101
|
Wang S, Sun H, Tanowitz M, Liang XH, Crooke ST. Intra-endosomal trafficking mediated by lysobisphosphatidic acid contributes to intracellular release of phosphorothioate-modified antisense oligonucleotides. Nucleic Acids Res 2017; 45:5309-5322. [PMID: 28379543 PMCID: PMC5605259 DOI: 10.1093/nar/gkx231] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 03/27/2017] [Indexed: 12/12/2022] Open
Abstract
Antisense oligonucleotides (ASOs) with phosphorothioate (PS) linkages are broadly used as research tools and therapeutic agents. Chemically modified PS-ASOs can mediate efficient target reduction by site-specific cleavage of RNA through RNase H1. PS-ASOs are known to be internalized via a number of endocytotic pathways and are released from membrane-enclosed endocytotic organelles, mainly late endosomes (LEs). This study was focused on the details of PS-ASO trafficking through endocytic pathways. It was found that lysobisphosphatidic acid (LBPA) is required for release of PS-ASOs from LEs. PS-ASOs exited early endosomes (EEs) rapidly after internalization and became co-localized with LBPA by 2 hours in LEs. Inside LEs, PS-ASOs and LBPA were co-localized in punctate, dot-like structures, likely intraluminal vesicles (ILVs). Deactivation of LBPA using anti-LBPA antibody significantly decreased PS-ASO activities without affecting total PS-ASO uptake. Reduction of Alix also substantially decreased PS-ASO activities without affecting total PS-ASO uptake. Furthermore, Alix reduction decreased LBPA levels and limited co-localization of LBPA with PS-ASOs at ILVs inside LEs. Thus, the fusion properties of ILVs, which are supported by LBPA, contribute to PS-ASO intracellular release from LEs.
Collapse
Affiliation(s)
- Shiyu Wang
- Department of Core Antisense Research, Ionis Pharmaceuticals, Inc. 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - Hong Sun
- Department of Core Antisense Research, Ionis Pharmaceuticals, Inc. 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - Michael Tanowitz
- Department of Medicinal Chemistry, Ionis Pharmaceuticals, Inc. 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - Xue-Hai Liang
- Department of Core Antisense Research, Ionis Pharmaceuticals, Inc. 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - Stanley T Crooke
- Department of Core Antisense Research, Ionis Pharmaceuticals, Inc. 2855 Gazelle Court, Carlsbad, CA 92010, USA
| |
Collapse
|
102
|
Buckley CM, King JS. Drinking problems: mechanisms of macropinosome formation and maturation. FEBS J 2017; 284:3778-3790. [DOI: 10.1111/febs.14115] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 04/25/2017] [Accepted: 05/17/2017] [Indexed: 11/30/2022]
Affiliation(s)
- Catherine M. Buckley
- Department of Biomedical Sciences Centre for Membrane Interactions and Dynamics University of Sheffield UK
- Bateson Centre University of Sheffield UK
| | - Jason S. King
- Department of Biomedical Sciences Centre for Membrane Interactions and Dynamics University of Sheffield UK
- Bateson Centre University of Sheffield UK
| |
Collapse
|
103
|
Carpaneto A, Boccaccio A, Lagostena L, Di Zanni E, Scholz-Starke J. The signaling lipid phosphatidylinositol-3,5-bisphosphate targets plant CLC-a anion/H + exchange activity. EMBO Rep 2017; 18:1100-1107. [PMID: 28536248 DOI: 10.15252/embr.201643814] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 04/20/2017] [Accepted: 04/21/2017] [Indexed: 12/21/2022] Open
Abstract
Phosphatidylinositol-3,5-bisphosphate (PI(3,5)P2) is a low-abundance signaling lipid associated with endo-lysosomal and vacuolar membranes in eukaryotic cells. Recent studies on Arabidopsis indicated a critical role of PI(3,5)P2 in vacuolar acidification and morphology during ABA-induced stomatal closure, but the molecular targets in plant cells remained unknown. By using patch-clamp recordings on Arabidopsis vacuoles, we show here that PI(3,5)P2 does not affect the activity of vacuolar H+-pyrophosphatase or vacuolar H+-ATPase. Instead, PI(3,5)P2 at low nanomolar concentrations inhibited an inwardly rectifying conductance, which appeared upon vacuolar acidification elicited by prolonged H+ pumping activity. We provide evidence that this novel conductance is mediated by chloride channel a (CLC-a), a member of the anion/H+ exchanger family formerly implicated in stomatal movements in Arabidopsis H+-dependent currents were absent in clc-a knock-out vacuoles, and canonical CLC-a-dependent nitrate/H+ antiport was inhibited by low concentrations of PI(3,5)P2 Finally, using the pH indicator probe BCECF, we show that CLC-a inhibition contributes to vacuolar acidification. These data provide a mechanistic explanation for the essential role of PI(3,5)P2 and advance our knowledge about the regulation of vacuolar ion transport.
Collapse
Affiliation(s)
- Armando Carpaneto
- Institute of Biophysics, Consiglio Nazionale delle Ricerche, Genova, Italy
| | - Anna Boccaccio
- Institute of Biophysics, Consiglio Nazionale delle Ricerche, Genova, Italy
| | - Laura Lagostena
- Institute of Biophysics, Consiglio Nazionale delle Ricerche, Genova, Italy
| | - Eleonora Di Zanni
- Institute of Biophysics, Consiglio Nazionale delle Ricerche, Genova, Italy
| | | |
Collapse
|
104
|
Schulze U, Vollenbröker B, Kühnl A, Granado D, Bayraktar S, Rescher U, Pavenstädt H, Weide T. Cellular vacuolization caused by overexpression of the PIKfyve-binding deficient Vac14L156R is rescued by starvation and inhibition of vacuolar-ATPase. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:749-759. [DOI: 10.1016/j.bbamcr.2017.02.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 02/09/2017] [Accepted: 02/15/2017] [Indexed: 12/28/2022]
|
105
|
Nelson EA, Dyall J, Hoenen T, Barnes AB, Zhou H, Liang JY, Michelotti J, Dewey WH, DeWald LE, Bennett RS, Morris PJ, Guha R, Klumpp-Thomas C, McKnight C, Chen YC, Xu X, Wang A, Hughes E, Martin S, Thomas C, Jahrling PB, Hensley LE, Olinger GG, White JM. The phosphatidylinositol-3-phosphate 5-kinase inhibitor apilimod blocks filoviral entry and infection. PLoS Negl Trop Dis 2017; 11:e0005540. [PMID: 28403145 PMCID: PMC5402990 DOI: 10.1371/journal.pntd.0005540] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 04/24/2017] [Accepted: 03/30/2017] [Indexed: 12/12/2022] Open
Abstract
Phosphatidylinositol-3-phosphate 5-kinase (PIKfyve) is a lipid kinase involved in endosome maturation that emerged from a haploid genetic screen as being required for Ebola virus (EBOV) infection. Here we analyzed the effects of apilimod, a PIKfyve inhibitor that was reported to be well tolerated in humans in phase 2 clinical trials, for its effects on entry and infection of EBOV and Marburg virus (MARV). We first found that apilimod blocks infections by EBOV and MARV in Huh 7, Vero E6 and primary human macrophage cells, with notable potency in the macrophages (IC50, 10 nM). We next observed that similar doses of apilimod block EBOV-glycoprotein-virus like particle (VLP) entry and transcription-replication competent VLP infection, suggesting that the primary mode of action of apilimod is as an entry inhibitor, preventing release of the viral genome into the cytoplasm to initiate replication. After providing evidence that the anti-EBOV action of apilimod is via PIKfyve, we showed that it blocks trafficking of EBOV VLPs to endolysosomes containing Niemann-Pick C1 (NPC1), the intracellular receptor for EBOV. Concurrently apilimod caused VLPs to accumulate in early endosome antigen 1-positive endosomes. We did not detect any effects of apilimod on bulk endosome acidification, on the activity of cathepsins B and L, or on cholesterol export from endolysosomes. Hence by antagonizing PIKfyve, apilimod appears to block EBOV trafficking to its site of fusion and entry into the cytoplasm. Given the drug's observed anti-filoviral activity, relatively unexplored mechanism of entry inhibition, and reported tolerability in humans, we propose that apilimod be further explored as part of a therapeutic regimen to treat filoviral infections.
Collapse
Affiliation(s)
- Elizabeth A. Nelson
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Julie Dyall
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Thomas Hoenen
- Laboratory of Virology, Division of Intramural Research, National Institutes of Health, Hamilton, Montana, United States of America
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald–Insel Riems, Germany
| | - Alyson B. Barnes
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Huanying Zhou
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Janie Y. Liang
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Julia Michelotti
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - William H. Dewey
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Lisa Evans DeWald
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Richard S. Bennett
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Patrick J. Morris
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Rajarshi Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Carleen Klumpp-Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Crystal McKnight
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yu-Chi Chen
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Xin Xu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Amy Wang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Emma Hughes
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Scott Martin
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Craig Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Peter B. Jahrling
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Lisa E. Hensley
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Gene G. Olinger
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Judith M. White
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
106
|
Kerr MC, Gomez GA, Ferguson C, Tanzer MC, Murphy JM, Yap AS, Parton RG, Huston WM, Teasdale RD. Laser-mediated rupture of chlamydial inclusions triggers pathogen egress and host cell necrosis. Nat Commun 2017; 8:14729. [PMID: 28281536 PMCID: PMC5353685 DOI: 10.1038/ncomms14729] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 01/25/2017] [Indexed: 12/21/2022] Open
Abstract
Remarkably little is known about how intracellular pathogens exit the host cell in order to infect new hosts. Pathogenic chlamydiae egress by first rupturing their replicative niche (the inclusion) before rapidly lysing the host cell. Here we apply a laser ablation strategy to specifically disrupt the chlamydial inclusion, thereby uncoupling inclusion rupture from the subsequent cell lysis and allowing us to dissect the molecular events involved in each step. Pharmacological inhibition of host cell calpains inhibits inclusion rupture, but not subsequent cell lysis. Further, we demonstrate that inclusion rupture triggers a rapid necrotic cell death pathway independent of BAK, BAX, RIP1 and caspases. Both processes work sequentially to efficiently liberate the pathogen from the host cytoplasm, promoting secondary infection. These results reconcile the pathogen's known capacity to promote host cell survival and induce cell death.
Collapse
Affiliation(s)
- Markus C. Kerr
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Guillermo A. Gomez
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Charles Ferguson
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Maria C. Tanzer
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - James M. Murphy
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Alpha S. Yap
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Robert G. Parton
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Wilhelmina M. Huston
- School of Life Sciences, University of Technology Sydney, Ultimo, New South Wales 2007, Australia
| | - Rohan D Teasdale
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| |
Collapse
|
107
|
Hasegawa J, Strunk BS, Weisman LS. PI5P and PI(3,5)P 2: Minor, but Essential Phosphoinositides. Cell Struct Funct 2017; 42:49-60. [PMID: 28302928 DOI: 10.1247/csf.17003] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In most eukaryotes, phosphoinositides (PIs) have crucial roles in multiple cellular functions. Although the cellular levels of phosphatidylinositol 5-phosphate (PI5P) and phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2) are extremely low relative to some other PIs, emerging evidence demonstrates that both lipids are crucial for the endocytic pathway, intracellular signaling, and adaptation to stress. Mutations that causes defects in the biosynthesis of PI5P and PI(3,5)P2 are linked to human diseases including neurodegenerative disorders. Here, we review recent findings on cellular roles of PI5P and PI(3,5)P2, as well as the pathophysiological importance of these lipids.Key words: Phosphoinositides, Membrane trafficking, Endocytosis, Vacuoles/Lysosomes, Fab1/PIKfyve.
Collapse
|
108
|
Peterson BG, Tan KW, Osa-Andrews B, Iram SH. High-content screening of clinically tested anticancer drugs identifies novel inhibitors of human MRP1 (ABCC1). Pharmacol Res 2017; 119:313-326. [PMID: 28258008 DOI: 10.1016/j.phrs.2017.02.024] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 02/23/2017] [Accepted: 02/27/2017] [Indexed: 02/06/2023]
Abstract
Multidrug resistance protein 1 (MRP1/ABCC1), an integral transmembrane efflux transporter, belongs to the ATP-binding cassette (ABC) protein superfamily. MRP1 governs the absorption and disposition of a wide variety of endogenous and xenobiotic substrates including various drugs across organs and physiological barriers. Additionally, its overexpression has been implicated in multidrug resistance in chemotherapy of multiple cancers. Here, we describe the development of a high content imaging-based screening assay for MRP1 activity. This live cell-based automated microscopy assay is very robust and allows simultaneous detection of cell permeable, non-toxic and potent inhibitors. The validity of the assay was demonstrated by profiling a library of 386 anti-cancer compounds, which are under clinical trials, for interactions with MRP1. The assay identified 12 potent inhibitors including two known MRP1 inhibitors, cyclosporine A and rapamycin. On the other hand, MRP1-inhibitory activity of tipifarnib, AZD1208, deforolimus, everolimus, temsirolimus, HS-173, YM201636, ESI-09, TAK-733, and CX-6258 has not been previously reported. Inhibition of MRP1 activity was further validated using flow cytometry and confocal microscopy for the respective detection of calcein and doxorubicin in MRP1-overexpressing cells. Among the identified compounds, tipifarnib, AZD1208, rapamycin, deforolimus, everolimus, TAK-733, and temsirolimus resensitized MRP1-overexpressing H69AR cells towards vincristine, a cytotoxic chemotherapeutic agent, by 2-6-fold. Using purified HEK293 membrane vesicles overexpressing MRP1, MRP2, MRP3, and MRP4, we also demonstrated that the identified compounds exert differential and selective response on the uptake of estradiol glucuronide, an endogenous MRP substrate. In summary, we demonstrated the effectiveness of the high content imaging-based high-throughput assay for profiling compound interaction with MRP1.
Collapse
Affiliation(s)
- Brian G Peterson
- Department of Chemistry & Biochemistry, College of Arts and Sciences, South Dakota State University, Brookings, SD, USA
| | - Kee W Tan
- Department of Chemistry & Biochemistry, College of Arts and Sciences, South Dakota State University, Brookings, SD, USA
| | - Bremansu Osa-Andrews
- Department of Chemistry & Biochemistry, College of Arts and Sciences, South Dakota State University, Brookings, SD, USA
| | - Surtaj H Iram
- Department of Chemistry & Biochemistry, College of Arts and Sciences, South Dakota State University, Brookings, SD, USA.
| |
Collapse
|
109
|
Reggiori F, Ungermann C. Autophagosome Maturation and Fusion. J Mol Biol 2017; 429:486-496. [DOI: 10.1016/j.jmb.2017.01.002] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/03/2017] [Accepted: 01/04/2017] [Indexed: 02/07/2023]
|
110
|
Hirano T, Munnik T, Sato MH. Inhibition of phosphatidylinositol 3,5-bisphosphate production has pleiotropic effects on various membrane trafficking routes in Arabidopsis. PLANT & CELL PHYSIOLOGY 2017; 58:120-129. [PMID: 27803131 DOI: 10.1093/pcp/pcw164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 09/15/2016] [Indexed: 06/06/2023]
Abstract
Phosphoinositides play an important role in various membrane trafficking events in eukaryotes. One of them, however, phosphatidylinositol 3,5-bisphosphate [PI(3,5)P2], has not been studied widely in plants. Using a combination of fluorescent reporter proteins and the PI(3,5)P2-specific inhibitor YM202636, here we demonstrated that in Arabidopsis thaliana, PI(3,5)P2 affects various membrane trafficking events, mostly in the post-Golgi routes. We found that YM201636 treatment effectively reduced PI(3,5)P2 concentration not only in the wild type but also in FAB1A-overexpressing Arabidopsis plants. In particular, reduced PI(3,5)P2 levels caused abnormal membrane dynamics of plasma membrane proteins, AUX1 and BOR1, with different trafficking patterns. Secretion and morphological characteristics of late endosomes and vacuoles were also affected by the decreased PI(3,5)P2 production. These pleiotropic defects in the post-Golgi trafficking events were caused by the inhibition of PI(3,5)P2 production. This effect is probably mediated by the inhibition of maturation of FAB1-positive late endosomes, thereby impairing late endosome function. In conclusion, our results imply that in Arabidopsis, late endosomes are involved in multiple post-Golgi membrane trafficking routes including not only vacuolar trafficking and endocytosis but also secretion.
Collapse
Affiliation(s)
- Tomoko Hirano
- Laboratory of Cellular Dynamics, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Teun Munnik
- Plant Physiology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Masa H Sato
- Laboratory of Cellular Dynamics, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| |
Collapse
|
111
|
Uchida Y, Rutaganira FU, Jullié D, Shokat KM, von Zastrow M. Endosomal Phosphatidylinositol 3-Kinase Is Essential for Canonical GPCR Signaling. Mol Pharmacol 2017; 91:65-73. [PMID: 27821547 PMCID: PMC5198513 DOI: 10.1124/mol.116.106252] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 11/04/2016] [Indexed: 02/04/2023] Open
Abstract
G protein-coupled receptors (GPCRs), the largest family of signaling receptors, are critically regulated by endosomal trafficking, suggesting that endosomes might provide new strategies for manipulating GPCR signaling. Here we test this hypothesis by focusing on class III phosphatidylinositol 3-kinase (Vps34), which is an essential regulator of endosomal trafficking. We verify that Vps34 is required for recycling of the β2-adrenoceptor (β2AR), a prototypical GPCR, and then investigate the effects of Vps34 inhibition on the canonical cAMP response elicited by β2AR activation. Vps34 inhibition impairs the ability of cells to recover this response after prolonged activation, which is in accord with the established role of recycling in GPCR resensitization. In addition, Vps34 inhibition also attenuates the short-term cAMP response, and its effect begins several minutes after initial agonist application. These results establish Vps34 as an essential determinant of both short-term and long-term canonical GPCR signaling, and support the potential utility of the endosomal system as a druggable target for signaling.
Collapse
Affiliation(s)
- Yasunori Uchida
- Department of Psychiatry (Y.U., D.J., and M.Z.), Department of Cellular and Molecular Pharmacology (F.U.R., K.M.S., and M.Z.), and Howard Hughes Medical Institute (F.U.R. and K.M.S.), University of California, San Francisco, San Francisco, California
| | - Florentine U Rutaganira
- Department of Psychiatry (Y.U., D.J., and M.Z.), Department of Cellular and Molecular Pharmacology (F.U.R., K.M.S., and M.Z.), and Howard Hughes Medical Institute (F.U.R. and K.M.S.), University of California, San Francisco, San Francisco, California
| | - Damien Jullié
- Department of Psychiatry (Y.U., D.J., and M.Z.), Department of Cellular and Molecular Pharmacology (F.U.R., K.M.S., and M.Z.), and Howard Hughes Medical Institute (F.U.R. and K.M.S.), University of California, San Francisco, San Francisco, California
| | - Kevan M Shokat
- Department of Psychiatry (Y.U., D.J., and M.Z.), Department of Cellular and Molecular Pharmacology (F.U.R., K.M.S., and M.Z.), and Howard Hughes Medical Institute (F.U.R. and K.M.S.), University of California, San Francisco, San Francisco, California
| | - Mark von Zastrow
- Department of Psychiatry (Y.U., D.J., and M.Z.), Department of Cellular and Molecular Pharmacology (F.U.R., K.M.S., and M.Z.), and Howard Hughes Medical Institute (F.U.R. and K.M.S.), University of California, San Francisco, San Francisco, California
| |
Collapse
|
112
|
Hessvik NP, Øverbye A, Brech A, Torgersen ML, Jakobsen IS, Sandvig K, Llorente A. PIKfyve inhibition increases exosome release and induces secretory autophagy. Cell Mol Life Sci 2016; 73:4717-4737. [PMID: 27438886 PMCID: PMC11108566 DOI: 10.1007/s00018-016-2309-8] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 07/08/2016] [Accepted: 07/11/2016] [Indexed: 01/08/2023]
Abstract
Exosomes are vesicles released from cells by fusion of multivesicular bodies (MVBs) with the plasma membrane. This study aimed to investigate whether the phosphoinositide kinase PIKfyve affects this process. Our results show that in PC-3 cells inhibition of PIKfyve by apilimod or depletion by siRNA increased the secretion of the exosomal fraction. Moreover, quantitative electron microscopy analysis showed that cells treated with apilimod contained more MVBs per cell and more intraluminal vesicles per MVB. Interestingly, mass spectrometry analysis revealed a considerable enrichment of autophagy-related proteins (NBR1, p62, LC3, WIPI2) in exosomal fractions released by apilimod-treated cells, a result that was confirmed by immunoblotting. When the exosome preparations were investigated by electron microscopy a small population of p62-labelled electron dense structures was observed together with CD63-containing exosomes. The p62-positive structures were found in less dense fractions than exosomes in density gradients. Inside the cells, p62 and CD63 were found in the same MVB-like organelles. Finally, both the degradation of EGF and long-lived proteins were shown to be reduced by apilimod. In conclusion, inhibition of PIKfyve increases secretion of exosomes and induces secretory autophagy, showing that these pathways are closely linked. We suggest this is due to impaired fusion of lysosomes with both MVBs and autophagosomes, and possibly increased fusion of MVBs with autophagosomes, and that the cells respond by secreting the content of these organelles to maintain cellular homeostasis.
Collapse
Affiliation(s)
- Nina Pettersen Hessvik
- Department of Molecular Cell Biology, The Norwegian Radium Hospital, Institute for Cancer Research, Oslo University Hospital, 0379, Oslo, Norway
- Centre for Cancer Biomedicine, University of Oslo, 0379, Oslo, Norway
| | - Anders Øverbye
- Department of Molecular Cell Biology, The Norwegian Radium Hospital, Institute for Cancer Research, Oslo University Hospital, 0379, Oslo, Norway
- Centre for Cancer Biomedicine, University of Oslo, 0379, Oslo, Norway
| | - Andreas Brech
- Department of Molecular Cell Biology, The Norwegian Radium Hospital, Institute for Cancer Research, Oslo University Hospital, 0379, Oslo, Norway
- Centre for Cancer Biomedicine, University of Oslo, 0379, Oslo, Norway
- Department of Biosciences, University of Oslo, 0316, Oslo, Norway
| | - Maria Lyngaas Torgersen
- Department of Molecular Cell Biology, The Norwegian Radium Hospital, Institute for Cancer Research, Oslo University Hospital, 0379, Oslo, Norway
- Centre for Cancer Biomedicine, University of Oslo, 0379, Oslo, Norway
| | - Ida Seim Jakobsen
- Department of Molecular Cell Biology, The Norwegian Radium Hospital, Institute for Cancer Research, Oslo University Hospital, 0379, Oslo, Norway
- Centre for Cancer Biomedicine, University of Oslo, 0379, Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, The Norwegian Radium Hospital, Institute for Cancer Research, Oslo University Hospital, 0379, Oslo, Norway
- Centre for Cancer Biomedicine, University of Oslo, 0379, Oslo, Norway
- Department of Biosciences, University of Oslo, 0316, Oslo, Norway
| | - Alicia Llorente
- Department of Molecular Cell Biology, The Norwegian Radium Hospital, Institute for Cancer Research, Oslo University Hospital, 0379, Oslo, Norway.
- Centre for Cancer Biomedicine, University of Oslo, 0379, Oslo, Norway.
| |
Collapse
|
113
|
Kanoh N, Mano K, Saigusa D, Usui T, Iwabuchi Y. Design and synthesis of the penta(acetoxymethyl) ester of dioctanoyl phosphatidylinositol-3,5-bisphosphate. Bioorg Med Chem Lett 2016; 26:5770-5772. [PMID: 28029511 DOI: 10.1016/j.bmcl.2016.10.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 10/07/2016] [Accepted: 10/14/2016] [Indexed: 11/24/2022]
Abstract
Extracellular administration of water-soluble and membrane-permeant analogs of phosphatidylinositol phosphates (PIPs) is a useful strategy for understanding the cellular roles of PIPs as well as the mode of action of drugs whose biological activity is associated with PIPs. We herein established the synthetic route to the dioctanoyl analogue of phosphatidylinositol 3,5-bisphosphate (di-C8-PI(3,5)P2) and its penta(acetoxymethyl) ester (di-C8-PI(3,5)P2/5AM).
Collapse
Affiliation(s)
- Naoki Kanoh
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aza-Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Kosuke Mano
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aza-Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Daisuke Saigusa
- Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8573, Japan
| | - Takeo Usui
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8572, Japan
| | - Yoshiharu Iwabuchi
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aza-Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| |
Collapse
|
114
|
Kim SM, Roy SG, Chen B, Nguyen TM, McMonigle RJ, McCracken AN, Zhang Y, Kofuji S, Hou J, Selwan E, Finicle BT, Nguyen TT, Ravi A, Ramirez MU, Wiher T, Guenther GG, Kono M, Sasaki AT, Weisman LS, Potma EO, Tromberg BJ, Edwards RA, Hanessian S, Edinger AL. Targeting cancer metabolism by simultaneously disrupting parallel nutrient access pathways. J Clin Invest 2016; 126:4088-4102. [PMID: 27669461 PMCID: PMC5096903 DOI: 10.1172/jci87148] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 08/16/2016] [Indexed: 12/23/2022] Open
Abstract
Oncogenic mutations drive anabolic metabolism, creating a dependency on nutrient influx through transporters, receptors, and macropinocytosis. While sphingolipids suppress tumor growth by downregulating nutrient transporters, macropinocytosis and autophagy still provide cancer cells with fuel. Therapeutics that simultaneously disrupt these parallel nutrient access pathways have potential as powerful starvation agents. Here, we describe a water-soluble, orally bioavailable synthetic sphingolipid, SH-BC-893, that triggers nutrient transporter internalization and also blocks lysosome-dependent nutrient generation pathways. SH-BC-893 activated protein phosphatase 2A (PP2A), leading to mislocalization of the lipid kinase PIKfyve. The concomitant mislocalization of the PIKfyve product PI(3,5)P2 triggered cytosolic vacuolation and blocked lysosomal fusion reactions essential for LDL, autophagosome, and macropinosome degradation. By simultaneously limiting access to both extracellular and intracellular nutrients, SH-BC-893 selectively killed cells expressing an activated form of the anabolic oncogene Ras in vitro and in vivo. However, slower-growing, autochthonous PTEN-deficient prostate tumors that did not exhibit a classic Warburg phenotype were equally sensitive. Remarkably, normal proliferative tissues were unaffected by doses of SH-BC-893 that profoundly inhibited tumor growth. These studies demonstrate that simultaneously blocking parallel nutrient access pathways with sphingolipid-based drugs is broadly effective and cancer selective, suggesting a potential strategy for overcoming the resistance conferred by tumor heterogeneity.
Collapse
Affiliation(s)
- Seong M. Kim
- Department of Developmental and Cell Biology, University of California Irvine (UCI), Irvine, California, USA
| | - Saurabh G. Roy
- Department of Developmental and Cell Biology, University of California Irvine (UCI), Irvine, California, USA
| | - Bin Chen
- Department of Chemistry, Université de Montréal, Montréal, Québec, Canada
| | - Tiffany M. Nguyen
- Department of Developmental and Cell Biology, University of California Irvine (UCI), Irvine, California, USA
| | - Ryan J. McMonigle
- Department of Developmental and Cell Biology, University of California Irvine (UCI), Irvine, California, USA
| | - Alison N. McCracken
- Department of Developmental and Cell Biology, University of California Irvine (UCI), Irvine, California, USA
| | - Yanling Zhang
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Satoshi Kofuji
- Departments of Internal Medicine, Neurosurgery, and Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jue Hou
- Department of Biomedical Engineering, UCI, Irvine, California, USA
| | - Elizabeth Selwan
- Department of Developmental and Cell Biology, University of California Irvine (UCI), Irvine, California, USA
| | - Brendan T. Finicle
- Department of Developmental and Cell Biology, University of California Irvine (UCI), Irvine, California, USA
| | - Tricia T. Nguyen
- Department of Developmental and Cell Biology, University of California Irvine (UCI), Irvine, California, USA
| | - Archna Ravi
- Department of Developmental and Cell Biology, University of California Irvine (UCI), Irvine, California, USA
| | - Manuel U. Ramirez
- Department of Developmental and Cell Biology, University of California Irvine (UCI), Irvine, California, USA
| | - Tim Wiher
- Department of Developmental and Cell Biology, University of California Irvine (UCI), Irvine, California, USA
| | - Garret G. Guenther
- Department of Developmental and Cell Biology, University of California Irvine (UCI), Irvine, California, USA
| | - Mari Kono
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Bethesda, Maryland, USA
| | - Atsuo T. Sasaki
- Departments of Internal Medicine, Neurosurgery, and Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Lois S. Weisman
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Eric O. Potma
- Department of Biomedical Engineering, UCI, Irvine, California, USA
| | | | - Robert A. Edwards
- Department of Pathology, University of California Irvine School of Medicine, Irvine, California, USA
| | - Stephen Hanessian
- Department of Chemistry, Université de Montréal, Montréal, Québec, Canada
- Department of Pharmaceutical Sciences, UCI, Irvine, California, USA
| | - Aimee L. Edinger
- Department of Developmental and Cell Biology, University of California Irvine (UCI), Irvine, California, USA
| |
Collapse
|
115
|
Phosphatidylinositol 3,5-bisphosphate: regulation of cellular events in space and time. Biochem Soc Trans 2016; 44:177-84. [PMID: 26862203 DOI: 10.1042/bst20150174] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Phosphorylated phosphatidylinositol lipids are crucial for most eukaryotes and have diverse cellular functions. The low-abundance signalling lipid phosphatidylinositol 3,5-bisphosphate [PI(3,5)P2] is critical for cellular homoeostasis and adaptation to stimuli. A large complex of proteins that includes the lipid kinase Fab1-PIKfyve, dynamically regulates the levels of PI(3,5)P2. Deficiencies in PI(3,5)P2 are linked to some human diseases, especially those of the nervous system. Future studies will probably determine new, undiscovered regulatory roles of PI(3,5)P2, as well as uncover mechanistic insights into how PI(3,5)P2 contributes to normal human physiology.
Collapse
|
116
|
Krishna S, Palm W, Lee Y, Yang W, Bandyopadhyay U, Xu H, Florey O, Thompson CB, Overholtzer M. PIKfyve Regulates Vacuole Maturation and Nutrient Recovery following Engulfment. Dev Cell 2016; 38:536-47. [PMID: 27623384 PMCID: PMC5046836 DOI: 10.1016/j.devcel.2016.08.001] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 05/20/2016] [Accepted: 08/08/2016] [Indexed: 12/20/2022]
Abstract
The scavenging of extracellular macromolecules by engulfment can sustain cell growth in a nutrient-depleted environment. Engulfed macromolecules are contained within vacuoles that are targeted for lysosome fusion to initiate degradation and nutrient export. We have shown that vacuoles containing engulfed material undergo mTORC1-dependent fission that redistributes degraded cargo back into the endosomal network. Here we identify the lipid kinase PIKfyve as a regulator of an alternative pathway that distributes engulfed contents in support of intracellular macromolecular synthesis during macropinocytosis, entosis, and phagocytosis. We find that PIKfyve regulates vacuole size in part through its downstream effector, the cationic transporter TRPML1. Furthermore, PIKfyve promotes recovery of nutrients from vacuoles, suggesting a potential link between PIKfyve activity and lysosomal nutrient export. During nutrient depletion, PIKfyve activity protects Ras-mutant cells from starvation-induced cell death and supports their proliferation. These data identify PIKfyve as a critical regulator of vacuole maturation and nutrient recovery during engulfment.
Collapse
Affiliation(s)
- Shefali Krishna
- Gerstner Sloan Kettering Graduate School of Biomedical Sciences, New York, NY 10065, USA; Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Wilhelm Palm
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yongchan Lee
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Wendy Yang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Urmi Bandyopadhyay
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Haoxing Xu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 3089 National Science Building (Kraus), 830 North University, Ann Arbor, MI 48109, USA
| | - Oliver Florey
- Signalling Programme, The Babraham Institute, Cambridge CB22 3AT, UK
| | - Craig B Thompson
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Michael Overholtzer
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
117
|
Targeting TRPM2 in ROS-Coupled Diseases. Pharmaceuticals (Basel) 2016; 9:ph9030057. [PMID: 27618067 PMCID: PMC5039510 DOI: 10.3390/ph9030057] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 08/05/2016] [Accepted: 09/05/2016] [Indexed: 12/15/2022] Open
Abstract
Under pathological conditions such as inflammation and ischemia-reperfusion injury large amounts of reactive oxygen species (ROS) are generated which, in return, contribute to the development and exacerbation of disease. The second member of the transient receptor potential (TRP) melastatin subfamily, TRPM2, is a Ca(2+)-permeable non-selective cation channel, activated by ROS in an ADP-ribose mediated fashion. In other words, TRPM2 functions as a transducer that converts oxidative stress into Ca(2+) signaling. There is good evidence that TRPM2 plays an important role in ROS-coupled diseases. For example, in monocytes the influx of Ca(2+) through TRPM2 activated by ROS contributes to the aggravation of inflammation via chemokine production. In this review, the focus is on TRPM2 as a molecular linker between ROS and Ca(2+) signaling in ROS-coupled diseases.
Collapse
|
118
|
Zondler L, Müller K, Khalaji S, Bliederhäuser C, Ruf WP, Grozdanov V, Thiemann M, Fundel-Clemes K, Freischmidt A, Holzmann K, Strobel B, Weydt P, Witting A, Thal DR, Helferich AM, Hengerer B, Gottschalk KE, Hill O, Kluge M, Ludolph AC, Danzer KM, Weishaupt JH. Peripheral monocytes are functionally altered and invade the CNS in ALS patients. Acta Neuropathol 2016; 132:391-411. [PMID: 26910103 DOI: 10.1007/s00401-016-1548-y] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 02/15/2016] [Accepted: 02/16/2016] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating progressive neurodegenerative disease affecting primarily the upper and lower motor neurons. A common feature of all ALS cases is a well-characterized neuroinflammatory reaction within the central nervous system (CNS). However, much less is known about the role of the peripheral immune system and its interplay with CNS resident immune cells in motor neuron degeneration. Here, we characterized peripheral monocytes in both temporal and spatial dimensions of ALS pathogenesis. We found the circulating monocytes to be deregulated in ALS regarding subtype constitution, function and gene expression. Moreover, we show that CNS infiltration of peripheral monocytes correlates with improved motor neuron survival in a genetic ALS mouse model. Furthermore, application of human immunoglobulins or fusion proteins containing only the human Fc, but not the Fab antibody fragment, increased CNS invasion of peripheral monocytes and delayed the disease onset. Our results underline the importance of peripheral monocytes in ALS pathogenesis and are in agreement with a protective role of monocytes in the early phase of the disease. The possibility to boost this beneficial function of peripheral monocytes by application of human immunoglobulins should be evaluated in clinical trials.
Collapse
Affiliation(s)
- Lisa Zondler
- Department of Neurology, Ulm University, Albert-Einstein Allee 11, O25, Niveau 5, 89081, Ulm, Germany
| | - Kathrin Müller
- Department of Neurology, Ulm University, Albert-Einstein Allee 11, O25, Niveau 5, 89081, Ulm, Germany
| | - Samira Khalaji
- Department of Experimental Physics, Ulm University, Ulm, Germany
| | - Corinna Bliederhäuser
- Department of Neurology, Ulm University, Albert-Einstein Allee 11, O25, Niveau 5, 89081, Ulm, Germany
| | - Wolfgang P Ruf
- Department of Neurology, Ulm University, Albert-Einstein Allee 11, O25, Niveau 5, 89081, Ulm, Germany
| | - Veselin Grozdanov
- Department of Neurology, Ulm University, Albert-Einstein Allee 11, O25, Niveau 5, 89081, Ulm, Germany
| | | | | | - Axel Freischmidt
- Department of Neurology, Ulm University, Albert-Einstein Allee 11, O25, Niveau 5, 89081, Ulm, Germany
| | | | | | - Patrick Weydt
- Department of Neurology, Ulm University, Albert-Einstein Allee 11, O25, Niveau 5, 89081, Ulm, Germany
| | - Anke Witting
- Department of Neurology, Ulm University, Albert-Einstein Allee 11, O25, Niveau 5, 89081, Ulm, Germany
| | - Dietmar R Thal
- Department of Neurology, Ulm University, Albert-Einstein Allee 11, O25, Niveau 5, 89081, Ulm, Germany
| | - Anika M Helferich
- Department of Neurology, Ulm University, Albert-Einstein Allee 11, O25, Niveau 5, 89081, Ulm, Germany
| | | | | | | | | | - Albert C Ludolph
- Department of Neurology, Ulm University, Albert-Einstein Allee 11, O25, Niveau 5, 89081, Ulm, Germany
| | - Karin M Danzer
- Department of Neurology, Ulm University, Albert-Einstein Allee 11, O25, Niveau 5, 89081, Ulm, Germany
| | - Jochen H Weishaupt
- Department of Neurology, Ulm University, Albert-Einstein Allee 11, O25, Niveau 5, 89081, Ulm, Germany.
| |
Collapse
|
119
|
Dolat L, Spiliotis ET. Septins promote macropinosome maturation and traffic to the lysosome by facilitating membrane fusion. J Cell Biol 2016; 214:517-27. [PMID: 27551056 PMCID: PMC5004444 DOI: 10.1083/jcb.201603030] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 07/21/2016] [Indexed: 11/22/2022] Open
Abstract
How macropinosomes traffic to lysosomes is poorly understood. Dolat and Spiliotis show that septins associate preferentially with mature macropinosomes in a PI(3,5)P2-dependent manner and regulate fluid-phase cargo traffic to lysosomes by promoting macropinosome/endosome fusion. Macropinocytosis, the internalization of extracellular fluid and material by plasma membrane ruffles, is critical for antigen presentation, cell metabolism, and signaling. Macropinosomes mature through homotypic and heterotypic fusion with endosomes and ultimately merge with lysosomes. The molecular underpinnings of this clathrin-independent endocytic pathway are largely unknown. Here, we show that the filamentous septin GTPases associate preferentially with maturing macropinosomes in a phosphatidylinositol 3,5-bisphosphate–dependent manner and localize to their contact/fusion sites with macropinosomes/endosomes. Septin knockdown results in large clusters of docked macropinosomes, which persist longer and exhibit fewer fusion events. Septin depletion and overexpression down-regulates and enhances, respectively, the delivery of fluid-phase cargo to lysosomes, without affecting Rab5 and Rab7 recruitment to macropinosomes/endosomes. In vitro reconstitution assays show that fusion of macropinosomes/endosomes is abrogated by septin immunodepletion and function-blocking antibodies and is induced by recombinant septins in the absence of cytosol and polymerized actin. Thus, septins regulate fluid-phase cargo traffic to lysosomes by promoting macropinosome maturation and fusion with endosomes/lysosomes.
Collapse
Affiliation(s)
- Lee Dolat
- Department of Biology, Drexel University, Philadelphia, PA 19104
| | | |
Collapse
|
120
|
Lenk GM, Szymanska K, Debska-Vielhaber G, Rydzanicz M, Walczak A, Bekiesinska-Figatowska M, Vielhaber S, Hallmann K, Stawinski P, Buehring S, Hsu DA, Kunz WS, Meisler MH, Ploski R. Biallelic Mutations of VAC14 in Pediatric-Onset Neurological Disease. Am J Hum Genet 2016; 99:188-94. [PMID: 27292112 DOI: 10.1016/j.ajhg.2016.05.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 05/02/2016] [Indexed: 01/29/2023] Open
Abstract
In the PI(3,5)P2 biosynthetic complex, the lipid kinase PIKFYVE and the phosphatase FIG4 are bound to the dimeric scaffold protein VAC14, which is composed of multiple heat-repeat domains. Mutations of FIG4 result in the inherited disorders Charcot-Marie-Tooth disease type 4J, Yunis-Varón syndrome, and polymicrogyria with seizures. We here describe inherited variants of VAC14 in two unrelated children with sudden onset of a progressive neurological disorder and regression of developmental milestones. Both children developed impaired movement with dystonia, became nonambulatory and nonverbal, and exhibited striatal abnormalities on MRI. A diagnosis of Leigh syndrome was rejected due to normal lactate profiles. Exome sequencing identified biallelic variants of VAC14 that were inherited from unaffected heterozygous parents in both families. Proband 1 inherited a splice-site variant that results in skipping of exon 13, p.Ile459Profs(∗)4 (not reported in public databases), and the missense variant p.Trp424Leu (reported in the ExAC database in a single heterozygote). Proband 2 inherited two missense variants in the dimerization domain of VAC14, p.Ala582Ser and p.Ser583Leu, that have not been previously reported. Cultured skin fibroblasts exhibited the accumulation of vacuoles that is characteristic of PI(3,5)P2 deficiency. Vacuolization of fibroblasts was rescued by transfection of wild-type VAC14 cDNA. The similar age of onset and neurological decline in the two unrelated children define a recessive disorder resulting from compound heterozygosity for deleterious variants of VAC14.
Collapse
Affiliation(s)
- Guy M Lenk
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109-5618, USA
| | - Krystyna Szymanska
- Department of Child Psychiatry, Warsaw Medical University, 02-106 Warsaw, Poland; Department of Experimental and Clinical Neuropathology, Medical Research Center, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | | | - Malgorzata Rydzanicz
- Department of Medical Genetics, Warsaw Medical University, 02-106 Warsaw, Poland
| | - Anna Walczak
- Department of Medical Genetics, Warsaw Medical University, 02-106 Warsaw, Poland
| | | | - Stefan Vielhaber
- Klinik für Neurologie, Universitätsklinikum Magdeburg, 39120 Magdeburg, Germany
| | - Kerstin Hallmann
- Klinik für Epileptologie and Life&Brain Center, Universitätsklinikum Bonn, 53105 Bonn, Germany
| | - Piotr Stawinski
- Department of Medical Genetics, Warsaw Medical University, 02-106 Warsaw, Poland; Institute of Physiology and Pathology of Hearing, 05-830 Nadarzyn, Poland
| | | | - David A Hsu
- Department of Neurology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Wolfram S Kunz
- Klinik für Epileptologie and Life&Brain Center, Universitätsklinikum Bonn, 53105 Bonn, Germany
| | - Miriam H Meisler
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109-5618, USA.
| | - Rafal Ploski
- Department of Medical Genetics, Warsaw Medical University, 02-106 Warsaw, Poland.
| |
Collapse
|
121
|
Hasegawa J, Iwamoto R, Otomo T, Nezu A, Hamasaki M, Yoshimori T. Autophagosome-lysosome fusion in neurons requires INPP5E, a protein associated with Joubert syndrome. EMBO J 2016; 35:1853-67. [PMID: 27340123 DOI: 10.15252/embj.201593148] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 05/27/2016] [Indexed: 12/20/2022] Open
Abstract
Autophagy is a multistep membrane traffic pathway. In contrast to autophagosome formation, the mechanisms underlying autophagosome-lysosome fusion remain largely unknown. Here, we describe a novel autophagy regulator, inositol polyphosphate-5-phosphatase E (INPP5E), involved in autophagosome-lysosome fusion process. In neuronal cells, INPP5E knockdown strongly inhibited autophagy by impairing the fusion step. A fraction of INPP5E is localized to lysosomes, and its membrane anchoring and enzymatic activity are necessary for autophagy. INPP5E decreases lysosomal phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2), one of the substrates of the phosphatase, that counteracts cortactin-mediated actin filament stabilization on lysosomes. Lysosomes require actin filaments on their surface for fusing with autophagosomes. INPP5E is one of the genes responsible for Joubert syndrome, a rare brain abnormality, and mutations found in patients with this disease caused defects in autophagy. Taken together, our data reveal a novel role of phosphoinositide on lysosomes and an association between autophagy and neuronal disease.
Collapse
Affiliation(s)
- Junya Hasegawa
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences Osaka University, Osaka, Japan Department of Genetics, Graduate School of Medicine Osaka University, Osaka, Japan
| | - Ryo Iwamoto
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences Osaka University, Osaka, Japan
| | - Takanobu Otomo
- Department of Genetics, Graduate School of Medicine Osaka University, Osaka, Japan
| | - Akiko Nezu
- Department of Genetics, Graduate School of Medicine Osaka University, Osaka, Japan
| | - Maho Hamasaki
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences Osaka University, Osaka, Japan Department of Genetics, Graduate School of Medicine Osaka University, Osaka, Japan
| | - Tamotsu Yoshimori
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences Osaka University, Osaka, Japan Department of Genetics, Graduate School of Medicine Osaka University, Osaka, Japan
| |
Collapse
|
122
|
Compton LM, Ikonomov OC, Sbrissa D, Garg P, Shisheva A. Active vacuolar H+ ATPase and functional cycle of Rab5 are required for the vacuolation defect triggered by PtdIns(3,5)P2 loss under PIKfyve or Vps34 deficiency. Am J Physiol Cell Physiol 2016; 311:C366-77. [PMID: 27335171 DOI: 10.1152/ajpcell.00104.2016] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/17/2016] [Indexed: 01/12/2023]
Abstract
The two evolutionarily conserved mammalian lipid kinases Vps34 and PIKfyve are involved in an important physiological relationship, whereby the former produces phosphatidylinositol (PtdIns) 3P that is used as a substrate for PtdIns(3,5)P2 synthesis by the latter. Reduced production of PtdIns(3,5)P2 in proliferating mammalian cells is phenotypically manifested by the formation of multiple translucent cytoplasmic vacuoles, readily rescued upon exogenous delivery of PtdIns(3,5)P2 or overproduction of PIKfyve. Although the aberrant vacuolation phenomenon has been frequently used as a sensitive functional measure of localized PtdIns(3,5)P2 reduction, cellular factors governing the appearance of cytoplasmic vacuoles under PtdIns3P-PtdIns(3,5)P2 loss remain elusive. To gain further mechanistic insight about the vacuolation process following PtdIns(3,5)P2 reduction, in this study we sought for cellular mechanisms required for manifestation of the aberrant endomembrane vacuoles triggered by PIKfyve or Vps34 dysfunction. The latter was achieved by various means such as pharmacological inhibition, gene disruption, or dominant-interference in several proliferating mammalian cell types. We report here that inhibition of V-ATPase with bafilomycin A1 as well as inactivation of the GTP-GDP cycle of Rab5a GTPase phenotypically rescued or completely precluded the cytoplasmic vacuolization despite the continued presence of inactivated PIKfyve or Vps34. Bafilomycin A1 also restored the aberrant EEA1-positive endosomes, enlarged upon short PIKfyve inhibition with YM201636. Together, our work identifies for the first time that factors such as active V-ATPase or functional Rab5a cycle are acting coincidentally with the PtdIns(3,5)P2 reduction in triggering formation of aberrant cytoplasmic vacuoles under PIKfyve or Vps34 dysfunction.
Collapse
Affiliation(s)
- Lauren M Compton
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan; and
| | - Ognian C Ikonomov
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan; and
| | - Diego Sbrissa
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan; and
| | - Puneet Garg
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Assia Shisheva
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan; and
| |
Collapse
|
123
|
Inhibition of c-Rel DNA binding is critical for the anti-inflammatory effects of novel PIKfyve inhibitor. Eur J Pharmacol 2016; 780:93-105. [DOI: 10.1016/j.ejphar.2016.03.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/14/2016] [Accepted: 03/18/2016] [Indexed: 01/15/2023]
|
124
|
Coxiella burnetii effector CvpB modulates phosphoinositide metabolism for optimal vacuole development. Proc Natl Acad Sci U S A 2016; 113:E3260-9. [PMID: 27226300 DOI: 10.1073/pnas.1522811113] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The Q fever bacterium Coxiella burnetii replicates inside host cells within a large Coxiella-containing vacuole (CCV) whose biogenesis relies on the Dot/Icm-dependent secretion of bacterial effectors. Several membrane trafficking pathways contribute membranes, proteins, and lipids for CCV biogenesis. These include the endocytic and autophagy pathways, which are characterized by phosphatidylinositol 3-phosphate [PI(3)P]-positive membranes. Here we show that the C. burnetii secreted effector Coxiella vacuolar protein B (CvpB) binds PI(3)P and phosphatidylserine (PS) on CCVs and early endosomal compartments and perturbs the activity of the phosphatidylinositol 5-kinase PIKfyve to manipulate PI(3)P metabolism. CvpB association to early endosome triggers vacuolation and clustering, leading to the channeling of large PI(3)P-positive membranes to CCVs for vacuole expansion. At CCVs, CvpB binding to early endosome- and autophagy-derived PI(3)P and the concomitant inhibition of PIKfyve favor the association of the autophagosomal machinery to CCVs for optimal homotypic fusion of the Coxiella-containing compartments. The importance of manipulating PI(3)P metabolism is highlighted by mutations in cvpB resulting in a multivacuolar phenotype, rescuable by gene complementation, indicative of a defect in CCV biogenesis. Using the insect model Galleria mellonella, we demonstrate the in vivo relevance of defective CCV biogenesis by highlighting an attenuated virulence phenotype associated with cvpB mutations.
Collapse
|
125
|
Garrity AG, Wang W, Collier CM, Levey SA, Gao Q, Xu H. The endoplasmic reticulum, not the pH gradient, drives calcium refilling of lysosomes. eLife 2016; 5. [PMID: 27213518 PMCID: PMC4909396 DOI: 10.7554/elife.15887] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/20/2016] [Indexed: 12/15/2022] Open
Abstract
Impaired homeostasis of lysosomal Ca2+ causes lysosome dysfunction and lysosomal storage diseases (LSDs), but the mechanisms by which lysosomes acquire and refill Ca2+ are not known. We developed a physiological assay to monitor lysosomal Ca2+ store refilling using specific activators of lysosomal Ca2+ channels to repeatedly induce lysosomal Ca2+ release. In contrast to the prevailing view that lysosomal acidification drives Ca2+ into the lysosome, inhibiting the V-ATPase H+ pump did not prevent Ca2+ refilling. Instead, pharmacological depletion or chelation of Endoplasmic Reticulum (ER) Ca2+ prevented lysosomal Ca2+ stores from refilling. More specifically, antagonists of ER IP3 receptors (IP3Rs) rapidly and completely blocked Ca2+ refilling of lysosomes, but not in cells lacking IP3Rs. Furthermore, reducing ER Ca2+ or blocking IP3Rs caused a dramatic LSD-like lysosome storage phenotype. By closely apposing each other, the ER may serve as a direct and primary source of Ca2+for the lysosome. DOI:http://dx.doi.org/10.7554/eLife.15887.001
Collapse
Affiliation(s)
- Abigail G Garrity
- Neuroscience Program, University of Michigan, Ann Arbor, United States
| | - Wuyang Wang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Crystal Md Collier
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Sara A Levey
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Qiong Gao
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Haoxing Xu
- Neuroscience Program, University of Michigan, Ann Arbor, United States.,Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, United States
| |
Collapse
|
126
|
Vacuolin-1 inhibits autophagy by impairing lysosomal maturation via PIKfyve inhibition. FEBS Lett 2016; 590:1576-85. [DOI: 10.1002/1873-3468.12195] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/07/2016] [Accepted: 04/14/2016] [Indexed: 12/29/2022]
|
127
|
A cell-permeable tool for analysing APP intracellular domain function and manipulation of PIKfyve activity. Biosci Rep 2016; 36:BSR20160040. [PMID: 26934981 PMCID: PMC5293579 DOI: 10.1042/bsr20160040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 03/02/2016] [Indexed: 12/24/2022] Open
Abstract
The mechanisms for regulating PIKfyve complex activity are currently emerging. The PIKfyve complex, consisting of the phosphoinositide kinase PIKfyve (also known as FAB1), VAC14 and FIG4, is required for the production of phosphatidylinositol 3,5-bisphosphate [PI(3,5)P2]. PIKfyve function is required for homoeostasis of the endo/lysosomal system and is crucially implicated in neuronal function and integrity, as loss of function mutations in the PIKfyve complex lead to neurodegeneration in mouse models and human patients. Our recent work has shown that the intracellular domain of the amyloid precursor protein (APP), a molecule central to the aetiology of Alzheimer's disease binds to VAC14 and enhances PIKfyve function. In the present study, we utilize this recent advance to create an easy-to-use tool for increasing PIKfyve activity in cells. We fused APP intracellular domain (AICD) to the HIV TAT domain, a cell-permeable peptide allowing proteins to penetrate cells. The resultant TAT-AICD fusion protein is cell permeable and triggers an increase in PI(3,5)P2 Using the PI(3,5)P2 specific GFP-ML1Nx2 probe, we show that cell-permeable AICD alters PI(3,5)P2 dynamics. TAT-AICD also provides partial protection from pharmacological inhibition of PIKfyve. All three lines of evidence show that the AICD activates the PIKfyve complex in cells, a finding that is important for our understanding of the mechanism of neurodegeneration in Alzheimer's disease.
Collapse
|
128
|
A molecular mechanism to regulate lysosome motility for lysosome positioning and tubulation. Nat Cell Biol 2016; 18:404-17. [PMID: 26950892 PMCID: PMC4871318 DOI: 10.1038/ncb3324] [Citation(s) in RCA: 299] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 02/04/2016] [Indexed: 12/12/2022]
Abstract
To mediate the degradation of bio-macromolecules, lysosomes must traffic towards cargo-carrying vesicles for subsequent membrane fusion or fission. Mutations of the lysosomal Ca2+ channel TRPML1 cause lysosome storage disease (LSD) characterized by disordered lysosomal membrane trafficking in cells. Here we show that TRPML1 activity is required to promote Ca2+-dependent centripetal movement of lysosomes towards the perinuclear region, where autophagosomes accumulate, upon autophagy induction. ALG-2, an EF-hand-containing protein, serves as a lysosomal Ca2+ sensor that associates physically with the minus-end directed dynactin-dynein motor, while PI(3,5)P2, a lysosome-localized phosphoinositide, acts upstream of TRPML1. Furthermore, the PI(3,5)P2-TRPML1-ALG-2-dynein signaling is necessary for lysosome tubulation and reformation. In contrast, the TRPML1 pathway is not required for the perinuclear accumulation of lysosomes observed in many LSDs, which is instead likely caused by secondary cholesterol accumulation that constitutively activates Rab7-RILP-dependent retrograde transport. Collectively, Ca2+ release from lysosomes provides an on-demand mechanism regulating lysosome motility, positioning, and tubulation.
Collapse
|
129
|
Matsuzawa K, Akita H, Watanabe T, Kakeno M, Matsui T, Wang S, Kaibuchi K. PAR3-aPKC regulates Tiam1 by modulating suppressive internal interactions. Mol Biol Cell 2016; 27:1511-23. [PMID: 26941335 PMCID: PMC4850038 DOI: 10.1091/mbc.e15-09-0670] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 02/24/2016] [Indexed: 01/19/2023] Open
Abstract
The Rac1 activator Tiam1 is inhibited by internal interactions. Phosphorylation of Tiam1 by aPKC lends bias to an “open” conformation, potentiating its activation and allowing for its localization through protein–protein interactions. Tiam1 is one of the most extensively analyzed activators of the small GTPase Rac. However, fundamental aspects of its regulation are poorly understood. Here we demonstrate that Tiam1 is functionally suppressed by internal interactions and that the PAR complex participates in its full activation. The N-terminal region of Tiam1 binds to the protein-binding and catalytic domains to inhibit its localization and activation. Atypical PKCs phosphorylate Tiam1 to relieve its intramolecular interactions, and the subsequent stabilization of its interaction with PAR3 allows it to exert localized activity. By analyzing Tiam1 regulation by PAR3-aPKC within the context of PDGF signaling, we also show that PAR3 directly binds PDGF receptor β. Thus we provide the first evidence for the negative regulation of Tiam1 by internal interactions, elucidate the nature of Tiam1 regulation by the PAR complex, and reveal a novel role for the PAR complex in PDGF signaling.
Collapse
Affiliation(s)
- Kenji Matsuzawa
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Hiroki Akita
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Takashi Watanabe
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Mai Kakeno
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Toshinori Matsui
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Shujie Wang
- Department of Neural Regeneration and Cell Communication, Graduate School of Medicine, Mie University, Tsu 514-8507, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
130
|
Proikas-Cezanne T, Takacs Z, Dönnes P, Kohlbacher O. WIPI proteins: essential PtdIns3P effectors at the nascent autophagosome. J Cell Sci 2016; 128:207-17. [PMID: 25568150 DOI: 10.1242/jcs.146258] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Autophagy is a pivotal cytoprotective process that secures cellular homeostasis, fulfills essential roles in development, immunity and defence against pathogens, and determines the lifespan of eukaryotic organisms. However, autophagy also crucially contributes to the development of age-related human pathologies, including cancer and neurodegeneration. Macroautophagy (hereafter referred to as autophagy) clears the cytoplasm by stochastic or specific cargo recognition and destruction, and is initiated and executed by autophagy related (ATG) proteins functioning in dynamical hierarchies to form autophagosomes. Autophagosomes sequester cytoplasmic cargo material, including proteins, lipids and organelles, and acquire acidic hydrolases from the lysosomal compartment for cargo degradation. Prerequisite and essential for autophagosome formation is the production of phosphatidylinositol 3-phosphate (PtdIns3P) by phosphatidylinositol 3-kinase class III (PI3KC3, also known as PIK3C3) in complex with beclin 1, p150 (also known as PIK3R4; Vps15 in yeast) and ATG14L. Members of the human WD-repeat protein interacting with phosphoinositides (WIPI) family play an important role in recognizing and decoding the PtdIns3P signal at the nascent autophagosome, and hence function as autophagy-specific PtdIns3P-binding effectors, similar to their ancestral yeast Atg18 homolog. The PtdIns3P effector function of human WIPI proteins appears to be compromised in cancer and neurodegeneration, and WIPI genes and proteins might present novel targets for rational therapies. Here, we summarize the current knowledge on the roles of the four human WIPI proteins, WIPI1-4, in autophagy. This article is part of a Focus on Autophagosome biogenesis. For further reading, please see related articles: 'ERES: sites for autophagosome biogenesis and maturation?' by Jana Sanchez-Wandelmer et al. (J. Cell Sci. 128, 185-192) and 'Membrane dynamics in autophagosome biogenesis' by Sven R. Carlsson and Anne Simonsen (J. Cell Sci. 128, 193-205).
Collapse
Affiliation(s)
- Tassula Proikas-Cezanne
- Autophagy Laboratory, Department of Molecular Biology, Interfaculty Institute of Cell Biology, Eberhard Karls University Tuebingen, Auf der Morgenstelle 15, 72076 Tuebingen, Germany International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Developmental Biology and Eberhard Karls University Tuebingen, Spemannstr. 35 - 39, 72076 Tuebingen, Germany
| | - Zsuzsanna Takacs
- Autophagy Laboratory, Department of Molecular Biology, Interfaculty Institute of Cell Biology, Eberhard Karls University Tuebingen, Auf der Morgenstelle 15, 72076 Tuebingen, Germany International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Developmental Biology and Eberhard Karls University Tuebingen, Spemannstr. 35 - 39, 72076 Tuebingen, Germany
| | - Pierre Dönnes
- Applied Bioinformatics Group, Center for Bioinformatics, Quantitative Biology Center, and Department of Computer Science, Eberhard Karls University Tuebingen, Sand 14, 72076 Tuebingen, Germany
| | - Oliver Kohlbacher
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Developmental Biology and Eberhard Karls University Tuebingen, Spemannstr. 35 - 39, 72076 Tuebingen, Germany Applied Bioinformatics Group, Center for Bioinformatics, Quantitative Biology Center, and Department of Computer Science, Eberhard Karls University Tuebingen, Sand 14, 72076 Tuebingen, Germany
| |
Collapse
|
131
|
Abstract
TRPML1 is a ubiquitously expressed cation channel found on lysosomes and late endosomes. Mutations in TRPML1 cause mucolipidosis type IV and it has been implicated in Alzheimer's disease and HIV. However, the mechanisms by which TRPML1 activity is regulated are not well understood. This review summarizes the current understanding of TRPML1 activation and regulation.
Collapse
|
132
|
Vergés M. Retromer in Polarized Protein Transport. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 323:129-79. [PMID: 26944621 DOI: 10.1016/bs.ircmb.2015.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Retromer is an evolutionary conserved protein complex required for endosome-to-Golgi retrieval of receptors for lysosomal hydrolases. It is constituted by a heterotrimer encoded by the vacuolar protein sorting (VPS) gene products Vps26, Vps35, and Vps29, which selects cargo, and a dimer of phosphoinositide-binding sorting nexins, which deforms the membrane. Recent progress in the mechanism of retromer assembly and functioning has strengthened the link between sorting at the endosome and cytoskeleton dynamics. Retromer is implicated in endosomal sorting of many cargos and plays an essential role in plant and animal development. Although it is best known for endosome sorting to the trans-Golgi network, it also intervenes in recycling to the plasma membrane. In polarized cells, such as epithelial cells and neurons, retromer may also be utilized for transcytosis and long-range transport. Considerable evidence implicates retromer in establishment and maintenance of cell polarity. That includes sorting of the apical polarity module Crumbs; regulation of retromer function by the basolateral polarity module Scribble; and retromer-dependent recycling of various cargoes to a certain surface domain, thus controlling polarized location and cell homeostasis. Importantly, altered retromer function has been linked to neurodegeneration, such as in Alzheimer's or Parkinson's disease. This review will underline how alterations in retromer localization and function may affect polarized protein transport and polarity establishment, thereby causing developmental defects and disease.
Collapse
Affiliation(s)
- Marcel Vergés
- Cardiovascular Genetics Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain; Medical Sciences Department, University of Girona, Girona, Spain.
| |
Collapse
|
133
|
Nishiyama Y, Ohmichi T, Kazami S, Iwasaki H, Mano K, Nagumo Y, Kudo F, Ichikawa S, Iwabuchi Y, Kanoh N, Eguchi T, Osada H, Usui T. Vicenistatin induces early endosome-derived vacuole formation in mammalian cells. Biosci Biotechnol Biochem 2016; 80:902-10. [PMID: 27104762 DOI: 10.1080/09168451.2015.1132152] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Homotypic fusion of early endosomes is important for efficient protein trafficking and sorting. The key controller of this process is Rab5 which regulates several effectors and PtdInsPs levels, but whose mechanisms are largely unknown. Here, we report that vicenistatin, a natural product, enhanced homotypic fusion of early endosomes and induced the formation of large vacuole-like structures in mammalian cells. Unlike YM201636, another early endosome vacuolating compound, vicenistatin did not inhibit PIKfyve activity in vitro but activated Rab5-PAS pathway in cells. Furthermore, vicenistatin increased the membrane surface fluidity of cholesterol-containing liposomes in vitro, and cholesterol deprivation from the plasma membrane stimulated vicenistatin-induced vacuolation in cells. These results suggest that vicenistatin is a novel compound that induces the formation of vacuole-like structures by activating Rab5-PAS pathway and increasing membrane fluidity.
Collapse
Affiliation(s)
- Yuko Nishiyama
- a Graduate School of Life and Environmental Sciences , University of Tsukuba , Tsukuba , Japan
| | - Tomohiro Ohmichi
- a Graduate School of Life and Environmental Sciences , University of Tsukuba , Tsukuba , Japan
| | - Sayaka Kazami
- b Chemical Biology Research Group, RIKEN CSRS , Wako, Japan
| | - Hiroki Iwasaki
- a Graduate School of Life and Environmental Sciences , University of Tsukuba , Tsukuba , Japan
| | - Kousuke Mano
- c Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai , Japan
| | - Yoko Nagumo
- a Graduate School of Life and Environmental Sciences , University of Tsukuba , Tsukuba , Japan
| | - Fumitaka Kudo
- d Graduate School of Science and Engineering , Tokyo Institute of Technology , Tokyo , Japan
| | - Sosaku Ichikawa
- e Faculty of Life and Environmental Sciences , University of Tsukuba , Tsukuba , Japan
| | - Yoshiharu Iwabuchi
- c Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai , Japan
| | - Naoki Kanoh
- c Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai , Japan
| | - Tadashi Eguchi
- d Graduate School of Science and Engineering , Tokyo Institute of Technology , Tokyo , Japan
| | - Hiroyuki Osada
- b Chemical Biology Research Group, RIKEN CSRS , Wako, Japan
| | - Takeo Usui
- e Faculty of Life and Environmental Sciences , University of Tsukuba , Tsukuba , Japan
| |
Collapse
|
134
|
Currinn H, Guscott B, Balklava Z, Rothnie A, Wassmer T. APP controls the formation of PI(3,5)P(2) vesicles through its binding of the PIKfyve complex. Cell Mol Life Sci 2016; 73:393-408. [PMID: 26216398 PMCID: PMC4706845 DOI: 10.1007/s00018-015-1993-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 06/26/2015] [Accepted: 07/16/2015] [Indexed: 12/05/2022]
Abstract
Phosphoinositides are signalling lipids that are crucial for major signalling events as well as established regulators of membrane trafficking. Control of endosomal sorting and endosomal homeostasis requires phosphatidylinositol-3-phosphate (PI(3)P) and phosphatidylinositol-3,5-bisphosphate (PI(3,5)P2), the latter a lipid of low abundance but significant physiological relevance. PI(3,5)P2 is formed by phosphorylation of PI(3)P by the PIKfyve complex which is crucial for maintaining endosomal homeostasis. Interestingly, loss of PIKfyve function results in dramatic neurodegeneration. Despite the significance of PIKfyve, its regulation is still poorly understood. Here we show that the Amyloid Precursor Protein (APP), a central molecule in Alzheimer's disease, associates with the PIKfyve complex (consisting of Vac14, PIKfyve and Fig4) and that the APP intracellular domain directly binds purified Vac14. We also show that the closely related APP paralogues, APLP1 and 2 associate with the PIKfyve complex. Whether APP family proteins can additionally form direct protein-protein interaction with PIKfyve or Fig4 remains to be explored. We show that APP binding to the PIKfyve complex drives formation of PI(3,5)P2 positive vesicles and that APP gene family members are required for supporting PIKfyve function. Interestingly, the PIKfyve complex is required for APP trafficking, suggesting a feedback loop in which APP, by binding to and stimulating PI(3,5)P2 vesicle formation may control its own trafficking. These data suggest that altered APP processing, as observed in Alzheimer's disease, may disrupt PI(3,5)P2 metabolism, endosomal sorting and homeostasis with important implications for our understanding of the mechanism of neurodegeneration in Alzheimer's disease.
Collapse
Affiliation(s)
- Heather Currinn
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham, B4 7ET, UK
| | - Benjamin Guscott
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham, B4 7ET, UK
| | - Zita Balklava
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham, B4 7ET, UK
| | - Alice Rothnie
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham, B4 7ET, UK
| | - Thomas Wassmer
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham, B4 7ET, UK.
| |
Collapse
|
135
|
Hong NH, Qi A, Weaver AM. PI(3,5)P2 controls endosomal branched actin dynamics by regulating cortactin-actin interactions. J Cell Biol 2015; 210:753-69. [PMID: 26323691 PMCID: PMC4555817 DOI: 10.1083/jcb.201412127] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The late endosomal lipid PI(3,5)P2 binds to cortactin through the filamentous actin (F-actin) binding domain of cortactin, leading to removal of cortactin from endosomal actin networks and inhibition of cortactin-mediated branched actin nucleation and stabilization. Branched actin critically contributes to membrane trafficking by regulating membrane curvature, dynamics, fission, and transport. However, how actin dynamics are controlled at membranes is poorly understood. Here, we identify the branched actin regulator cortactin as a direct binding partner of phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2) and demonstrate that their interaction promotes turnover of late endosomal actin. In vitro biochemical studies indicated that cortactin binds PI(3,5)P2 via its actin filament-binding region. Furthermore, PI(3,5)P2 competed with actin filaments for binding to cortactin, thereby antagonizing cortactin activity. These findings suggest that PI(3,5)P2 formation on endosomes may remove cortactin from endosome-associated branched actin. Indeed, inhibition of PI(3,5)P2 production led to cortactin accumulation and actin stabilization on Rab7+ endosomes. Conversely, inhibition of Arp2/3 complex activity greatly reduced cortactin localization to late endosomes. Knockdown of cortactin reversed PI(3,5)P2-inhibitor–induced actin accumulation and stabilization on endosomes. These data suggest a model in which PI(3,5)P2 binding removes cortactin from late endosomal branched actin networks and thereby promotes net actin turnover.
Collapse
Affiliation(s)
- Nan Hyung Hong
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Aidong Qi
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Alissa M Weaver
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232 Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232 Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| |
Collapse
|
136
|
Abstract
Existing analgesics are not efficacious in treating all patients with chronic pain and have harmful side effects when used long term. A deeper understanding of pain signaling and sensitization could lead to the development of more efficacious analgesics. Nociceptor sensitization occurs under conditions of inflammation and nerve injury where diverse chemicals are released and signal through receptors to reduce the activation threshold of ion channels, leading to an overall increase in neuronal excitability. Drugs that inhibit specific receptors have so far been unsuccessful in alleviating pain, possibly because they do not simultaneously target the diverse receptors that contribute to nociceptor sensitization. Hence, the focus has shifted toward targeting downstream convergence points of nociceptive signaling. Lipid mediators, including phosphatidylinositol 4,5-bisphosphate (PIP2), are attractive targets, as these molecules are required for signaling downstream of G-protein-coupled receptors and receptor tyrosine kinases. Furthermore, PIP2 regulates the activity of various ion channels. Thus, PIP2 sits at a critical convergence point for multiple receptors, ion channels, and signaling pathways that promote and maintain chronic pain. Decreasing the amount of PIP2 in neurons was recently shown to attenuate pronociceptive signaling and could provide a novel approach for treating pain. Here, we review the lipid kinases that are known to regulate pain signaling and sensitization and speculate on which additional lipid kinases might regulate signaling in nociceptive neurons.
Collapse
|
137
|
Hirano T, Munnik T, Sato MH. Phosphatidylinositol 3-Phosphate 5-Kinase, FAB1/PIKfyve Kinase Mediates Endosome Maturation to Establish Endosome-Cortical Microtubule Interaction in Arabidopsis. PLANT PHYSIOLOGY 2015; 169:1961-74. [PMID: 26353760 PMCID: PMC4634102 DOI: 10.1104/pp.15.01368] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 09/09/2015] [Indexed: 05/19/2023]
Abstract
Phosphatidylinositol 3,5-bisphosphate [PtdIns(3,5)P2] is an important lipid in membrane trafficking in animal and yeast systems; however, its role is still largely obscure in plants. Here, we demonstrate that the phosphatidylinositol 3-phosphate 5-kinase, formation of aploid and binucleate cells1 (FAB1)/FYVE finger-containing phosphoinositide kinase (PIKfyve), and its product, PtdIns(3,5)P2, are essential for the maturation process of endosomes to mediate cortical microtubule association of endosomes, thereby controlling proper PIN-FORMED protein trafficking in young cortical and stele cells of root. We found that FAB1 predominantly localizes on the Sorting Nexin1 (SNX1)-residing late endosomes, and a loss of FAB1 function causes the release of late endosomal proteins, Ara7, and SNX1 from the endosome membrane, indicating that FAB1, or its product PtdIns(3,5)P2, mediates the maturation process of the late endosomes. We also found that loss of FAB1 function causes the release of endosomes from cortical microtubules and disturbs proper cortical microtubule organization.
Collapse
Affiliation(s)
- Tomoko Hirano
- Laboratory of Cellular Dynamics, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan (T.H., M.H.S.); andSection Plant Physiology, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands (T.M.)
| | - Teun Munnik
- Laboratory of Cellular Dynamics, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan (T.H., M.H.S.); andSection Plant Physiology, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands (T.M.)
| | - Masa H Sato
- Laboratory of Cellular Dynamics, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan (T.H., M.H.S.); andSection Plant Physiology, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands (T.M.)
| |
Collapse
|
138
|
The ML1Nx2 Phosphatidylinositol 3,5-Bisphosphate Probe Shows Poor Selectivity in Cells. PLoS One 2015; 10:e0139957. [PMID: 26460749 PMCID: PMC4604148 DOI: 10.1371/journal.pone.0139957] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 09/17/2015] [Indexed: 01/15/2023] Open
Abstract
Phosphatidylinositol (3,5)-bisphosphate (PtdIns(3,5)P2) is a quantitatively minor phospholipid in eukaryotic cells that plays a fundamental role in regulating endocytic membrane traffic. Despite its clear importance for cellular function and organism physiology, mechanistic details of its biology have so far not been fully elucidated. In part, this is due to a lack of experimental tools that specifically probe for PtdIns(3,5)P2 in cells to unambiguously identify its dynamics and site(s) of action. In this study, we have evaluated a recently reported PtdIns(3,5)P2 biosensor, GFP-ML1Nx2, for its veracity as such a probe. We report that, in live cells, the localization of this biosensor to sub-cellular compartments is largely independent of PtdIns(3,5)P2, as assessed after pharmacological, chemical genetic or genomic interventions that block the lipid's synthesis. We therefore conclude that it is unwise to interpret the localization of ML1Nx2 as a true and unbiased biosensor for PtdIns(3,5)P2.
Collapse
|
139
|
Messenger SW, Thomas DD, Cooley MM, Jones EK, Falkowski MA, August BK, Fernandez LA, Gorelick FS, Groblewski GE. Early to Late Endosome Trafficking Controls Secretion and Zymogen Activation in Rodent and Human Pancreatic Acinar Cells. Cell Mol Gastroenterol Hepatol 2015; 1:695-709. [PMID: 26618189 PMCID: PMC4657148 DOI: 10.1016/j.jcmgh.2015.08.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND & AIMS Pancreatic acinar cells have an expanded apical endosomal system, the physiological and pathophysiological significance of which is still emerging. Phosphatidylinositol-3,5-bisphosphate (PI(3,5)P2) is an essential phospholipid generated by PIKfyve, which phosphorylates phosphatidylinositol-3-phosphate (PI(3)P). PI(3,5)P2 is necessary for maturation of early endosomes (EE) to late endosomes (LE). Inhibition of EE to LE trafficking enhances anterograde endosomal trafficking and secretion at the plasma membrane by default through a recycling endosome (RE) intermediate. We assessed the effects of modulating PIKfyve activity on apical trafficking and pancreatitis responses in pancreatic acinar cells. METHODS Inhibition of EE to LE trafficking was achieved using pharmacological inhibitors of PIKfyve, expression of dominant negative PIKfyve K1877E, or constitutively active Rab5-GTP Q79L. Anterograde endosomal trafficking was manipulated by expression of constitutively active and dominant negative Rab11a mutants. The effects of these agents on secretion, endolysosomal exocytosis of lysosome associated membrane protein (LAMP1), and trypsinogen activation in response to high-dose CCK-8, bile acids and cigarette toxin was determined. RESULTS PIKfyve inhibition increased basal and stimulated secretion. Adenoviral overexpression of PIKfyve decreased secretion leading to cellular death. Expression of Rab5-GTP Q79L or Rab11a-GTP Q70L enhanced secretion. Conversely, dominant-negative Rab11a-GDP S25N reduced secretion. High-dose CCK inhibited endolysosomal exocytosis that was reversed by PIKfyve inhibition. PIKfyve inhibition blocked intracellular trypsin accumulation and cellular damage responses to high CCK-8, tobacco toxin, and bile salts in both rodent and human acini. CONCLUSIONS These data demonstrate that EE-LE trafficking acutely controls acinar secretion and the intracellular activation of zymogens leading to the pathogenicity of acute pancreatitis.
Collapse
Affiliation(s)
- Scott W. Messenger
- Department of Nutritional Sciences, University of Wisconsin, Madison, Wisconsin
| | - Diana D.H. Thomas
- Department of Nutritional Sciences, University of Wisconsin, Madison, Wisconsin
| | - Michelle M. Cooley
- Department of Nutritional Sciences, University of Wisconsin, Madison, Wisconsin
| | - Elaina K. Jones
- Department of Nutritional Sciences, University of Wisconsin, Madison, Wisconsin
| | | | - Benjamin K. August
- Department of Nutritional Sciences, University of Wisconsin, Madison, Wisconsin
| | | | - Fred S. Gorelick
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut,Department of Cell Biology, School of Medicine, Yale University, New Haven, Connecticut,Veterans Administration Connecticut Healthcare, West Haven, Connecticut
| | - Guy E. Groblewski
- Department of Nutritional Sciences, University of Wisconsin, Madison, Wisconsin,Correspondence Address correspondence to: Guy E. Groblewski, PhD, University of Wisconsin–Madison, Department of Nutritional Sciences, 1415 Linden Drive, Madison, Wisconsin 53706. fax: (608) 262-5860.University of Wisconsin–MadisonDepartment of Nutritional Sciences1415 Linden DriveMadisonWisconsin 53706
| |
Collapse
|
140
|
Munson MJ, Allen GF, Toth R, Campbell DG, Lucocq JM, Ganley IG. mTOR activates the VPS34-UVRAG complex to regulate autolysosomal tubulation and cell survival. EMBO J 2015; 34:2272-90. [PMID: 26139536 PMCID: PMC4585463 DOI: 10.15252/embj.201590992] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 05/16/2015] [Accepted: 06/04/2015] [Indexed: 02/01/2023] Open
Abstract
Lysosomes are essential organelles that function to degrade and recycle unwanted, damaged and toxic biological components. Lysosomes also act as signalling platforms in activating the nutrient-sensing kinase mTOR. mTOR regulates cellular growth, but it also helps to maintain lysosome identity by initiating lysosomal tubulation through a process termed autophagosome-lysosome reformation (ALR). Here we identify a lysosomal pool of phosphatidylinositol 3-phosphate that, when depleted by specific inhibition of the class III phosphoinositide 3-kinase VPS34, results in prolonged lysosomal tubulation. This tubulation requires mTOR activity, and we identified two direct mTOR phosphorylation sites on UVRAG (S550 and S571) that activate VPS34. Loss of these phosphorylation sites reduced VPS34 lipid kinase activity and resulted in an increase in number and length of lysosomal tubules. In cells in which phosphorylation at these UVRAG sites is disrupted, the result of impaired lysosomal tubulation alongside ALR activation is massive cell death. Our data imply that ALR is critical for cell survival under nutrient stress and that VPS34 is an essential regulatory element in this process.
Collapse
Affiliation(s)
- Michael J Munson
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences University of Dundee, Dundee, UK
| | - George Fg Allen
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences University of Dundee, Dundee, UK
| | - Rachel Toth
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences University of Dundee, Dundee, UK
| | - David G Campbell
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences University of Dundee, Dundee, UK
| | - John M Lucocq
- School of Medicine University of St Andrews, St Andrews, UK
| | - Ian G Ganley
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences University of Dundee, Dundee, UK
| |
Collapse
|
141
|
Hayashi K, Sasai M, Iwasaki A. Toll-like receptor 9 trafficking and signaling for type I interferons requires PIKfyve activity. Int Immunol 2015; 27:435-45. [PMID: 25925170 PMCID: PMC4560039 DOI: 10.1093/intimm/dxv021] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 04/17/2015] [Indexed: 12/17/2022] Open
Abstract
Toll-like receptors (TLRs) traffic to distinct membranes for signaling. TLR7 and TLR9 recognize viral nucleic acids in the endosomes and induce robust anti-viral program. Signaling from these TLRs bifurcate at the level of distinct endosomal compartments, namely VAMP3(+) and LAMP(+) endosomes, to mediate the induction of cytokine and type I interferon (IFN) genes, respectively. The formation of the TLR9 endosome competent for IFNs induction requires AP-3. Phosphoinositides (PIs) mark distinct subcellular membranes and control membrane trafficking. However, their role in TLR trafficking and signaling in different dendritic cell (DC) subsets remains unclear. Here, we examined the role of phosphatidylinositol 3P 5-kinase, PIKfyve, in TLR9 trafficking and signaling. We demonstrate that inhibition of PIKfyve activity preferentially blocks TLR9 signaling for type I IFN induction in FLT3L-bone marrow-derived DCs. By confocal microscopy using RAW264.7 cells, we show that trafficking of both TLR9 and CpG to the LAMP1(+) compartment was blocked by PIKfyve inhibitor treatment, whereas their trafficking to the VAMP3(+) endosome remained intact. Further, AP-3 recruitment to TLR9 endosomes was impaired by PIKfyve inhibition. These data indicate that PIKfyve provides critical PIs necessary for the formation of endosome from which TLR9 signals to induce type I IFNs.
Collapse
Affiliation(s)
- Kachiko Hayashi
- Howard Hughes Medical Institute, Department of Immunobiology, Yale University School of Medicine, 300 Cedar Street, TAC S655B, New Haven, CT 06520, USA
| | - Miwa Sasai
- Howard Hughes Medical Institute, Department of Immunobiology, Yale University School of Medicine, 300 Cedar Street, TAC S655B, New Haven, CT 06520, USA Present address: Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Akiko Iwasaki
- Howard Hughes Medical Institute, Department of Immunobiology, Yale University School of Medicine, 300 Cedar Street, TAC S655B, New Haven, CT 06520, USA
| |
Collapse
|
142
|
PIKfyve mediates the motility of late endosomes and lysosomes in neuronal dendrites. Neurosci Lett 2015; 605:18-23. [PMID: 26232680 DOI: 10.1016/j.neulet.2015.07.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 07/14/2015] [Accepted: 07/20/2015] [Indexed: 12/22/2022]
Abstract
The endosome/lysosome system in the nervous system is critically important for a variety of neuronal functions such as neurite outgrowth, retrograde transport, and synaptic plasticity. In neurons, the endosome/lysosome system is crucial for the activity-dependent internalization of membrane proteins and contributes to the regulation of lipid level on the plasma membrane. Although homeostasis of membrane dynamics plays important roles in the properties of central nervous systems, it has not been elucidated how endosome/lysosome system is regulated. Here, we report that phosphatidylinositol 3-phosphate 5-kinase (PIKfyve) mediates the motility of late endosomes and lysosomes in neuronal dendrites. Endosomes and lysosomes are highly motile in resting neurons, however knockdown of PIKfyve led to a significant reduction in late endosomes and lysosomes motility. We also found that vesicle acidification is crucial for their motility and PIKfyve is associated with this process indirectly. These data suggest that PIKfyve mediates vesicle motility through the regulation of vesicle integrity in neurons.
Collapse
|
143
|
Balklava Z, Niehage C, Currinn H, Mellor L, Guscott B, Poulin G, Hoflack B, Wassmer T. The Amyloid Precursor Protein Controls PIKfyve Function. PLoS One 2015; 10:e0130485. [PMID: 26125944 PMCID: PMC4488396 DOI: 10.1371/journal.pone.0130485] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 05/20/2015] [Indexed: 12/27/2022] Open
Abstract
While the Amyloid Precursor Protein (APP) plays a central role in Alzheimer's disease, its cellular function still remains largely unclear. It was our goal to establish APP function which will provide insights into APP's implication in Alzheimer's disease. Using our recently developed proteo-liposome assay we established the interactome of APP's intracellular domain (known as AICD), thereby identifying novel APP interactors that provide mechanistic insights into APP function. By combining biochemical, cell biological and genetic approaches we validated the functional significance of one of these novel interactors. Here we show that APP binds the PIKfyve complex, an essential kinase for the synthesis of the endosomal phosphoinositide phosphatidylinositol-3,5-bisphosphate. This signalling lipid plays a crucial role in endosomal homeostasis and receptor sorting. Loss of PIKfyve function by mutation causes profound neurodegeneration in mammals. Using C. elegans genetics we demonstrate that APP functionally cooperates with PIKfyve in vivo. This regulation is required for maintaining endosomal and neuronal function. Our findings establish an unexpected role for APP in the regulation of endosomal phosphoinositide metabolism with dramatic consequences for endosomal biology and important implications for our understanding of Alzheimer's disease.
Collapse
Affiliation(s)
- Zita Balklava
- Aston University, School of Life and Health Sciences, Aston Triangle, Birmingham, B4 7ET, United Kingdom
| | - Christian Niehage
- Biotechnologisches Zentrum, TU-Dresden, Tatzberg 47–49, 01307 Dresden, Germany
| | - Heather Currinn
- Aston University, School of Life and Health Sciences, Aston Triangle, Birmingham, B4 7ET, United Kingdom
| | - Laura Mellor
- University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Benjamin Guscott
- Aston University, School of Life and Health Sciences, Aston Triangle, Birmingham, B4 7ET, United Kingdom
| | - Gino Poulin
- University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Bernard Hoflack
- Biotechnologisches Zentrum, TU-Dresden, Tatzberg 47–49, 01307 Dresden, Germany
| | - Thomas Wassmer
- Aston University, School of Life and Health Sciences, Aston Triangle, Birmingham, B4 7ET, United Kingdom
- * E-mail:
| |
Collapse
|
144
|
Mandelbaum J, Rollins N, Shah P, Bowman D, Lee JY, Tayber O, Bernard H, LeRoy P, Li P, Koenig E, Brownell JE, D'Amore N. Identification of a lung cancer cell line deficient in atg7-dependent autophagy. Autophagy 2015:0. [PMID: 26090719 DOI: 10.1080/15548627.2015.1056966] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Autophagy is a major cellular process for bulk degradation of proteins and organelles in order to maintain metabolic homeostasis, and it represents an emerging target area for cancer. Initially proposed to be a cancer-restricting process for tumor initiation, recent studies suggest that autophagy can also promote cell survival in established tumors. ATG7 is an essential autophagy gene that encodes the E1 enzyme necessary for the lipidation of the LC3 family of ubiquitin-like proteins and autophagosome formation. In this study we identified a rare case of a cancer cell line, H1650 lung adenocarcinoma, which has lost ATG7 expression due to a focal biallelic deletion within the ATG7 locus. These cells displayed no evidence of ATG7 pathway activity; however, reconstituting the cells with wild-type ATG7 restored both LC3 lipidation and downstream autophagic consumption of autophagy substrates such as the SQSTM1/p62 protein. We characterized several phenotypes reported to be influenced by autophagy, and observed an ATG7-dependent increase in cell growth and clearance of proteasome-inhibitor induced protein aggregates. Cellular changes in mitochondrial metabolism or response to nutrient starvation were unaffected by ATG7 expression. In addition, parental H1650 cells that lacked ATG7 were still able to consume autophagy substrates SQSTM1, NBR1 and TAX1BP1 via a bafilomycin A1-sensitive pathway, suggesting that these proteins were not exclusively degraded by autophagy. Overall, these findings highlight a unique outlier instance of complete loss of ATG7-dependent autophagy in a cancer cell line. The H1650 cell line may be a useful system for future studies to further understand the role of autophagy in tumorigenesis and potential redundant pathways that allow cells to circumvent the loss of ATG7-dependent autophagy in cancer.
Collapse
Key Words
- ACTB, actin, beta
- ATG, autophagy related
- Atg7
- BAF, bafilomyin A1
- ECAR, extracellular acidification rate
- GABARAP, GABA(A) receptor-associated protein
- HCQ, hydroxychloroquine
- LC3, microtubule-associated protein 1 light chain 3
- MTOR, mechanistic target of rapamycin (serine/threonine kinase)
- NBR1, neighbor of BRCA1 gene 1
- OCR, oxygen consumption rate
- PI, proteasome inhibitor
- SQSTM1, sequestosome 1
- TAX1BP1, Tax1 (human T-cell leukemia virus type I) binding protein 1
- UB, ubiquitin
- Ubl, ubiquitin-like protein
- WT, wild-type
- lung cancer
- metabolism
- mitochondria
- proteasome
- ubiquitin
Collapse
Affiliation(s)
- Jonathan Mandelbaum
- a Oncology Drug Discovery Unit, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited , Cambridge , Massachusetts , USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Dayam RM, Saric A, Shilliday RE, Botelho RJ. The Phosphoinositide-Gated Lysosomal Ca2+Channel, TRPML1, Is Required for Phagosome Maturation. Traffic 2015; 16:1010-26. [DOI: 10.1111/tra.12303] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 05/15/2015] [Accepted: 05/17/2015] [Indexed: 12/27/2022]
Affiliation(s)
- Roya M. Dayam
- Department of Chemistry and Biology; Ryerson University; Toronto Ontario M5B2K3 Canada
- Molecular Science Program; Ryerson University; Toronto Ontario M5B2K3 Canada
| | - Amra Saric
- Department of Chemistry and Biology; Ryerson University; Toronto Ontario M5B2K3 Canada
- Molecular Science Program; Ryerson University; Toronto Ontario M5B2K3 Canada
| | - Ryan E. Shilliday
- Department of Chemistry and Biology; Ryerson University; Toronto Ontario M5B2K3 Canada
| | - Roberto J. Botelho
- Department of Chemistry and Biology; Ryerson University; Toronto Ontario M5B2K3 Canada
- Molecular Science Program; Ryerson University; Toronto Ontario M5B2K3 Canada
| |
Collapse
|
146
|
Up-regulation of lysosomal TRPML1 channels is essential for lysosomal adaptation to nutrient starvation. Proc Natl Acad Sci U S A 2015; 112:E1373-81. [PMID: 25733853 DOI: 10.1073/pnas.1419669112] [Citation(s) in RCA: 182] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Upon nutrient starvation, autophagy digests unwanted cellular components to generate catabolites that are required for housekeeping biosynthesis processes. A complete execution of autophagy demands an enhancement in lysosome function and biogenesis to match the increase in autophagosome formation. Here, we report that mucolipin-1 (also known as TRPML1 or ML1), a Ca(2+) channel in the lysosome that regulates many aspects of lysosomal trafficking, plays a central role in this quality-control process. By using Ca(2+) imaging and whole-lysosome patch clamping, lysosomal Ca(2+) release and ML1 currents were detected within hours of nutrient starvation and were potently up-regulated. In contrast, lysosomal Na(+)-selective currents were not up-regulated. Inhibition of mammalian target of rapamycin (mTOR) or activation of transcription factor EB (TFEB) mimicked a starvation effect in fed cells. The starvation effect also included an increase in lysosomal proteostasis and enhanced clearance of lysosomal storage, including cholesterol accumulation in Niemann-Pick disease type C (NPC) cells. However, this effect was not observed when ML1 was pharmacologically inhibited or genetically deleted. Furthermore, overexpression of ML1 mimicked the starvation effect. Hence, lysosomal adaptation to environmental cues such as nutrient levels requires mTOR/TFEB-dependent, lysosome-to-nucleus regulation of lysosomal ML1 channels and Ca(2+) signaling.
Collapse
|
147
|
Shubin AV, Demidyuk IV, Lunina NA, Komissarov AA, Roschina MP, Leonova OG, Kostrov SV. Protease 3C of hepatitis A virus induces vacuolization of lysosomal/endosomal organelles and caspase-independent cell death. BMC Cell Biol 2015; 16:4. [PMID: 25886889 PMCID: PMC4355371 DOI: 10.1186/s12860-015-0050-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 01/26/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND 3C proteases, the main proteases of picornaviruses, play the key role in viral life cycle by processing polyproteins. In addition, 3C proteases digest certain host cell proteins to suppress antiviral defense, transcription, and translation. The activity of 3C proteases per se induces host cell death, which makes them critical factors of viral cytotoxicity. To date, cytotoxic effects have been studied for several 3C proteases, all of which induce apoptosis. This study for the first time describes the cytotoxic effect of 3C protease of human hepatitis A virus (3Cpro), the only proteolytic enzyme of the virus. RESULTS Individual expression of 3Cpro induced catalytic activity-dependent cell death, which was not abrogated by the pan-caspase inhibitor (z-VAD-fmk) and was not accompanied by phosphatidylserine externalization in contrast to other picornaviral 3C proteases. The cell survival was also not affected by the inhibitors of cysteine proteases (z-FA-fmk) and RIP1 kinase (necrostatin-1), critical enzymes involved in non-apoptotic cell death. A substantial fraction of dying cells demonstrated numerous non-acidic cytoplasmic vacuoles with not previously described features and originating from several types of endosomal/lysosomal organelles. The lysosomal protein Lamp1 and GTPases Rab5, Rab7, Rab9, and Rab11 were associated with the vacuolar membranes. The vacuolization was completely blocked by the vacuolar ATPase inhibitor (bafilomycin A1) and did not depend on the activity of the principal factors of endosomal transport, GTPases Rab5 and Rab7, as well as on autophagy and macropinocytosis. CONCLUSIONS 3Cpro, apart from other picornaviral 3C proteases, induces caspase-independent cell death, accompanying by cytoplasmic vacuolization. 3Cpro-induced vacuoles have unique properties and are formed from several organelle types of the endosomal/lysosomal compartment. The data obtained demonstrate previously undocumented morphological characters of the 3Cpro-induced cell death, which can reflect unknown aspects of the human hepatitis A virus-host cell interaction.
Collapse
Affiliation(s)
- Andrey V Shubin
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Russian Academy of Science, Moscow, 123182, Russia.
| | - Ilya V Demidyuk
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Russian Academy of Science, Moscow, 123182, Russia.
| | - Nataliya A Lunina
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Russian Academy of Science, Moscow, 123182, Russia.
| | - Alexey A Komissarov
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Russian Academy of Science, Moscow, 123182, Russia.
| | - Marina P Roschina
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Russian Academy of Science, Moscow, 123182, Russia.
| | - Olga G Leonova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119992, Russia.
| | - Sergey V Kostrov
- Laboratory of Protein Engineering, Institute of Molecular Genetics, Russian Academy of Science, Moscow, 123182, Russia.
- National Research Center "Kurchatov Institute", Moscow, 123182, Russia.
| |
Collapse
|
148
|
Ho CY, Choy CH, Wattson CA, Johnson DE, Botelho RJ. The Fab1/PIKfyve phosphoinositide phosphate kinase is not necessary to maintain the pH of lysosomes and of the yeast vacuole. J Biol Chem 2015; 290:9919-28. [PMID: 25713145 DOI: 10.1074/jbc.m114.613984] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Indexed: 12/30/2022] Open
Abstract
Lysosomes and the yeast vacuole are degradative and acidic organelles. Phosphatidylinositol 3,5-bisphosphate (PtdIns(3,5)P2), a master architect of endolysosome and vacuole identity, is thought to be necessary for vacuolar acidification in yeast. There is also evidence that PtdIns(3,5)P2 may play a role in lysosomal acidification in higher eukaryotes. Nevertheless, these conclusions rely on qualitative assays of lysosome/vacuole pH. For example, quinacrine, an acidotropic fluorescent base, does not accumulate in the vacuoles of fab1Δ yeast. Fab1, along with its mammalian ortholog PIKfyve, is the lipid kinase responsible for synthesizing PtdIns(3,5)P2. In this study, we employed several assays that quantitatively assessed the lysosomal and vacuolar pH in PtdIns(3,5)P2-depleted cells. Using ratiometric imaging, we conclude that lysosomes retain a pH < 5 in PIKfyve-inhibited mammalian cells. In addition, quantitative fluorescence microscopy of vacuole-targeted pHluorin, a pH-sensitive GFP variant, indicates that fab1Δ vacuoles are as acidic as wild-type yeast. Importantly, we also employed fluorimetry of vacuoles loaded with cDCFDA, a pH-sensitive dye, to show that both wild-type and fab1Δ vacuoles have a pH < 5.0. In comparison, the vacuolar pH of the V-ATPase mutant vph1Δ or vph1Δ fab1Δ double mutant was 6.1. Although the steady-state vacuolar pH is not affected by PtdIns(3,5)P2 depletion, it may have a role in stabilizing the vacuolar pH during salt shock. Overall, we propose a model in which PtdIns(3,5)P2 does not govern the steady-state pH of vacuoles or lysosomes.
Collapse
Affiliation(s)
- Cheuk Y Ho
- From the Department of Chemistry and Biology and the Molecular Science Program, Ryerson University, Toronto, Ontario M5B2K3, Canada and
| | - Christopher H Choy
- From the Department of Chemistry and Biology and the Molecular Science Program, Ryerson University, Toronto, Ontario M5B2K3, Canada and
| | | | - Danielle E Johnson
- the Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario M5G1X8, Canada
| | - Roberto J Botelho
- From the Department of Chemistry and Biology and the Molecular Science Program, Ryerson University, Toronto, Ontario M5B2K3, Canada and the Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario M5G1X8, Canada
| |
Collapse
|
149
|
Ikonomov OC, Sbrissa D, Venkatareddy M, Tisdale E, Garg P, Shisheva A. Class III PI 3-kinase is the main source of PtdIns3P substrate and membrane recruitment signal for PIKfyve constitutive function in podocyte endomembrane homeostasis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1240-50. [PMID: 25619930 DOI: 10.1016/j.bbamcr.2015.01.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/22/2014] [Accepted: 01/16/2015] [Indexed: 11/19/2022]
Abstract
The evolutionarily conserved PIKfyve, which synthesizes PtdIns5P from PtdIns, and PtdIns(3,5)P2 from PtdIns3P, requires PtdIns3P as both an enzyme substrate and a membrane recruitment signal. Whereas the PtdIns3P source is undetermined, class III PI3K (Vps34), the only evolutionarily conserved of the eight mammalian PI3Ks, is presumed as a main candidate. A hallmark of PIKfyve deficiency is formation of multiple translucent cytoplasmic vacuoles seen by light microscopy in cells cultured in complete media. Such an aberrant phenotype is often observed in cells from conditional Vps34 knockout (KO) mice. To clarify the mechanism of Vps34 KO-triggered vacuolation and the PtdIns3P source for PIKfyve functionality, here we have characterized a podocyte cell type derived from Vps34fl/fl mice, which, upon Cre-mediated gene KO, robustly formed cytoplasmic vacuoles resembling those in PikfyveKO MEFs. Vps34wt, expressed in Vps34KO podocytes restored the normal morphology, but only if the endogenous PIKfyve activity was intact. Conversely, expressed PIKfyvewt rescued completely the vacuolation only in PikfyveKO MEFs but not in Vps34KO podocytes. Analyses of phosphoinositide profiles by HPLC and localization patterns by a PtdIns3P biosensor revealed that Vps34 is the main supplier of localized PtdIns3P not only for PIKfyve activity but also for membrane recruitment. Concordantly, Vps34KO podocytes had severely reduced steady-state levels of both PtdIns(3,5)P2 and PtdIns5P, along with PtdIns3P. We further revealed a plausible physiologically-relevant Vps34-independent PtdIns3P supply for PIKfyve, operating through activated class I PI3Ks. Our data provide the first evidence that the vacuolation phenotype in Vps34KO podocytes is due to PIKfyve dysfunction and that Vps34 is a main PtdIns3P source for constitutive PIKfyve functionality.
Collapse
Affiliation(s)
- Ognian C Ikonomov
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Diego Sbrissa
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | - Ellen Tisdale
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Puneet Garg
- Division of Nephrology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Assia Shisheva
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
150
|
PI(5)P regulates autophagosome biogenesis. Mol Cell 2015; 57:219-34. [PMID: 25578879 PMCID: PMC4306530 DOI: 10.1016/j.molcel.2014.12.007] [Citation(s) in RCA: 204] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 10/22/2014] [Accepted: 11/25/2014] [Indexed: 01/12/2023]
Abstract
Phosphatidylinositol 3-phosphate (PI(3)P), the product of class III PI3K VPS34, recruits specific autophagic effectors, like WIPI2, during the initial steps of autophagosome biogenesis and thereby regulates canonical autophagy. However, mammalian cells can produce autophagosomes through enigmatic noncanonical VPS34-independent pathways. Here we show that PI(5)P can regulate autophagy via PI(3)P effectors and thereby identify a mechanistic explanation for forms of noncanonical autophagy. PI(5)P synthesis by the phosphatidylinositol 5-kinase PIKfyve was required for autophagosome biogenesis, and it increased levels of PI(5)P, stimulated autophagy, and reduced the levels of autophagic substrates. Inactivation of VPS34 impaired recruitment of WIPI2 and DFCP1 to autophagic precursors, reduced ATG5-ATG12 conjugation, and compromised autophagosome formation. However, these phenotypes were rescued by PI(5)P in VPS34-inactivated cells. These findings provide a mechanistic framework for alternative VPS34-independent autophagy-initiating pathways, like glucose starvation, and unravel a cytoplasmic function for PI(5)P, which previously has been linked predominantly to nuclear roles. PI(5)P positively regulates autophagy PI(5)P is associated with autophagy effectors that bind PI(3)P PI(5)P sustains noncanonical autophagy in PI(3)P-depleted cells PI(5)P is essential for VPS34-independent, glucose-starvation-induced autophagy
Collapse
|