, Munich, Germany
| | - Angelika M Vollmar
- Department of Pharmacy, Center for Drug Research and Center for Integrated Protein Science Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Wolfgang Sippl
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Martin Luther University of Halle-Wittenberg, Halle, Germany
| | - Rosa Puertollano
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - Diego Medina
- Telethon Institute of Genetics and Medicine, Naples, Italy
| | - Martin Biel
- Department of Pharmacy, Center for Drug Research and Center for Integrated Protein Science Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | | | - Franz Bracher
- Department of Pharmacy, Center for Drug Research and Center for Integrated Protein Science Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Christian Grimm
- Department of Pharmacology and Toxicology, Medical Faculty, Ludwig Maximilian University of Munich, Munich, Germany
| |
Collapse
|
224
|
Wang WA, Agellon LB, Michalak M. Endoplasmic reticulum calcium dictates the distribution of intracellular unesterified cholesterol. Cell Calcium 2018; 76:116-121. [PMID: 30463032 DOI: 10.1016/j.ceca.2018.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/07/2018] [Accepted: 11/07/2018] [Indexed: 01/06/2023]
Abstract
Endoplasmic reticulum (ER) luminal Ca2+ influences many functions of this organelle, notably the synthesis and quality control of proteins and lipids. Cholesterol is an essential component of biological membranes and a precursor for many biologically important signaling molecules. The sterol regulatory element-binding proteins (SREBPs) are key regulators of lipid metabolism. These transcription factors are synthesized as ER membrane-bound precursor proteins that are proteolytically processed in response to cellular cholesterol status. Recently, ER Ca2+ status was shown to be an important determinant of the basal sensitivity of the sterol sensing mechanism inherent to the SREBP processing pathway. This article discusses the emerging relationship between cellular Ca2+ and cholesterol metabolism.
Collapse
Affiliation(s)
- Wen-An Wang
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2S7, Canada
| | - Luis B Agellon
- School of Human Nutrition, McGill University, Ste. Anne de Bellevue, Quebec, H9X 3V9, Canada.
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2S7, Canada.
| |
Collapse
|
225
|
Gadolinium Chloride Rescues Niemann⁻Pick Type C Liver Damage. Int J Mol Sci 2018; 19:ijms19113599. [PMID: 30441844 PMCID: PMC6274821 DOI: 10.3390/ijms19113599] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 11/01/2018] [Accepted: 11/02/2018] [Indexed: 02/07/2023] Open
Abstract
Niemann–Pick type C (NPC) disease is a rare neurovisceral cholesterol storage disorder that arises from loss of function mutations in the NPC1 or NPC2 genes. Soon after birth, some patients present with an aggressive hepatosplenomegaly and cholestatic signs. Histopathologically, the liver presents with large numbers of foam cells; however, their role in disease pathogenesis has not been explored in depth. Here, we studied the consequences of gadolinium chloride (GdCl3) treatment, a well-known Kupffer/foam cell inhibitor, at late stages of NPC liver disease and compared it with NPC1 genetic rescue in hepatocytes in vivo. GdCl3 treatment successfully blocked the endocytic capacity of hepatic Kupffer/foam measured by India ink endocytosis, decreased the levels CD68—A marker of Kupffer cells in the liver—and normalized the transaminase levels in serum of NPC mice to a similar extent to those obtained by genetic Npc1 rescue of liver cells. Gadolinium salts are widely used as magnetic resonance imaging (MRI) contrasts. This study opens the possibility of targeting foam cells with gadolinium or by other means for improving NPC liver disease. Synopsis: Gadolinium chloride can effectively rescue some parameters of liver dysfunction in NPC mice and its potential use in patients should be carefully evaluated.
Collapse
|
226
|
Zhang X, Hu M, Yang Y, Xu H. Organellar TRP channels. Nat Struct Mol Biol 2018; 25:1009-1018. [PMID: 30374082 DOI: 10.1038/s41594-018-0148-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 09/28/2018] [Indexed: 02/08/2023]
Abstract
Mammalian transient receptor potential (TRP) channels mediate Ca2+ flux and voltage changes across membranes in response to environmental and cellular signals. At the plasma membrane, sensory TRPs act as neuronal detectors of physical and chemical environmental signals, and receptor-operated (metabotropic) TRPs decode extracellular neuroendocrine cues to control body homeostasis. In intracellular membranes, such as those in lysosomes, organellar TRPs respond to compartment-derived signals to control membrane trafficking, signal transduction, and organelle function. Complementing mouse and human genetics and high-resolution structural approaches, physiological studies employing natural agonists and synthetic inhibitors have become critical in resolving the in vivo functions of metabotropic, sensory, and organellar TRPs.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Meiqin Hu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.,Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Yexin Yang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Haoxing Xu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
227
|
Fine M, Schmiege P, Li X. Structural basis for PtdInsP 2-mediated human TRPML1 regulation. Nat Commun 2018; 9:4192. [PMID: 30305615 PMCID: PMC6180102 DOI: 10.1038/s41467-018-06493-7] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 09/05/2018] [Indexed: 12/27/2022] Open
Abstract
Transient receptor potential mucolipin 1 (TRPML1), a lysosomal channel, maintains the low pH and calcium levels for lysosomal function. Several small molecules modulate TRPML1 activity. ML-SA1, a synthetic agonist, binds to the pore region and phosphatidylinositol-3,5-bisphosphate (PtdIns(3,5)P2), a natural lipid, stimulates channel activity to a lesser extent than ML-SA1; moreover, PtdIns(4,5)P2, another natural lipid, prevents TRPML1-mediated calcium release. Notably, PtdIns(3,5)P2 and ML-SA1 cooperate further increasing calcium efflux. Here we report the structures of human TRPML1 at pH 5.0 with PtdIns(3,5)P2, PtdIns(4,5)P2, or ML-SA1 and PtdIns(3,5)P2, revealing a unique lipid-binding site. PtdIns(3,5)P2 and PtdIns(4,5)P2 bind to the extended helices of S1, S2, and S3. The phosphate group of PtdIns(3,5)P2 induces Y355 to form a π-cation interaction with R403, moving the S4-S5 linker, thus allosterically activating the channel. Our structures and electrophysiological characterizations reveal an allosteric site and provide molecular insight into how lipids regulate TRP channels.
Collapse
Affiliation(s)
- Michael Fine
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Philip Schmiege
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Xiaochun Li
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
228
|
Superiority of SpiroZin2 Versus FluoZin-3 for monitoring vesicular Zn 2+ allows tracking of lysosomal Zn 2+ pools. Sci Rep 2018; 8:15034. [PMID: 30302024 PMCID: PMC6177427 DOI: 10.1038/s41598-018-33102-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 09/17/2018] [Indexed: 12/28/2022] Open
Abstract
Small-molecule fluorescent probes are powerful and ubiquitous tools for measuring the concentration and distribution of analytes in living cells. However, accurate characterization of these analytes requires rigorous evaluation of cell-to-cell heterogeneity in fluorescence intensities and intracellular distribution of probes. In this study, we perform a parallel and systematic comparison of two small-molecule fluorescent vesicular Zn2+ probes, FluoZin-3 AM and SpiroZin2, to evaluate each probe for measurement of vesicular Zn2+ pools. Our results reveal that SpiroZin2 is a specific lysosomal vesicular Zn2+ probe and affords uniform measurement of resting Zn2+ levels at the single cell level with proper calibration. In contrast, FluoZin-3 AM produces highly variable fluorescence intensities and non-specifically localizes in the cytosol and multiple vesicular compartments. We further applied SpiroZin2 to lactating mouse mammary epithelial cells and detected a transient increase of lysosomal free Zn2+ at 24-hour after lactation hormone treatment, which implies that lysosomes play a role in the regulation of Zn2+ homeostasis during lactation. This study demonstrates the need for critical characterization of small-molecule fluorescent probes to define the concentration and localization of analytes in different cell populations, and reveals SpiroZin2 to be capable of reporting diverse perturbations to lysosomal Zn2+.
Collapse
|
229
|
Kim SW, Kim DH, Park KS, Kim MK, Park YM, Muallem S, So I, Kim HJ. Palmitoylation controls trafficking of the intracellular Ca 2+ channel MCOLN3/TRPML3 to regulate autophagy. Autophagy 2018; 15:327-340. [PMID: 30215288 PMCID: PMC6333453 DOI: 10.1080/15548627.2018.1518671] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
MCOLN3/TRPML3 is a Ca2+-permeable cation channel that is expressed in multiple subcellular compartments with dynamic localization. Our previous studies suggest that upon macroautophagy/autophagy induction MCOLN3/TRPML3 is recruited and provides Ca2+ for the fusion process in autophagosome biogenesis. However, how intracellular trafficking and the Ca2+ channel function of MCOLN3/TRPML3 are related to autophagy are not known. Here we report that MCOLN3/TRPML3 undergoes palmitoylation at its C-terminal region, which is required for dynamic trafficking and cellular function of MCOLN3/TRPML3 in autophagy. Palmitoylation regulated MCOLN3/TRPML3 surface expression and trafficking, but not channel properties or localization and function of intracellular MCOLN3/TRPML3. Activation of intracellular MCOLN3/TRPML3 induced robust Ca2+ release, which solely increased autophagy in Ca2+- and palmitoylation-dependent manners. Palmitoylation regulated not only intracellular MCOLN3/TRPML3 trafficking to autophagic structures but also autophagic flux in induced autophagy. Importantly, nutrient starvation activated MCOLN3/TRPML3 to release Ca2+ and increased the level of MCOLN3/TRPML3 palmitoylation. Disruption of MCOLN3/TRPML3 palmitoylation, however, abolished the starvation-induced MCOLN3/TRPML3 activation without affecting channel activity. These results suggest that trafficking and channel function of MCOLN3/TRPML3 are regulated in the context of autophagy, and palmitoylation is a prerequisite for the function of MCOLN3/TRPML3 as a Ca2+ channel in autophagosome formation by controlling its trafficking between subcellular compartments. Abbreviations: 17-ODYA, 17-octadecynoic acid; 2-BP, 2-bromopalmitate; BFA, brefeldin A; DN, dominant-negative; GPN, glycyl-L-phenylalanine-beta-naphthylamide; HN, hydroxylamine; KD, knockdown; MCOLN3/TRPML3, mucolipin 3; MS, mass spectrometry; PAT, palmitoyl acyltransferase; PM, plasma membrane; WT, wild type; ZDHHC, a zinc-finger motif and an Asp-His-His-Cys sequence.
Collapse
Affiliation(s)
- So Woon Kim
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Dong Hyun Kim
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Kyoung Sun Park
- Wide River Institute of Immunology, Seoul National University College of Medicine, Gangwon-do, Korea
| | - Mi Kyung Kim
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Yun Min Park
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Shmuel Muallem
- Epithelial Signaling and Transport Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Insuk So
- Department of Physiology and Biophysics, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun Jin Kim
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea,Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea,CONTACT Hyun Jin Kim Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea; Insuk So Department of Physiology and Biophysics, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
230
|
NAADP-dependent Ca 2+ signaling regulates Middle East respiratory syndrome-coronavirus pseudovirus translocation through the endolysosomal system. Cell Calcium 2018; 75:30-41. [PMID: 30121440 PMCID: PMC6251489 DOI: 10.1016/j.ceca.2018.08.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/07/2018] [Accepted: 08/07/2018] [Indexed: 11/09/2022]
Abstract
Nicotinic acid adenine dinucleotide phosphate (NAADP) releases Ca2+ from acidic organelles. Middle East Respiratory Syndrome coronavirus (MERS-CoV) traffics through host-cell acidic organelles. Blockers of NAADP action inhibited pseudotyped MERS-CoV infectivity. Knockdown of two-pore channels (TPCs), a target of NAADP, also blocked MERS-CoV infectivity.
Middle East Respiratory Syndrome coronavirus (MERS-CoV) infections are associated with a significant mortality rate, and existing drugs show poor efficacy. Identifying novel targets/pathways required for MERS infectivity is therefore important for developing novel therapeutics. As an enveloped virus, translocation through the endolysosomal system provides one pathway for cellular entry of MERS-CoV. In this context, Ca2+-permeable channels within the endolysosomal system regulate both the luminal environment and trafficking events, meriting investigation of their role in regulating processing and trafficking of MERS-CoV. Knockdown of endogenous two-pore channels (TPCs), targets for the Ca2+ mobilizing second messenger NAADP, impaired infectivity in a MERS-CoV spike pseudovirus particle translocation assay. This effect was selective as knockdown of the lysosomal cation channel mucolipin-1 (TRPML1) was without effect. Pharmacological inhibition of NAADP-evoked Ca2+ release using several bisbenzylisoquinoline alkaloids also blocked MERS pseudovirus translocation. Knockdown of TPC1 (biased endosomally) or TPC2 (biased lysosomally) decreased the activity of furin, a protease which facilitates MERS fusion with cellular membranes. Pharmacological or genetic inhibition of TPC1 activity also inhibited endosomal motility impairing pseudovirus progression through the endolysosomal system. Overall, these data support a selective, spatially autonomous role for TPCs within acidic organelles to support MERS-CoV translocation.
Collapse
|
231
|
Bretou M, Sáez PJ, Sanséau D, Maurin M, Lankar D, Chabaud M, Spampanato C, Malbec O, Barbier L, Muallem S, Maiuri P, Ballabio A, Helft J, Piel M, Vargas P, Lennon-Duménil AM. Lysosome signaling controls the migration of dendritic cells. Sci Immunol 2018; 2:2/16/eaak9573. [PMID: 29079589 DOI: 10.1126/sciimmunol.aak9573] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 05/26/2017] [Accepted: 09/20/2017] [Indexed: 12/14/2022]
Abstract
Dendritic cells (DCs) patrol their environment by linking antigen acquisition by macropinocytosis to cell locomotion. DC activation upon bacterial sensing inhibits macropinocytosis and increases DC migration, thus promoting the arrival of DCs to lymph nodes for antigen presentation to T cells. The signaling events that trigger such changes are not fully understood. We show that lysosome signaling plays a critical role in this process. Upon bacterial sensing, lysosomal calcium is released by the ionic channel TRPML1 (transient receptor potential cation channel, mucolipin subfamily, member 1), which activates the actin-based motor protein myosin II at the cell rear, promoting fast and directional migration. Lysosomal calcium further induces the activation of the transcription factor EB (TFEB), which translocates to the nucleus to maintain TRPML1 expression. We found that the TRPML1-TFEB axis results from the down-regulation of macropinocytosis after bacterial sensing by DCs. Lysosomal signaling therefore emerges as a hitherto unexpected link between macropinocytosis, actomyosin cytoskeleton organization, and DC migration.
Collapse
Affiliation(s)
- Marine Bretou
- INSERM U932 Immunité et Cancer, Institut Curie, Paris Sciences & Lettres Research University, F-75248 Paris, Cedex 05, France.
| | - Pablo J Sáez
- INSERM U932 Immunité et Cancer, Institut Curie, Paris Sciences & Lettres Research University, F-75248 Paris, Cedex 05, France.,Institut Curie, Paris Sciences & Lettres Research University, CNRS, UMR 144, F-75005 Paris, France.,Institut Pierre-Gilles de Gennes, Paris Sciences & Lettres Research University, F-75005 Paris, France
| | - Doriane Sanséau
- INSERM U932 Immunité et Cancer, Institut Curie, Paris Sciences & Lettres Research University, F-75248 Paris, Cedex 05, France
| | - Mathieu Maurin
- INSERM U932 Immunité et Cancer, Institut Curie, Paris Sciences & Lettres Research University, F-75248 Paris, Cedex 05, France
| | - Danielle Lankar
- INSERM U932 Immunité et Cancer, Institut Curie, Paris Sciences & Lettres Research University, F-75248 Paris, Cedex 05, France
| | - Melanie Chabaud
- INSERM U932 Immunité et Cancer, Institut Curie, Paris Sciences & Lettres Research University, F-75248 Paris, Cedex 05, France
| | - Carmine Spampanato
- Telethon Institute of Genetics and Medicine (TIGEM), I-80078 Pozzuoli, Naples, Italy
| | - Odile Malbec
- INSERM U932 Immunité et Cancer, Institut Curie, Paris Sciences & Lettres Research University, F-75248 Paris, Cedex 05, France
| | - Lucie Barbier
- Institut Curie, Paris Sciences & Lettres Research University, CNRS, UMR 144, F-75005 Paris, France.,Institut Pierre-Gilles de Gennes, Paris Sciences & Lettres Research University, F-75005 Paris, France.,Université Paris-Sud, Université Paris-Saclay, F-91405 Orsay, France
| | - Shmuel Muallem
- Epithelial Signaling and Transport Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Paolo Maiuri
- Institut Curie, Paris Sciences & Lettres Research University, CNRS, UMR 144, F-75005 Paris, France.,Institut Pierre-Gilles de Gennes, Paris Sciences & Lettres Research University, F-75005 Paris, France.,Institute FIRC (Italian Foundation for Cancer Research) of Molecular Oncology (IFOM-FIRC), I-20139 Milano, Italy.,Istituto di Genetica Molecolare-Consiglio Nazionale delle Ricerche (IGM-CNR), Via Abbiategrasso 207, I-27100 Pavia, Italy
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), I-80078 Pozzuoli, Naples, Italy.,Medical Genetics, Department of Translational Medicine, Federico II University, I-80131 Naples, Italy.,Department of Molecular and Human Genetics, Baylor College of Medicine and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Julie Helft
- INSERM U932 Immunité et Cancer, Institut Curie, Paris Sciences & Lettres Research University, F-75248 Paris, Cedex 05, France
| | - Matthieu Piel
- Institut Curie, Paris Sciences & Lettres Research University, CNRS, UMR 144, F-75005 Paris, France.,Institut Pierre-Gilles de Gennes, Paris Sciences & Lettres Research University, F-75005 Paris, France
| | - Pablo Vargas
- INSERM U932 Immunité et Cancer, Institut Curie, Paris Sciences & Lettres Research University, F-75248 Paris, Cedex 05, France. .,Institut Curie, Paris Sciences & Lettres Research University, CNRS, UMR 144, F-75005 Paris, France.,Institut Pierre-Gilles de Gennes, Paris Sciences & Lettres Research University, F-75005 Paris, France
| | - Ana-Maria Lennon-Duménil
- INSERM U932 Immunité et Cancer, Institut Curie, Paris Sciences & Lettres Research University, F-75248 Paris, Cedex 05, France.
| |
Collapse
|
232
|
Abstract
One of the fundamental properties of the cell is the capability to digest and remodel its own components according to metabolic and developmental needs. This is accomplished via the autophagy-lysosome system, a pathway of critical importance in the brain, where it contributes to neuronal plasticity and must protect nonreplaceable neurons from the potentially harmful accumulation of cellular waste. The study of lysosomal biogenesis and function in the context of common and rare neurodegenerative diseases has revealed that a dysfunctional autophagy-lysosome system is the shared nexus where multiple, interconnected pathogenic events take place. The characterization of pathways and mechanisms regulating the lysosomal system and autophagic clearance offers unprecedented opportunities for the development of polyvalent therapeutic strategies based on the enhancement of the autophagy-lysosome pathway to maintain cellular homeostasis and achieve neuroprotection.
Collapse
Affiliation(s)
- Jaiprakash Sharma
- Department of Molecular and Human Genetics, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA;
| | - Alberto di Ronza
- Department of Molecular and Human Genetics, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA;
| | - Parisa Lotfi
- Department of Molecular and Human Genetics, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA;
| | - Marco Sardiello
- Department of Molecular and Human Genetics, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA;
| |
Collapse
|
233
|
Schmiege P, Fine M, Li X. The regulatory mechanism of mammalian TRPMLs revealed by cryo-EM. FEBS J 2018; 285:2579-2585. [PMID: 29577631 DOI: 10.1111/febs.14443] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/15/2018] [Accepted: 03/20/2018] [Indexed: 01/18/2023]
Abstract
Transient receptor potential mucolipin (TRPML) channels are the most recently identified subfamily of TRP channels and have seen a surge of new reports revealing both structural and functional insight. In 2017, several groups published multiple conformations of TRPML channels using cryo-EM. Similar to other TRP channels, the ML subfamily consists of six transmembrane helices (S1-S6), and a pore region including S5, S6, and two pore helices (PH1 and PH2). However, these reports also reveal distinct structural characteristics of the ML subfamily. Asp residues within the luminal pore may function to control calcium/pH regulation. A synthetic agonist, ML-SA1, can bind to the pore region of TRPMLs to force a direct dilation of the lower gate. Finally, biophysical and electrophysiological characterizations reveal another natural agonist binding site in the unique domain of TRPMLs, presumably regulating the conformation of the S4-S5 linker to open the channel. This work elucidates the molecular architecture and provides insights into how multiple ligands regulate TRPMLs.
Collapse
Affiliation(s)
- Philip Schmiege
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michael Fine
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaochun Li
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
234
|
Beckel JM, Gómez NM, Lu W, Campagno KE, Nabet B, Albalawi F, Lim JC, Boesze-Battaglia K, Mitchell CH. Stimulation of TLR3 triggers release of lysosomal ATP in astrocytes and epithelial cells that requires TRPML1 channels. Sci Rep 2018; 8:5726. [PMID: 29636491 PMCID: PMC5893592 DOI: 10.1038/s41598-018-23877-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 03/16/2018] [Indexed: 01/25/2023] Open
Abstract
Cross-reactions between innate immunity, lysosomal function, and purinergic pathways may link signaling systems in cellular pathologies. We found activation of toll-like receptor 3 (TLR3) triggers lysosomal ATP release from both astrocytes and retinal pigmented epithelial (RPE) cells. ATP efflux was accompanied by lysosomal acid phosphatase and beta hexosaminidase release. Poly(I:C) alkalinized lysosomes, and lysosomal alkalization with bafilomycin or chloroquine triggered ATP release. Lysosomal rupture with glycyl-L-phenylalanine-2-naphthylamide (GPN) eliminated both ATP and acid phosphatase release. Secretory lysosome marker LAMP3 colocalized with VNUT, while MANT-ATP colocalized with LysoTracker. Unmodified membrane-impermeant 21-nt and "non-targeting" scrambled 21-nt siRNA triggered ATP and acid phosphatase release, while smaller 16-nt RNA was ineffective. Poly(I:C)-dependent ATP release was reduced by TBK-1 block and in TRPML1-/- cells, while TRPML activation with ML-SA1 was sufficient to release both ATP and acid phosphatase. The ability of poly(I:C) to raise cytoplasmic Ca2+ was abolished by removing extracellular ATP with apyrase, suggesting ATP release by poly(I:C) increased cellular signaling. Starvation but not rapamycin prevented lysosomal ATP release. In summary, stimulation of TLR3 triggers lysosomal alkalization and release of lysosomal ATP through activation of TRPML1; this links innate immunity to purinergic signaling via lysosomal physiology, and suggests even scrambled siRNA can influence these pathways.
Collapse
Affiliation(s)
- Jonathan M Beckel
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Néstor Más Gómez
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Wennan Lu
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Keith E Campagno
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Bardia Nabet
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Farraj Albalawi
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Orthodontics, University of Pennsylvania, Philadelphia, PA, USA
| | - Jason C Lim
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Claire H Mitchell
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Physiology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
235
|
19q13.12 microdeletion syndrome fibroblasts display abnormal storage of cholesterol and sphingolipids in the endo-lysosomal system. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2108-2118. [PMID: 29580926 DOI: 10.1016/j.bbadis.2018.03.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 03/20/2018] [Accepted: 03/21/2018] [Indexed: 11/23/2022]
Abstract
Microdeletions in 19q12q13.12 cause a rare and complex haploinsufficiency syndrome characterized by intellectual deficiency, developmental delays, and neurological movement disorders. Variability in the size and interval of the deletions makes it difficult to attribute the complex clinical phenotype of this syndrome to an underlying gene(s). As an alternate approach, we examined the biochemical and metabolic features of fibroblasts from an affected individual to derive clues as to the molecular basis for the syndrome. Immunofluorescence and electron microscopy of affected fibroblasts revealed an abnormal endo-lysosomal compartment that was characterized by rapid accumulation of lysosomotropic dyes, elevated LAMP1 and LAMP2 expression and vacuoles containing membrane whorls, common features of lysosomal lipid storage disorders. The late endosomes-lysosomes (LE/LY) of affected fibroblasts accumulated low-density lipoprotein cholesterol, and displayed reduced cholesterol esterification and increased de novo cholesterol synthesis, indicative of defective cholesterol transport to the endoplasmic reticulum. Affected fibroblasts also had increased ceramide and sphingolipid mass, altered glycosphingolipid species and accumulation of a fluorescent lactosylceramide probe in LE/LY. Autophagosomes also accumulated in affected fibroblasts because of decreased fusion with autolysosomes, a defect associated with other lysosomal storage diseases. Attempts to correct the cholesterol/sphingolipid storage defect in fibroblasts with cyclodextrin, sphingolipid synthesis inhibitors or by altering ion transport were unsuccessful. Our data show that 19q13.12 deletion fibroblasts have abnormal accumulation of cholesterol and sphingolipids in the endo-lysosomal system that compromises organelle function and could be an underlying cause of the clinical features of the syndrome.
Collapse
|
236
|
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
|
237
|
Sterea AM, Almasi S, El Hiani Y. The hidden potential of lysosomal ion channels: A new era of oncogenes. Cell Calcium 2018; 72:91-103. [PMID: 29748137 DOI: 10.1016/j.ceca.2018.02.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/28/2018] [Accepted: 02/28/2018] [Indexed: 01/14/2023]
Abstract
Lysosomes serve as the control centre for cellular clearance. These membrane-bound organelles receive biomolecules destined for degradation from intracellular and extracellular pathways; thus, facilitating the production of energy and shaping the fate of the cell. At the base of their functionality are the lysosomal ion channels which mediate the function of the lysosome through the modulation of ion influx and efflux. Ion channels form pores in the membrane of lysosomes and allow the passage of ions, a seemingly simple task which harbours the potential of overthrowing the cell's stability. Considered the master regulators of ion homeostasis, these integral membrane proteins enable the proper operation of the lysosome. Defects in the structure or function of these ion channels lead to the development of lysosomal storage diseases, neurodegenerative diseases and cancer. Although more than 50 years have passed since their discovery, lysosomes are not yet fully understood, with their ion channels being even less well characterized. However, significant improvements have been made in the development of drugs targeted against these ion channels as a means of combating diseases. In this review, we will examine how Ca2+, K+, Na+ and Cl- ion channels affect the function of the lysosome, their involvement in hereditary and spontaneous diseases, and current ion channel-based therapies.
Collapse
Affiliation(s)
- Andra M Sterea
- Departments of Physiology & Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Shekoufeh Almasi
- Departments of Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Yassine El Hiani
- Departments of Physiology & Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
238
|
Viaud M, Ivanov S, Vujic N, Duta-Mare M, Aira LE, Barouillet T, Garcia E, Orange F, Dugail I, Hainault I, Stehlik C, Marchetti S, Boyer L, Guinamard R, Foufelle F, Bochem A, Hovingh KG, Thorp EB, Gautier EL, Kratky D, Dasilva-Jardine P, Yvan-Charvet L. Lysosomal Cholesterol Hydrolysis Couples Efferocytosis to Anti-Inflammatory Oxysterol Production. Circ Res 2018. [PMID: 29523554 DOI: 10.1161/circresaha.117.312333] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Macrophages face a substantial amount of cholesterol after the ingestion of apoptotic cells, and the LIPA (lysosomal acid lipase) has a major role in hydrolyzing cholesteryl esters in the endocytic compartment. OBJECTIVE Here, we directly investigated the role of LIPA-mediated clearance of apoptotic cells both in vitro and in vivo. METHODS AND RESULTS We show that LIPA inhibition causes a defective efferocytic response because of impaired generation of 25-hydroxycholesterol and 27-hydroxycholesterol. Reduced synthesis of 25-hydroxycholesterol after LIPA inhibition contributed to defective mitochondria-associated membrane leading to mitochondrial oxidative stress-induced NLRP3 (NOD-like receptor family, pyrin domain containing) inflammasome activation and caspase-1-dependent Rac1 (Ras-related C3 botulinum toxin substrate 1) degradation. A secondary event consisting of failure to appropriately activate liver X receptor-mediated pathways led to mitigation of cholesterol efflux and apoptotic cell clearance. In mice, LIPA inhibition caused defective clearance of apoptotic lymphocytes and stressed erythrocytes by hepatic and splenic macrophages, culminating in splenomegaly and splenic iron accumulation under hypercholesterolemia. CONCLUSIONS Our findings position lysosomal cholesterol hydrolysis as a critical process that prevents metabolic inflammation by enabling efficient macrophage apoptotic cell clearance.
Collapse
Affiliation(s)
- Manon Viaud
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | - Stoyan Ivanov
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | - Nemanja Vujic
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria (N.V., M.D.-M., D.K.)
| | - Madalina Duta-Mare
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria (N.V., M.D.-M., D.K.)
| | - Lazaro-Emilio Aira
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | | | - Elsa Garcia
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | - Francois Orange
- UFR Sciences, Faculté des Sciences de l'Université de Nice-Sophia Antipolis, France (F.O.)
| | - Isabelle Dugail
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1166, Pierre & Marie Curie University, ICAN Institute of Cardiometabolism & Nutrition, Hôpital de la Pitié, Boulevard de l'Hôpital, Paris, France (I.D., E.L.G.)
| | - Isabelle Hainault
- Institut National de la Santé et de la Recherche Médicale (Inserm) UMRS 1138, Centre de Recherche des Cordeliers, Paris, France (I.H., F.F.)
| | - Christian Stehlik
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL (C.S., E.B.T.)
| | - Sandrine Marchetti
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | - Laurent Boyer
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | - Rodolphe Guinamard
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| | - Fabienne Foufelle
- Institut National de la Santé et de la Recherche Médicale (Inserm) UMRS 1138, Centre de Recherche des Cordeliers, Paris, France (I.H., F.F.)
| | | | | | - Edward B Thorp
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL (C.S., E.B.T.)
| | - Emmanuel L Gautier
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1166, Pierre & Marie Curie University, ICAN Institute of Cardiometabolism & Nutrition, Hôpital de la Pitié, Boulevard de l'Hôpital, Paris, France (I.D., E.L.G.)
| | - Dagmar Kratky
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, Austria (N.V., M.D.-M., D.K.)
| | - Paul Dasilva-Jardine
- Academic Medical Center, Amsterdam, The Netherlands; and Staten Biotechnology, Nijmegen, The Netherlands (P.D.-J.)
| | - Laurent Yvan-Charvet
- From the Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France (M.V., S.I., L.-E.A., E.G., S.M., L.B., R.G., L.Y.-C.)
| |
Collapse
|
239
|
Chloroquine modulates antitumor immune response by resetting tumor-associated macrophages toward M1 phenotype. Nat Commun 2018; 9:873. [PMID: 29491374 PMCID: PMC5830447 DOI: 10.1038/s41467-018-03225-9] [Citation(s) in RCA: 304] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 01/30/2018] [Indexed: 02/08/2023] Open
Abstract
Resetting tumor-associated macrophages (TAMs) is a promising strategy to ameliorate the immunosuppressive tumor microenvironment and improve innate and adaptive antitumor immunity. Here we show that chloroquine (CQ), a proven anti-malarial drug, can function as an antitumor immune modulator that switches TAMs from M2 to tumor-killing M1 phenotype. Mechanistically, CQ increases macrophage lysosomal pH, causing Ca2+ release via the lysosomal Ca2+ channel mucolipin-1 (Mcoln1), which induces the activation of p38 and NF-κB, thus polarizing TAMs to M1 phenotype. In parallel, the released Ca2+ activates transcription factor EB (TFEB), which reprograms the metabolism of TAMs from oxidative phosphorylation to glycolysis. As a result, CQ-reset macrophages ameliorate tumor immune microenvironment by decreasing immunosuppressive infiltration of myeloid-derived suppressor cells and Treg cells, thus enhancing antitumor T-cell immunity. These data illuminate a previously unrecognized antitumor mechanism of CQ, suggesting a potential new macrophage-based tumor immunotherapeutic modality. Tumour-associated macrophages (TAMs) display an M2 phenotype that promote tumour immune escape. Here the authors show that Chloroquine (CQ), a lysosome inhibitor used against malaria, inhibits tumour growth by switching TAMs into an M1 tumor-killing phenotype by repolarizing macrophages metabolism.
Collapse
|
240
|
Sun X, Yang Y, Zhong XZ, Cao Q, Zhu XH, Zhu X, Dong XP. A negative feedback regulation of MTORC1 activity by the lysosomal Ca 2+ channel MCOLN1 (mucolipin 1) using a CALM (calmodulin)-dependent mechanism. Autophagy 2018; 14:38-52. [PMID: 29460684 DOI: 10.1080/15548627.2017.1389822] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Macroautophagy/autophagy is an evolutionarily conserved pathway that is required for cellular homeostasis, growth and survival. The lysosome plays an essential role in autophagy regulation. For example, the activity of MTORC1, a master regulator of autophagy, is regulated by nutrients within the lysosome. Starvation inhibits MTORC1 causing autophagy induction. Given that MTORC1 is critical for protein synthesis and cellular homeostasis, a feedback regulatory mechanism must exist to restore MTORC1 during starvation. However, the molecular mechanism underlying this feedback regulation is unclear. In this study, we report that starvation activates the lysosomal Ca2+ release channel MCOLN1 (mucolipin 1) by relieving MTORC1's inhibition of the channel. Activated MCOLN1 in turn facilitates MTORC1 activity that requires CALM (calmodulin). Moreover, both MCOLN1 and CALM are necessary for MTORC1 reactivation during prolonged starvation. Our data suggest that lysosomal Ca2+ signaling is an essential component of the canonical MTORC1-dependent autophagy pathway and MCOLN1 provides a negative feedback regulation of MTORC1 to prevent excessive loss of MTORC1 function during starvation. The feedback regulation may be important for maintaining cellular homeostasis during starvation, as well as many other stressful or disease conditions.
Collapse
Affiliation(s)
- Xue Sun
- a Department of Physiology and Biophysics , Dalhousie University, Sir Charles Tupper Medical Building , Halifax , Nova Scotia, Canada.,d Key Laboratory of Molecular Epigenetics of Ministry of Education , Institute of Cytology and Genetics, Northeast Normal University , Changchun , Jilin , China
| | - Yiming Yang
- a Department of Physiology and Biophysics , Dalhousie University, Sir Charles Tupper Medical Building , Halifax , Nova Scotia, Canada
| | - Xi Zoë Zhong
- a Department of Physiology and Biophysics , Dalhousie University, Sir Charles Tupper Medical Building , Halifax , Nova Scotia, Canada
| | - Qi Cao
- a Department of Physiology and Biophysics , Dalhousie University, Sir Charles Tupper Medical Building , Halifax , Nova Scotia, Canada
| | - Xin-Hong Zhu
- b Institute of Mental Health, Southern Medical University , Guangzhou , China.,c Key Laboratory of Psychiatric Disorders of Guangdong Province , Guangzhou , China
| | - Xiaojuan Zhu
- d Key Laboratory of Molecular Epigenetics of Ministry of Education , Institute of Cytology and Genetics, Northeast Normal University , Changchun , Jilin , China
| | - Xian-Ping Dong
- a Department of Physiology and Biophysics , Dalhousie University, Sir Charles Tupper Medical Building , Halifax , Nova Scotia, Canada
| |
Collapse
|
241
|
Devany J, Chakraborty K, Krishnan Y. Subcellular Nanorheology Reveals Lysosomal Viscosity as a Reporter for Lysosomal Storage Diseases. NANO LETTERS 2018; 18:1351-1359. [PMID: 29313356 DOI: 10.1021/acs.nanolett.7b05040] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
We describe a new method to measure viscosity within subcellular organelles of a living cell using nanorheology. We demonstrate proof of concept by measuring viscosity in lysosomes in multiple cell types and disease models. The lysosome is an organelle responsible for the breakdown of complex biomolecules. When different lysosomal proteins are defective, they are unable to break down specific biological substrates, which get stored within the lysosome, causing about 70 fatal diseases called lysosomal storage disorders (LSDs). Although the buildup of storage material is critical to the pathology of these diseases, methods to monitor cargo accumulation in the lysosome are lacking for most LSDs. Using passive particle tracking nanorheology and fluorescence recovery after photobleaching, we report that viscosity in the lysosome increases significantly during cargo accumulation in several LSD models. In a mammalian cell culture model of Niemann Pick C, lysosomal viscosity directly correlates with the levels of accumulated cholesterol. We also observed increased viscosity in diverse LSD models in Caenorhabditis elegans, revealing that lysosomal viscosity is a powerful reporter with which to monitor substrate accumulation in LSDs for new diagnostics or to assay therapeutic efficacy.
Collapse
Affiliation(s)
- John Devany
- Department of Physics, ‡Department of Chemistry, and §Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, University of Chicago , Chicago, Illinois 60637, United States
| | - Kasturi Chakraborty
- Department of Physics, ‡Department of Chemistry, and §Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, University of Chicago , Chicago, Illinois 60637, United States
| | - Yamuna Krishnan
- Department of Physics, ‡Department of Chemistry, and §Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, University of Chicago , Chicago, Illinois 60637, United States
| |
Collapse
|
242
|
Gómez NM, Lu W, Lim JC, Kiselyov K, Campagno KE, Grishchuk Y, Slaugenhaupt SA, Pfeffer BA, Fliesler SJ, Mitchell CH. Robust lysosomal calcium signaling through channel TRPML1 is impaired by lysosomal lipid accumulation. FASEB J 2018; 32:782-794. [PMID: 29030399 PMCID: PMC5888396 DOI: 10.1096/fj.201700220rr] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 09/26/2017] [Indexed: 12/20/2022]
Abstract
The transient receptor potential cation channel mucolipin 1 (TRPML1) channel is a conduit for lysosomal calcium efflux, and channel activity may be affected by lysosomal contents. The lysosomes of retinal pigmented epithelial (RPE) cells are particularly susceptible to build-up of lysosomal waste products because they must degrade the outer segments phagocytosed daily from adjacent photoreceptors; incomplete degradation leads to accumulation of lipid waste in lysosomes. This study asks whether stimulation of TRPML1 can release lysosomal calcium in RPE cells and whether such release is affected by lysosomal accumulations. The TRPML agonist ML-SA1 raised cytoplasmic calcium levels in mouse RPE cells, hesRPE cells, and ARPE-19 cells; this increase was rapid, robust, reversible, and reproducible. The increase was not altered by extracellular calcium removal or by thapsigargin but was eliminated by lysosomal rupture with glycyl-l-phenylalanine-β-naphthylamide. Treatment with desipramine to inhibit acid sphingomyelinase or YM201636 to inhibit PIKfyve also reduced the cytoplasmic calcium increase triggered by ML-SA1, whereas RPE cells from TRPML1-/- mice showed no response to ML-SA1. Cotreatment with chloroquine and U18666A induced formation of neutral, autofluorescent lipid in RPE lysosomes and decreased lysosomal Ca2+ release. Lysosomal Ca2+ release was also impaired in RPE cells from the ATP-binding cassette, subfamily A, member 4-/- mouse model of Stargardt's retinal dystrophy. Neither TRPML1 mRNA nor total lysosomal calcium levels were altered in these models, suggesting a more direct effect on the channel. In summary, stimulation of TRPML1 elevates cytoplasmic calcium levels in RPE cells, but this response is reduced by lysosomal accumulation.-Gómez, N. M., Lu, W. Lim, J. C., Kiselyov, K., Campagno, K. E., Grishchuk, Y., Slaugenhaupt, S. A., Pfeffer, B., Fliesler, S. J., Mitchell, C. H. Robust lysosomal calcium signaling through channel TRPML1 is impaired by lysosomal lipid accumulation.
Collapse
Affiliation(s)
- Néstor Más Gómez
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Wennan Lu
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jason C. Lim
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kirill Kiselyov
- Department of Biological Science, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Keith E. Campagno
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yulia Grishchuk
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Susan A. Slaugenhaupt
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Bruce A. Pfeffer
- Department of Ophthalmology, Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York (SUNY)–University at Buffalo, Buffalo, New York, USA
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York (SUNY)–University at Buffalo, Buffalo, New York, USA
- State University of New York (SUNY)–Eye Institute, Buffalo, New York, USA
- Research Service, Veterans Affairs Western New York Healthcare System, Buffalo, New York, USA
| | - Steven J. Fliesler
- Department of Ophthalmology, Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York (SUNY)–University at Buffalo, Buffalo, New York, USA
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York (SUNY)–University at Buffalo, Buffalo, New York, USA
- State University of New York (SUNY)–Eye Institute, Buffalo, New York, USA
- Research Service, Veterans Affairs Western New York Healthcare System, Buffalo, New York, USA
| | - Claire H. Mitchell
- Department of Anatomy and Cell Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
243
|
|
244
|
Patel S. Ins and outs of Ca 2+ transport by acidic organelles and cell migration. Commun Integr Biol 2018. [PMCID: PMC5824967 DOI: 10.1080/19420889.2017.1331800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Much contemporary evidence underscores the pathophysiological importance of Ca2+ handling by acidic organelles such as lysosomes. Whereas our knowledge of how Ca2+ is released from these acidic Ca2+ stores (the ‘outs’) is advancing, we know relatively little about how Ca2+ uptake is effected (the ‘ins’). Here I highlight new work identifying animal Ca2+/H+ (CAX) exchangers that localize to acidic organelles, mediate Ca2+ uptake and regulate cell migration in vivo. Continued molecular definition of the acidic Ca2+ store toolkit provides new insight into Ca2+-dependent function.
Collapse
Affiliation(s)
- Sandip Patel
- Department of Cell and Developmental Biology, University College London, London, UK
| |
Collapse
|
245
|
Chang TY, Liu KL, Chang CS, Su CT, Chen SH, Lee YC, Chang JS. Ferric Citrate Supplementation Reduces Red-Blood-Cell Aggregation and Improves CD163+ Macrophage-Mediated Hemoglobin Metabolism in a Rat Model of High-Fat-Diet-Induced Obesity. Mol Nutr Food Res 2017; 62. [PMID: 29064631 DOI: 10.1002/mnfr.201700442] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 10/09/2017] [Indexed: 12/13/2022]
Abstract
SCOPE In adults, >90% of the daily iron requirement is derived from macrophage-mediated heme iron, recycling from senescent red blood cells (RBCs) or free hemoglobin (Hb). Currently, the effects of pharmacological doses of iron supplementation on RBCs and heme iron recycling in obesity are unclear. METHODS AND RESULTS Sprague Dawley rats are fed a standard diet or a 50% high-fat diet (HFD) with (0.25, 1, and 2 g of ferric iron per kg diet) or without ferric citrate supplementation for 12 weeks. Ferric iron increases hepatic iron accumulation in macrophages and hepatocyte-like cells. Compared with rats that received the standard diet, HFD-fed rats exhibit higher RBC aggregation and serum-free Hb levels but lower LVV-hemorphin-7 levels. These effects are reversed by ferric citrate supplementation. Immunofluorescent staining reveals that ferric iron increases the expression of hepatic CD163+ macrophages and heme oxygenase (HO)-1. A further analysis reveals the dose-related effects of ferric iron on hepatic globin degradation proteins (cathepsin D and glyoxalase 1), cytochrome p450 reductase expression, and HO-1 enzyme activity. CONCLUSIONS Ferric citrate supplementation reduces RBC aggregation and improves CD163+ macrophage-mediated Hb metabolism in HFD-induced obese rats. These findings suggest that ferric citrate may be explored as an alternative treatment method for RBC dysfunction.
Collapse
Affiliation(s)
- Ting-Yun Chang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, Taiwan
| | - Kai-Li Liu
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan.,Department of Dietitian, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Cheng Sheng Chang
- Department of Family Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chien-Tien Su
- Department of Family Medicine, Taipei Medical University Hospital, Taipei, Taiwan.,School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Seu-Hwa Chen
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chieh Lee
- Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, Taiwan
| | - Jung-Su Chang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, Taiwan.,Nutrition Research Center, Taipei Medical University Hospital, Taipei, Taiwan.,Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
246
|
Davis W, Tew KD. ATP-binding cassette transporter-2 (ABCA2) as a therapeutic target. Biochem Pharmacol 2017; 151:188-200. [PMID: 29223352 DOI: 10.1016/j.bcp.2017.11.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 11/27/2017] [Indexed: 12/28/2022]
Abstract
The ATP binding cassette transporter ABCA2 is primarily an endolysosomal membrane protein that demonstrates pleiotropic functionalities, coalescing around the maintenance of homeostasis of sterols, sphingolipids and cholesterol. It is most highly expressed in brain tissue and ABCA2 knockout mice express neurological defects consistent with aberrant myelination. Increased expression of the transporter has been linked with resistance to cancer drugs, particularly those possessing a steroid backbone and gene expression (in concert with other genes involved in cholesterol metabolism) was found to be regulated by sterols. Moreover, in macrophages ABCA2 is influenced by sterols and has a role in regulating cholesterol sequestration, potentially important in cardiovascular disease. Accumulating data indicate the critical importance of ABCA2 in mediating movement of sphingolipids within cellular compartments and these have been implicated in various aspects of cholesterol trafficking. Perhaps because the functions of ABCA2 are linked with membrane building blocks, there are reports linking it with human pathologies, including, cholesterolemias and cardiovascular disease, Alzheimer's and cancer. The present review addresses whether there is now sufficient information to consider ABCA2 as a plausible therapeutic target.
Collapse
Affiliation(s)
- Warren Davis
- Dept. of Cell & Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue, BSB, MSC 509, Charleston, SC 29425, United States
| | - Kenneth D Tew
- Dept. of Cell & Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue, BSB, MSC 509, Charleston, SC 29425, United States.
| |
Collapse
|
247
|
Zhou X, Li M, Su D, Jia Q, Li H, Li X, Yang J. Cryo-EM structures of the human endolysosomal TRPML3 channel in three distinct states. Nat Struct Mol Biol 2017; 24:1146-1154. [PMID: 29106414 PMCID: PMC5747366 DOI: 10.1038/nsmb.3502] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/10/2017] [Indexed: 12/23/2022]
Abstract
TRPML3 channels are mainly localized to endolysosomes and play a critical role in the endocytic pathway. Their dysfunction causes deafness and pigmentation defects in mice. TRPML3 activity is inhibited by low endolysosomal pH. Here we present cryo-electron microscopy (cryo-EM) structures of human TRPML3 in the closed, agonist-activated, and low-pH-inhibited states, with resolutions of 4.06, 3.62, and 4.65 Å, respectively. The agonist ML-SA1 lodges between S5 and S6 and opens an S6 gate. A polycystin-mucolipin domain (PMD) forms a luminal cap. S1 extends into this cap, forming a 'gating rod' that connects directly to a luminal pore loop, which undergoes dramatic conformational changes in response to low pH. S2 extends intracellularly and interacts with several intracellular regions to form a 'gating knob'. These unique structural features, combined with the results of electrophysiological studies, indicate a new mechanism by which luminal pH and other physiological modulators such as PIP2 regulate TRPML3 by changing S1 and S2 conformations.
Collapse
Affiliation(s)
- Xiaoyuan Zhou
- Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Minghui Li
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Deyuan Su
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, and Ion Channel Research and Drug Development Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Qi Jia
- Department of Orthopedic Oncology, Shanghai Changzheng Hospital, The Second Military Medical University, Shanghai 200003, China
| | - Huan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, and Ion Channel Research and Drug Development Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - Xueming Li
- Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jian Yang
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, and Ion Channel Research and Drug Development Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
248
|
Torres S, Balboa E, Zanlungo S, Enrich C, Garcia-Ruiz C, Fernandez-Checa JC. Lysosomal and Mitochondrial Liaisons in Niemann-Pick Disease. Front Physiol 2017; 8:982. [PMID: 29249985 PMCID: PMC5714892 DOI: 10.3389/fphys.2017.00982] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 11/16/2017] [Indexed: 12/28/2022] Open
Abstract
Lysosomal storage disorders (LSD) are characterized by the accumulation of diverse lipid species in lysosomes. Niemann-Pick type A/B (NPA/B) and type C diseases Niemann-Pick type C (NPC) are progressive LSD caused by loss of function of distinct lysosomal-residing proteins, acid sphingomyelinase and NPC1, respectively. While the primary cause of these diseases differs, both share common biochemical features, including the accumulation of sphingolipids and cholesterol, predominantly in endolysosomes. Besides these alterations in lysosomal homeostasis and function due to accumulation of specific lipid species, the lysosomal functional defects can have far-reaching consequences, disrupting intracellular trafficking of sterols, lipids and calcium through membrane contact sites (MCS) of apposed compartments. Although MCS between endoplasmic reticulum and mitochondria have been well studied and characterized in different contexts, emerging evidence indicates that lysosomes also exhibit close proximity with mitochondria, which translates in their mutual functional regulation. Indeed, as best illustrated in NPC disease, alterations in the lysosomal-mitochondrial liaisons underlie the secondary accumulation of specific lipids, such as cholesterol in mitochondria, resulting in mitochondrial dysfunction and defective antioxidant defense, which contribute to disease progression. Thus, a better understanding of the lysosomal and mitochondrial interactions and trafficking may identify novel targets for the treatment of Niemann-Pick disease.
Collapse
Affiliation(s)
- Sandra Torres
- Department of Cell Death and Proliferation, Intituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, Barcelona, Spain.,Liver Unit and Hospital Clinc I Provincial, Centro de Investigación Biomédica en Red (CIBEREHD), Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Elisa Balboa
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Silvana Zanlungo
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos Enrich
- Departamento de Biomedicina, Unidad de Biología Celular, Centro de Investigación Biomédica CELLEX, Facultad de Medicina y Ciencias de la Salud, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Universidad de Barcelona, Barcelona, Spain
| | - Carmen Garcia-Ruiz
- Department of Cell Death and Proliferation, Intituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, Barcelona, Spain.,Liver Unit and Hospital Clinc I Provincial, Centro de Investigación Biomédica en Red (CIBEREHD), Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain.,Southern California Research Center for ALDP and Cirrhosis, Los Angeles, CA, United States
| | - Jose C Fernandez-Checa
- Department of Cell Death and Proliferation, Intituto de Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, Barcelona, Spain.,Liver Unit and Hospital Clinc I Provincial, Centro de Investigación Biomédica en Red (CIBEREHD), Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain.,Southern California Research Center for ALDP and Cirrhosis, Los Angeles, CA, United States
| |
Collapse
|
249
|
Schmiege P, Fine M, Blobel G, Li X. Human TRPML1 channel structures in open and closed conformations. Nature 2017; 550:366-370. [PMID: 29019983 DOI: 10.1038/nature24036] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 08/24/2017] [Indexed: 12/20/2022]
Abstract
Transient receptor potential mucolipin 1 (TRPML1) is a Ca2+-releasing cation channel that mediates the calcium signalling and homeostasis of lysosomes. Mutations in TRPML1 lead to mucolipidosis type IV, a severe lysosomal storage disorder. Here we report two electron cryo-microscopy structures of full-length human TRPML1: a 3.72-Å apo structure at pH 7.0 in the closed state, and a 3.49-Å agonist-bound structure at pH 6.0 in an open state. Several aromatic and hydrophobic residues in pore helix 1, helices S5 and S6, and helix S6 of a neighbouring subunit, form a hydrophobic cavity to house the agonist, suggesting a distinct agonist-binding site from that found in TRPV1, a TRP channel from a different subfamily. The opening of TRPML1 is associated with distinct dilations of its lower gate together with a slight structural movement of pore helix 1. Our work reveals the regulatory mechanism of TRPML channels, facilitates better understanding of TRP channel activation, and provides insights into the molecular basis of mucolipidosis type IV pathogenesis.
Collapse
Affiliation(s)
- Philip Schmiege
- Laboratory of Cell Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, New York 10065, USA.,Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Michael Fine
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Günter Blobel
- Laboratory of Cell Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, New York 10065, USA
| | - Xiaochun Li
- Laboratory of Cell Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, New York 10065, USA.,Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
250
|
Chen Q, She J, Zeng W, Guo J, Xu H, Bai XC, Jiang Y. Structure of mammalian endolysosomal TRPML1 channel in nanodiscs. Nature 2017; 550:415-418. [PMID: 29019981 PMCID: PMC5901962 DOI: 10.1038/nature24035] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/24/2017] [Indexed: 12/14/2022]
Abstract
Transient receptor potential mucolipin 1 (TRPML1) is an endo/lysosomal cation channel ubiquitously expressed in mammalian cells1,2 and its loss-of-function mutations are the direct cause of Type IV mucolipidosis (MLIV), an autosomal recessive lysosomal storage disease3-6. Here we present the single particle cryo-electron microscopy (cryo-EM) structure of the mouse TRPML1 channel embedded in nanodiscs. Combined with mutagenesis, the TRPML1 structure reveals that phosphatidylinositol bisphosphate (PIP2) binds to the N-terminus of the channel – distal from the pore – and the helix-turn-helix extension between S2 and S3 likely couples ligand binding to pore opening. The tightly packed selectivity filter contains multiple ion binding sites and the conserved acidic residues form the luminal Ca2+ blocking site that confers luminal pH and Ca2+ modulation on channel conductance. A luminal linker domain forms a fenestrated canopy atop the channel, providing multiple luminal ion passages to the pore and also creating a negative electrostatic trap – preferably for divalent cations at the luminal entrance. The structure also reveals two equally distributed S4-S5 linker conformations in the closed channel, providing structural implication for the S4-S5 linker-mediated PIP2 gating mechanism among TRPML channels7,8.
Collapse
Affiliation(s)
- Qingfeng Chen
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040, USA.,Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8816, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040, USA
| | - Ji She
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040, USA.,Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8816, USA
| | - Weizhong Zeng
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040, USA.,Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8816, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040, USA
| | - Jiangtao Guo
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040, USA.,Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8816, USA
| | - Haoxing Xu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109-1048, USA
| | - Xiao-Chen Bai
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8816, USA
| | - Youxing Jiang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040, USA.,Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8816, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9040, USA
| |
Collapse
|