1
|
Jerabkova-Roda K, Peralta M, Huang KJ, Mousson A, Bourgeat Maudru C, Bochler L, Busnelli I, Karali R, Justiniano H, Lisii LM, Carl P, Mittelheisser V, Asokan N, Larnicol A, Lefebvre O, Lachuer H, Pichot A, Stemmelen T, Molitor A, Scheid L, Frenger Q, Gros F, Hirschler A, Delalande F, Sick E, Carapito R, Carapito C, Lipsker D, Schauer K, Rondé P, Hyenne V, Goetz JG. Peripheral positioning of lysosomes supports melanoma aggressiveness. Nat Commun 2025; 16:3375. [PMID: 40204688 PMCID: PMC11982396 DOI: 10.1038/s41467-025-58528-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
Emerging evidence suggests that the function and position of organelles are pivotal for tumor cell dissemination. Among them, lysosomes stand out as they integrate metabolic sensing with gene regulation and secretion of proteases. Yet, how their function is linked to their position and how this controls metastasis remains elusive. Here, we analyze lysosome subcellular distribution in patient-derived melanoma cells and patient biopsies and show that lysosome spreading scales with melanoma aggressiveness. Peripheral lysosomes promote matrix degradation and cell invasion which is directly linked to the lysosomal and cell transcriptional programs. Using chemo-genetical control of lysosome positioning, we demonstrate that perinuclear clustering impairs lysosome secretion, matrix degradation and invasion. Impairing lysosome spreading significantly reduces invasive outgrowth in two in vivo models, mouse and zebrafish. Our study provides a direct demonstration that lysosome positioning controls cell invasion, illustrating the importance of organelle adaptation in carcinogenesis and suggesting its potential utility for diagnosis of metastatic melanoma.
Collapse
Affiliation(s)
- Katerina Jerabkova-Roda
- Tumor Biomechanics, Strasbourg, France.
- INSERM UMR_S1109, Strasbourg, France.
- Université de Strasbourg, Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France.
- Institut Curie, PSL, CNRS, UMR144, Paris, France.
| | - Marina Peralta
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
- Epigenetics and Neurobiology Unit, European Molecular Biology Laboratory, 00015, Rome, Italy
| | - Kuang-Jing Huang
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Antoine Mousson
- Université de Strasbourg, Strasbourg, France
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France
| | - Clara Bourgeat Maudru
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Louis Bochler
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Ignacio Busnelli
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Rabia Karali
- Université de Strasbourg, Strasbourg, France
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France
| | - Hélène Justiniano
- Université de Strasbourg, Strasbourg, France
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France
| | - Lucian-Mihai Lisii
- Université de Strasbourg, Strasbourg, France
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France
| | - Philippe Carl
- Université de Strasbourg, Strasbourg, France
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France
| | - Vincent Mittelheisser
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Nandini Asokan
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Annabel Larnicol
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Olivier Lefebvre
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Hugo Lachuer
- Institut Curie, PSL, CNRS, UMR144, Paris, France
- Institut Gustave Roussy, INSERM UMR1279, Université Paris-Saclay, Villejuif, France
- Université de Paris, CNRS, Institut Jacques Monod, 75013, Paris, France
| | - Angélique Pichot
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Plateforme GENOMAX, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
| | - Tristan Stemmelen
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Plateforme GENOMAX, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
| | - Anne Molitor
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Plateforme GENOMAX, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
- Service d'Immunologie Biologique, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, 1 Place de l'Hôpital, 67091, Strasbourg, France
| | - Léa Scheid
- Faculté de Médecine, Université de Strasbourg et Clinique Dermatologique, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Quentin Frenger
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Frédéric Gros
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Aurélie Hirschler
- Laboratoire de Spectrométrie de Masse Bio-Organique (LSMBO), IPHC, UMR 7178, CNRS, Université de Strasbourg, Infrastructure Nationale de Protéomique ProFI, FR2048, Strasbourg, France
| | - François Delalande
- Laboratoire de Spectrométrie de Masse Bio-Organique (LSMBO), IPHC, UMR 7178, CNRS, Université de Strasbourg, Infrastructure Nationale de Protéomique ProFI, FR2048, Strasbourg, France
| | - Emilie Sick
- Université de Strasbourg, Strasbourg, France
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France
| | - Raphaël Carapito
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Plateforme GENOMAX, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
- Service d'Immunologie Biologique, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, 1 Place de l'Hôpital, 67091, Strasbourg, France
| | - Christine Carapito
- Laboratoire de Spectrométrie de Masse Bio-Organique (LSMBO), IPHC, UMR 7178, CNRS, Université de Strasbourg, Infrastructure Nationale de Protéomique ProFI, FR2048, Strasbourg, France
| | - Dan Lipsker
- Faculté de Médecine, Université de Strasbourg et Clinique Dermatologique, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Kristine Schauer
- Institut Curie, PSL, CNRS, UMR144, Paris, France.
- Institut Gustave Roussy, INSERM UMR1279, Université Paris-Saclay, Villejuif, France.
| | - Philippe Rondé
- Université de Strasbourg, Strasbourg, France.
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France.
| | - Vincent Hyenne
- Tumor Biomechanics, Strasbourg, France.
- INSERM UMR_S1109, Strasbourg, France.
- Université de Strasbourg, Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France.
- CNRS, SNC5055, Strasbourg, France.
| | - Jacky G Goetz
- Tumor Biomechanics, Strasbourg, France.
- INSERM UMR_S1109, Strasbourg, France.
- Université de Strasbourg, Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France.
| |
Collapse
|
2
|
Afghah Z, Khan N, Datta G, Halcrow PW, Geiger JD, Chen X. Involvement of Endolysosomes and Aurora Kinase A in the Regulation of Amyloid β Protein Levels in Neurons. Int J Mol Sci 2024; 25:6200. [PMID: 38892390 PMCID: PMC11172969 DOI: 10.3390/ijms25116200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/28/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Aurora kinase A (AURKA) is a serine/threonine-protein kinase that regulates microtubule organization during neuron migration and neurite formation. Decreased activity of AURKA was found in Alzheimer's disease (AD) brain samples, but little is known about the role of AURKA in AD pathogenesis. Here, we demonstrate that AURKA is expressed in primary cultured rat neurons, neurons from adult mouse brains, and neurons in postmortem human AD brains. AURKA phosphorylation, which positively correlates with its activity, is reduced in human AD brains. In SH-SY5Y cells, pharmacological activation of AURKA increased AURKA phosphorylation, acidified endolysosomes, decreased the activity of amyloid beta protein (Aβ) generating enzyme β-site amyloid precursor protein cleaving enzyme (BACE-1), increased the activity of the Aβ degrading enzyme cathepsin D, and decreased the intracellular and secreted levels of Aβ. Conversely, pharmacological inhibition of AURKA decreased AURKA phosphorylation, de-acidified endolysosomes, decreased the activity of cathepsin D, and increased intracellular and secreted levels of Aβ. Thus, reduced AURKA activity in AD may contribute to the development of intraneuronal accumulations of Aβ and extracellular amyloid plaque formation.
Collapse
Affiliation(s)
| | | | | | | | | | - Xuesong Chen
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA; (Z.A.); (N.K.); (G.D.); (P.W.H.); (J.D.G.)
| |
Collapse
|
3
|
Luo X, Zhang J, Tolö J, Kügler S, Michel U, Bähr M, Koch JC. Axonal autophagic vesicle transport in the rat optic nerve in vivo under normal conditions and during acute axonal degeneration. Acta Neuropathol Commun 2024; 12:82. [PMID: 38812004 PMCID: PMC11134632 DOI: 10.1186/s40478-024-01791-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 05/08/2024] [Indexed: 05/31/2024] Open
Abstract
Neurons pose a particular challenge to degradative processes like autophagy due to their long and thin processes. Autophagic vesicles (AVs) are formed at the tip of the axon and transported back to the soma. This transport is essential since the final degradation of the vesicular content occurs only close to or in the soma. Here, we established an in vivo live-imaging model in the rat optic nerve using viral vector mediated LC3-labeling and two-photon-microscopy to analyze axonal transport of AVs. Under basal conditions in vivo, 50% of the AVs are moving with a majority of 85% being transported in the retrograde direction. Transport velocity is higher in the retrograde than in the anterograde direction. A crush lesion of the optic nerve results in a rapid breakdown of retrograde axonal transport while the anterograde transport stays intact over several hours. Close to the lesion site, the formation of AVs is upregulated within the first 6 h after crush, but the clearance of AVs and the levels of lysosomal markers in the adjacent axon are reduced. Expression of p150Glued, an adaptor protein of dynein, is significantly reduced after crush lesion. In vitro, fusion and colocalization of the lysosomal marker cathepsin D with AVs are reduced after axotomy. Taken together, we present here the first in vivo analysis of axonal AV transport in the mammalian CNS using live-imaging. We find that axotomy leads to severe defects of retrograde motility and a decreased clearance of AVs via the lysosomal system.
Collapse
Affiliation(s)
- Xiaoyue Luo
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - Jiong Zhang
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - Johan Tolö
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - Sebastian Kügler
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - Uwe Michel
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - Mathias Bähr
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - Jan Christoph Koch
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany.
| |
Collapse
|
4
|
Yin Q, Yang C. Exploring lysosomal biology: current approaches and methods. BIOPHYSICS REPORTS 2024; 10:111-120. [PMID: 38774350 PMCID: PMC11103719 DOI: 10.52601/bpr.2023.230028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 01/04/2024] [Indexed: 05/24/2024] Open
Abstract
Lysosomes are the degradation centers and signaling hubs in the cell. Lysosomes undergo adaptation to maintain cell homeostasis in response to a wide variety of cues. Dysfunction of lysosomes leads to aging and severe diseases including lysosomal storage diseases (LSDs), neurodegenerative disorders, and cancer. To understand the complexity of lysosome biology, many research approaches and tools have been developed to investigate lysosomal functions and regulatory mechanisms in diverse experimental systems. This review summarizes the current approaches and tools adopted for studying lysosomes, and aims to provide a methodological overview of lysosomal research and related fields.
Collapse
Affiliation(s)
- Qiuyuan Yin
- State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan and Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Chonglin Yang
- State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan and Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650091, China
- Southwest United Graduate School, Kunming 650092, China
| |
Collapse
|
5
|
Bose S, de Heus C, Kennedy ME, Wang F, Jentsch TJ, Klumperman J, Stauber T. Impaired Autophagic Clearance with a Gain-of-Function Variant of the Lysosomal Cl -/H + Exchanger ClC-7. Biomolecules 2023; 13:1799. [PMID: 38136669 PMCID: PMC10742274 DOI: 10.3390/biom13121799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/04/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
ClC-7 is a ubiquitously expressed voltage-gated Cl-/H+ exchanger that critically contributes to lysosomal ion homeostasis. Together with its β-subunit Ostm1, ClC-7 localizes to lysosomes and to the ruffled border of osteoclasts, where it supports the acidification of the resorption lacuna. Loss of ClC-7 or Ostm1 leads to osteopetrosis accompanied by accumulation of storage material in lysosomes and neurodegeneration. Interestingly, not all osteopetrosis-causing CLCN7 mutations from patients are associated with a loss of ion transport. Some rather result in an acceleration of voltage-dependent ClC-7 activation. Recently, a gain-of-function variant, ClC-7Y715C, that yields larger ion currents upon heterologous expression, was identified in two patients with neurodegeneration, organomegaly and albinism. However, neither the patients nor a mouse model that carried the equivalent mutation developed osteopetrosis, although expression of ClC-7Y715C induced the formation of enlarged intracellular vacuoles. Here, we investigated how, in transfected cells with mutant ClC-7, the substitution of this tyrosine impinged on the morphology and function of lysosomes. Combinations of the tyrosine mutation with mutations that either uncouple Cl- from H+ counter-transport or strongly diminish overall ion currents were used to show that increased ClC-7 Cl-/H+ exchange activity is required for the formation of enlarged vacuoles by membrane fusion. Degradation of endocytosed material was reduced in these compartments and resulted in an accumulation of lysosomal storage material. In cells expressing the ClC-7 gain-of-function mutant, autophagic clearance was largely impaired, resulting in a build-up of autophagic material.
Collapse
Affiliation(s)
- Shroddha Bose
- Institute for Molecular Medicine, MSH Medical School Hamburg, 20457 Hamburg, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Cecilia de Heus
- Center for Molecular Medicine/Cell Biology, University Medical Center (UMC), 3584 CX Utrecht, The Netherlands
| | - Mary E. Kennedy
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Fan Wang
- Institute for Molecular Medicine, MSH Medical School Hamburg, 20457 Hamburg, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Thomas J. Jentsch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Judith Klumperman
- Center for Molecular Medicine/Cell Biology, University Medical Center (UMC), 3584 CX Utrecht, The Netherlands
| | - Tobias Stauber
- Institute for Molecular Medicine, MSH Medical School Hamburg, 20457 Hamburg, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| |
Collapse
|
6
|
Cook AA, Leung TCS, Rice M, Nachman M, Zadigue-Dube É, Watt AJ. Endosomal dysfunction contributes to cerebellar deficits in spinocerebellar ataxia type 6. eLife 2023; 12:RP90510. [PMID: 38084749 PMCID: PMC10715727 DOI: 10.7554/elife.90510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023] Open
Abstract
Spinocerebellar ataxia type 6 (SCA6) is a rare disease that is characterized by cerebellar dysfunction. Patients have progressive motor coordination impairment, and postmortem brain tissue reveals degeneration of cerebellar Purkinje cells and a reduced level of cerebellar brain-derived neurotrophic factor (BDNF). However, the pathophysiological changes underlying SCA6 are not fully understood. We carried out RNA-sequencing of cerebellar vermis tissue in a mouse model of SCA6, which revealed widespread dysregulation of genes associated with the endo-lysosomal system. Since disruption to endosomes or lysosomes could contribute to cellular deficits, we examined the endo-lysosomal system in SCA6. We identified alterations in multiple endosomal compartments in the Purkinje cells of SCA6 mice. Early endosomes were enlarged, while the size of the late endosome compartment was reduced. We also found evidence for impaired trafficking of cargo to the lysosomes. As the proper functioning of the endo-lysosomal system is crucial for the sorting and trafficking of signaling molecules, we wondered whether these changes could contribute to previously identified deficits in signaling by BDNF and its receptor tropomyosin kinase B (TrkB) in SCA6. Indeed, we found that the enlarged early endosomes in SCA6 mice accumulated both BDNF and TrkB. Furthermore, TrkB recycling to the cell membrane in recycling endosomes was reduced, and the late endosome transport of BDNF for degradation was impaired. Therefore, mis-trafficking due to aberrant endo-lysosomal transport and function could contribute to SCA6 pathophysiology through alterations to BDNF-TrkB signaling, as well as mishandling of other signaling molecules. Deficits in early endosomes and BDNF localization were rescued by chronic administration of a TrkB agonist, 7,8-dihydroxyflavone, that we have previously shown restores motor coordination and cerebellar TrkB expression. The endo-lysosomal system is thus both a novel locus of pathophysiology in SCA6 and a promising therapeutic target.
Collapse
Affiliation(s)
- Anna A Cook
- Biology Department, McGill UniversityMontrealCanada
| | | | - Max Rice
- Biology Department, McGill UniversityMontrealCanada
- Department of Biological Sciences, Columbia UniversityNew YorkUnited States
| | - Maya Nachman
- Biology Department, McGill UniversityMontrealCanada
| | | | | |
Collapse
|
7
|
Rodriguez-Rios M, McHugh BJ, Liang Z, Megia-Fernandez A, Lilienkampf A, Dockrell D, Bradley M. A fluorogenic, peptide-based probe for the detection of Cathepsin D in macrophages. Commun Chem 2023; 6:237. [PMID: 37919467 PMCID: PMC10622513 DOI: 10.1038/s42004-023-01035-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 10/20/2023] [Indexed: 11/04/2023] Open
Abstract
Cathepsin D is a protease that is an effector in the immune response of macrophages, yet to date, only a limited number of probes have been developed for its detection. Herein, we report a water soluble, highly sensitive, pH insensitive fluorescent probe for the detection of Cathepsin D activity that provides a strong OFF/ON signal upon activation and with bright emission at 515 nm. The probe was synthesised using a combination of solid and solution-phase chemistries, with probe optimisation to increase its water solubility and activation kinetics by addition of a long PEG chain (5 kDa) at the C-terminus. A BODIPY fluorophore allowed detection of Cathepsin D across a wide pH range, important as the protease is active both at the low pH found in lysosomes and also in higher pH phagolysosomes, and in the cytosol. The probe was successfully used to detect Cathepsin D activity in macrophages challenged by exposure to bacteria.
Collapse
Affiliation(s)
- Maria Rodriguez-Rios
- School of Chemistry, University of Edinburgh, David Brewster Road, EH9 3FJ, Edinburgh, UK
| | - Brian J McHugh
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh, EH16 4TJ, UK
| | - Zhengqi Liang
- School of Chemistry, University of Edinburgh, David Brewster Road, EH9 3FJ, Edinburgh, UK
| | - Alicia Megia-Fernandez
- School of Chemistry, University of Edinburgh, David Brewster Road, EH9 3FJ, Edinburgh, UK
- Organic Chemistry Department, Faculty of Sciences, University of Granada, Avda. Fuente Nueva S/N, Granada, 18071, Spain
| | - Annamaria Lilienkampf
- School of Chemistry, University of Edinburgh, David Brewster Road, EH9 3FJ, Edinburgh, UK
| | - David Dockrell
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh, EH16 4TJ, UK
| | - Mark Bradley
- Precision Healthcare University Research Institute, Queen Mary University of London, Empire House, 67-75 New Road, London, E1 1HH, UK.
| |
Collapse
|
8
|
Song P, Zhang X, Wang S, Xu W, Wang F, Fu R, Wei F. Microbial proteases and their applications. Front Microbiol 2023; 14:1236368. [PMID: 37779686 PMCID: PMC10537240 DOI: 10.3389/fmicb.2023.1236368] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/30/2023] [Indexed: 10/03/2023] Open
Abstract
Proteases (proteinases or peptidases) are a class of hydrolases that cleave peptide chains in proteins. Endopeptidases are a type of protease that hydrolyze the internal peptide bonds of proteins, forming shorter peptides; exopeptidases hydrolyze the terminal peptide bonds from the C-terminal or N-terminal, forming free amino acids. Microbial proteases are a popular instrument in many industrial applications. In this review, the classification, detection, identification, and sources of microbial proteases are systematically introduced, as well as their applications in food, detergents, waste treatment, and biotechnology processes in the industry fields. In addition, recent studies on techniques used to express heterologous microbial proteases are summarized to describe the process of studying proteases. Finally, future developmental trends for microbial proteases are discussed.
Collapse
Affiliation(s)
- Peng Song
- College of Life Sciences, Liaocheng University, Liaocheng, China
- Shandong Aobo Biotech Co. Ltd., Liaocheng, China
- Jiangxi Zymerck Biotech Co. Ltd., Nanchang, China
| | - Xue Zhang
- College of Life Sciences, Liaocheng University, Liaocheng, China
| | - Shuhua Wang
- Shandong Aobo Biotech Co. Ltd., Liaocheng, China
| | - Wei Xu
- College of Life Sciences, Liaocheng University, Liaocheng, China
| | - Fei Wang
- College of Life Sciences, Liaocheng University, Liaocheng, China
| | - Rongzhao Fu
- Jiangxi Zymerck Biotech Co. Ltd., Nanchang, China
| | - Feng Wei
- College of Life Sciences, Liaocheng University, Liaocheng, China
| |
Collapse
|
9
|
Grochowska KM, Sperveslage M, Raman R, Failla AV, Głów D, Schulze C, Laprell L, Fehse B, Kreutz MR. Chaperone-mediated autophagy in neuronal dendrites utilizes activity-dependent lysosomal exocytosis for protein disposal. Cell Rep 2023; 42:112998. [PMID: 37590146 DOI: 10.1016/j.celrep.2023.112998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 06/16/2023] [Accepted: 08/01/2023] [Indexed: 08/19/2023] Open
Abstract
The complex morphology of neurons poses a challenge for proteostasis because the majority of lysosomal degradation machinery is present in the cell soma. In recent years, however, mature lysosomes were identified in dendrites, and a fraction of those appear to fuse with the plasma membrane and release their content to the extracellular space. Here, we report that dendritic lysosomes are heterogeneous in their composition and that only those containing lysosome-associated membrane protein (LAMP) 2A and 2B fuse with the membrane and exhibit activity-dependent motility. Exocytotic lysosomes dock in close proximity to GluN2B-containing N-methyl-D-aspartate-receptors (NMDAR) via an association of LAMP2B to the membrane-associated guanylate kinase family member SAP102/Dlg3. NMDAR-activation decreases lysosome motility and promotes membrane fusion. We find that chaperone-mediated autophagy is a supplier of content that is released to the extracellular space via lysosome exocytosis. This mechanism enables local disposal of aggregation-prone proteins like TDP-43 and huntingtin.
Collapse
Affiliation(s)
- Katarzyna M Grochowska
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany.
| | - Marit Sperveslage
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Rajeev Raman
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Antonio V Failla
- UKE Microscopic Imaging Facility (umif), University Medical Center Eppendorf, 20251 Hamburg, Germany
| | - Dawid Głów
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Centre Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany
| | - Christian Schulze
- Institute of Synaptic Physiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Laura Laprell
- Institute of Synaptic Physiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Centre Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany
| | - Michael R Kreutz
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto von Guericke University, 39120 Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany.
| |
Collapse
|
10
|
Sánchez-Vidaña DI, Li J, Abokyi S, Chan JNM, Ngai SPC, Lau BWM. In vitro methods in autophagy research: Applications in neurodegenerative diseases and mood disorders. Front Mol Neurosci 2023; 16:1168948. [PMID: 37122628 PMCID: PMC10130388 DOI: 10.3389/fnmol.2023.1168948] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 03/14/2023] [Indexed: 05/02/2023] Open
Abstract
Background Autophagy is a conserved physiological intracellular mechanism responsible for the degradation and recycling of cytoplasmic constituents (e.g., damaged organelles, and protein aggregates) to maintain cell homeostasis. Aberrant autophagy has been observed in neurodegenerative diseases, including Alzheimer's Disease (AD), Parkinson's Disease (PD), Amyotrophic Lateral Sclerosis (ALS), and Huntington's Disease (HD), and recently aberrant autophagy has been associated with mood disorders, such as depression. Several in vitro methods have been developed to study the complex and tightly regulated mechanisms of autophagy. In vitro methods applied to autophagy research are used to identify molecular key players involved in dysfunctional autophagy and to screen autophagy regulators with therapeutic applications in neurological diseases and mood disorders. Therefore, the aims of this narrative review are (1) to compile information on the cell-based methods used in autophagy research, (2) to discuss their application, and (3) to create a catalog of traditional and novel in vitro methods applied in neurodegenerative diseases and depression. Methods Pubmed and Google Scholar were used to retrieve relevant in vitro studies on autophagy mechanisms in neurological diseases and depression using a combination of search terms per mechanism and disease (e.g., "macroautophagy" and "Alzheimer's disease"). A total of 37 studies were included (14 in PD, 8 in AD, 5 in ALS, 5 in %, and 5 in depression). Results A repertoire of traditional and novel approaches and techniques was compiled and discussed. The methods used in autophagy research focused on the mechanisms of macroautophagy, microautophagy, and chaperone-mediated autophagy. The in vitro tools presented in this review can be applied to explore pathophysiological mechanisms at a molecular level and to screen for potential therapeutic agents and their mechanism of action, which can be of great importance to understanding disease biology and potential therapeutic options in the context of neurodegenerative disorders and depression. Conclusion This is the first review to compile, discuss, and provide a catalog of traditional and novel in vitro models applied to neurodegenerative disorders and depression.
Collapse
Affiliation(s)
- Dalinda Isabel Sánchez-Vidaña
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Mental Health Research Centre, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Jingjing Li
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Samuel Abokyi
- School of Optometry, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Jackie Ngai-Man Chan
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Shirley Pui-Ching Ngai
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Benson Wui-Man Lau
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Mental Health Research Centre, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| |
Collapse
|
11
|
Somogyi A, Kirkham ED, Lloyd-Evans E, Winston J, Allen ND, Mackrill JJ, Anderson KE, Hawkins PT, Gardiner SE, Waller-Evans H, Sims R, Boland B, O'Neill C. The synthetic TRPML1 agonist ML-SA1 rescues Alzheimer-related alterations of the endosomal-autophagic-lysosomal system. J Cell Sci 2023; 136:jcs259875. [PMID: 36825945 PMCID: PMC10112969 DOI: 10.1242/jcs.259875] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Abnormalities in the endosomal-autophagic-lysosomal (EAL) system are an early event in Alzheimer's disease (AD) pathogenesis. However, the mechanisms underlying these abnormalities are unclear. The transient receptor potential channel mucolipin 1(TRPML1, also known as MCOLN1), a vital endosomal-lysosomal Ca2+ channel whose loss of function leads to neurodegeneration, has not been investigated with respect to EAL pathogenesis in late-onset AD (LOAD). Here, we identify pathological hallmarks of TRPML1 dysregulation in LOAD neurons, including increased perinuclear clustering and vacuolation of endolysosomes. We reveal that induced pluripotent stem cell (iPSC)-derived human cortical neurons expressing APOE ε4, the strongest genetic risk factor for LOAD, have significantly diminished TRPML1-induced endolysosomal Ca2+ release. Furthermore, we found that blocking TRPML1 function in primary neurons by depleting the TRPML1 agonist PI(3,5)P2 via PIKfyve inhibition, recreated multiple features of EAL neuropathology evident in LOAD. This included increased endolysosomal Ca2+ content, enlargement and perinuclear clustering of endolysosomes, autophagic vesicle accumulation and early endosomal enlargement. Strikingly, these AD-like neuronal EAL defects were rescued by TRPML1 reactivation using its synthetic agonist ML-SA1. These findings implicate defects in TRPML1 in LOAD EAL pathogenesis and present TRPML1 as a potential therapeutic target.
Collapse
Affiliation(s)
- Aleksandra Somogyi
- School of Biochemistry and Cell Biology, BioSciences Institute, University College Cork, T12 YT20 Cork, Ireland
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, T12 XF62 Cork, Ireland
- Cork Neuroscience Centre (CNSC), University College Cork, T12 YT20 Cork, Ireland
| | - Emily D Kirkham
- School of Biosciences, Sir Martin Evans building, Cardiff University, CF10 3AX Cardiff, UK
| | - Emyr Lloyd-Evans
- School of Biosciences, Sir Martin Evans building, Cardiff University, CF10 3AX Cardiff, UK
| | - Jincy Winston
- UK Dementia Research Institute, Hadyn Ellis Building, Cardiff University, CF24 4HQ Cardiff, UK
| | - Nicholas D Allen
- School of Biosciences, Sir Martin Evans building, Cardiff University, CF10 3AX Cardiff, UK
| | - John J Mackrill
- Department of Physiology, School of Medicine, University College Cork, T12 YT20 Cork, Ireland
| | - Karen E Anderson
- The Babraham Institute, Babraham Research Campus, CB22 3AT Cambridge, UK
| | - Phillip T Hawkins
- The Babraham Institute, Babraham Research Campus, CB22 3AT Cambridge, UK
| | - Sian E Gardiner
- Medicines Discovery Institute, Main Building, Cardiff University, CF10 3AT Cardiff, UK
| | - Helen Waller-Evans
- Medicines Discovery Institute, Main Building, Cardiff University, CF10 3AT Cardiff, UK
| | - Rebecca Sims
- Division of Psychological Medicine and Clinical Neuroscience, Cardiff University, C14 4XN Cardiff, UK
| | - Barry Boland
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, T12 XF62 Cork, Ireland
- Cork Neuroscience Centre (CNSC), University College Cork, T12 YT20 Cork, Ireland
| | - Cora O'Neill
- School of Biochemistry and Cell Biology, BioSciences Institute, University College Cork, T12 YT20 Cork, Ireland
- Cork Neuroscience Centre (CNSC), University College Cork, T12 YT20 Cork, Ireland
| |
Collapse
|
12
|
Serra-Vinardell J, Sandler MB, De Pace R, Manzella-Lapeira J, Cougnoux A, Keyvanfar K, Introne WJ, Brzostowski JA, Ward ME, Gahl WA, Sharma P, Malicdan MCV. LYST deficiency impairs autophagic lysosome reformation in neurons and alters lysosome number and size. Cell Mol Life Sci 2023; 80:53. [PMID: 36707427 PMCID: PMC11072721 DOI: 10.1007/s00018-023-04695-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/11/2022] [Accepted: 01/08/2023] [Indexed: 01/29/2023]
Abstract
Chediak-Higashi syndrome (CHS) is a rare, autosomal recessive disorder caused by biallelic mutations in the lysosomal trafficking regulator (LYST) gene. Even though enlarged lysosomes and/or lysosome-related organelles (LROs) are the typical cellular hallmarks of CHS, they have not been investigated in human neuronal models. Moreover, how and why the loss of LYST function causes a lysosome phenotype in cells has not been elucidated. We report that the LYST-deficient human neuronal model exhibits lysosome depletion accompanied by hyperelongated tubules extruding from enlarged autolysosomes. These results have also been recapitulated in neurons differentiated from CHS patients' induced pluripotent stem cells (iPSCs), validating our model system. We propose that LYST ensures the correct fission/scission of the autolysosome tubules during autophagic lysosome reformation (ALR), a crucial process to restore the number of free lysosomes after autophagy. We further demonstrate that LYST is recruited to the lysosome membrane, likely to facilitate the fission of autolysosome tubules. Together, our results highlight the key role of LYST in maintaining lysosomal homeostasis following autophagy and suggest that ALR dysregulation is likely associated with the neurodegenerative CHS phenotype.
Collapse
Affiliation(s)
- Jenny Serra-Vinardell
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Maxwell B Sandler
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Raffaella De Pace
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Javier Manzella-Lapeira
- Twinbrook Imaging Facility, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20892, USA
| | - Antony Cougnoux
- Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Keyvan Keyvanfar
- National Heart, Lung, and Blood Institute, Flow Cytometry Facility, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Wendy J Introne
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Joseph A Brzostowski
- Twinbrook Imaging Facility, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20892, USA
| | - Michael E Ward
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892, USA
| | - William A Gahl
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Undiagnosed Diseases Program, National Human Genome Research Institute (NHGRI), National Institutes of Health (NIH), Common Fund, Office of the Director, NIH, Bethesda, MD, 20892, USA
| | - Prashant Sharma
- Undiagnosed Diseases Program, National Human Genome Research Institute (NHGRI), National Institutes of Health (NIH), Common Fund, Office of the Director, NIH, Bethesda, MD, 20892, USA.
| | - May Christine V Malicdan
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Undiagnosed Diseases Program, National Human Genome Research Institute (NHGRI), National Institutes of Health (NIH), Common Fund, Office of the Director, NIH, Bethesda, MD, 20892, USA
| |
Collapse
|
13
|
Mulligan RJ, Yap CC, Winckler B. Endosomal Transport to Lysosomes and the Trans-Golgi Network in Neurons and Other Cells: Visualizing Maturational Flux. Methods Mol Biol 2023; 2557:595-618. [PMID: 36512240 DOI: 10.1007/978-1-0716-2639-9_36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
High-level microscopy enables the comprehensive study of dynamic intracellular processes. Here we describe a toolkit of combinatorial approaches for fixed cell imaging and live cell imaging to investigate the interactions along the trans-Golgi network (TGN)-endosome-lysosome transport axis, which underlie the maturation of endosomal compartments and degradative flux. For fixed cell approaches, we specifically highlight how choices of permeabilization conditions, antibody selection, and antibody multiplexing affect interpretation of results. For live cell approaches, we emphasize the use of sensors that read out pH and degradative capacity in combination with endosomal identity for elucidating dynamic compartment changes.
Collapse
Affiliation(s)
| | - Chan Choo Yap
- Department of Cell Biology, University of Virginia, Charlottesville, VA, USA.
| | - Bettina Winckler
- Department of Cell Biology, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
14
|
Jo YR, Oh Y, Kim YH, Shin YK, Kim HR, Go H, Shin J, Park HJ, Koh H, Kim JK, Shin JE, Lee KE, Park HT. Adaptive autophagy reprogramming in Schwann cells during peripheral demyelination. Cell Mol Life Sci 2023; 80:34. [PMID: 36622429 PMCID: PMC9829575 DOI: 10.1007/s00018-022-04683-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/23/2022] [Accepted: 12/26/2022] [Indexed: 01/10/2023]
Abstract
The myelin sheath is an essential structure for the rapid transmission of electrical impulses through axons, and peripheral myelination is a well-programmed postnatal process of Schwann cells (SCs), the myelin-forming peripheral glia. SCs transdifferentiate into demyelinating SCs (DSCs) to remove the myelin sheath during Wallerian degeneration after axonal injury and demyelinating neuropathies, and macrophages are responsible for the degradation of myelin under both conditions. In this study, the mechanism by which DSCs acquire the ability of myelin exocytosis was investigated. Using serial ultrastructural evaluation, we found that autophagy-related gene 7-dependent formation of a "secretory phagophore (SP)" and tubular phagophore was necessary for exocytosis of large myelin chambers by DSCs. DSCs seemed to utilize myelin membranes for SP formation and employed p62/sequestosome-1 (p62) as an autophagy receptor for myelin excretion. In addition, the acquisition of the myelin exocytosis ability of DSCs was associated with the decrease in canonical autolysosomal flux and was demonstrated by p62 secretion. Finally, this SC demyelination mechanism appeared to also function in inflammatory demyelinating neuropathies. Our findings show a novel autophagy-mediated myelin clearance mechanism by DSCs in response to nerve damage.
Collapse
Affiliation(s)
- Young Rae Jo
- Peripheral Neuropathy Research Center (PNRC), Department of Molecular Neuroscience, College of Medicine, Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Dong-A University, Busan, 49201 Republic of Korea
| | - Yuna Oh
- Advanced Analysis Center, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792 Republic of Korea
| | - Young Hee Kim
- Peripheral Neuropathy Research Center (PNRC), Department of Molecular Neuroscience, College of Medicine, Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Dong-A University, Busan, 49201 Republic of Korea
| | - Yoon Kyung Shin
- Peripheral Neuropathy Research Center (PNRC), Department of Molecular Neuroscience, College of Medicine, Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Dong-A University, Busan, 49201 Republic of Korea
| | - Hye Ran Kim
- Peripheral Neuropathy Research Center (PNRC), Department of Molecular Neuroscience, College of Medicine, Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Dong-A University, Busan, 49201 Republic of Korea
| | - Hana Go
- Peripheral Neuropathy Research Center (PNRC), Department of Molecular Neuroscience, College of Medicine, Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Dong-A University, Busan, 49201 Republic of Korea
| | - Jaekyoon Shin
- Department of Molecular and Cellular Biology, College of Medicine, Sungkyunkwan University, Suwon-Si, 16419 Republic of Korea
| | - Hye Ji Park
- Department of Pharmacology, College of Medicine, Dong-A University, Busan, 49201 Republic of Korea
| | - Hyongjong Koh
- Department of Pharmacology, College of Medicine, Dong-A University, Busan, 49201 Republic of Korea
| | - Jong Kuk Kim
- Department of Neurology, College of Medicine, Dong-A University, Busan, 49201 Republic of Korea
| | - Jung Eun Shin
- Peripheral Neuropathy Research Center (PNRC), Department of Molecular Neuroscience, College of Medicine, Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Dong-A University, Busan, 49201 Republic of Korea
| | - Kyung Eun Lee
- Advanced Analysis Center, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792 Republic of Korea
| | - Hwan Tae Park
- Peripheral Neuropathy Research Center (PNRC), Department of Molecular Neuroscience, College of Medicine, Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Dong-A University, Busan, 49201 Republic of Korea
| |
Collapse
|
15
|
Parkinson's disease-risk protein TMEM175 is a proton-activated proton channel in lysosomes. Cell 2022; 185:2292-2308.e20. [PMID: 35750034 DOI: 10.1016/j.cell.2022.05.021] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 11/07/2021] [Accepted: 05/24/2022] [Indexed: 02/06/2023]
Abstract
Lysosomes require an acidic lumen between pH 4.5 and 5.0 for effective digestion of macromolecules. This pH optimum is maintained by proton influx produced by the V-ATPase and efflux through an unidentified "H+ leak" pathway. Here we show that TMEM175, a genetic risk factor for Parkinson's disease (PD), mediates the lysosomal H+ leak by acting as a proton-activated, proton-selective channel on the lysosomal membrane (LyPAP). Acidification beyond the normal range potently activated LyPAP to terminate further acidification of lysosomes. An endogenous polyunsaturated fatty acid and synthetic agonists also activated TMEM175 to trigger lysosomal proton release. TMEM175 deficiency caused lysosomal over-acidification, impaired proteolytic activity, and facilitated α-synuclein aggregation in vivo. Mutational and pH normalization analyses indicated that the channel's H+ conductance is essential for normal lysosome function. Thus, modulation of LyPAP by cellular cues may dynamically tune the pH optima of endosomes and lysosomes to regulate lysosomal degradation and PD pathology.
Collapse
|
16
|
Chin M, Ang KH, Davies J, Alquezar C, Garda VG, Rooney B, Leng K, Kampmann M, Arkin MR, Kao AW. Phenotypic Screening Using High-Content Imaging to Identify Lysosomal pH Modulators in a Neuronal Cell Model. ACS Chem Neurosci 2022; 13:1505-1516. [PMID: 35522480 PMCID: PMC9121341 DOI: 10.1021/acschemneuro.1c00804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 04/25/2022] [Indexed: 11/29/2022] Open
Abstract
Lysosomes are intracellular organelles responsible for the degradation of diverse macromolecules in a cell. A highly acidic pH is required for the optimal functioning of lysosomal enzymes. Loss of lysosomal intralumenal acidity can disrupt cellular protein homeostasis and is linked to age-related diseases such as neurodegeneration. Using a new robust lysosomal pH biosensor (FIRE-pHLy), we developed a cell-based fluorescence assay for high-throughput screening (HTS) and applied it to differentiated SH-SY5Y neuroblastoma cells. The goal of this study was twofold: (1) to screen for small molecules that acidify lysosomal pH and (2) to identify molecular targets and pathways that regulate lysosomal pH. We conducted a screen of 1835 bioactive compounds with annotated target information to identify lysosomal pH modulators (both acidifiers and alkalinizers). Forty-five compounds passed the initial hit selection criteria, using a combined analysis approach of population-based and object-based data. Twenty-three compounds were retested in dose-response assays and two compounds, OSI-027 and PP242, were identified as top acidifying hits. Overall, data from this phenotypic HTS screen may be used to explore novel regulatory pathways of lysosomal pH regulation. Additionally, OSI-027 and PP242 may serve as useful tool compounds to enable mechanistic studies of autophagy activation and lysosomal acidification as potential therapeutic pathways for neurodegenerative diseases.
Collapse
Affiliation(s)
- Marcus
Y. Chin
- Memory
and Aging Center, Department of Neurology, University of California, San
Francisco, California, California 94158, United States
- Small
Molecule Discovery Center, Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, United States
| | - Kean-Hooi Ang
- Small
Molecule Discovery Center, Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, United States
| | - Julia Davies
- Small
Molecule Discovery Center, Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, United States
| | - Carolina Alquezar
- Memory
and Aging Center, Department of Neurology, University of California, San
Francisco, California, California 94158, United States
| | - Virginia G. Garda
- Memory
and Aging Center, Department of Neurology, University of California, San
Francisco, California, California 94158, United States
- Small
Molecule Discovery Center, Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, United States
| | - Brendan Rooney
- Institute
for Neurodegenerative Diseases, Department of Biochemistry and Biophysics, University of California, San Francisco, California 94158, United States
| | - Kun Leng
- Institute
for Neurodegenerative Diseases, Department of Biochemistry and Biophysics, University of California, San Francisco, California 94158, United States
- Biomedical
Sciences Graduate Program, University of
California, San Francisco, California 94158, United States
- Medical
Scientist Training Program, University of
California, San Francisco, California 94158, United States
| | - Martin Kampmann
- Institute
for Neurodegenerative Diseases, Department of Biochemistry and Biophysics, University of California, San Francisco, California 94158, United States
| | - Michelle R. Arkin
- Small
Molecule Discovery Center, Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, United States
| | - Aimee W. Kao
- Memory
and Aging Center, Department of Neurology, University of California, San
Francisco, California, California 94158, United States
| |
Collapse
|
17
|
Wróbel M, Cendrowski J, Szymańska E, Grębowicz-Maciukiewicz M, Budick-Harmelin N, Macias M, Szybińska A, Mazur M, Kolmus K, Goryca K, Dąbrowska M, Paziewska A, Mikula M, Miączyńska M. ESCRT-I fuels lysosomal degradation to restrict TFEB/TFE3 signaling via the Rag-mTORC1 pathway. Life Sci Alliance 2022; 5:5/7/e202101239. [PMID: 35354596 PMCID: PMC8967991 DOI: 10.26508/lsa.202101239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/13/2022] Open
Abstract
ESCRT-I deficiency impairs lysosome membrane turnover and induces homeostatic responses to lysosomal nutrient starvation including activation of MiT-TFE signaling caused by inhibition of the substrate-specific mTORC1 pathway. Within the endolysosomal pathway in mammalian cells, ESCRT complexes facilitate degradation of proteins residing in endosomal membranes. Here, we show that mammalian ESCRT-I restricts the size of lysosomes and promotes degradation of proteins from lysosomal membranes, including MCOLN1, a Ca2+ channel protein. The altered lysosome morphology upon ESCRT-I depletion coincided with elevated expression of genes annotated to biogenesis of lysosomes due to prolonged activation of TFEB/TFE3 transcription factors. Lack of ESCRT-I also induced transcription of cholesterol biosynthesis genes, in response to inefficient delivery of cholesterol from endolysosomal compartments. Among factors that could possibly activate TFEB/TFE3 signaling upon ESCRT-I deficiency, we excluded lysosomal cholesterol accumulation and Ca2+-mediated dephosphorylation of TFEB/TFE3. However, we discovered that this activation occurs due to the inhibition of Rag GTPase–dependent mTORC1 pathway that specifically reduced phosphorylation of TFEB at S122. Constitutive activation of the Rag GTPase complex in cells lacking ESCRT-I restored S122 phosphorylation and prevented TFEB/TFE3 activation. Our results indicate that ESCRT-I deficiency evokes a homeostatic response to counteract lysosomal nutrient starvation, that is, improper supply of nutrients derived from lysosomal degradation.
Collapse
Affiliation(s)
- Marta Wróbel
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Jarosław Cendrowski
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Ewelina Szymańska
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | | | - Noga Budick-Harmelin
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Matylda Macias
- Microscopy and Cytometry Facility, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Aleksandra Szybińska
- Microscopy and Cytometry Facility, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Michał Mazur
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Krzysztof Kolmus
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Krzysztof Goryca
- Department of Genetics, Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Michalina Dąbrowska
- Department of Genetics, Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Agnieszka Paziewska
- Department of Gastroenterology, Hepatology and Clinical Oncology, Medical Center for Postgraduate Education, Warsaw, Poland
| | - Michał Mikula
- Department of Genetics, Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Marta Miączyńska
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| |
Collapse
|
18
|
Lysosomal ATP Transporter SLC17A9 Controls Cell Viability via Regulating Cathepsin D. Cells 2022; 11:cells11050887. [PMID: 35269509 PMCID: PMC8909234 DOI: 10.3390/cells11050887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 12/04/2022] Open
Abstract
SLC17A9 (solute carrier family 17 member 9) functions as an ATP transporter in lysosomes as well as other secretory vesicles. SLC17A9 inhibition or silence leads to cell death. However, the molecular mechanisms causing cell death are unclear. In this study, we report that cell death induced by SLC17A9 deficiency is rescued by the transcription factor EB (TFEB), a master gene for lysosomal protein expression, suggesting that SLC17A9 deficiency may be the main cause of lysosome dysfunction, subsequently leading to cell death. Interestingly, Cathepsin D, a lysosomal aspartic protease, is inhibited by SLC17A9 deficiency. Heterologous expression of Cathepsin D successfully rescues lysosomal dysfunction and cell death induced by SLC17A9 deficiency. On the other hand, the activity of Cathepsin B, a lysosomal cysteine protease, is not altered by SLC17A9 deficiency, and Cathepsin B overexpression does not rescue lysosomal dysfunction and cell death induced by SLC17A9 deficiency. Our data suggest that lysosomal ATP and SLC17A9 play critical roles in lysosomal function and cell viability by regulating Cathepsin D activity.
Collapse
|
19
|
Badr A, Eltobgy M, Krause K, Hamilton K, Estfanous S, Daily KP, Abu Khweek A, Hegazi A, Anne MNK, Carafice C, Robledo-Avila F, Saqr Y, Zhang X, Bonfield TL, Gavrilin MA, Partida-Sanchez S, Seveau S, Cormet-Boyaka E, Amer AO. CFTR Modulators Restore Acidification of Autophago-Lysosomes and Bacterial Clearance in Cystic Fibrosis Macrophages. Front Cell Infect Microbiol 2022; 12:819554. [PMID: 35252032 PMCID: PMC8890004 DOI: 10.3389/fcimb.2022.819554] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 01/19/2022] [Indexed: 12/17/2022] Open
Abstract
Cystic fibrosis (CF) human and mouse macrophages are defective in their ability to clear bacteria such as Burkholderia cenocepacia. The autophagy process in CF (F508del) macrophages is halted, and the underlying mechanism remains unclear. Furthermore, the role of CFTR in maintaining the acidification of endosomal and lysosomal compartments in CF cells has been a subject of debate. Using 3D reconstruction of z-stack confocal images, we show that CFTR is recruited to LC3-labeled autophagosomes harboring B. cenocepacia. Using several complementary approaches, we report that CF macrophages display defective lysosomal acidification and degradative function for cargos destined to autophagosomes, whereas non-autophagosomal cargos are effectively degraded within acidic compartments. Notably, treatment of CF macrophages with CFTR modulators (tezacaftor/ivacaftor) improved the autophagy flux, lysosomal acidification and function, and bacterial clearance. In addition, CFTR modulators improved CFTR function as demonstrated by patch-clamp. In conclusion, CFTR regulates the acidification of a specific subset of lysosomes that specifically fuse with autophagosomes. Therefore, our study describes a new biological location and function for CFTR in autophago-lysosomes and clarifies the long-standing discrepancies in the field.
Collapse
Affiliation(s)
- Asmaa Badr
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
- Clinical Pathology Department, College of Medicine, Mansoura University, Mansoura, Egypt
| | - Mostafa Eltobgy
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Kathrin Krause
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
- Max Planck Unit for the Science of Pathogens, Berlin, Germany
| | - Kaitlin Hamilton
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Shady Estfanous
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Kylene P. Daily
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Arwa Abu Khweek
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
- Department of Biology and Biochemistry, Birzeit University, West Bank, Palestine
| | - Ahmad Hegazi
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Midhun N. K. Anne
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Cierra Carafice
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Frank Robledo-Avila
- Center for Microbial Pathogenesis, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Youssra Saqr
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Xiaoli Zhang
- Center for Biostatistics, Ohio State University, Columbus, OH, United States
| | - Tracey L. Bonfield
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Mikhail A. Gavrilin
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Columbus, OH, United States
| | | | - Stephanie Seveau
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Estelle Cormet-Boyaka
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Amal O. Amer
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
20
|
Roney JC, Cheng XT, Sheng ZH. Neuronal endolysosomal transport and lysosomal functionality in maintaining axonostasis. J Cell Biol 2022; 221:213000. [PMID: 35142819 PMCID: PMC8932522 DOI: 10.1083/jcb.202111077] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 02/08/2023] Open
Abstract
Lysosomes serve as degradation hubs for the turnover of endocytic and autophagic cargos, which is essential for neuron function and survival. Deficits in lysosome function result in progressive neurodegeneration in most lysosomal storage disorders and contribute to the pathogenesis of aging-related neurodegenerative diseases. Given their size and highly polarized morphology, neurons face exceptional challenges in maintaining cellular homeostasis in regions far removed from the cell body where mature lysosomes are enriched. Neurons therefore require coordinated bidirectional intracellular transport to sustain efficient clearance capacity in distal axonal regions. Emerging lines of evidence have started to uncover mechanisms and signaling pathways regulating endolysosome transport and maturation to maintain axonal homeostasis, or “axonostasis,” that is relevant to a range of neurologic disorders. In this review, we discuss recent advances in how axonal endolysosomal trafficking, distribution, and lysosomal functionality support neuronal health and become disrupted in several neurodegenerative diseases.
Collapse
Affiliation(s)
- Joseph C Roney
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Xiu-Tang Cheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Zu-Hang Sheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| |
Collapse
|
21
|
Wang N, Hao Y, Feng X, Zhu H, Zhang D, Wang T, Cui X. Silicon-substituted rhodamines for stimulated emission depletion fluorescence nanoscopy. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.06.075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
22
|
Loss of Christianson Syndrome Na +/H + Exchanger 6 (NHE6) Causes Abnormal Endosome Maturation and Trafficking Underlying Lysosome Dysfunction in Neurons. J Neurosci 2021; 41:9235-9256. [PMID: 34526390 PMCID: PMC8570832 DOI: 10.1523/jneurosci.1244-20.2021] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/05/2021] [Accepted: 08/07/2021] [Indexed: 01/08/2023] Open
Abstract
Loss-of-function mutations in endosomal Na+/H+ exchanger 6 (NHE6) cause the X-linked neurologic disorder Christianson syndrome. Patients exhibit symptoms associated with both neurodevelopmental and neurodegenerative abnormalities. While loss of NHE6 has been shown to overacidify the endosome lumen, and is associated with endolysosome neuropathology, NHE6-mediated mechanisms in endosome trafficking and lysosome function have been understudied. Here, we show that NHE6-null mouse neurons demonstrate worsening lysosome function with time in culture, likely as a result of defective endosome trafficking. NHE6-null neurons exhibit overall reduced lysosomal proteolysis despite overacidification of the endosome and lysosome lumen. Akin to Nhx1 mutants in Saccharomyces cerevisiae, we observe decreased endosome-lysosome fusion in NHE6-null neurons. Also, we find premature activation of pH-dependent cathepsin D (CatD) in endosomes. While active CatD is increased in endosomes, CatD activation and CatD protein levels are reduced in the lysosome. Protein levels of another mannose 6-phosphate receptor (M6PR)-dependent enzyme, β-N-acetylglucosaminidase, were also decreased in lysosomes of NHE6-null neurons. M6PRs accumulate in late endosomes, suggesting defective M6PR recycling and retromer function in NHE6-null neurons. Finally, coincident with decreased endosome-lysosome fusion, using total internal reflection fluorescence, we also find a prominent increase in fusion between endosomal multivesicular bodies and the plasma membrane, indicating enhanced exosome secretion from NHE6-null neurons. In summary, in addition to overacidification of endosomes and lysosomes, loss of NHE6 leads to defects in endosome maturation and trafficking, including enhanced exosome release, contributing to lysosome deficiency and potentially leading to neurodegenerative disease. SIGNIFICANCE STATEMENT Loss-of-function mutations in the endosomal Na+/H+ exchanger 6 (NHE6) cause Christianson syndrome, an X-linked neurologic disorder. Loss of NHE6 has been shown to overacidify endosomes; however, endosome trafficking mechanisms have been understudied, and the mechanisms leading to neurodegeneration are largely unknown. In NHE6-null mouse neurons in vitro, we find worsening lysosome function with days in culture. Notably, pH-dependent lysosome enzymes, such as cathepsin D, have reduced activity in lysosomes yet increased, precocious activity in endosomes in NHE6-null neurons. Further, endosomes show reduced fusion to lysosomes, and increased fusion to the plasma membrane with increased exosome release. This study identifies new mechanisms involving defective endosome maturation and trafficking that impair lysosome function in Christianson syndrome, likely contributing to neurodegeneration.
Collapse
|
23
|
Liu K, Kong L, Graham DB, Carey KL, Xavier RJ. SAC1 regulates autophagosomal phosphatidylinositol-4-phosphate for xenophagy-directed bacterial clearance. Cell Rep 2021; 36:109434. [PMID: 34320354 PMCID: PMC8327279 DOI: 10.1016/j.celrep.2021.109434] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/21/2020] [Accepted: 07/01/2021] [Indexed: 02/07/2023] Open
Abstract
Phosphoinositides are important molecules in lipid signaling, membrane identity, and trafficking that are spatiotemporally controlled by factors from both mammalian cells and intracellular pathogens. Here, using small interfering RNA (siRNA) directed against phosphoinositide kinases and phosphatases, we screen for regulators of the host innate defense response to intracellular bacterial replication. We identify SAC1, a transmembrane phosphoinositide phosphatase, as an essential regulator of xenophagy. Depletion or inactivation of SAC1 compromises fusion between Salmonella-containing autophagosomes and lysosomes, leading to increased bacterial replication. Mechanistically, the loss of SAC1 results in aberrant accumulation of phosphatidylinositol-4-phosphate [PI(4)P] on Salmonella-containing autophagosomes, thus facilitating recruitment of SteA, a PI(4)P-binding Salmonella effector protein, which impedes lysosomal fusion. Replication of Salmonella lacking SteA is suppressed by SAC-1-deficient cells, however, demonstrating bacterial adaptation to xenophagy. Our findings uncover a paradigm in which a host protein regulates the level of its substrate and impairs the function of a bacterial effector during xenophagy.
Collapse
Affiliation(s)
- Kai Liu
- Center for Computational and Integrative Biology, Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lingjia Kong
- Center for Computational and Integrative Biology, Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Daniel B Graham
- Center for Computational and Integrative Biology, Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Ramnik J Xavier
- Center for Computational and Integrative Biology, Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
24
|
Born JR, Chenniappan VK, Davis DP, Dahlin JL, Marugan JJ, Patnaik S. The Impact of Assay Design on Medicinal Chemistry: Case Studies. SLAS DISCOVERY 2021; 26:1243-1255. [PMID: 34225522 DOI: 10.1177/24725552211026238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
GRAPHICAL ABSTRACT
Collapse
Affiliation(s)
- Joshua R Born
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Vinoth Kumar Chenniappan
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Danielle P Davis
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Jayme L Dahlin
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Juan J Marugan
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Samarjit Patnaik
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
25
|
Grimm F, Rehman J, Stoldt S, Khan TA, Schlötel JG, Nizamov S, John M, Belov VN, Hell SW. Rhodamines with a Chloronicotinic Acid Fragment for Live Cell Superresolution STED Microscopy*. Chemistry 2021; 27:6070-6076. [PMID: 33496998 PMCID: PMC8048976 DOI: 10.1002/chem.202005134] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Indexed: 12/22/2022]
Abstract
Formylation of 2,6-dichloro-5-R-nicotinic acids at C-4 followed by condensation with 3-hydroxy-N,N-dimethylaniline gave analogs of the popular TAMRA fluorescent dye with a 2,6-dichloro-5-R-nicotinic acid residues (R=H, F). The following reaction with thioglycolic acid is selective, involves only one chlorine atom at the carbon between pyridine nitrogen and the carboxylic acid group and affords new rhodamine dyes absorbing at 564/ 573 nm and emitting at 584/ 597 nm (R=H/ F, in aq. PBS). Conjugates of the dyes with "small molecules" provided specific labeling (covalent and non-covalent) of organelles as well as of components of the cytoskeleton in living cells and were combined with fluorescent probes prepared from 610CP and SiR dyes and applied in two-color STED microscopy with a 775 nm STED laser.
Collapse
Affiliation(s)
- Florian Grimm
- Abberior GmbHHans Adolf Krebs Weg 137077GöttingenGermany
| | - Jasmin Rehman
- Abberior GmbHHans Adolf Krebs Weg 137077GöttingenGermany
| | - Stefan Stoldt
- Department of NanobiophotonicsMax Planck Institute for Biophysical Chemistry (MPIBPC)Am Fassberg 1137077GöttingenGermany
| | - Taukeer A. Khan
- Department of NanobiophotonicsMax Planck Institute for Biophysical Chemistry (MPIBPC)Am Fassberg 1137077GöttingenGermany
| | - Jan Gero Schlötel
- Abberior-Instruments GmbHHans Adolf Krebs Weg 137077GöttingenGermany
| | - Shamil Nizamov
- Abberior GmbHHans Adolf Krebs Weg 137077GöttingenGermany
| | - Michael John
- Institute of Organic and Biomolecular ChemistryGeorg-August UniversityTammannstr. 237077GöttingenGermany
| | - Vladimir N. Belov
- Department of NanobiophotonicsMax Planck Institute for Biophysical Chemistry (MPIBPC)Am Fassberg 1137077GöttingenGermany
| | - Stefan W. Hell
- Department of NanobiophotonicsMax Planck Institute for Biophysical Chemistry (MPIBPC)Am Fassberg 1137077GöttingenGermany
| |
Collapse
|
26
|
Yin Q, Jian Y, Xu M, Huang X, Wang N, Liu Z, Li Q, Li J, Zhou H, Xu L, Wang Y, Yang C. CDK4/6 regulate lysosome biogenesis through TFEB/TFE3. J Cell Biol 2021; 219:151944. [PMID: 32662822 PMCID: PMC7401801 DOI: 10.1083/jcb.201911036] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 04/15/2020] [Accepted: 05/06/2020] [Indexed: 12/14/2022] Open
Abstract
Lysosomes are degradation and signaling organelles that adapt their biogenesis to meet many different cellular demands; however, it is unknown how lysosomes change their numbers for cell division. Here, we report that the cyclin-dependent kinases CDK4/6 regulate lysosome biogenesis during the cell cycle. Chemical or genetic inactivation of CDK4/6 increases lysosomal numbers by activating the lysosome and autophagy transcription factors TFEB and TFE3. CDK4/6 interact with and phosphorylate TFEB/TFE3 in the nucleus, thereby inactivating them by promoting their shuttling to the cytoplasm. During the cell cycle, lysosome numbers increase in S and G2/M phases when cyclin D turnover diminishes CDK4/6 activity. These findings not only uncover the molecular events that direct the nuclear export of TFEB/TFE3, but also suggest a mechanism that controls lysosome biogenesis in the cell cycle. CDK4/6 inhibitors promote autophagy and lysosome-dependent degradation, which has important implications for the therapy of cancer and lysosome-related disorders.
Collapse
Affiliation(s)
- Qiuyuan Yin
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan and Center for Life Science, School of Life Sciences, Yunnan University, Kunming, China
| | - Youli Jian
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Meng Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Xiahe Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Niya Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Zhifang Liu
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan and Center for Life Science, School of Life Sciences, Yunnan University, Kunming, China
| | - Qian Li
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan and Center for Life Science, School of Life Sciences, Yunnan University, Kunming, China
| | - Jinglin Li
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan and Center for Life Science, School of Life Sciences, Yunnan University, Kunming, China
| | - Hejiang Zhou
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan and Center for Life Science, School of Life Sciences, Yunnan University, Kunming, China
| | - Lin Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Chonglin Yang
- State Key Laboratory of Conservation and Utilization of Bio-Resources in Yunnan and Center for Life Science, School of Life Sciences, Yunnan University, Kunming, China
| |
Collapse
|
27
|
Wu K, Seylani A, Wu J, Wu X, Bleck CK, Sack MN. BLOC1S1/GCN5L1/BORCS1 is a critical mediator for the initiation of autolysosomal tubulation. Autophagy 2021; 17:3707-3724. [DOI: 10.1080/15548627.2021.1894759] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- Kaiyuan Wu
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, National Institutes of Health, Bethesda, Maryland
| | - Allen Seylani
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, National Institutes of Health, Bethesda, Maryland
| | - Jing Wu
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, National Institutes of Health, Bethesda, Maryland
| | - Xufeng Wu
- Light Microscopy Core, National Heart, Lung and Blood Institute, NIH, Bethesda, USA
| | - Christopher K.E Bleck
- Electron Microcopy Core, National Heart, Lung and Blood Institute, NIH, Bethesda, USA
| | - Michael N Sack
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
28
|
Kim D, Hwang HY, Ji ES, Kim JY, Yoo JS, Kwon HJ. Activation of mitochondrial TUFM ameliorates metabolic dysregulation through coordinating autophagy induction. Commun Biol 2021; 4:1. [PMID: 33398033 PMCID: PMC7782552 DOI: 10.1038/s42003-020-01566-0] [Citation(s) in RCA: 253] [Impact Index Per Article: 63.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 12/02/2020] [Indexed: 12/14/2022] Open
Abstract
Disorders of autophagy, a key regulator of cellular homeostasis, cause a number of human diseases. Due to the role of autophagy in metabolic dysregulation, there is a need to identify autophagy regulators as therapeutic targets. To address this need, we conducted an autophagy phenotype-based screen and identified the natural compound kaempferide (Kaem) as an autophagy enhancer. Kaem promoted autophagy through translocation of transcription factor EB (TFEB) without MTOR perturbation, suggesting it is safe for administration. Moreover, Kaem accelerated lipid droplet degradation in a lysosomal activity-dependent manner in vitro and ameliorated metabolic dysregulation in a diet-induced obesity mouse model. To elucidate the mechanism underlying Kaem’s biological activity, the target protein was identified via combined drug affinity responsive target stability and LC–MS/MS analyses. Kaem directly interacted with the mitochondrial elongation factor TUFM, and TUFM absence reversed Kaem-induced autophagy and lipid degradation. Kaem also induced mitochondrial reactive oxygen species (mtROS) to sequentially promote lysosomal Ca2+ efflux, TFEB translocation and autophagy induction, suggesting a role of TUFM in mtROS regulation. Collectively, these results demonstrate that Kaem is a potential therapeutic candidate/chemical tool for treating metabolic dysregulation and reveal a role for TUFM in autophagy for metabolic regulation with lipid overload. Kim, Hwang et al. use in vitro and in vivo models of autophagy disorder/metabolic dysfunction to show that in this context, the natural compound kaempferide is an autophagy enhancer and reveal that one of the underlying mechanisms governing this is mediated by the mitochondrial elongation factor TUFM. This insight may have therapeutic value in the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Dasol Kim
- Chemical Genomics Global Research Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hui-Yun Hwang
- Chemical Genomics Global Research Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Eun Sun Ji
- Biomedical Omics Group, Korea Basic Science Institute, Ochang, Chungbuk, 28119, Republic of Korea
| | - Jin Young Kim
- Biomedical Omics Group, Korea Basic Science Institute, Ochang, Chungbuk, 28119, Republic of Korea
| | - Jong Shin Yoo
- Biomedical Omics Group, Korea Basic Science Institute, Ochang, Chungbuk, 28119, Republic of Korea
| | - Ho Jeong Kwon
- Chemical Genomics Global Research Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
29
|
Farfel-Becker T, Roney JC, Cheng XT, Li S, Cuddy SR, Sheng ZH. Neuronal Soma-Derived Degradative Lysosomes Are Continuously Delivered to Distal Axons to Maintain Local Degradation Capacity. Cell Rep 2020; 28:51-64.e4. [PMID: 31269450 PMCID: PMC6696943 DOI: 10.1016/j.celrep.2019.06.013] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/12/2019] [Accepted: 06/04/2019] [Indexed: 12/19/2022] Open
Abstract
Neurons face the challenge of maintaining cellular homeostasis through lysosomal degradation. While enzymatically active degradative lysosomes are enriched in the soma, their axonal trafficking and positioning and impact on axonal physiology remain elusive. Here, we characterized axon-targeted delivery of degradative lysosomes by applying fluorescent probes that selectively label active forms of lysosomal cathepsins D, B, L, and GCase. By time-lapse imaging of cortical neurons in microfluidic devices and standard dishes, we reveal that soma-derived degradative lysosomes rapidly influx into distal axons and target to autophagosomes and Parkinson disease-related α-synuclein cargos for local degradation. Impairing lysosome axonal delivery induces an aberrant accumulation of autophagosomes and α-synuclein cargos in distal axons. Our study demonstrates that the axon is an active compartment for local degradation and reveals fundamental aspects of axonal lysosomal delivery and maintenance. Our work establishes a foundation for investigations into axonal lysosome trafficking and functionality in neurodegenerative diseases.
Collapse
Affiliation(s)
- Tamar Farfel-Becker
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA
| | - Joseph C Roney
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA
| | - Xiu-Tang Cheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA
| | - Sunan Li
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA
| | - Sean R Cuddy
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA
| | - Zu-Hang Sheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA.
| |
Collapse
|
30
|
Wen X, An P, Li H, Zhou Z, Sun Y, Wang J, Ma L, Lu B. Tau Accumulation via Reduced Autophagy Mediates GGGGCC Repeat Expansion-Induced Neurodegeneration in Drosophila Model of ALS. Neurosci Bull 2020; 36:1414-1428. [PMID: 32500377 DOI: 10.1007/s12264-020-00518-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 03/21/2020] [Indexed: 12/21/2022] Open
Abstract
Expansions of trinucleotide or hexanucleotide repeats lead to several neurodegenerative disorders, including Huntington disease [caused by expanded CAG repeats (CAGr) in the HTT gene], and amyotrophic lateral sclerosis [ALS, possibly caused by expanded GGGGCC repeats (G4C2r) in the C9ORF72 gene], of which the molecular mechanisms remain unclear. Here, we demonstrated that lowering the Drosophila homologue of tau protein (dtau) significantly rescued in vivo neurodegeneration, motor performance impairments, and the shortened life-span in Drosophila expressing expanded CAGr or expanded G4C2r. Expression of human tau (htau4R) restored the disease-related phenotypes that had been mitigated by the loss of dtau, suggesting an evolutionarily-conserved role of tau in neurodegeneration. We further revealed that G4C2r expression increased tau accumulation by inhibiting autophagosome-lysosome fusion, possibly due to lowering the level of BAG3, a regulator of autophagy and tau. Taken together, our results reveal a novel mechanism by which expanded G4C2r causes neurodegeneration via an evolutionarily-conserved mechanism. Our findings provide novel autophagy-related mechanistic insights into C9ORF72-ALS and possible entry points to disease treatment.
Collapse
Affiliation(s)
- Xue Wen
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Ping An
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Hexuan Li
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Zijian Zhou
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Yimin Sun
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Jian Wang
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai, 200438, China.
| | - Lixiang Ma
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| | - Boxun Lu
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, School of Life Sciences, Fudan University, Shanghai, 200438, China.
| |
Collapse
|
31
|
Han J, Goldstein LA, Hou W, Watkins SC, Rabinowich H. Involvement of CASP9 (caspase 9) in IGF2R/CI-MPR endosomal transport. Autophagy 2020; 17:1393-1409. [PMID: 32397873 DOI: 10.1080/15548627.2020.1761742] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Recently, we reported that increased expression of CASP9 pro-domain, at the endosomal membrane in response to HSP90 inhibition, mediates a cell-protective effect that does not involve CASP9 apoptotic activity. We report here that a non-apoptotic activity of endosomal membrane CASP9 facilitates the retrograde transport of IGF2R/CI-MPR from the endosomes to the trans-Golgi network, indicating the involvement of CASP9 in endosomal sorting and lysosomal biogenesis. CASP9-deficient cells demonstrate the missorting of CTSD (cathepsin D) and other acid hydrolases, accumulation of late endosomes, and reduced degradation of bafilomycin A1-sensitive proteins. In the absence of CASP9, IGF2R undergoes significant degradation, and its rescue is achieved by the re-expression of a non-catalytic CASP9 mutant. This endosomal activity of CASP9 is potentially mediated by herein newly identified interactions of CASP9 with the components of the endosomal membrane transport complexes. These endosomal complexes include the retromer VPS35 and the SNX dimers, SNX1-SNX5 and SNX2-SNX6, which are involved in the IGF2R retrieval mechanism. Additionally, CASP9 interacts with HGS/HRS/ESCRT-0 and the CLTC (clathrin heavy chain) that participate in the initiation of the endosomal ESCRT degradation pathway. We propose that endosomal CASP9 inhibits the endosomal membrane degradative subdomain(s) from initiating the ESCRT-mediated degradation of IGF2R, allowing its retrieval to transport-designated endosomal membrane subdomain(s). These findings are the first to identify a cell survival, non-apoptotic function for CASP9 at the endosomal membrane, a site distinctly removed from the cytoplasmic apoptosome. Via its non-apoptotic endosomal function, CASP9 impacts the retrograde transport of IGF2R and, consequently, lysosomal biogenesis.Abbreviations: ACTB: actin beta; ATG7: autophagy related 7; BafA1: bafilomycin A1; CASP: caspase; CLTC/CHC: clathrin, heavy chain; CTSD: cathepsin D; ESCRT: endosomal sorting complexes required for transport; HEXB: hexosaminidase subunit beta; HGS/HRS/ESCRT-0: hepatocyte growth factor-regulated tyrosine kinase substrate; IGF2R/CI-MPR: insulin like growth factor 2 receptor; ILV: intraluminal vesicles; KD: knockdown; KO: knockout; M6PR/CD-MPR: mannose-6-phosphate receptor, cation dependent; MEF: murine embryonic fibroblasts; MWU: Mann-Whitney U test; PepA: pepstatin A; RAB7A: RAB7, member RAS oncogene family; SNX-BAR: sorting nexin dimers with a Bin/Amphiphysin/Rvs (BAR) domain each; TGN: trans-Golgi network; TUBB: tubulin beta; VPS26: VPS26 retromer complex component; VPS29: VPS29 retromer complex component; VPS35: VPS35 retromer complex component.
Collapse
Affiliation(s)
- Jie Han
- Departments of Pathology, University of Pittsburgh School of Medicine and the University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Leslie A Goldstein
- Departments of Pathology, University of Pittsburgh School of Medicine and the University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Wen Hou
- Departments of Pathology, University of Pittsburgh School of Medicine and the University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Simon C Watkins
- Cell Biology, University of Pittsburgh School of Medicine and the University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Hannah Rabinowich
- Departments of Pathology, University of Pittsburgh School of Medicine and the University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
32
|
Shao Q, Yang M, Liang C, Ma L, Zhang W, Jiang Z, Luo J, Lee JK, Liang C, Chen JF. C9orf72 and smcr8 mutant mice reveal MTORC1 activation due to impaired lysosomal degradation and exocytosis. Autophagy 2019; 16:1635-1650. [PMID: 31847700 DOI: 10.1080/15548627.2019.1703353] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
How lysosome and MTORC1 signaling interact remains elusive in terminally differentiated cells. A G4C2 repeat expansion in C9orf72 is the most common cause of familial amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) (C9ALS-FTD). We previously identified a C9orf72-SMCR8-containing complex. Here we found that c9orf72 and smcr8 double-knockout (dKO) mice exhibit similar but more severe immune defects than the individual knockouts. In c9orf72 or smcr8 mutant macrophages, lysosomal degradation and exocytosis were impaired due to the disruption of autolysosome acidification. As a result of impaired lysosomal degradation, MTOR protein was aberrantly increased, resulting in MTORC1 signaling overactivation. Inhibition of hyperactive MTORC1 partially rescued macrophage dysfunction, splenomegaly and lymphadenopathy in c9orf72 or smcr8 mutant mice. Pharmacological inhibition of lysosomal degradation upregulated MTOR protein and MTORC1 signaling in differentiated wild-type macrophages, which resemble phenotypes in KO mice. In contrast, C9orf72 or Smcr8 depletion in proliferating macrophages decreased MTORC1 signaling. Our studies causatively link C9orf72-SMCR8's cellular functions in lysosomal degradation, exocytosis, and MTORC1 signaling with their organism-level immune regulation, suggesting cell state (proliferation vs. differentiation)-dependent regulation of MTOR signaling via lysosomes.Abbreviations: ALS: amyotrophic lateral sclerosis; ATG13: autophagy related 13; BMDMs: bone marrow-derived macrophages; BafA1: bafilomycin A1; C9orf72: C9orf72, member of C9orf72-SMCR8 complex; CD68: CD68 antigen; ConA: concanamycin A; dKO: double knockout; DENN: differentially expressed in normal and neoplastic cells; FTD: frontotemporal dementia; GEF: guanine nucleotide exchange factor; IFNB1: interferon beta 1, fibroblast; IFNG: interferon gamma; IL1B/IL-1β: interleukin 1 beta; IL6: interleukin 6; iPSCs: induced pluripotent stem cells; LAMP1: lysosomal-associated membrane protein 1; LPOs: LAMP1-positive organelles; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; LPS: lipopolysaccharide; MTORC1: mechanistic target of rapamycin kinase complex 1; MEFs: mouse embryonic fibroblasts; MNs: motor neurons; NOS2/iNOS: nitric oxide synthase 2, inducible; RAN: repeat-associated non-AUG; RB1CC1/FIP200: RB1-inducible coiled-coil 1; RPS6/S6: ribosomal protein S6; RPS6KB1/S6K1: ribosomal protein S6 kinase, polypeptide 1; SMCR8: Smith-Magenis syndrome chromosome region, candidate 8; SQSTM1/p62: sequestosome 1; TFEB: transcription factor EB; TNF: tumor necrosis factor; TSC1: TSC complex subunit 1; ULK1: unc-51 like kinase 1; v-ATPase: vacuolar-type H⁺-translocating ATPase.
Collapse
Affiliation(s)
- Qiang Shao
- Center for Craniofacial Molecular Biology, University of Southern California , Los Angeles, CA, USA
| | - Mei Yang
- Center for Craniofacial Molecular Biology, University of Southern California , Los Angeles, CA, USA.,Center for Life Sciences, School of Life Sciences, Yunnan University , Kunming, P.R. China
| | - Chen Liang
- Department of Cellular Biology, University of Georgia , Athens, GA, USA
| | - Li Ma
- Center for Craniofacial Molecular Biology, University of Southern California , Los Angeles, CA, USA
| | - Wei Zhang
- Center for Craniofacial Molecular Biology, University of Southern California , Los Angeles, CA, USA
| | - Zhiwen Jiang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research, Guangdong Medical University , Dongguan, Guangdong, P.R. China
| | - Jun Luo
- Division of VIP Center, Stomatological Hospital of Chongqing Medical University , Chongqing, P.R. China
| | - Jae-Kyung Lee
- Department of Physiology and Pharmacology, University of Georgia , Athens, GA, USA
| | - Chengyu Liang
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California , Los Angeles, CA, USA
| | - Jian-Fu Chen
- Center for Craniofacial Molecular Biology, University of Southern California , Los Angeles, CA, USA
| |
Collapse
|
33
|
Altered γ-Secretase Processing of APP Disrupts Lysosome and Autophagosome Function in Monogenic Alzheimer's Disease. Cell Rep 2019; 25:3647-3660.e2. [PMID: 30590039 PMCID: PMC6315085 DOI: 10.1016/j.celrep.2018.11.095] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/02/2018] [Accepted: 11/27/2018] [Indexed: 01/01/2023] Open
Abstract
Abnormalities of the endolysosomal and autophagy systems are found in Alzheimer’s disease, but it is not clear whether defects in these systems are a cause or consequence of degenerative processes in the disease. In human neuronal models of monogenic Alzheimer’s disease, APP and PSEN1 mutations disrupt lysosome function and autophagy, leading to impaired lysosomal proteolysis and defective autophagosome clearance. Processing of APP by γ-secretase is central to the pathogenic changes in the lysosome-autophagy system caused by PSEN1 and APP mutations: reducing production of C-terminal APP by inhibition of BACE1 rescued these phenotypes in both APP and PSEN1 mutant neurons, whereas inhibition of γ-secretase induced lysosomal and autophagic pathology in healthy neurons. Defects in lysosomes and autophagy due to PSEN1 mutations are rescued by CRISPR-knockout of APP. These data demonstrate a key role for proteolysis of the C-terminal of APP by γ-secretase in neuronal dysfunction in monogenic Alzheimer’s disease. APP and PSEN1 mutant neurons have deficits in lysosome proteolysis BACE1 inhibition rescues lysosome and autophagy defects PSEN1 mutant phenotypes are rescued by genetic deletion of APP Lysosome and autophagy defects are causes of neuronal dysfunction in AD
Collapse
|
34
|
Miller HE, Hoyt FH, Heinzen RA. Replication of Coxiella burnetii in a Lysosome-Like Vacuole Does Not Require Lysosomal Hydrolases. Infect Immun 2019; 87:e00493-19. [PMID: 31405956 PMCID: PMC6803326 DOI: 10.1128/iai.00493-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/06/2019] [Indexed: 01/03/2023] Open
Abstract
Coxiella burnetii is an intracellular bacterium that causes query, or Q fever, a disease that typically manifests as a severe flu-like illness. The initial target of C. burnetii is the alveolar macrophage. Here, it regulates vesicle trafficking pathways and fusion events to establish a large replication vacuole called the Coxiella-containing vacuole (CCV). Similar to a phagolysosome, the CCV has an acidic pH and contains lysosomal hydrolases obtained via fusion with late endocytic vesicles. Lysosomal hydrolases break down various lipids, carbohydrates, and proteins; thus, it is assumed C. burnetii derives nutrients for growth from these degradation products. To investigate this possibility, we utilized a GNPTAB-/- HeLa cell line that lacks lysosomal hydrolases in endocytic compartments. Unexpectedly, examination of C. burnetii growth in GNPTAB-/- HeLa cells revealed replication and viability are not impaired, indicating C. burnetii does not require by-products of hydrolase degradation to survive and grow in the CCV. However, although bacterial growth was normal, CCVs were abnormal, appearing dark and condensed rather than clear and spacious. Lack of degradation within CCVs allowed waste products to accumulate, including intraluminal vesicles, autophagy protein LC3, and cholesterol. The build-up of waste products coincided with an altered CCV membrane, where LAMP1 was decreased and CD63 and LAMP1 redistributed from a punctate to uniform localization. This disruption of CCV membrane organization may account for the decreased CCV size due to impaired fusion with late endocytic vesicles. Collectively, these results demonstrate lysosomal hydrolases are not required for C. burnetii survival and growth but are needed for normal CCV development. These data provide insight into mechanisms of CCV biogenesis while raising the important question of how C. burnetii obtains essential nutrients from its host.
Collapse
Affiliation(s)
- Heather E Miller
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Forrest H Hoyt
- Electron Microscopy Unit, Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Robert A Heinzen
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| |
Collapse
|
35
|
Grimm F, Nizamov S, Belov VN. Green-Emitting Rhodamine Dyes for Vital Labeling of Cell Organelles Using STED Super-Resolution Microscopy. Chembiochem 2019; 20:2248-2254. [PMID: 31050112 DOI: 10.1002/cbic.201900177] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/02/2019] [Indexed: 12/12/2022]
Abstract
Fluorescence microscopy reveals the localization, spatial distribution, and temporal dynamics of the specifically labeled organelles in living cells. Labeling with exogenous conjugates prepared from fluorescent dyes and small molecules (ligands) is an attractive alternative to the use of fluorescent proteins, but proved to be challenging due to insufficient cell-permeability of the probes, unspecific staining, or low dye brightness. We evaluated four green-emitting rhodamine dyes and their conjugates intended for the specific labeling of lysosomes, mitochondria, tubulin, and actin in living cells. The imaging performance of the probes in living human fibroblasts has been studied by using confocal and stimulated emission depletion (STED) super-resolution microscopy with a commercial 595 nm STED laser. Two bright and photostable dyes (LIVE 510 and LIVE 515) provide specific and versatile staining.
Collapse
Affiliation(s)
- Florian Grimm
- Abberior GmbH, Hans-Adolf-Krebs-Weg 1, 37077, Göttingen, Germany
| | - Shamil Nizamov
- Abberior GmbH, Hans-Adolf-Krebs-Weg 1, 37077, Göttingen, Germany
| | - Vladimir N Belov
- Department of Nanobiophotonics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| |
Collapse
|
36
|
Sharma G, Guardia CM, Roy A, Vassilev A, Saric A, Griner LN, Marugan J, Ferrer M, Bonifacino JS, DePamphilis ML. A family of PIKFYVE inhibitors with therapeutic potential against autophagy-dependent cancer cells disrupt multiple events in lysosome homeostasis. Autophagy 2019; 15:1694-1718. [PMID: 30806145 DOI: 10.1080/15548627.2019.1586257] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
High-throughput screening identified 5 chemical analogs (termed the WX8-family) that disrupted 3 events in lysosome homeostasis: (1) lysosome fission via tubulation without preventing homotypic lysosome fusion; (2) trafficking of molecules into lysosomes without altering lysosomal acidity, and (3) heterotypic fusion between lysosomes and autophagosomes. Remarkably, these compounds did not prevent homotypic fusion between lysosomes, despite the fact that homotypic fusion required some of the same machinery essential for heterotypic fusion. These effects varied 400-fold among WX8-family members, were time and concentration dependent, reversible, and resulted primarily from their ability to bind specifically to the PIKFYVE phosphoinositide kinase. The ability of the WX8-family to prevent lysosomes from participating in macroautophagy/autophagy suggested they have therapeutic potential in treating autophagy-dependent diseases. In fact, the most potent family member (WX8) was 100-times more lethal to 'autophagy-addicted' melanoma A375 cells than the lysosomal inhibitors hydroxychloroquine and chloroquine. In contrast, cells that were insensitive to hydroxychloroquine and chloroquine were also insensitive to WX8. Therefore, the WX8-family of PIKFYVE inhibitors provides a basis for developing drugs that could selectively kill autophagy-dependent cancer cells, as well as increasing the effectiveness of established anti-cancer therapies through combinatorial treatments. Abbreviations: ACTB: actin beta; Baf: bafilomycin A1; BECN1: beclin 1; BODIPY: boron-dipyrromethene; BORC: BLOC-1 related complex; BRAF: B-Raf proto-oncogene, serine/threonine kinase; BSA: bovine serum albumin; CTSD: cathepsin D; CQ: chloroquine; DNA: deoxyribonucleic acid; EC50: half maximal effective concentration; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; HCQ: hydroxychloroquine; HOPS complex: homotypic fusion and protein sorting complex; Kd: equilibrium binding constant; IC50: half maximal inhibitory concentration; KO: knockout; LAMP1: lysosomal associated membrane protein 1; MAP1LC3A: microtubule associated protein 1 light chain 3 alpha; MES: 2-(N-morpholino)ethanesulphonic acid; MTOR: mechanistic target of rapamycin kinase; μM: micromolar; NDF: 3-methylbenzaldehyde (2,6-dimorpholin-4-ylpyrimidin-4-yl)hydrazine;NEM: N-ethylmaleimide; NSF: N-ethylmaleimide sensitive factor; PBS: phosphate-buffered saline; PIKFYVE: phosphoinositide kinase, FYVE-type zinc finger containing; PIP4K2C: phosphatidylinositol-5-phosphate 4-kinase type 2 gamma; PtdIns3P: phosphatidylinositol 3-phosphate; PtdIns(3,5)P2: phosphatidylinositol 3,5-biphosphate; RFP: red fluorescent protein; RPS6: ribosomal protein S6; RPS6KB1: ribosomal protein S6 kinase B1; SQSTM1: sequestosome 1; TWEEN 20: polysorbate 20; V-ATPase: vacuolar-type H+-translocating ATPase; VPS39: VPS39 subunit of HOPS complex; VPS41: VPS41 subunit of HOPS complex; WWL: benzaldehyde [2,6-di(4-morpholinyl)-4-pyrimidinyl]hydrazone; WX8: 1H-indole-3-carbaldehyde [4-anilino-6-(4-morpholinyl)-1,3,5-triazin-2-yl]hydrazine; XBA: N-(3-chloro-4-fluorophenyl)-4,6-dimorpholino-1,3,5-triazin-2-amine hydrochloride; XB6: N-(4-ethylphenyl)-4,6-dimorpholino-1,3,5-triazin-2-amine hydrochloride.
Collapse
Affiliation(s)
- Gaurav Sharma
- Division of Developmental Biology, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Carlos M Guardia
- Cell Biology and Neurobiology Branch, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Ajit Roy
- Division of Developmental Biology, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Alex Vassilev
- Division of Developmental Biology, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Amra Saric
- Cell Biology and Neurobiology Branch, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Lori N Griner
- Division of Developmental Biology, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Juan Marugan
- Division of Pre-Clinical Innovation, NIH Chemical Genomics Center, National Center for Advancing Translational Sciences, National Institutes of Health , Rockville , MD , USA
| | - Marc Ferrer
- Division of Pre-Clinical Innovation, NIH Chemical Genomics Center, National Center for Advancing Translational Sciences, National Institutes of Health , Rockville , MD , USA
| | - Juan S Bonifacino
- Cell Biology and Neurobiology Branch, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| | - Melvin L DePamphilis
- Division of Developmental Biology, National Institute of Child Health & Human Development, National Institutes of Health , Bethesda , MD , USA
| |
Collapse
|
37
|
Zhao X, Wei S, Li Z, Lin C, Zhu Z, Sun D, Bai R, Qian J, Gao X, Chen G, Xu Z. Autophagic flux blockage in alveolar epithelial cells is essential in silica nanoparticle-induced pulmonary fibrosis. Cell Death Dis 2019; 10:127. [PMID: 30755584 PMCID: PMC6372720 DOI: 10.1038/s41419-019-1340-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/18/2018] [Accepted: 01/07/2019] [Indexed: 01/02/2023]
Abstract
Silica nanoparticles (SiNPs) have been reported to induce pulmonary fibrosis (PF) with an unknown mechanism. Recently, the activation of autophagy, a lysosome-dependent cell degradation pathway, by SiNPs has been identified in alveolar epithelial cells (AECs). However, the underlying mechanism and the relevance of SiNPs-induced autophagy to the development of PF remain elusive. Here, we report that autophagy dysfunction and subsequent apoptosis in AECs are involved in SiNPs-induced PF. SiNPs engulfed by AECs enhance autophagosome accumulation and apoptosis both in vivo and in vitro. Mechanically, SiNPs block autophagy flux through impairing lysosomal degradation via acidification inhibition. Lysosomal reacidification by cyclic-3',5'-adenosine monophosphate (cAMP) significantly enhances autophagic degradation and attenuate apoptosis. Importantly, enhancement of autophagic degradation by rapamycin protects AECs from apoptosis and attenuates SiNPs-induced PF in the mouse model. Altogether, our data demonstrate a repressive effect of SiNPs on lysosomal acidification, contributing to the decreased autophagic degradation in AECs, thus leading to apoptosis and subsequent PF. These findings may provide an improved understanding of SiNPs-induced PF and molecular targets to antagonize it.
Collapse
Affiliation(s)
- Xinyuan Zhao
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong Unversity, Nantong, 226019, China
| | - Saisai Wei
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Zhijian Li
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Chen Lin
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Zhenfeng Zhu
- State Key Laboratory of Modern Optical Instrumentation, Centre for Optical and Electromagnetic Research, JORCEP (Sino-Swedish Joint Research Center of Photonics), Zhejiang University, Hangzhou, 310058, China
| | - Desen Sun
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Rongpan Bai
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jun Qian
- State Key Laboratory of Modern Optical Instrumentation, Centre for Optical and Electromagnetic Research, JORCEP (Sino-Swedish Joint Research Center of Photonics), Zhejiang University, Hangzhou, 310058, China
| | - Xiangwei Gao
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Guangdi Chen
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Zhengping Xu
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
38
|
Li Y, Zhang Y, Gan Q, Xu M, Ding X, Tang G, Liang J, Liu K, Liu X, Wang X, Guo L, Gao Z, Hao X, Yang C. C. elegans-based screen identifies lysosome-damaging alkaloids that induce STAT3-dependent lysosomal cell death. Protein Cell 2018; 9:1013-1026. [PMID: 29611115 PMCID: PMC6251801 DOI: 10.1007/s13238-018-0520-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 01/16/2018] [Indexed: 01/13/2023] Open
Abstract
Lysosomes are degradation and signaling centers within the cell, and their dysfunction impairs a wide variety of cellular processes. To understand the cellular effect of lysosome damage, we screened natural small-molecule compounds that induce lysosomal abnormality using Caenorhabditis elegans (C. elegans) as a model system. A group of vobasinyl-ibogan type bisindole alkaloids (ervachinines A-D) were identified that caused lysosome enlargement in C. elegans macrophage-like cells. Intriguingly, these compounds triggered cell death in the germ line independently of the canonical apoptosis pathway. In mammalian cells, ervachinines A-D induced lysosomal enlargement and damage, leading to leakage of cathepsin proteases, inhibition of autophagosome degradation and necrotic cell death. Further analysis revealed that this ervachinine-induced lysosome damage and lysosomal cell death depended on STAT3 signaling, but not RIP1 or RIP3 signaling. These findings suggest that lysosome-damaging compounds are promising reagents for dissecting signaling mechanisms underlying lysosome homeostasis and lysosome-related human disorders.
Collapse
Affiliation(s)
- Yang Li
- Department of Pharmacology, Key Laboratory of Metabolism and Molecular Medicine (The Ministry of Education), School of Basic Medical Science, Fudan University, Shanghai, 200032, China. .,State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Yu Zhang
- State Key Laboratory of Phytochemistry and Plant Resources in Western China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650021, China
| | - Qiwen Gan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Meng Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiao Ding
- State Key Laboratory of Phytochemistry and Plant Resources in Western China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650021, China
| | - Guihua Tang
- State Key Laboratory of Phytochemistry and Plant Resources in Western China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650021, China
| | - Jingjing Liang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Kai Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xuezhao Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xin Wang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650091, China
| | - Lingli Guo
- State Key Laboratory of Phytochemistry and Plant Resources in Western China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650021, China
| | - Zhiyang Gao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiaojiang Hao
- State Key Laboratory of Phytochemistry and Plant Resources in Western China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650021, China. .,The Key Laboratory of Chemistry for Natural Product of Guizhou Province, Chinese Academy of Science, Guiyang, 550002, China.
| | - Chonglin Yang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China. .,State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650091, China.
| |
Collapse
|
39
|
Hybrid 2D/3D-quantitative structure-activity relationship and modeling studies perspectives of pepstatin A analogs as cathepsin D inhibitors. Future Med Chem 2017; 10:5-26. [PMID: 29235371 DOI: 10.4155/fmc-2017-0077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
AIM Cathepsin D, one of the attractive targets in the treatment of breast cancer, has been implicated in HIV neuropathogenesis with potential proteolytic effects on chemokines. Methodology/result: Diverse modeling tools were used to reveal the key structural features affecting the inhibitory activities of 78 pepstatin A analogs. Analyses were performed to investigate the stability, rationality and fluctuation of the analogs. Results showed a clear correlation between the experimental and predicted activities of the analogs as well as the variation in their activities relative to structural modifications. CONCLUSION The insight gained from this study offers theoretical references for understanding the mechanism of action of cathepsin D and will aid in the design of more potent and clinically-relevant drugs. Graphical abstract [Formula: see text].
Collapse
|
40
|
Aghdassi AA, John DS, Sendler M, Weiss FU, Reinheckel T, Mayerle J, Lerch MM. Cathepsin D regulates cathepsin B activation and disease severity predominantly in inflammatory cells during experimental pancreatitis. J Biol Chem 2017; 293:1018-1029. [PMID: 29229780 DOI: 10.1074/jbc.m117.814772] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 11/03/2017] [Indexed: 12/27/2022] Open
Abstract
Acute pancreatitis is a complex disorder involving both premature intracellular protease activation and inflammatory cell invasion. An initiating event is the intracellular activation of trypsinogen by cathepsin B (CTSB), which can be induced directly via G protein-coupled receptors on acinar cells or through inflammatory cells. Here, we studied CTSB regulation by another lysosomal hydrolase, cathepsin D (CTSD), using mice with a complete (CTSD-/-) or pancreas-specific conditional CTSD knockout (KO) (CTSDf/f/p48Cre/+). We induced acute pancreatitis by repeated caerulein injections and isolated acinar and bone marrow cells for ex vivo studies. Supramaximal caerulein stimulation induced subcellular redistribution of CTSD from the lysosomal to the zymogen-containing subcellular compartment of acinar cells and activation of CTSD, CTSB, and trypsinogen. Of note, the CTSD KO greatly reduced CTSB and trypsinogen activation in acinar cells, and CTSD directly activated CTSB but not trypsinogen in vitro During pancreatitis in pancreas-specific CTSDf/f/p48Cre/+ animals, markers of severity were reduced only at 1 h, whereas in the complete KO, this effect also included the late disease phase (8 h), indicating an important effect of extra-acinar CTSD on course of the disease. CTSD-/- leukocytes exhibited reduced cytokine release after lipopolysaccharide (LPS) stimulation, and CTSD KO also reduced caspase-3 activation and apoptosis in acinar cells stimulated with the intestinal hormone cholecystokinin. In summary, CTSD is expressed in pancreatic acinar and inflammatory cells, undergoes subcellular redistribution and activation during experimental pancreatitis, and regulates disease severity by potently activating CTSB. Its impact is only minimal and transient in the early, acinar cell-dependent phase of pancreatitis and much greater in the later, inflammatory cell-dependent phase of the disease.
Collapse
Affiliation(s)
- Ali A Aghdassi
- From the Department of Medicine A, University Medicine Greifswald, D-17475 Greifswald, Germany,
| | - Daniel S John
- From the Department of Medicine A, University Medicine Greifswald, D-17475 Greifswald, Germany
| | - Matthias Sendler
- From the Department of Medicine A, University Medicine Greifswald, D-17475 Greifswald, Germany
| | - F Ulrich Weiss
- From the Department of Medicine A, University Medicine Greifswald, D-17475 Greifswald, Germany
| | - Thomas Reinheckel
- the Institute of Molecular Medicine and Cell Research, Medical Faculty, Albert-Ludwigs-University Freiburg, D-79104 Freiburg, Germany, and
| | - Julia Mayerle
- From the Department of Medicine A, University Medicine Greifswald, D-17475 Greifswald, Germany.,the Department of Medicine II, Ludwigs-Maximilians University Munich, 80539 Munich, Germany
| | - Markus M Lerch
- From the Department of Medicine A, University Medicine Greifswald, D-17475 Greifswald, Germany
| |
Collapse
|
41
|
Jia S, Wang Y, You Z, Liu B, Gao J, Liu W. Mammalian Atg9 contributes to the post-Golgi transport of lysosomal hydrolases by interacting with adaptor protein-1. FEBS Lett 2017; 591:4027-4038. [PMID: 29156099 DOI: 10.1002/1873-3468.12916] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 10/10/2017] [Accepted: 10/21/2017] [Indexed: 11/11/2022]
Abstract
Accumulating evidence has indicated a role for autophagy-related (Atgs) proteins in cell regulation which is independent of their autophagic activities. As the only known transmembrane protein essential for autophagy, Atg9 cycles between the trans-Golgi network (TGN) and endosomes. Here, we report a function for mammalian Atg9 (mAtg9) in the transport of lysosomal hydrolases which impacts the lysosomal degradation capacity. Depletion of mAtg9 inhibits the degradation of epidermal growth factor receptor and the maturation of cathepsin D and cathepsin L. mAtg9 interacts with adaptor protein-1 (AP1) and the cation-independent mannose-6-phosphate receptor, facilitating AP1 polymerization and the transport of cathepsin D from the TGN. These results suggest that mAtg9 may serve as a coreceptor of lysosomal hydrolases for their TGN export by cycling between the TGN and endosomes.
Collapse
Affiliation(s)
- Shu Jia
- Department of Biochemistry and Molecular Biology, Program in Molecular Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Yusha Wang
- Department of Biochemistry and Molecular Biology, Program in Molecular Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiyuan You
- Department of Biochemistry and Molecular Biology, Program in Molecular Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Bo Liu
- Department of Biochemistry and Molecular Biology, Program in Molecular Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinfeng Gao
- Department of Biochemistry and Molecular Biology, Program in Molecular Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Liu
- Department of Biochemistry and Molecular Biology, Program in Molecular Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
42
|
Rahman N, Ramos-Espiritu L, Milner TA, Buck J, Levin LR. Soluble adenylyl cyclase is essential for proper lysosomal acidification. J Gen Physiol 2017; 148:325-39. [PMID: 27670898 PMCID: PMC5037342 DOI: 10.1085/jgp.201611606] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/08/2016] [Indexed: 01/07/2023] Open
Abstract
Lysosomes, the degradative organelles of the endocytic and autophagic pathways, function at an acidic pH. Lysosomes are acidified by the proton-pumping vacuolar ATPase (V-ATPase), but the molecular processes that set the organelle's pH are not completely understood. In particular, pH-sensitive signaling enzymes that can regulate lysosomal acidification in steady-state physiological conditions have yet to be identified. Soluble adenylyl cyclase (sAC) is a widely expressed source of cAMP that serves as a physiological pH sensor in cells. For example, in proton-secreting epithelial cells, sAC is responsible for pH-dependent translocation of V-ATPase to the luminal surface. Here we show genetically and pharmacologically that sAC is also essential for lysosomal acidification. In the absence of sAC, V-ATPase does not properly localize to lysosomes, lysosomes fail to fully acidify, lysosomal degradative capacity is diminished, and autophagolysosomes accumulate.
Collapse
Affiliation(s)
- Nawreen Rahman
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065 Graduate Program in Neuroscience, Weill Cornell Medical College, New York, NY 10065
| | | | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY 10065 Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065
| | - Jochen Buck
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065
| | - Lonny R Levin
- Department of Pharmacology, Weill Cornell Medical College, New York, NY 10065
| |
Collapse
|
43
|
Sun H, Liu Y, Zhang L, Shao X, Liu K, Ding Z, Liu X, Jiang C, Li H, Li H. Numb positively regulates autophagic flux via regulating lysosomal function. Biochem Biophys Res Commun 2017; 491:780-786. [PMID: 28720501 DOI: 10.1016/j.bbrc.2017.07.084] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 07/14/2017] [Indexed: 11/15/2022]
Abstract
Autophagy is a lysosome-dependent catabolic process involving in the degradation and recycling of unnecessary or damaged proteins and organelles. Emerging evidence indicates that autophagy dysfunction is closely related to various human diseases including cancer, aging, myopathies and neurodegenerative disorders. Here, using genetic knockdown, we uncover the role of Numb, an endocytic adaptor protein, in regulating the late steps of autophagy. We found that Numb depletion led to the accumulation of autophagic vacuole, as verified by RFP-LC3 staining combined with transmission electron microscopy. Further investigation indicated that Numb depletion impaired autophagic degradation through inhibiting the activities of lysosomal enzymes (Cathepsin D, β-glucuronidase and β-glucosidase). Moreover, Numb depletion induced elevation of lysosomal pH values and decrease of glycosylated lysosome-associated membrane proteins. We further observed that Rab7 activity was inhibited in Numb-depleted cells. Together, our findings revealed a novel function of Numb and its likely mechanism in regulation of autophagy events.
Collapse
Affiliation(s)
- Haiyan Sun
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Liu
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Lei Zhang
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Ximing Shao
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Ke Liu
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Zhihao Ding
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Xianming Liu
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; College of Life Science and Technology, Jinan University, 601 West Huangpu Blvd., Guangzhou 510632, China
| | - Changan Jiang
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Huashun Li
- SARITEX Center for Stem Cell Engineering Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Advanced Institute of Translational Medicine, Shanghai 200123, China; ATCG Corporation, BioBay, Suzhou Industrial Park, Suzhou, Jiangsu 215123, China
| | - Hongchang Li
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
44
|
Butkevich AN, Lukinavičius G, D’Este E, Hell SW. Cell-Permeant Large Stokes Shift Dyes for Transfection-Free Multicolor Nanoscopy. J Am Chem Soc 2017; 139:12378-12381. [DOI: 10.1021/jacs.7b06412] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Alexey N. Butkevich
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| | - Gražvydas Lukinavičius
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| | - Elisa D’Este
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| | - Stefan W. Hell
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| |
Collapse
|
45
|
Qiao ZY, Lai WJ, Lin YX, Li D, Nan XH, Wang Y, Wang H, Fang QJ. Polymer–KLAK Peptide Conjugates Induce Cancer Cell Death through Synergistic Effects of Mitochondria Damage and Autophagy Blockage. Bioconjug Chem 2017; 28:1709-1721. [DOI: 10.1021/acs.bioconjchem.7b00176] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
| | | | - Yao-Xin Lin
- University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Dan Li
- University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xiao-Hui Nan
- University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yi Wang
- University of Chinese Academy of Sciences, Beijing 100049, PR China
| | | | - Qiao-Jun Fang
- University of Chinese Academy of Sciences, Beijing 100049, PR China
| |
Collapse
|
46
|
Kang HT, Park JT, Choi K, Kim Y, Choi HJC, Jung CW, Lee YS, Park SC. Chemical screening identifies ATM as a target for alleviating senescence. Nat Chem Biol 2017; 13:616-623. [DOI: 10.1038/nchembio.2342] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 12/21/2016] [Indexed: 12/19/2022]
|
47
|
Zuhl AM, Nolan CE, Brodney MA, Niessen S, Atchison K, Houle C, Karanian DA, Ambroise C, Brulet JW, Beck EM, Doran SD, O'Neill BT, Am Ende CW, Chang C, Geoghegan KF, West GM, Judkins JC, Hou X, Riddell DR, Johnson DS. Chemoproteomic profiling reveals that cathepsin D off-target activity drives ocular toxicity of β-secretase inhibitors. Nat Commun 2016; 7:13042. [PMID: 27727204 PMCID: PMC5062570 DOI: 10.1038/ncomms13042] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 08/26/2016] [Indexed: 01/18/2023] Open
Abstract
Inhibition of β-secretase BACE1 is considered one of the most promising approaches for treating Alzheimer's disease. Several structurally distinct BACE1 inhibitors have been withdrawn from development after inducing ocular toxicity in animal models, but the target mediating this toxicity has not been identified. Here we use a clickable photoaffinity probe to identify cathepsin D (CatD) as a principal off-target of BACE1 inhibitors in human cells. We find that several BACE1 inhibitors blocked CatD activity in cells with much greater potency than that displayed in cell-free assays with purified protein. Through a series of exploratory toxicology studies, we show that quantifying CatD target engagement in cells with the probe is predictive of ocular toxicity in vivo. Taken together, our findings designate off-target inhibition of CatD as a principal driver of ocular toxicity for BACE1 inhibitors and more generally underscore the power of chemical proteomics for discerning mechanisms of drug action. Several β-secretase (BACE) inhibitors exhibit unexplained ocular toxicity in preclinical studies. Here the authors generate a clickable photoaffinity probe to interrogate off-targets in cells and animals, and identify inhibition of cathepsin D as a driver of ocular toxicity.
Collapse
Affiliation(s)
- Andrea M Zuhl
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, USA.,Worldwide Medicinal Chemistry
| | - Charles E Nolan
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, USA.,Neuroscience Research Unit
| | - Michael A Brodney
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, USA.,Worldwide Medicinal Chemistry
| | - Sherry Niessen
- Worldwide Medicinal Chemistry.,Pfizer Worldwide Research and Development, San Diego, California 92121, USA
| | - Kevin Atchison
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, USA.,Neuroscience Research Unit
| | - Christopher Houle
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, USA.,Drug Safety Research and Development
| | - David A Karanian
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, USA.,Drug Safety Research and Development
| | - Claude Ambroise
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, USA.,Neuroscience Research Unit
| | - Jeffrey W Brulet
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, USA.,Worldwide Medicinal Chemistry
| | - Elizabeth M Beck
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, USA.,Worldwide Medicinal Chemistry
| | - Shawn D Doran
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, USA.,Pharmacokinetics, Dynamics and Metabolism
| | - Brian T O'Neill
- Worldwide Medicinal Chemistry.,Pfizer Worldwide Research and Development, Groton, Connecticut 06340, USA
| | - Christopher W Am Ende
- Worldwide Medicinal Chemistry.,Pfizer Worldwide Research and Development, Groton, Connecticut 06340, USA
| | - Cheng Chang
- Pfizer Worldwide Research and Development, Groton, Connecticut 06340, USA.,Pharmacokinetics, Dynamics and Metabolism
| | - Kieran F Geoghegan
- Worldwide Medicinal Chemistry.,Pfizer Worldwide Research and Development, Groton, Connecticut 06340, USA.,Structural Biology and Biophysics Group
| | - Graham M West
- Worldwide Medicinal Chemistry.,Pfizer Worldwide Research and Development, Groton, Connecticut 06340, USA.,Structural Biology and Biophysics Group
| | - Joshua C Judkins
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, USA.,Worldwide Medicinal Chemistry
| | - Xinjun Hou
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, USA.,Worldwide Medicinal Chemistry
| | - David R Riddell
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, USA.,Neuroscience Research Unit
| | - Douglas S Johnson
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, USA.,Worldwide Medicinal Chemistry
| |
Collapse
|
48
|
Li Y, Xu M, Ding X, Yan C, Song Z, Chen L, Huang X, Wang X, Jian Y, Tang G, Tang C, Di Y, Mu S, Liu X, Liu K, Li T, Wang Y, Miao L, Guo W, Hao X, Yang C. Protein kinase C controls lysosome biogenesis independently of mTORC1. Nat Cell Biol 2016; 18:1065-77. [PMID: 27617930 DOI: 10.1038/ncb3407] [Citation(s) in RCA: 268] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 08/11/2016] [Indexed: 12/13/2022]
Abstract
Lysosomes respond to environmental cues by controlling their own biogenesis, but the underlying mechanisms are poorly understood. Here we describe a protein kinase C (PKC)-dependent and mTORC1-independent mechanism for regulating lysosome biogenesis, which provides insights into previously reported effects of PKC on lysosomes. By identifying lysosome-inducing compounds we show that PKC couples activation of the TFEB transcription factor with inactivation of the ZKSCAN3 transcriptional repressor through two parallel signalling cascades. Activated PKC inactivates GSK3β, leading to reduced phosphorylation, nuclear translocation and activation of TFEB, while PKC activates JNK and p38 MAPK, which phosphorylate ZKSCAN3, leading to its inactivation by translocation out of the nucleus. PKC activation may therefore mediate lysosomal adaptation to many extracellular cues. PKC activators facilitate clearance of aggregated proteins and lipid droplets in cell models and ameliorate amyloid β plaque formation in APP/PS1 mouse brains. Thus, PKC activators are viable treatment options for lysosome-related disorders.
Collapse
Affiliation(s)
- Yang Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No.1 West Beichen Road, Chaoyang District, Beijing 100101, China
| | - Meng Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No.1 West Beichen Road, Chaoyang District, Beijing 100101, China.,Graduate University of Chinese Academy of Sciences, Beijing 100039, China
| | - Xiao Ding
- State Key Laboratory of Phytochemistry and Plant Resources in Western China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650021, China
| | - Chen Yan
- The Key Laboratory of Chemistry for Natural Product of Guizhou Province and Chinese Academy of Science, Guiyang 550002, China
| | - Zhiqin Song
- The Key Laboratory of Chemistry for Natural Product of Guizhou Province and Chinese Academy of Science, Guiyang 550002, China
| | - Lianwan Chen
- Key Laboratory of RNA Biology, Beijing Key Laboratory of Noncoding RNA, Institute of Biophysics, Chinese Academy of Sciences, No.15 Datun Road, Chaoyang District, Beijing 100101, China
| | - Xiahe Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No.1 West Beichen Road, Chaoyang District, Beijing 100101, China
| | - Xin Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No.1 West Beichen Road, Chaoyang District, Beijing 100101, China
| | - Youli Jian
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No.1 West Beichen Road, Chaoyang District, Beijing 100101, China
| | - Guihua Tang
- State Key Laboratory of Phytochemistry and Plant Resources in Western China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650021, China
| | - Changyong Tang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No.1 West Beichen Road, Chaoyang District, Beijing 100101, China
| | - Yingtong Di
- State Key Laboratory of Phytochemistry and Plant Resources in Western China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650021, China
| | - Shuzhen Mu
- The Key Laboratory of Chemistry for Natural Product of Guizhou Province and Chinese Academy of Science, Guiyang 550002, China
| | - Xuezhao Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No.1 West Beichen Road, Chaoyang District, Beijing 100101, China.,Graduate University of Chinese Academy of Sciences, Beijing 100039, China
| | - Kai Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No.1 West Beichen Road, Chaoyang District, Beijing 100101, China.,Graduate University of Chinese Academy of Sciences, Beijing 100039, China
| | - Ting Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No.1 West Beichen Road, Chaoyang District, Beijing 100101, China
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No.1 West Beichen Road, Chaoyang District, Beijing 100101, China
| | - Long Miao
- Key Laboratory of RNA Biology, Beijing Key Laboratory of Noncoding RNA, Institute of Biophysics, Chinese Academy of Sciences, No.15 Datun Road, Chaoyang District, Beijing 100101, China
| | - Weixiang Guo
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No.1 West Beichen Road, Chaoyang District, Beijing 100101, China
| | - Xiaojiang Hao
- State Key Laboratory of Phytochemistry and Plant Resources in Western China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650021, China.,The Key Laboratory of Chemistry for Natural Product of Guizhou Province and Chinese Academy of Science, Guiyang 550002, China
| | - Chonglin Yang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No.1 West Beichen Road, Chaoyang District, Beijing 100101, China
| |
Collapse
|
49
|
Lukinavičius G, Reymond L, Umezawa K, Sallin O, D'Este E, Göttfert F, Ta H, Hell SW, Urano Y, Johnsson K. Fluorogenic Probes for Multicolor Imaging in Living Cells. J Am Chem Soc 2016; 138:9365-8. [PMID: 27420907 DOI: 10.1021/jacs.6b04782] [Citation(s) in RCA: 179] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Here we present a far-red, silicon-rhodamine-based fluorophore (SiR700) for live-cell multicolor imaging. SiR700 has excitation and emission maxima at 690 and 715 nm, respectively. SiR700-based probes for F-actin, microtubules, lysosomes, and SNAP-tag are fluorogenic, cell-permeable, and compatible with superresolution microscopy. In conjunction with probes based on the previously introduced carboxy-SiR650, SiR700-based probes permit multicolor live-cell superresolution microscopy in the far-red, thus significantly expanding our capacity for imaging living cells.
Collapse
Affiliation(s)
- Gražvydas Lukinavičius
- Institute of Bioengineering, NCCR in Chemical Biology, Institute of Chemical Sciences and Engineering (ISIC), Ecole Polytechnique Fédérale de Lausanne (EPFL) , 1015 Lausanne, Switzerland.,Department of NanoBiophotonics, Max-Planck-Institute for Biophysical Chemistry , Am Fassberg 11, 37077 Göttingen, Germany
| | - Luc Reymond
- Institute of Bioengineering, NCCR in Chemical Biology, Institute of Chemical Sciences and Engineering (ISIC), Ecole Polytechnique Fédérale de Lausanne (EPFL) , 1015 Lausanne, Switzerland
| | - Keitaro Umezawa
- Graduate School of Pharmaceutical Sciences, The University of Tokyo , 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Olivier Sallin
- Institute of Bioengineering, NCCR in Chemical Biology, Institute of Chemical Sciences and Engineering (ISIC), Ecole Polytechnique Fédérale de Lausanne (EPFL) , 1015 Lausanne, Switzerland
| | - Elisa D'Este
- Department of NanoBiophotonics, Max-Planck-Institute for Biophysical Chemistry , Am Fassberg 11, 37077 Göttingen, Germany
| | - Fabian Göttfert
- Department of NanoBiophotonics, Max-Planck-Institute for Biophysical Chemistry , Am Fassberg 11, 37077 Göttingen, Germany
| | - Haisen Ta
- Department of NanoBiophotonics, Max-Planck-Institute for Biophysical Chemistry , Am Fassberg 11, 37077 Göttingen, Germany
| | - Stefan W Hell
- Department of NanoBiophotonics, Max-Planck-Institute for Biophysical Chemistry , Am Fassberg 11, 37077 Göttingen, Germany
| | - Yasuteru Urano
- Graduate School of Pharmaceutical Sciences, The University of Tokyo , 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Kai Johnsson
- Institute of Bioengineering, NCCR in Chemical Biology, Institute of Chemical Sciences and Engineering (ISIC), Ecole Polytechnique Fédérale de Lausanne (EPFL) , 1015 Lausanne, Switzerland
| |
Collapse
|
50
|
Carrasco S, Benito-Peña E, Walt DR, Moreno-Bondi MC. Fiber-optic array using molecularly imprinted microspheres for antibiotic analysis. Chem Sci 2015; 6:3139-3147. [PMID: 29142687 PMCID: PMC5657405 DOI: 10.1039/c5sc00115c] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 03/18/2015] [Indexed: 12/12/2022] Open
Abstract
In this article we describe a new class of high-density optical microarrays based on molecularly imprinted microsphere sensors that directly incorporate specific recognition capabilities to detect enrofloxacin (ENRO), an antibiotic widely used for both human and veterinary applications. This approach involves the preparation of highly cross-linked polymer microspheres by thermal precipitation-polymerization in the presence and absence of the target analyte ENRO to generate either molecularly imprinted (MIP) or non-imprinted polymer (NIP) microspheres, respectively. Each polymer type of tailor-made microsphere is fluorescently encoded with either coumarin-30 or tris(4,7-diphenyl-1,10-phenanthroline)ruthenium(ii) dichloride [Ru(dip)3]Cl2 to enable the microspheres to be distinguished. The new MIP-based sensing platform utilizes an optical fiber bundle containing approximately 50 000 individual 3.1 μm diameter fibers that are chemically etched to create microwells in which MIP and NIP microspheres can be deposited and imaged using an epi-fluorescence microscope. The method enables multiplexed detection by independently addressing both types of beads through their separate light channels. The unique response to the presence of ENRO is manifested on the basis of a competitive immunoassay. A red-fluorescent dye-tagged ENRO, labeled with BODIPY® TR Cadaverine, competes with ENRO for specific binding sites. The developed immuno-like assay displayed a limit of detection (LOD) of 0.04 μM (10% binding inhibition) and a dynamic range of 0.29-21.54 μM (20-80% binding inhibition). The selectivity of the assay was evaluated by measuring the cross-reactivity of other fluoroquinolones (ciprofloxacin, norfloxacin, danofloxacin, and flumequine) and non-related antibiotics (penicillin G and doxycycline). This work demonstrates, for the first time, the applicability of MIPs, as an alternative to biomolecule receptors, for the development of multiplexed detection fiber-optic microarrays paving the way for a new generation of biomimetic sensors.
Collapse
Affiliation(s)
- Sergio Carrasco
- Department of Analytical Chemistry , Faculty of Chemistry , Complutense University , Ciudad Universitaria s/n , Madrid 28040 , Spain . ; ; ; Tel: +34-91394-5147
| | - Elena Benito-Peña
- Department of Analytical Chemistry , Faculty of Chemistry , Complutense University , Ciudad Universitaria s/n , Madrid 28040 , Spain . ; ; ; Tel: +34-91394-5147
- Department of Chemistry , Tufts University , 62 Talbot Avenue , Medford , MA 02155 , USA . ; ; Tel: +1-617-627-3470
| | - David R Walt
- Department of Chemistry , Tufts University , 62 Talbot Avenue , Medford , MA 02155 , USA . ; ; Tel: +1-617-627-3470
| | - María C Moreno-Bondi
- Department of Analytical Chemistry , Faculty of Chemistry , Complutense University , Ciudad Universitaria s/n , Madrid 28040 , Spain . ; ; ; Tel: +34-91394-5147
| |
Collapse
|