1
|
Gregori M, Pereira GJ, Allen R, West N, Chau KY, Cai X, Bostock MP, Bolsover SR, Keller M, Lee CY, Lei SH, Harvey K, Bracher F, Grimm C, Hasan G, Gegg ME, Schapira AH, Sweeney ST, Patel S. Lysosomal TPC2 channels disrupt Ca2+ entry and dopaminergic function in models of LRRK2-Parkinson's disease. J Cell Biol 2025; 224:e202412055. [PMID: 40279672 PMCID: PMC12029513 DOI: 10.1083/jcb.202412055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 04/27/2025] Open
Abstract
Parkinson's disease results from degeneration of dopaminergic neurons in the midbrain, but the underlying mechanisms are unclear. Here, we identify novel crosstalk between depolarization-induced entry of Ca2+ and lysosomal cation release in maintaining dopaminergic neuronal function. The common disease-causing G2019S mutation in LRRK2 selectively exaggerated Ca2+ entry in vitro. Chemical and molecular strategies inhibiting the lysosomal ion channel TPC2 reversed this. Using Drosophila, which lack TPCs, we show that the expression of human TPC2 phenocopied LRRK2 G2019S in perturbing dopaminergic-dependent vision and movement in vivo. Mechanistically, dysfunction required an intact pore, correct subcellular targeting and Rab interactivity of TPC2. Reducing Ca2+ permeability with a novel biased TPC2 agonist corrected deviant Ca2+ entry and behavioral defects. Thus, both inhibition and select activation of TPC2 are beneficial. Functional coupling between lysosomal cation release and Ca2+ influx emerges as a potential druggable node in Parkinson's disease.
Collapse
Affiliation(s)
- Martina Gregori
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Gustavo J.S. Pereira
- Department of Cell and Developmental Biology, University College London, London, UK
- Department of Pharmacology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Robert Allen
- Department of Biology, University of York, York, UK
| | | | - Kai-Yin Chau
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Xinjiang Cai
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Matthew P. Bostock
- Centre for Developmental Neurobiology and MRC Centre for Neurodevelopmental Disorders, King’s College London, London, UK
| | - Stephen R. Bolsover
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Marco Keller
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilian University, Munich, Germany
| | - Chiao-Yin Lee
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Si Hang Lei
- Department of Pharmacology, UCL School of Pharmacy, London, UK
| | - Kirsten Harvey
- Department of Pharmacology, UCL School of Pharmacy, London, UK
| | - Franz Bracher
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilian University, Munich, Germany
| | - Christian Grimm
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilian University, Munich, Germany
- Immunology, Infection and Pandemic Research IIP, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt, Germany
| | - Gaiti Hasan
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Matthew E. Gegg
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Anthony H.V. Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | | | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, London, UK
| |
Collapse
|
2
|
Aynaci A, Toussaint M, Gilis F, Albert M, Gaussin JF, Jadot M, Boonen M. Disruption of Man-6-P-Dependent Sorting to Lysosomes Confers IGF1R-Mediated Apoptosis Resistance. Int J Mol Sci 2025; 26:3586. [PMID: 40332073 PMCID: PMC12026698 DOI: 10.3390/ijms26083586] [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: 03/17/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Mutations in GNPTAB underlie mucolipidosis II and mucolipidosis III α/β, which are inherited lysosomal storage disorders caused by a defective UDP-N-acetylglucosamine:lysosomal-enzyme N-acetylglucosamine phosphotransferase. As a result, newly synthesized acid hydrolases fail to acquire Mannose-6-Phosphate (Man-6-P) sorting signals, or do so to a lesser extent, and exhibit an impaired trafficking to lysosomes. Interestingly, we found that GNPTAB knockout HeLa cells are resistant to several cytotoxic agents: doxorubicin, chloroquine, staurosporine and paclitaxel. While we detected an increased trapping of weak bases in the expanded lysosomal population of these cells, which could reduce the effect of doxorubicin and chloroquine; the decreased cell response to staurosporine and paclitaxel suggested the involvement of alternative resistance mechanisms. Indeed, further investigation revealed that the hyperactivation of the Insulin-like Growth Factor 1 Receptor (IGF1R) pathway is a central player in the apoptosis resistance exhibited by Man-6-P sorting deficient cells.
Collapse
Affiliation(s)
- Asena Aynaci
- Laboratory of Intracellular Trafficking Biology, URPhyM, NARILIS, UNamur, 61 rue de Bruxelles, 5000 Namur, Belgium; (A.A.); (M.T.); (F.G.); (M.A.); (J.-F.G.)
- Laboratory of Physiological Chemistry, URPhyM, NARILIS, UNamur, 61 rue de Bruxelles, 5000 Namur, Belgium;
| | - Maxence Toussaint
- Laboratory of Intracellular Trafficking Biology, URPhyM, NARILIS, UNamur, 61 rue de Bruxelles, 5000 Namur, Belgium; (A.A.); (M.T.); (F.G.); (M.A.); (J.-F.G.)
- Laboratory of Physiological Chemistry, URPhyM, NARILIS, UNamur, 61 rue de Bruxelles, 5000 Namur, Belgium;
| | - Florentine Gilis
- Laboratory of Intracellular Trafficking Biology, URPhyM, NARILIS, UNamur, 61 rue de Bruxelles, 5000 Namur, Belgium; (A.A.); (M.T.); (F.G.); (M.A.); (J.-F.G.)
- Laboratory of Physiological Chemistry, URPhyM, NARILIS, UNamur, 61 rue de Bruxelles, 5000 Namur, Belgium;
| | - Martine Albert
- Laboratory of Intracellular Trafficking Biology, URPhyM, NARILIS, UNamur, 61 rue de Bruxelles, 5000 Namur, Belgium; (A.A.); (M.T.); (F.G.); (M.A.); (J.-F.G.)
| | - Jean-François Gaussin
- Laboratory of Intracellular Trafficking Biology, URPhyM, NARILIS, UNamur, 61 rue de Bruxelles, 5000 Namur, Belgium; (A.A.); (M.T.); (F.G.); (M.A.); (J.-F.G.)
| | - Michel Jadot
- Laboratory of Physiological Chemistry, URPhyM, NARILIS, UNamur, 61 rue de Bruxelles, 5000 Namur, Belgium;
| | - Marielle Boonen
- Laboratory of Intracellular Trafficking Biology, URPhyM, NARILIS, UNamur, 61 rue de Bruxelles, 5000 Namur, Belgium; (A.A.); (M.T.); (F.G.); (M.A.); (J.-F.G.)
| |
Collapse
|
3
|
Krogsaeter EK, McKetney J, Valiente-Banuet L, Marquez A, Willis A, Cakir Z, Stevenson E, Jang GM, Rao A, Li E, Zhou A, Attili A, Chang TS, Kampmann M, Huang Y, Krogan NJ, Swaney DL. Lysosomal proteomics reveals mechanisms of neuronal apoE4-associated lysosomal dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.02.560519. [PMID: 37873080 PMCID: PMC10592882 DOI: 10.1101/2023.10.02.560519] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
ApoE4 is the primary risk factor for Alzheimer Disease (AD). Early AD pathological events first affect the neuronal endolysosomal system, which in turn causes neuronal protein aggregation and cell death. Despite the crucial influence of lysosomes upon AD pathophysiology, and that apoE4 localizes to lysosomes, the influence of apoE4 on lysosomal function remains unexplored. We find that expression of apoE4 in neuronal cell lines results in lysosomal alkalinization and impaired lysosomal function. To identify driving factors for these defects, we performed quantitative lysosomal proteome profiling. This revealed that apoE4 expression results in differential regulation of numerous lysosomal proteins, correlating with apoE allele status and disease severity in AD brains. In particular, apoE4 expression results in the depletion of lysosomal Lgals3bp and the accumulation of lysosomal Tmed5. We additionally validated that these lysosomal protein changes can be targeted to modulate lysosomal function. Taken together, this work thereby reveals that apoE4 causes widespread lysosomal defects through remodeling the lysosomal proteome, with the lysosomal Tmed5 accumulation and Lgals3bp depletion manifesting as lysosomal alkalinization in apoE4 neurons.
Collapse
Affiliation(s)
- Einar K. Krogsaeter
- Gladstone Data Science and Biotechnology Institute, The J. David Gladstone Institutes, San Francisco, California, USA
- Quantitative Bioscience Institute, University of California, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
- These authors contributed equally
| | - Justin McKetney
- Gladstone Data Science and Biotechnology Institute, The J. David Gladstone Institutes, San Francisco, California, USA
- Quantitative Bioscience Institute, University of California, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
- These authors contributed equally
| | - Leopoldo Valiente-Banuet
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Angelica Marquez
- Gladstone Data Science and Biotechnology Institute, The J. David Gladstone Institutes, San Francisco, California, USA
- Quantitative Bioscience Institute, University of California, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
| | - Alexandra Willis
- Gladstone Data Science and Biotechnology Institute, The J. David Gladstone Institutes, San Francisco, California, USA
- Quantitative Bioscience Institute, University of California, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
| | - Zeynep Cakir
- Gladstone Data Science and Biotechnology Institute, The J. David Gladstone Institutes, San Francisco, California, USA
- Quantitative Bioscience Institute, University of California, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
| | - Erica Stevenson
- Gladstone Data Science and Biotechnology Institute, The J. David Gladstone Institutes, San Francisco, California, USA
- Quantitative Bioscience Institute, University of California, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
| | - Gwendolyn M. Jang
- Gladstone Data Science and Biotechnology Institute, The J. David Gladstone Institutes, San Francisco, California, USA
- Quantitative Bioscience Institute, University of California, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
| | - Antara Rao
- Gladstone Institute of Neurological Disease, The J. David Gladstone Institutes, San Francisco, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, USA
| | - Emmy Li
- Institute for Neurodegenerative Diseases, University of California, San Francisco, California, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, USA
| | - Anton Zhou
- Gladstone Institute of Neurological Disease, The J. David Gladstone Institutes, San Francisco, USA
| | - Anjani Attili
- Institute for Neurodegenerative Diseases, University of California, San Francisco, California, USA
- Biosciences Internship Program, City College of San Francisco, USA
| | - Timothy S. Chang
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, University of California, San Francisco, California, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, California, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, The J. David Gladstone Institutes, San Francisco, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, USA
- Neuroscience Graduate Program, University of California, San Francisco, USA
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, USA
- Departments of Neurology and Pathology, University of California, San Francisco, USA
| | - Nevan J. Krogan
- Gladstone Data Science and Biotechnology Institute, The J. David Gladstone Institutes, San Francisco, California, USA
- Quantitative Bioscience Institute, University of California, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
| | - Danielle L. Swaney
- Gladstone Data Science and Biotechnology Institute, The J. David Gladstone Institutes, San Francisco, California, USA
- Quantitative Bioscience Institute, University of California, San Francisco, California, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, USA
| |
Collapse
|
4
|
Bjørnestad SA, Solbakken MH, Krokene P, Thiede B, Hylland K, Jakobsen KS, Jentoft S, Bakke O, Progida C. The Atlantic Cod MHC I compartment has the properties needed for cross-presentation in the absence of MHC II. Sci Rep 2024; 14:25404. [PMID: 39455705 PMCID: PMC11511864 DOI: 10.1038/s41598-024-76225-z] [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: 02/06/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Atlantic cod has a peculiar immune system, characterized by the loss of Major Histocompatibility Complex (MHC) class II pathway, and an extreme expansion of the MHC class I gene repertoire. This has led to the hypothesis that some of the MHC I variants have replaced MHC II by presenting exogenous-peptides in a process similar to cross-presentation. In mammals, MHC I loads endogenous antigens in the endoplasmic reticulum, but we recently found that different Atlantic cod MHC I gene variants traffic to endolysosomes. There, they colocalize with Tapasin and other components of the peptide-loading complex, indicating a plausible peptide-loading system outside the endoplasmic reticulum. In this study, we further characterize the identity of the Atlantic cod MHC I compartment (cMIC). We found that, similarly to mammalian MHC II compartment, cMIC contains late endosomal markers such as Rab7, LAMP1 and CD63. Furthermore, we identified Hsp90b1 (also known as grp94) and LRP1 (also known as CD91) as interactors of MHC I by mass spectrometry. As these two proteins are involved in cross-presentation in mammals, this further suggests that Atlantic cod MHC I might use a similar mechanism to present exogenous peptides, thus, compensating for the absence of MHC II.
Collapse
Affiliation(s)
| | - Monica Hongrø Solbakken
- Department of Biosciences, University of Oslo, Oslo, Norway
- Norwegian University of Life Sciences, Ås, Norway
| | - Pia Krokene
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Bernd Thiede
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Ketil Hylland
- Department of Biosciences, University of Oslo, Oslo, Norway
| | | | - Sissel Jentoft
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Oddmund Bakke
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Cinzia Progida
- Department of Biosciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
5
|
Pasqualotto BA, Nelson A, Deheshi S, Sheldon CA, Vogl AW, Rintoul GL. Impaired mitochondrial morphological plasticity and failure of mitophagy associated with the G11778A mutation of LHON. Biochem Biophys Res Commun 2024; 721:150119. [PMID: 38768545 DOI: 10.1016/j.bbrc.2024.150119] [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: 03/04/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 05/22/2024]
Abstract
Mitochondrial dynamics were examined in human dermal fibroblasts biopsied from a confirmed Leber's Hereditary Optic Neuropathy (LHON) patient with a homoplasmic G11778A mutation of the mitochondrial genome. Expression of the G11778A mutation did not impart any discernible difference in mitochondrial network morphology using widefield fluorescence microscopy. However, at the ultrastructural level, cells expressing this mutation exhibited an impairment of mitochondrial morphological plasticity when forced to utilize oxidative phosphorylation (OXPHOS) by transition to glucose-free, galactose-containing media. LHON fibroblasts also displayed a transient increase in mitophagy upon transition to galactose media. These results provide new insights into the consequences of the G11778A mutation of LHON and the pathological mechanisms underlying this disease.
Collapse
Affiliation(s)
- Bryce A Pasqualotto
- Centre for Cell Biology, Development, and Disease, and the Department of Biological Sciences, Simon Fraser University, Canada
| | - Alexa Nelson
- Centre for Cell Biology, Development, and Disease, and the Department of Biological Sciences, Simon Fraser University, Canada
| | - Samineh Deheshi
- Centre for Cell Biology, Development, and Disease, and the Department of Biological Sciences, Simon Fraser University, Canada
| | - Claire A Sheldon
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Canada
| | - A Wayne Vogl
- Life Sciences Institute and the Department of Cellular & Physiological Sciences, University of British Columbia, Canada
| | - Gordon L Rintoul
- Centre for Cell Biology, Development, and Disease, and the Department of Biological Sciences, Simon Fraser University, Canada.
| |
Collapse
|
6
|
Lesport E, Commeau L, Genet M, Baulieu EE, Tawk M, Giustiniani J. A decrease in Fkbp52 alters autophagosome maturation and A152T-tau clearance in vivo. Front Cell Neurosci 2024; 18:1425222. [PMID: 39119047 PMCID: PMC11306173 DOI: 10.3389/fncel.2024.1425222] [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: 04/29/2024] [Accepted: 07/15/2024] [Indexed: 08/10/2024] Open
Abstract
The failure of the autophagy-lysosomal pathway to clear the pathogenic forms of Tau exacerbates the pathogenesis of tauopathies. We have previously shown that the immunophilin FKBP52 interacts both physically and functionally with Tau, and that a decrease in FKBP52 protein levels is associated with Tau deposition in affected human brains. We have also shown that FKBP52 is physiologically present within the lysosomal system in healthy human neurons and that a decrease in FKBP52 expression alters perinuclear lysosomal positioning and Tau clearance during Tau-induced proteotoxic stress in vitro. In this study, we generate a zebrafish fkbp4 loss of function mutant and show that axonal retrograde trafficking of Lamp1 vesicles is altered in this mutant. Moreover, using our transgenic HuC::mCherry-EGFP-LC3 line, we demonstrate that the autophagic flux is impaired in fkbp4 mutant embryos, suggesting a role for Fkbp52 in the maturation of autophagic vesicles. Alterations in both axonal transport and autophagic flux are more evident in heterozygous rather than homozygous fkbp4 mutants. Finally, taking advantage of the previously described A152T-Tau transgenic fish, we show that the clearance of pathogenic A152T-Tau mutant proteins is slower in fkbp4 +/- mutants in comparison to fkbp4 +/+ larvae. Altogether, these results indicate that Fkbp52 is required for the normal trafficking and maturation of lysosomes and autophagic vacuoles along axons, and that its decrease is sufficient to hinder the clearance of pathogenic Tau in vivo.
Collapse
Affiliation(s)
- Emilie Lesport
- Institut Professeur Baulieu, INSERM U1195, Kremlin-Bicêtre, France
- INSERM U1195, Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Lucie Commeau
- Institut Professeur Baulieu, INSERM U1195, Kremlin-Bicêtre, France
| | - Mélanie Genet
- Institut Professeur Baulieu, INSERM U1195, Kremlin-Bicêtre, France
| | - Etienne-Emile Baulieu
- Institut Professeur Baulieu, INSERM U1195, Kremlin-Bicêtre, France
- INSERM U1195, Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Marcel Tawk
- INSERM U1195, Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Julien Giustiniani
- Institut Professeur Baulieu, INSERM U1195, Kremlin-Bicêtre, France
- INSERM U1195, Université Paris-Saclay, Kremlin-Bicêtre, France
| |
Collapse
|
7
|
Eysert F, Kinoshita PF, Lagarde J, Lacas-Gervais S, Xicota L, Dorothée G, Bottlaender M, Checler F, Potier MC, Sarazin M, Chami M. Mitochondrial alterations in fibroblasts from sporadic Alzheimer's disease (AD) patients correlate with AD-related clinical hallmarks. Acta Neuropathol Commun 2024; 12:90. [PMID: 38851733 PMCID: PMC11161956 DOI: 10.1186/s40478-024-01807-x] [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: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024] Open
Abstract
Mitochondrial dysfunctions are key features of Alzheimer's disease (AD). The occurrence of these disturbances in the peripheral cells of AD patients and their potential correlation with disease progression are underinvestigated. We studied mitochondrial structure, function and mitophagy in fibroblasts from healthy volunteers and AD patients at the prodromal (AD-MCI) or demented (AD-D) stages. We carried out correlation studies with clinical cognitive scores, namely, (i) Mini-Mental State Examination (MMSE) and (ii) Dementia Rating-Scale Sum of Boxes (CDR-SOB), and with (iii) amyloid beta (Aβ) plaque burden (PiB-PET imaging) and (iv) the accumulation of peripheral amyloid precursor protein C-terminal fragments (APP-CTFs). We revealed alterations in mitochondrial structure as well as specific mitochondrial dysfunction signatures in AD-MCI and AD-D fibroblasts and revealed that defective mitophagy and autophagy are linked to impaired lysosomal activity in AD-D fibroblasts. We reported significant correlations of a subset of these dysfunctions with cognitive decline, AD-related clinical hallmarks and peripheral APP-CTFs accumulation. This study emphasizes the potential use of peripheral cells for investigating AD pathophysiology.
Collapse
Affiliation(s)
- Fanny Eysert
- INSERM, CNRS, Institute of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, 660 Route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| | - Paula-Fernanda Kinoshita
- INSERM, CNRS, Institute of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, 660 Route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| | - Julien Lagarde
- Department of Neurology of Memory and Language, GHU Paris Psychiatrie & Neurosciences, Hôpital Sainte Anne, 75014, Paris, France
- Université Paris-Cité, 75006, Paris, France
- BioMaps, Service Hospitalier Frédéric Joliot CEA, CNRS, Inserm, Université Paris-Saclay, 91401, Orsay, France
| | - Sandra Lacas-Gervais
- Centre Commun de Microscopie Appliquée, Université de Nice Côte d'Azur, 06108, Nice, France
| | - Laura Xicota
- UPMC University Paris 06, UMRS 1127, Sorbonne Universités, Paris, France
- ICM Research Center, CNRS UMR 7225, Paris, France
| | - Guillaume Dorothée
- Inserm, Centre de Recherche Saint-Antoine, CRSA, Immune System and Neuroinflammation Laboratory, Hôpital Saint-Antoine, Sorbonne Université, 75012, Paris, France
| | - Michel Bottlaender
- BioMaps, Service Hospitalier Frédéric Joliot CEA, CNRS, Inserm, Université Paris-Saclay, 91401, Orsay, France
- UNIACT, Neurospin, Joliot Institute, CEA, Université Paris-Saclay, 91140, Gif sur Yvette, France
| | - Frédéric Checler
- INSERM, CNRS, Institute of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, 660 Route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| | - Marie-Claude Potier
- UPMC University Paris 06, UMRS 1127, Sorbonne Universités, Paris, France
- ICM Research Center, CNRS UMR 7225, Paris, France
| | - Marie Sarazin
- Department of Neurology of Memory and Language, GHU Paris Psychiatrie & Neurosciences, Hôpital Sainte Anne, 75014, Paris, France
- Université Paris-Cité, 75006, Paris, France
- BioMaps, Service Hospitalier Frédéric Joliot CEA, CNRS, Inserm, Université Paris-Saclay, 91401, Orsay, France
| | - Mounia Chami
- INSERM, CNRS, Institute of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, 660 Route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France.
| |
Collapse
|
8
|
Schrӧder LF, Peng W, Gao G, Wong YC, Schwake M, Krainc D. VPS13C regulates phospho-Rab10-mediated lysosomal function in human dopaminergic neurons. J Cell Biol 2024; 223:e202304042. [PMID: 38358348 PMCID: PMC10868123 DOI: 10.1083/jcb.202304042] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 12/14/2023] [Accepted: 01/31/2024] [Indexed: 02/16/2024] Open
Abstract
Loss-of-function mutations in VPS13C are linked to early-onset Parkinson's disease (PD). While VPS13C has been previously studied in non-neuronal cells, the neuronal role of VPS13C in disease-relevant human dopaminergic neurons has not been elucidated. Using live-cell microscopy, we investigated the role of VPS13C in regulating lysosomal dynamics and function in human iPSC-derived dopaminergic neurons. Loss of VPS13C in dopaminergic neurons disrupts lysosomal morphology and dynamics with increased inter-lysosomal contacts, leading to impaired lysosomal motility and cellular distribution, as well as defective lysosomal hydrolytic activity and acidification. We identified Rab10 as a phospho-dependent interactor of VPS13C on lysosomes and observed a decreased phospho-Rab10-mediated lysosomal stress response upon loss of VPS13C. These findings highlight an important role of VPS13C in regulating lysosomal homeostasis in human dopaminergic neurons and suggest that disruptions in Rab10-mediated lysosomal stress response contribute to disease pathogenesis in VPS13C-linked PD.
Collapse
Affiliation(s)
- Leonie F. Schrӧder
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Biochemistry III/Faculty of Chemistry, Bielefeld University, Bielefeld, Germany
| | - Wesley Peng
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ge Gao
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yvette C. Wong
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Michael Schwake
- Biochemistry III/Faculty of Chemistry, Bielefeld University, Bielefeld, Germany
| | - Dimitri Krainc
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
9
|
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
|
10
|
Yuan Y, Arige V, Saito R, Mu Q, Brailoiu GC, Pereira GJS, Bolsover SR, Keller M, Bracher F, Grimm C, Brailoiu E, Marchant JS, Yule DI, Patel S. Two-pore channel-2 and inositol trisphosphate receptors coordinate Ca 2+ signals between lysosomes and the endoplasmic reticulum. Cell Rep 2024; 43:113628. [PMID: 38160394 PMCID: PMC10931537 DOI: 10.1016/j.celrep.2023.113628] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/13/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024] Open
Abstract
Lysosomes and the endoplasmic reticulum (ER) are Ca2+ stores mobilized by the second messengers NAADP and IP3, respectively. Here, we establish Ca2+ signals between the two sources as fundamental building blocks that couple local release to global changes in Ca2+. Cell-wide Ca2+ signals evoked by activation of endogenous NAADP-sensitive channels on lysosomes comprise both local and global components and exhibit a major dependence on ER Ca2+ despite their lysosomal origin. Knockout of ER IP3 receptor channels delays these signals, whereas expression of lysosomal TPC2 channels accelerates them. High-resolution Ca2+ imaging reveals elementary events upon TPC2 opening and signals coupled to IP3 receptors. Biasing TPC2 activation to a Ca2+-permeable state sensitizes local Ca2+ signals to IP3. This increases the potency of a physiological agonist to evoke global Ca2+ signals and activate a downstream target. Our data provide a conceptual framework to understand how Ca2+ release from physically separated stores is coordinated.
Collapse
Affiliation(s)
- Yu Yuan
- Department of Cell and Developmental Biology, University College London, Gower Street, WC1E 6BT London, UK
| | - Vikas Arige
- Department of Pharmacology and Physiology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Ryo Saito
- Department of Cell and Developmental Biology, University College London, Gower Street, WC1E 6BT London, UK; Department of Dermatology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Qianru Mu
- Department of Cell and Developmental Biology, University College London, Gower Street, WC1E 6BT London, UK
| | - Gabriela C Brailoiu
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, 901 Walnut Street, Philadelphia, PA 19107, USA
| | - Gustavo J S Pereira
- Department of Cell and Developmental Biology, University College London, Gower Street, WC1E 6BT London, UK; Department of Pharmacology, Federal University of São Paulo (UNIFESP), São Paulo 04044-020, Brazil
| | - Stephen R Bolsover
- Department of Cell and Developmental Biology, University College London, Gower Street, WC1E 6BT London, UK
| | - Marco Keller
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilian University, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Franz Bracher
- Department of Pharmacy-Center for Drug Research, Ludwig-Maximilian University, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Christian Grimm
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilian University, Nussbaumstrasse 26, 80336 Munich, Germany; Immunology, Infection and Pandemic Research IIP, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt, Germany
| | - Eugen Brailoiu
- Department of Neural Sciences and Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Jonathan S Marchant
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - David I Yule
- Department of Pharmacology and Physiology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA.
| | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, Gower Street, WC1E 6BT London, UK.
| |
Collapse
|
11
|
Wible DJ, Parikh Z, Cho EJ, Chen MD, Jeter CR, Mukhopadhyay S, Dalby KN, Varadarajan S, Bratton SB. Unexpected inhibition of the lipid kinase PIKfyve reveals an epistatic role for p38 MAPKs in endolysosomal fission and volume control. Cell Death Dis 2024; 15:80. [PMID: 38253602 PMCID: PMC10803372 DOI: 10.1038/s41419-024-06423-0] [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: 05/09/2023] [Revised: 12/19/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024]
Abstract
p38 mitogen-activated protein kinases (MAPKs) participate in autophagic signaling; and previous reports suggest that pyridinyl imidazole p38 MAPK inhibitors, including SB203580 and SB202190, induce cell death in some cancer cell-types through unrestrained autophagy. Subsequent studies, however, have suggested that the associated cytoplasmic vacuolation resulted from off-target inhibition of an unidentified enzyme. Herein, we report that SB203580-induced vacuolation is rapid, reversible, and relies on the class III phosphatidylinositol 3-kinase (PIK3C3) complex and the production of phosphatidylinositol 3-phosphate [PI(3)P] but not on autophagy per se. Rather, vacuolation resulted from the accumulation of Rab7 on late endosome and lysosome (LEL) membranes, combined with an osmotic imbalance that triggered severe swelling in these organelles. Inhibition of PIKfyve, the lipid kinase that converts PI(3)P to PI(3,5)P2 on LEL membranes, produced a similar phenotype in cells; therefore, we performed in vitro kinase assays and discovered that both SB203580 and SB202190 directly inhibited recombinant PIKfyve. Cancer cells treated with either drug likewise displayed significant reductions in the endogenous levels of PI(3,5)P2. Despite these results, SB203580-induced vacuolation was not entirely due to off-target inhibition of PIKfyve, as a drug-resistant p38α mutant suppressed vacuolation; and combined genetic deletion of both p38α and p38β dramatically sensitized cells to established PIKfyve inhibitors, including YM201636 and apilimod. The rate of vacuole dissolution (i.e., LEL fission), following the removal of apilimod, was also significantly reduced in cells treated with BIRB-796, a structurally unrelated p38 MAPK inhibitor. Thus, our studies indicate that pyridinyl imidazole p38 MAPK inhibitors induce cytoplasmic vacuolation through the combined inhibition of both PIKfyve and p38 MAPKs, and more generally, that p38 MAPKs act epistatically to PIKfyve, most likely to promote LEL fission.
Collapse
Affiliation(s)
- Daric J Wible
- Department of Epigenetics & Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Zalak Parikh
- Department of Epigenetics & Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Eun Jeong Cho
- Targeted Therapeutic Drug Discovery and Development Program, Division of Chemical Biology & Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Miao-Der Chen
- Department of Epigenetics & Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Collene R Jeter
- Department of Epigenetics & Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Somshuvra Mukhopadhyay
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Kevin N Dalby
- Targeted Therapeutic Drug Discovery and Development Program, Division of Chemical Biology & Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Shankar Varadarajan
- Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - Shawn B Bratton
- Department of Epigenetics & Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA.
| |
Collapse
|
12
|
Davis LC, Morgan AJ, Galione A. Optical profiling of autonomous Ca 2+ nanodomains generated by lysosomal TPC2 and TRPML1. Cell Calcium 2023; 116:102801. [PMID: 37742482 DOI: 10.1016/j.ceca.2023.102801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/30/2023] [Accepted: 09/17/2023] [Indexed: 09/26/2023]
Abstract
Multiple families of Ca2+-permeable channels co-exist on lysosomal Ca2+ stores but how each family couples to its own unique downstream physiology is unclear. We have therefore investigated the Ca2+-signalling architecture underpinning different channels on the same vesicle that drive separate pathways, using phagocytosis as a physiological stimulus. Lysosomal Ca2+-channels are a major Ca2+ source driving particle uptake in macrophages, but different channels drive different aspects of Fc-receptor-mediated phagocytosis: TPC2 couples to dynamin activation, whilst TRPML1 couples to lysosomal exocytosis. We hypothesised that they are driven by discrete local plumes of Ca2+ around open channels (Ca2+ nanodomains). To test this, we optimized Ca2+-nanodomain recordings by screening panels of genetically encoded Ca2+ indicators (GECIs) fused to TPC2 to monitor the [Ca2+] next to the channel. Signal calibration accounting for the distance of the GECI from the channel mouth reveals that, during phagocytosis, TPC2 generates local Ca2+ nanodomains around itself of up to 42 µM, nearly a hundred-fold greater than the global cytosolic [Ca2+] rise. We further show that TPC2 and TRPML1, though on the same lysosomes, generate autonomous Ca2+ nanodomains of high [Ca2+] that are largely insulated from one another, a platform allowing their discrete Ca2+-decoding to promote unique respective physiologies.
Collapse
Affiliation(s)
- Lianne C Davis
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Anthony J Morgan
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK.
| |
Collapse
|
13
|
Soha SA, Santhireswaran A, Huq S, Casimir-Powell J, Jenkins N, Hodgson GK, Sugiyama M, Antonescu CN, Impellizzeri S, Botelho RJ. Improved imaging and preservation of lysosome dynamics using silver nanoparticle-enhanced fluorescence. Mol Biol Cell 2023; 34:ar96. [PMID: 37405751 PMCID: PMC10551705 DOI: 10.1091/mbc.e22-06-0200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/06/2023] Open
Abstract
The dynamics of living cells can be studied by live-cell fluorescence microscopy. However, this requires the use of excessive light energy to obtain good signal-to-noise ratio, which can then photobleach fluorochromes, and more worrisomely, lead to phototoxicity. Upon light excitation, noble metal nanoparticles such as silver nanoparticles (AgNPs) generate plasmons, which can then amplify excitation in direct proximity of the nanoparticle's surface and couple to the oscillating dipole of nearby radiating fluorophores, modifying their rate of emission and thus, enhancing their fluorescence. Here, we show that AgNPs fed to cells to accumulate within lysosomes enhanced the fluorescence of lysosome-targeted Alexa488-conjugated dextran, BODIPY-cholesterol, and DQ-BSA. Moreover, AgNP increased the fluorescence of GFP fused to the cytosolic tail of LAMP1, showing that metal enhanced fluorescence can occur across the lysosomal membrane. The inclusion of AgNPs in lysosomes did not disturb lysosomal properties such as lysosomal pH, degradative capacity, autophagy and autophagic flux, and membrane integrity, though AgNP seemed to increase basal lysosome tubulation. Importantly, by using AgNP, we could track lysosome motility with reduced laser power without damaging and altering lysosome dynamics. Overall, AgNP-enhanced fluorescence may be a useful tool to study the dynamics of the endo-lysosomal pathway while minimizing phototoxicity.
Collapse
Affiliation(s)
- Sumaiya A. Soha
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
| | - Araniy Santhireswaran
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
| | - Saaimatul Huq
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
| | - Jayde Casimir-Powell
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
| | - Nicala Jenkins
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
| | - Gregory K. Hodgson
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
| | - Michael Sugiyama
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
| | - Costin N. Antonescu
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
| | - Stefania Impellizzeri
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
| | - Roberto J. Botelho
- Molecular Science Graduate Program, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada, M5B 2K3
| |
Collapse
|
14
|
Peng W, Schröder LF, Song P, Wong YC, Krainc D. Parkin regulates amino acid homeostasis at mitochondria-lysosome (M/L) contact sites in Parkinson's disease. SCIENCE ADVANCES 2023; 9:eadh3347. [PMID: 37467322 PMCID: PMC10355824 DOI: 10.1126/sciadv.adh3347] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/15/2023] [Indexed: 07/21/2023]
Abstract
Mutations in the E3 ubiquitin ligase parkin are the most common cause of early-onset Parkinson's disease (PD). Although parkin modulates mitochondrial and endolysosomal homeostasis during cellular stress, whether parkin regulates mitochondrial and lysosomal cross-talk under physiologic conditions remains unresolved. Using transcriptomics, metabolomics and super-resolution microscopy, we identify amino acid metabolism as a disrupted pathway in iPSC-derived dopaminergic neurons from patients with parkin PD. Compared to isogenic controls, parkin mutant neurons exhibit decreased mitochondria-lysosome contacts via destabilization of active Rab7. Subcellular metabolomics in parkin mutant neurons reveals amino acid accumulation in lysosomes and their deficiency in mitochondria. Knockdown of the Rab7 GTPase-activating protein TBC1D15 restores mitochondria-lysosome tethering and ameliorates cellular and subcellular amino acid profiles in parkin mutant neurons. Our data thus uncover a function of parkin in promoting mitochondrial and lysosomal amino acid homeostasis through stabilization of mitochondria-lysosome contacts and suggest that modulation of interorganelle contacts may serve as a potential target for ameliorating amino acid dyshomeostasis in disease.
Collapse
Affiliation(s)
| | - Leonie F. Schröder
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Pingping Song
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | | | | |
Collapse
|
15
|
Raj N, Greune L, Kahms M, Mildner K, Franzkoch R, Psathaki OE, Zobel T, Zeuschner D, Klingauf J, Gerke V. Early Endosomes Act as Local Exocytosis Hubs to Repair Endothelial Membrane Damage. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300244. [PMID: 36938863 PMCID: PMC10161044 DOI: 10.1002/advs.202300244] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/21/2023] [Indexed: 05/06/2023]
Abstract
The plasma membrane of a cell is subject to stresses causing ruptures that must be repaired immediately to preserve membrane integrity and ensure cell survival. Yet, the spatio-temporal membrane dynamics at the wound site and the source of the membrane required for wound repair are poorly understood. Here, it is shown that early endosomes, previously only known to function in the uptake of extracellular material and its endocytic transport, are involved in plasma membrane repair in human endothelial cells. Using live-cell imaging and correlative light and electron microscopy, it is demonstrated that membrane injury triggers a previously unknown exocytosis of early endosomes that is induced by Ca2+ entering through the wound. This exocytosis is restricted to the vicinity of the wound site and mediated by the endosomal soluble N-ethylmaleimide sensitive factor attachment protein receptor (SNARE) VAMP2, which is crucial for efficient membrane repair. Thus, the newly identified Ca2+ -evoked and localized exocytosis of early endosomes supplies the membrane material required for rapid resealing of a damaged plasma membrane, thereby providing the first line of defense against damage in mechanically challenged endothelial cells.
Collapse
Affiliation(s)
- Nikita Raj
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation (ZMBE), Cells in Motion Interfaculty Center, University of Münster, 48149, Münster, Germany
| | - Lilo Greune
- Institute of Infectiology, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, 48149, Münster, Germany
| | - Martin Kahms
- Institute of Medical Physics and Biophysics, University of Münster, 48149, Münster, Germany
| | - Karina Mildner
- Electron Microscopy Facility, Max Planck Institute for Molecular Biomedicine, 48149, Münster, Germany
| | - Rico Franzkoch
- Department of Biology, integrated Bioimaging Facility (iBiOs), Center of Cellular Nanoanalytics (CellNanO), University of Osnabrück, 49076, Osnabrück, Germany
| | - Olympia Ekaterini Psathaki
- Department of Biology, integrated Bioimaging Facility (iBiOs), Center of Cellular Nanoanalytics (CellNanO), University of Osnabrück, 49076, Osnabrück, Germany
| | - Thomas Zobel
- Imaging Network, Cells in Motion Interfaculty Centre, University of Münster, 48149, Münster, Germany
| | - Dagmar Zeuschner
- Electron Microscopy Facility, Max Planck Institute for Molecular Biomedicine, 48149, Münster, Germany
| | - Jürgen Klingauf
- Institute of Medical Physics and Biophysics, University of Münster, 48149, Münster, Germany
| | - Volker Gerke
- Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation (ZMBE), Cells in Motion Interfaculty Center, University of Münster, 48149, Münster, Germany
| |
Collapse
|
16
|
Tingskov SJ, D'Agostino M, Login FH, Tamma G, Nejsum LN, Nørregaard R. Tamoxifen Affects Aquaporin-3 Expression and Subcellular Localization in Rat and Human Renal Collecting Ducts. Cells 2023; 12:cells12081140. [PMID: 37190049 DOI: 10.3390/cells12081140] [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: 02/27/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Sex hormones play an important role in the regulation of water homeostasis, and we have previously shown that tamoxifen (TAM), a selective estrogen receptor modulator (SERM), affects the regulation of aquaporin (AQP)-2. In this study, we investigated the effect of TAM on the expression and localization of AQP3 in collecting ducts using various animal, tissue, and cell models. The impact of TAM on AQP3 regulation was studied in rats subjected to 7 days of unilateral ureteral obstruction (UUO), with the rats fed a lithium-containing diet to induce nephrogenic diabetes insipidus (NDI), as well as in human precision-cut kidney slices (PCKS). Moreover, intracellular trafficking of AQP3 after TAM treatment was investigated in Madin-Darby Canine Kidney (MDCK) cells stably expressing AQP3. In all models, the expression of AQP3 was evaluated by Western blotting, immunohistochemistry and qPCR. TAM administration attenuated UUO-induced downregulation of AQP3 and affected the localization of AQP3 in both the UUO model and the lithium-induced NDI model. In parallel, TAM also affected the expression profile of other basolateral proteins, including AQP4 and Na/K-ATPase. In addition, TGF-β and TGF-β+TAM treatment affected the localization of AQP3 in stably transfected MDCK cells, and TAM partly attenuated the reduced AQP3 expression in TGF-β exposed human tissue slices. These findings suggest that TAM attenuates the downregulation of AQP3 in a UUO model and a lithium-induced NDI model and affects the intracellular localization in the collecting ducts.
Collapse
Affiliation(s)
| | - Mariagrazia D'Agostino
- Department of Bioscience, Biotechnologies and Environment, University of Bari, 70125 Bari, Italy
| | - Frédéric H Login
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus N, Denmark
| | - Grazia Tamma
- Department of Bioscience, Biotechnologies and Environment, University of Bari, 70125 Bari, Italy
| | - Lene N Nejsum
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus N, Denmark
| | - Rikke Nørregaard
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus N, Denmark
- Department of Renal Medicine, Aarhus University Hospital, 8200 Aarhus N, Denmark
| |
Collapse
|
17
|
Wible DJ, Parikh Z, Cho EJ, Chen MD, Mukhopadhyay S, Dalby KN, Varadarajan S, Bratton SB. Unexpected inhibition of the lipid kinase PIKfyve reveals an epistatic role for p38 MAPKs in endolysosomal fission and volume control. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.13.532495. [PMID: 36993747 PMCID: PMC10054966 DOI: 10.1101/2023.03.13.532495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
p38 mitogen-activated protein kinases (MAPKs) regulate early endocytic trafficking, but their effects on late endocytic trafficking remain unclear. Herein, we report that the pyridinyl imidazole p38 MAPK inhibitors, SB203580 and SB202190, induce a rapid but reversible Rab7-dependent accumulation of large cytoplasmic vacuoles. While SB203580 did not induce canonical autophagy, phosphatidylinositol 3-phosphate [PI(3)P] accumulated on vacuole membranes, and inhibition of the class III PI3-kinase (PIK3C3/VPS34) suppressed vacuolation. Ultimately, vacuolation resulted from the fusion of ER/Golgi-derived membrane vesicles with late endosomes and lysosomes (LELs), combined with an osmotic imbalance in LELs that led to severe swelling and a decrease in LEL fission. Since PIKfyve inhibitors induce a similar phenotype by preventing the conversion of PI(3)P to PI(3,5)P2, we performed in vitro kinase assays and found that PIKfyve activity was unexpectedly inhibited by SB203580 and SB202190, corresponding to losses in endogenous PI(3,5)P2 levels in treated cells. However, vacuolation was not entirely due to 'off-target' inhibition of PIKfyve by SB203580, as a drug-resistant p38α mutant suppressed vacuolation. Moreover, genetic deletion of both p38α and p38β rendered cells dramatically more sensitive to PIKfyve inhibitors, including YM201636 and apilimod. In subsequent 'washout' experiments, the rate of vacuole dissolution upon the removal of apilimod was also significantly reduced in cells treated with BIRB-796, a structurally unrelated p38 MAPK inhibitor. Thus, p38 MAPKs act epistatically to PIKfyve to promote LEL fission; and pyridinyl imidazole p38 MAPK inhibitors induce cytoplasmic vacuolation through the combined inhibition of both PIKfyve and p38 MAPKs.
Collapse
|
18
|
Bjørnestad SA, Solbakken MH, Jakobsen KS, Jentoft S, Bakke O, Progida C. Atlantic cod ( Gadus morhua) MHC I localizes to endolysosomal compartments independently of cytosolic sorting signals. Front Cell Dev Biol 2023; 11:1050323. [PMID: 36760361 PMCID: PMC9905690 DOI: 10.3389/fcell.2023.1050323] [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: 09/21/2022] [Accepted: 01/11/2023] [Indexed: 01/26/2023] Open
Abstract
Major histocompatibility complex (MHC) class I and II are crucial for the adaptive immune system because they are involved in peptide presentation to T cells. Until recently, it was believed that MHC genes and their associated immune components had been conserved since their evolutionary emergence in jawed fish. However, sequencing of the Atlantic cod (Gadus morhua) genome revealed a loss of MHC class II genes, and an extreme expansion of MHC class I genes. These findings lead to the hypothesis that a loss of the MHC class II pathway coincided with a more versatile use of MHC class I, but so far there is no direct experimental evidence in support of this. To gain a deeper understanding of the function of the expanded MHC class I, we selected five MHC class I gene variants representing five of the six clades identified in previous studies and investigated their intracellular localization in human and Atlantic cod larval cells. Intriguingly, we uncovered that all selected MHC class I variants localize to endolysosomal compartments in Atlantic cod cells. Additionally, by introducing point mutations or deletions in the cytosolic tail, we found that hypothetical sorting signals in the MHC class I cytosolic tail do not influence MHC class I trafficking. Moreover, we demonstrated that in Atlantic cod, tapasin and MHC class I colocalize on endolysosomes suggesting that peptide-loading assistance and stabilization of MHC class I occurs outside the endoplasmic reticulum. Altogether, our results demonstrate that MHC class I from Atlantic cod is sorted to the endolysosomal system, which may indicate that it interacts with exogenous peptides for potential cross presentation.
Collapse
Affiliation(s)
- Synne Arstad Bjørnestad
- Section of Physiology and Cell Biology, Department of Biosciences, University of Oslo, Oslo, Norway
| | - Monica Hongrø Solbakken
- Centre for Ecological and Evolutionary Synthesis (CEES), Department of Biosciences, University of Oslo, Oslo, Norway
| | - Kjetill S. Jakobsen
- Centre for Ecological and Evolutionary Synthesis (CEES), Department of Biosciences, University of Oslo, Oslo, Norway
| | - Sissel Jentoft
- Centre for Ecological and Evolutionary Synthesis (CEES), Department of Biosciences, University of Oslo, Oslo, Norway
| | - Oddmund Bakke
- Section of Physiology and Cell Biology, Department of Biosciences, University of Oslo, Oslo, Norway
| | - Cinzia Progida
- Section of Physiology and Cell Biology, Department of Biosciences, University of Oslo, Oslo, Norway,*Correspondence: Cinzia Progida,
| |
Collapse
|
19
|
Wen QX, Luo B, Xie XY, Zhou GF, Chen J, Song L, Liu Y, Xie SQ, Chen L, Li KY, Xiang XJ, Chen GJ. AP2S1 regulates APP degradation through late endosome-lysosome fusion in cells and APP/PS1 mice. Traffic 2023; 24:20-33. [PMID: 36412210 PMCID: PMC10107530 DOI: 10.1111/tra.12874] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 09/08/2022] [Accepted: 11/14/2022] [Indexed: 11/23/2022]
Abstract
AP2S1 is the sigma 2 subunit of adaptor protein 2 (AP2) that is essential for endocytosis. In this study, we investigated the potential role of AP2S1 in intracellular processing of amyloid precursor protein (APP), which contributes to the pathogenesis of Alzheimer disease (AD) by generating the toxic β-amyloid peptide (Aβ). We found that knockdown or overexpression of AP2S1 decreased or increased the protein levels of APP and Aβ in cells stably expressing human full-length APP695, respectively. This effect was unrelated to endocytosis but involved lysosomal degradation. Morphological studies revealed that silencing of AP2S1 promoted the translocalization of APP from RAB9-positive late endosomes (LE) to LAMP1-positive lysosomes, which was paralleled by the enhanced LE-lysosome fusion. In support, silencing of vacuolar protein sorting-associated protein 41 (VPS41) that is implicated in LE-lyso fusion prevented AP2S1-mediated regulation of APP degradation and translocalization. In APP/PS1 mice, an animal model of AD, AAV-mediated delivery of AP2S1 shRNA in the hippocampus significantly reduced the protein levels of APP and Aβ, with the concomitant APP translocalization, LE-lyso fusion and the improved cognitive functions. Taken together, these data uncover a LE-lyso fusion mechanism in APP degradation and suggest a novel role for AP2S1 in the pathophysiology of AD.
Collapse
Affiliation(s)
- Qi-Xin Wen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Biao Luo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Xiao-Yong Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Gui-Feng Zhou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Jian Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Li Song
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Yue Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Shi-Qi Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Long Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Kun-Yi Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Xiao-Jiao Xiang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guo-Jun Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Major Neurological and Mental Disorders, Chongqing Key Laboratory of Neurology, Chongqing, China.,Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, China
| |
Collapse
|
20
|
Capitini C, Pesce L, Fani G, Mazzamuto G, Genovese M, Franceschini A, Paoli P, Pieraccini G, Zasloff M, Chiti F, Pavone FS, Calamai M. Studying the trafficking of labeled trodusquemine and its application as nerve marker for light-sheet and expansion microscopy. FASEB J 2022; 36:e22655. [PMID: 36421008 PMCID: PMC9827910 DOI: 10.1096/fj.202201276r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 11/25/2022]
Abstract
Trodusquemine is an aminosterol with a variety of biological and pharmacological functions, such as acting as an antimicrobial, stimulating body weight loss and interfering with the toxicity of proteins involved in the development of Alzheimer's and Parkinson's diseases. The mechanisms of interaction of aminosterols with cells are, however, still largely uncharacterized. Here, by using fluorescently labeled trodusquemine (TRO-A594 and TRO-ATTO565), we show that trodusquemine binds initially to the plasma membrane of living cells, that the binding affinity is dependent on cholesterol, and that trodusquemine is then internalized and mainly targeted to lysosomes after internalization. We also found that TRO-A594 is able to strongly and selectively bind to myelinated fibers in fixed mouse brain slices, and that it is a marker compatible with tissue clearing and light-sheet fluorescence microscopy or expansion microscopy. In conclusion, this work contributes to further characterize the biology of aminosterols and provides a new tool for nerve labeling suitable for the most advanced microscopy techniques.
Collapse
Affiliation(s)
- Claudia Capitini
- European Laboratory for Non‐Linear Spectroscopy (LENS)University of FlorenceSesto FiorentinoItaly,Department of PhysicsUniversity of FlorenceSesto FiorentinoItaly
| | - Luca Pesce
- European Laboratory for Non‐Linear Spectroscopy (LENS)University of FlorenceSesto FiorentinoItaly,Department of PhysicsUniversity of FlorenceSesto FiorentinoItaly
| | - Giulia Fani
- Department of Experimental and Clinical Biomedical Sciences, Section of BiochemistryUniversity of FlorenceFlorenceItaly
| | - Giacomo Mazzamuto
- European Laboratory for Non‐Linear Spectroscopy (LENS)University of FlorenceSesto FiorentinoItaly,Department of PhysicsUniversity of FlorenceSesto FiorentinoItaly,National Institute of Optics – National Research Council (CNR‐INO)Sesto FiorentinoItaly
| | - Massimo Genovese
- Department of Experimental and Clinical Biomedical Sciences, Section of BiochemistryUniversity of FlorenceFlorenceItaly
| | - Alessandra Franceschini
- European Laboratory for Non‐Linear Spectroscopy (LENS)University of FlorenceSesto FiorentinoItaly,Department of PhysicsUniversity of FlorenceSesto FiorentinoItaly
| | - Paolo Paoli
- Department of Experimental and Clinical Biomedical Sciences, Section of BiochemistryUniversity of FlorenceFlorenceItaly
| | | | - Michael Zasloff
- Enterin Inc.PhiladelphiaPennsylvaniaUSA,MedStar‐Georgetown Transplant InstituteGeorgetown University School of MedicineWashingtonDCUSA
| | - Fabrizio Chiti
- Department of Experimental and Clinical Biomedical Sciences, Section of BiochemistryUniversity of FlorenceFlorenceItaly
| | - Francesco S. Pavone
- European Laboratory for Non‐Linear Spectroscopy (LENS)University of FlorenceSesto FiorentinoItaly,Department of PhysicsUniversity of FlorenceSesto FiorentinoItaly,National Institute of Optics – National Research Council (CNR‐INO)Sesto FiorentinoItaly
| | - Martino Calamai
- European Laboratory for Non‐Linear Spectroscopy (LENS)University of FlorenceSesto FiorentinoItaly,National Institute of Optics – National Research Council (CNR‐INO)Sesto FiorentinoItaly
| |
Collapse
|
21
|
Wong YC, Kim S, Cisneros J, Molakal CG, Song P, Lubbe SJ, Krainc D. Mid51/Fis1 mitochondrial oligomerization complex drives lysosomal untethering and network dynamics. J Cell Biol 2022; 221:213434. [PMID: 36044022 PMCID: PMC9437119 DOI: 10.1083/jcb.202206140] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 11/22/2022] Open
Abstract
Lysosomes are highly dynamic organelles implicated in multiple diseases. Using live super-resolution microscopy, we found that lysosomal tethering events rarely undergo lysosomal fusion, but rather untether over time to reorganize the lysosomal network. Inter-lysosomal untethering events are driven by a mitochondrial Mid51/Fis1 complex that undergoes coupled oligomerization on the outer mitochondrial membrane. Importantly, Fis1 oligomerization mediates TBC1D15 (Rab7-GAP) mitochondrial recruitment to drive inter-lysosomal untethering via Rab7 GTP hydrolysis. Moreover, inhibiting Fis1 oligomerization by either mutant Fis1 or a Mid51 oligomerization mutant potentially associated with Parkinson’s disease prevents lysosomal untethering events, resulting in misregulated lysosomal network dynamics. In contrast, dominant optic atrophy–linked mutant Mid51, which does not inhibit Mid51/Fis1 coupled oligomerization, does not disrupt downstream lysosomal dynamics. As Fis1 conversely also regulates Mid51 oligomerization, our work further highlights an oligomeric Mid51/Fis1 mitochondrial complex that mechanistically couples together both Drp1 and Rab7 GTP hydrolysis machinery at mitochondria–lysosome contact sites. These findings have significant implications for organelle networks in cellular homeostasis and human disease.
Collapse
Affiliation(s)
- Yvette C Wong
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Soojin Kim
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Jasmine Cisneros
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Catherine G Molakal
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Pingping Song
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Steven J Lubbe
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Simpson Querrey Center for Neurogenetics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Dimitri Krainc
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL.,Simpson Querrey Center for Neurogenetics, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
22
|
Meiring JCM, Grigoriev I, Nijenhuis W, Kapitein LC, Akhmanova A. Opto-katanin, an optogenetic tool for localized, microtubule disassembly. Curr Biol 2022; 32:4660-4674.e6. [PMID: 36174574 DOI: 10.1016/j.cub.2022.09.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 08/01/2022] [Accepted: 09/06/2022] [Indexed: 11/30/2022]
Abstract
Microtubules are cytoskeletal polymers that separate chromosomes during mitosis and serve as rails for intracellular transport and organelle positioning. Manipulation of microtubules is widely used in cell and developmental biology, but tools for precise subcellular spatiotemporal control of microtubules are currently lacking. Here, we describe a light-activated system for localized recruitment of the microtubule-severing enzyme katanin. This system, named opto-katanin, uses targeted illumination with blue light to induce rapid, localized, and reversible microtubule depolymerization. This tool allows precise clearing of a subcellular region of microtubules while preserving the rest of the microtubule network, demonstrating that regulation of katanin recruitment to microtubules is sufficient to control its severing activity. The tool is not toxic in the absence of blue light and can be used to disassemble both dynamic and stable microtubules in primary neurons as well as in dividing cells. We show that opto-katanin can be used to locally block vesicle transport and to clarify the dependence of organelle morphology and dynamics on microtubules. Specifically, our data indicate that microtubules are not required for the maintenance of the Golgi stacks or the tubules of the endoplasmic reticulum but are needed for the formation of new membrane tubules. Finally, we demonstrate that this tool can be applied to study the contribution of microtubules to cell mechanics by showing that microtubule bundles can exert forces constricting the nucleus.
Collapse
Affiliation(s)
- Joyce C M Meiring
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan, Utrecht 3584 CS, the Netherlands
| | - Ilya Grigoriev
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan, Utrecht 3584 CS, the Netherlands
| | - Wilco Nijenhuis
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan, Utrecht 3584 CS, the Netherlands; Center for Living Technologies, Eindhoven-Wageningen-Utrecht Alliance, UMC Utrecht, Utrecht 3584 CB, the Netherlands
| | - Lukas C Kapitein
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan, Utrecht 3584 CS, the Netherlands; Center for Living Technologies, Eindhoven-Wageningen-Utrecht Alliance, UMC Utrecht, Utrecht 3584 CB, the Netherlands
| | - Anna Akhmanova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan, Utrecht 3584 CS, the Netherlands.
| |
Collapse
|
23
|
Rhys GG, Cross JA, Dawson WM, Thompson HF, Shanmugaratnam S, Savery NJ, Dodding MP, Höcker B, Woolfson DN. De novo designed peptides for cellular delivery and subcellular localisation. Nat Chem Biol 2022; 18:999-1004. [PMID: 35836017 DOI: 10.1038/s41589-022-01076-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 06/03/2022] [Indexed: 12/14/2022]
Abstract
Increasingly, it is possible to design peptide and protein assemblies de novo from first principles or computationally. This approach provides new routes to functional synthetic polypeptides, including designs to target and bind proteins of interest. Much of this work has been developed in vitro. Therefore, a challenge is to deliver de novo polypeptides efficiently to sites of action within cells. Here we describe the design, characterisation, intracellular delivery, and subcellular localisation of a de novo synthetic peptide system. This system comprises a dual-function basic peptide, programmed both for cell penetration and target binding, and a complementary acidic peptide that can be fused to proteins of interest and introduced into cells using synthetic DNA. The designs are characterised in vitro using biophysical methods and X-ray crystallography. The utility of the system for delivery into mammalian cells and subcellular targeting is demonstrated by marking organelles and actively engaging functional protein complexes.
Collapse
Affiliation(s)
- Guto G Rhys
- Department of Biochemistry, University of Bayreuth, Bayreuth, Germany
| | - Jessica A Cross
- School of Chemistry, University of Bristol, Bristol, UK.,School of Biochemistry, University of Bristol, Bristol, UK
| | | | - Harry F Thompson
- School of Chemistry, University of Bristol, Bristol, UK.,School of Biochemistry, University of Bristol, Bristol, UK
| | | | - Nigel J Savery
- School of Biochemistry, University of Bristol, Bristol, UK.,BrisSynBio, University of Bristol, Bristol, UK
| | - Mark P Dodding
- School of Biochemistry, University of Bristol, Bristol, UK
| | - Birte Höcker
- Department of Biochemistry, University of Bayreuth, Bayreuth, Germany.
| | - Derek N Woolfson
- School of Chemistry, University of Bristol, Bristol, UK. .,School of Biochemistry, University of Bristol, Bristol, UK. .,BrisSynBio, University of Bristol, Bristol, UK.
| |
Collapse
|
24
|
Segregated cation flux by TPC2 biases Ca 2+ signaling through lysosomes. Nat Commun 2022; 13:4481. [PMID: 35918320 PMCID: PMC9346130 DOI: 10.1038/s41467-022-31959-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 07/12/2022] [Indexed: 12/19/2022] Open
Abstract
Two-pore channels are endo-lysosomal cation channels with malleable selectivity filters that drive endocytic ion flux and membrane traffic. Here we show that TPC2 can differentially regulate its cation permeability when co-activated by its endogenous ligands, NAADP and PI(3,5)P2. Whereas NAADP rendered the channel Ca2+-permeable and PI(3,5)P2 rendered the channel Na+-selective, a combination of the two increased Ca2+ but not Na+ flux. Mechanistically, this was due to an increase in Ca2+ permeability independent of changes in ion selectivity. Functionally, we show that cell permeable NAADP and PI(3,5)P2 mimetics synergistically activate native TPC2 channels in live cells, globalizing cytosolic Ca2+ signals and regulating lysosomal pH and motility. Our data reveal that flux of different ions through the same pore can be independently controlled and identify TPC2 as a likely coincidence detector that optimizes lysosomal Ca2+ signaling. TPC2 is a lysosomal ion channel permeable to both calcium and sodium ions. Here, the authors show that TPC2 can selectively increase its calcium permeability when simultaneously challenged by both its natural activators- NAADP and PI(3,5)P2.
Collapse
|
25
|
Duan L, Zaepfel BL, Aksenova V, Dasso M, Rothstein JD, Kalab P, Hayes LR. Nuclear RNA binding regulates TDP-43 nuclear localization and passive nuclear export. Cell Rep 2022; 40:111106. [PMID: 35858577 PMCID: PMC9345261 DOI: 10.1016/j.celrep.2022.111106] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 03/26/2022] [Accepted: 06/27/2022] [Indexed: 11/27/2022] Open
Abstract
Nuclear clearance of the RNA-binding protein TDP-43 is a hallmark of neurodegeneration and an important therapeutic target. Our current understanding of TDP-43 nucleocytoplasmic transport does not fully explain its predominantly nuclear localization or mislocalization in disease. Here, we show that TDP-43 exits nuclei by passive diffusion, independent of facilitated mRNA export. RNA polymerase II blockade and RNase treatment induce TDP-43 nuclear efflux, suggesting that nuclear RNAs sequester TDP-43 in nuclei and limit its availability for passive export. Induction of TDP-43 nuclear efflux by short, GU-rich oligomers (presumably by outcompeting TDP-43 binding to endogenous nuclear RNAs), and nuclear retention conferred by splicing inhibition, demonstrate that nuclear TDP-43 localization depends on binding to GU-rich nuclear RNAs. Indeed, RNA-binding domain mutations markedly reduce TDP-43 nuclear localization and abolish transcription blockade-induced nuclear efflux. Thus, the nuclear abundance of GU-RNAs, dictated by the balance of transcription, pre-mRNA processing, and RNA export, regulates TDP-43 nuclear localization.
Collapse
Affiliation(s)
- Lauren Duan
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Benjamin L Zaepfel
- Biochemistry, Cellular and Molecular Biology Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Vasilisa Aksenova
- Division of Molecular and Cellular Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mary Dasso
- Division of Molecular and Cellular Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jeffrey D Rothstein
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Petr Kalab
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.
| | - Lindsey R Hayes
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
26
|
Jing Y, Kobayashi M, Vu HT, Kasahara A, Chen X, Pham LT, Kurayoshi K, Tadokoro Y, Ueno M, Todo T, Nakada M, Hirao A. Therapeutic advantage of targeting lysosomal membrane integrity supported by lysophagy in malignant glioma. Cancer Sci 2022; 113:2716-2726. [PMID: 35657693 PMCID: PMC9357661 DOI: 10.1111/cas.15451] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/26/2022] [Accepted: 05/31/2022] [Indexed: 12/01/2022] Open
Abstract
Lysosomes function as the digestive system of a cell and are involved in macromolecular recycling, vesicle trafficking, metabolic reprogramming, and progrowth signaling. Although quality control of lysosome biogenesis is thought to be a potential target for cancer therapy, practical strategies have not been established. Here, we show that lysosomal membrane integrity supported by lysophagy, a selective autophagy for damaged lysosomes, is a promising therapeutic target for glioblastoma (GBM). In this study, we found that ifenprodil, an FDA‐approved drug with neuromodulatory activities, efficiently inhibited spheroid formation of patient‐derived GBM cells in a combination with autophagy inhibition. Ifenprodil increased intracellular Ca2+ level, resulting in mitochondrial reactive oxygen species–mediated cytotoxicity. The ifenprodil‐induced Ca2+ elevation was due to Ca2+ release from lysosomes, but not endoplasmic reticulum, associated with galectin‐3 punctation as an indicator of lysosomal membrane damage. As the Ca2+ release was enhanced by ATG5 deficiency, autophagy protected against lysosomal membrane damage. By comparative analysis of 765 FDA‐approved compounds, we identified another clinically available drug for central nervous system (CNS) diseases, amoxapine, in addition to ifenprodil. Both compounds promoted degradation of lysosomal membrane proteins, indicating a critical role of lysophagy in quality control of lysosomal membrane integrity. Importantly, a synergistic inhibitory effect of ifenprodil and chloroquine, a clinically available autophagy inhibitor, on spheroid formation was remarkable in GBM cells, but not in nontransformed neural progenitor cells. Finally, chloroquine dramatically enhanced effects of the compounds inducing lysosomal membrane damage in a patient‐derived xenograft model. These data demonstrate a therapeutic advantage of targeting lysosomal membrane integrity in GBM.
Collapse
Affiliation(s)
- Yongwei Jing
- Division of Molecular Genetics Cancer Research Institute Kanazawa University, Kakuma‐machi, Kanazawa 920‐1192 Japan
| | - Masahiko Kobayashi
- Division of Molecular Genetics Cancer Research Institute Kanazawa University, Kakuma‐machi, Kanazawa 920‐1192 Japan
| | - Ha Thi Vu
- Division of Molecular Genetics Cancer Research Institute Kanazawa University, Kakuma‐machi, Kanazawa 920‐1192 Japan
- Present address: Department of Molecular Biology and Genetics Hanoi Medical University No1‐Ton That Tung street‐Dong Da district, Ha Noi Vietnam
| | - Atsuko Kasahara
- Institute for Frontier Science Initiative Kanazawa University, Kakuma‐machi, Kanazawa, 920‐1192 Japan
- WPI Nano Life Science Institute (WPI‐Nano LSI) Kanazawa University, Kakuma‐machi, Kanazawa 920‐1192 Japan
| | - Xi Chen
- Division of Molecular Genetics Cancer Research Institute Kanazawa University, Kakuma‐machi, Kanazawa 920‐1192 Japan
| | - Loc Thi Pham
- Division of Molecular Genetics Cancer Research Institute Kanazawa University, Kakuma‐machi, Kanazawa 920‐1192 Japan
| | - Kenta Kurayoshi
- Division of Molecular Genetics Cancer Research Institute Kanazawa University, Kakuma‐machi, Kanazawa 920‐1192 Japan
| | - Yuko Tadokoro
- Division of Molecular Genetics Cancer Research Institute Kanazawa University, Kakuma‐machi, Kanazawa 920‐1192 Japan
- WPI Nano Life Science Institute (WPI‐Nano LSI) Kanazawa University, Kakuma‐machi, Kanazawa 920‐1192 Japan
| | - Masaya Ueno
- Division of Molecular Genetics Cancer Research Institute Kanazawa University, Kakuma‐machi, Kanazawa 920‐1192 Japan
- WPI Nano Life Science Institute (WPI‐Nano LSI) Kanazawa University, Kakuma‐machi, Kanazawa 920‐1192 Japan
| | - Tomoki Todo
- Division of Innovative Cancer Therapy, Institute of Medical Science The University of Tokyo Tokyo Japan
| | - Mitsutoshi Nakada
- Department of Neurosurgery, Graduate School of Medical Science Kanazawa University Kanazawa Ishikawa Japan
| | - Atsushi Hirao
- Division of Molecular Genetics Cancer Research Institute Kanazawa University, Kakuma‐machi, Kanazawa 920‐1192 Japan
- WPI Nano Life Science Institute (WPI‐Nano LSI) Kanazawa University, Kakuma‐machi, Kanazawa 920‐1192 Japan
| |
Collapse
|
27
|
Belton TB, Leisten ED, Cisneros J, Wong YC. Live cell microscopy of mitochondria-lysosome contact site formation and tethering dynamics. STAR Protoc 2022; 3:101262. [PMID: 35330964 PMCID: PMC8938322 DOI: 10.1016/j.xpro.2022.101262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mitochondria-lysosome contact sites are critical for maintaining cellular homeostasis by regulating mitochondrial and lysosomal network dynamics and mediating metabolite exchange. Here, we present a protocol to quantitatively analyze the formation and tethering duration of mitochondria-lysosome contact sites by using time-lapse live confocal microscopy of LAMP1 and TOMM20. Although this protocol focuses on mammalian HeLa cells, it can be applied to other cell types for further studies on mitochondria-lysosome contact regulation and function, and elucidation of their role in human disorders. For complete details on the use and execution of this protocol, please refer to Wong et al. (2018) and Wong et al. (2019b). Visualization of mitochondrial and lysosomal dynamics by confocal microscopy Imaging of mitochondria-lysosome contact sites in live HeLa cells Quantification of contact site dynamics using TOMM20/LAMP1 Analysis of contact site tethering duration over time
Collapse
|
28
|
Frei MS, Tarnawski M, Roberti MJ, Koch B, Hiblot J, Johnsson K. Engineered HaloTag variants for fluorescence lifetime multiplexing. Nat Methods 2022; 19:65-70. [PMID: 34916672 PMCID: PMC8748199 DOI: 10.1038/s41592-021-01341-x] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 11/02/2021] [Indexed: 12/03/2022]
Abstract
Self-labeling protein tags such as HaloTag are powerful tools that can label fusion proteins with synthetic fluorophores for use in fluorescence microscopy. Here we introduce HaloTag variants with either increased or decreased brightness and fluorescence lifetime compared with HaloTag7 when labeled with rhodamines. Combining these HaloTag variants enabled live-cell fluorescence lifetime multiplexing of three cellular targets in one spectral channel using a single fluorophore and the generation of a fluorescence lifetime-based biosensor. Additionally, the brightest HaloTag variant showed up to 40% higher brightness in live-cell imaging applications.
Collapse
Affiliation(s)
- Michelle S Frei
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany
- Institute of Chemical Sciences and Engineering (ISIC), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Miroslaw Tarnawski
- Protein Expression and Characterization Facility, Max Planck Institute for Medical Research, Heidelberg, Germany
| | | | - Birgit Koch
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Julien Hiblot
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Kai Johnsson
- Department of Chemical Biology, Max Planck Institute for Medical Research, Heidelberg, Germany.
- Institute of Chemical Sciences and Engineering (ISIC), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
29
|
Möller K, Brambach M, Villani A, Gallo E, Gilmour D, Peri F. A role for the centrosome in regulating the rate of neuronal efferocytosis by microglia in vivo. eLife 2022; 11:82094. [PMID: 36398880 PMCID: PMC9674339 DOI: 10.7554/elife.82094] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/25/2022] [Indexed: 11/19/2022] Open
Abstract
During brain development, many newborn neurons undergo apoptosis and are engulfed by microglia, the tissue-resident phagocytes of the brain, in a process known as efferocytosis. A hallmark of microglia is their highly branched morphology characterized by the presence of numerous dynamic extensions that these cells use for scanning the brain parenchyma and engulfing unwanted material. The mechanisms driving branch formation and apoptotic cell engulfment in microglia are unclear. By taking a live-imaging approach in zebrafish, we show that while microglia generate multiple microtubule-based branches, they only successfully engulf one apoptotic neuron at a time. Further investigation into the mechanism underlying this sequential engulfment revealed that targeted migration of the centrosome into one branch is predictive of phagosome formation and polarized vesicular trafficking. Moreover, experimentally doubling centrosomal numbers in microglia increases the rate of engulfment and even allows microglia to remove two neurons simultaneously, providing direct supporting evidence for a model where centrosomal migration is a rate-limiting step in branch-mediated efferocytosis. Conversely, light-mediated depolymerization of microtubules causes microglia to lose their typical branched morphology and switch to an alternative mode of engulfment, characterized by directed migration towards target neurons, revealing unexpected plasticity in their phagocytic ability. Finally, building on work focusing on the establishment of the immunological synapse, we identified a conserved signalling pathway underlying centrosomal movement in engulfing microglia.
Collapse
Affiliation(s)
- Katrin Möller
- Department of Molecular Life Sciences, University of ZürichZürichSwitzerland
| | - Max Brambach
- Department of Molecular Life Sciences, University of ZürichZürichSwitzerland
| | - Ambra Villani
- Department of Molecular Life Sciences, University of ZürichZürichSwitzerland
| | - Elisa Gallo
- Department of Molecular Life Sciences, University of ZürichZürichSwitzerland
| | - Darren Gilmour
- Department of Molecular Life Sciences, University of ZürichZürichSwitzerland
| | - Francesca Peri
- Department of Molecular Life Sciences, University of ZürichZürichSwitzerland
| |
Collapse
|
30
|
Brandt JN, Voss L, Rambo FM, Nicholson K, Thein JR, Fairchild L, Seabrook L, Lewis D, Guevara-Hernandez L, White ML, Sax L, Eichten V, Harper L, Hermann GJ. Asymmetric organelle positioning during epithelial polarization of C. elegans intestinal cells. Dev Biol 2022; 481:75-94. [PMID: 34597675 PMCID: PMC8665101 DOI: 10.1016/j.ydbio.2021.09.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/11/2021] [Accepted: 09/24/2021] [Indexed: 01/03/2023]
Abstract
While the epithelial cell cortex displays profound asymmetries in protein distribution and morphology along the apico-basal axis, the extent to which the cytoplasm is similarly polarized within epithelial cells remains relatively unexplored. We show that cytoplasmic organelles within C. elegans embryonic intestinal cells develop extensive apico-basal polarity at the time they establish cortical asymmetry. Nuclei and conventional endosomes, including early endosomes, late endosomes, and lysosomes, become polarized apically. Lysosome-related gut granules, yolk platelets, and lipid droplets become basally enriched. Removal of par-3 activity does not disrupt organelle positioning, indicating that cytoplasmic apico-basal asymmetry is independent of the PAR polarity pathway. Blocking the apical migration of nuclei leads to the apical positioning of gut granules and yolk platelets, whereas the asymmetric localization of conventional endosomes and lipid droplets is unaltered. This suggests that nuclear positioning organizes some, but not all, cytoplasmic asymmetries in this cell type. We show that gut granules become apically enriched when WHT-2 and WHT-7 function is disrupted, identifying a novel role for ABCG transporters in gut granule positioning during epithelial polarization. Analysis of WHT-2 and WHT-7 ATPase mutants is consistent with a WHT-2/WHT-7 heterodimer acting as a transporter in gut granule positioning. In wht-2(-) mutants, the polarized distribution of other organelles is not altered and gut granules do not take on characteristics of conventional endosomes that could have explained their apical mispositioning. During epithelial polarization wht-2(-) gut granules exhibit a loss of the Rab32/38 family member GLO-1 and ectopic expression of GLO-1 is sufficient to rescue the basal positioning of wht-2(-) and wht-7(-) gut granules. Furthermore, depletion of GLO-1 causes the mislocalization of the endolysosomal RAB-7 to gut granules and RAB-7 drives the apical mispositioning of gut granules when GLO-1, WHT-2, or WHT-7 function is disrupted. We suggest that ABC transporters residing on gut granules can regulate Rab dynamics to control organelle positioning during epithelial polarization.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Greg J. Hermann
- Corresponding author. Department of Biology, Lewis & Clark College, Portland, OR, USA, (G.J. Hermann)
| |
Collapse
|
31
|
Podinovskaia M, Prescianotto-Baschong C, Buser DP, Spang A. A novel live-cell imaging assay reveals regulation of endosome maturation. eLife 2021; 10:e70982. [PMID: 34846303 PMCID: PMC8635980 DOI: 10.7554/elife.70982] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Cell-cell communication is an essential process in life, with endosomes acting as key organelles for regulating uptake and secretion of signaling molecules. Endocytosed material is accepted by the sorting endosome where it either is sorted for recycling or remains in the endosome as it matures to be degraded in the lysosome. Investigation of the endosome maturation process has been hampered by the small size and rapid movement of endosomes in most cellular systems. Here, we report an easy versatile live-cell imaging assay to monitor endosome maturation kinetics, which can be applied to a variety of mammalian cell types. Acute ionophore treatment led to enlarged early endosomal compartments that matured into late endosomes and fused with lysosomes to form endolysosomes. Rab5-to-Rab7 conversion and PI(3)P formation and turn over were recapitulated with this assay and could be observed with a standard widefield microscope. We used this approach to show that Snx1 and Rab11-positive recycling endosome recruitment occurred throughout endosome maturation and was uncoupled from Rab conversion. In contrast, efficient endosomal acidification was dependent on Rab conversion. The assay provides a powerful tool to further unravel various aspects of endosome maturation.
Collapse
Affiliation(s)
| | | | | | - Anne Spang
- Biozentrum, University of BaselBaselSwitzerland
| |
Collapse
|
32
|
George G, Ninagawa S, Yagi H, Furukawa JI, Hashii N, Ishii-Watabe A, Deng Y, Matsushita K, Ishikawa T, Mamahit YP, Maki Y, Kajihara Y, Kato K, Okada T, Mori K. Purified EDEM3 or EDEM1 alone produces determinant oligosaccharide structures from M8B in mammalian glycoprotein ERAD. eLife 2021; 10:70357. [PMID: 34698634 PMCID: PMC8570694 DOI: 10.7554/elife.70357] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 10/07/2021] [Indexed: 12/04/2022] Open
Abstract
Sequential mannose trimming of N-glycan, from M9 to M8B and then to oligosaccharides exposing the α1,6-linked mannosyl residue (M7A, M6, and M5), facilitates endoplasmic reticulum-associated degradation of misfolded glycoproteins (gpERAD). We previously showed that EDEM2 stably disulfide-bonded to the thioredoxin domain-containing protein TXNDC11 is responsible for the first step (George et al., 2020). Here, we show that EDEM3 and EDEM1 are responsible for the second step. Incubation of pyridylamine-labeled M8B with purified EDEM3 alone produced M7 (M7A and M7C), M6, and M5. EDEM1 showed a similar tendency, although much lower amounts of M6 and M5 were produced. Thus, EDEM3 is a major α1,2-mannosidase for the second step from M8B. Both EDEM3 and EDEM1 trimmed M8B from a glycoprotein efficiently. Our confirmation of the Golgi localization of MAN1B indicates that no other α1,2-mannosidase is required for gpERAD. Accordingly, we have established the entire route of oligosaccharide processing and the enzymes responsible.
Collapse
Affiliation(s)
- Ginto George
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan
| | - Satoshi Ninagawa
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan
| | - Hirokazu Yagi
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Jun-Ichi Furukawa
- Department of Advanced Clinical Glycobiology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Noritaka Hashii
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Kawasaki, Japan
| | - Akiko Ishii-Watabe
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Kawasaki, Japan
| | - Ying Deng
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan
| | - Kazutoshi Matsushita
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan
| | - Tokiro Ishikawa
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan
| | - Yugoviandi P Mamahit
- Department of Chemistry, Graduate School of Science, Osaka University, Toyonaka, Japan
| | - Yuta Maki
- Department of Chemistry, Graduate School of Science, Osaka University, Toyonaka, Japan.,Project Research Center for Fundamental Sciences, Graduate School of Science, Osaka University, Toyonaka, Japan
| | - Yasuhiro Kajihara
- Department of Chemistry, Graduate School of Science, Osaka University, Toyonaka, Japan.,Project Research Center for Fundamental Sciences, Graduate School of Science, Osaka University, Toyonaka, Japan
| | - Koichi Kato
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.,Exploratory Research Center on Life and Living Systems (ExCELLS) and Institute for Molecular Science, National Institutes of Natural Sciences, Okazaki, Japan
| | - Tetsuya Okada
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan
| | - Kazutoshi Mori
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan
| |
Collapse
|
33
|
Sousa D, Lima RT, Lopes-Rodrigues V, Gonzalez E, Royo F, Xavier CPR, Falcón-Pérez JM, Vasconcelos MH. Different Ability of Multidrug-Resistant and -Sensitive Counterpart Cells to Release and Capture Extracellular Vesicles. Cells 2021; 10:cells10112886. [PMID: 34831110 PMCID: PMC8616370 DOI: 10.3390/cells10112886] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer multidrug resistance (MDR) is one of the main challenges for cancer treatment efficacy. MDR is a phenomenon by which tumor cells become resistant to several unrelated drugs. Some studies have previously described the important role of extracellular vesicles (EVs) in the dissemination of a MDR phenotype. EVs’ cargo may include different players of MDR, such as microRNAS and drug-efflux pumps, which may be transferred from donor MDR cells to recipient drug-sensitive counterparts. The present work aimed to: (i) compare the ability of drug-sensitive and their MDR counterpart cells to release and capture EVs and (ii) study and relate those differences with possible distinct fate of the endocytic pathway in these counterpart cells. Our results showed that MDR cells released more EVs than their drug-sensitive counterparts and also that the drug-sensitive cells captured more EVs than their MDR counterparts. This difference in the release and capture of EVs may be associated with differences in the endocytic pathway between drug-sensitive and MDR cells. Importantly, manipulation of the recycling pathway influenced the response of drug-sensitive cells to doxorubicin treatment.
Collapse
Affiliation(s)
- Diana Sousa
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (D.S.); (R.T.L.); (V.L.-R.); (C.P.R.X.)
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, FFUP—Faculty of Pharmacy of the University of Porto, 4050-313 Porto, Portugal
| | - Raquel T. Lima
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (D.S.); (R.T.L.); (V.L.-R.); (C.P.R.X.)
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Department of Pathology, FMUP—Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
- Cancer Signaling & Metabolism Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Vanessa Lopes-Rodrigues
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (D.S.); (R.T.L.); (V.L.-R.); (C.P.R.X.)
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- ICBAS-UP—Institute of Biomedical Sciences Abel Salazar of the University of Porto, 4099-003 Porto, Portugal
| | - Esperanza Gonzalez
- Exosomes Lab. & Metabolomics Platform, CIC bioGUNE, CIBERehd, 28160 Derio, Spain; (E.G.); (F.R.); (J.M.F.-P.)
| | - Félix Royo
- Exosomes Lab. & Metabolomics Platform, CIC bioGUNE, CIBERehd, 28160 Derio, Spain; (E.G.); (F.R.); (J.M.F.-P.)
| | - Cristina P. R. Xavier
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (D.S.); (R.T.L.); (V.L.-R.); (C.P.R.X.)
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Juan M. Falcón-Pérez
- Exosomes Lab. & Metabolomics Platform, CIC bioGUNE, CIBERehd, 28160 Derio, Spain; (E.G.); (F.R.); (J.M.F.-P.)
- IKERBASQUE Basque Foundation for Science, 48013 Bilbao, Spain
| | - M. Helena Vasconcelos
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (D.S.); (R.T.L.); (V.L.-R.); (C.P.R.X.)
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, FFUP—Faculty of Pharmacy of the University of Porto, 4050-313 Porto, Portugal
- Correspondence: ; Tel.: +351-225-570-772
| |
Collapse
|
34
|
Iqbal S, Luo B, Melamed JR, Day ES. Critical Evaluation of Different Lysosomal Labeling Methods Used to Analyze RNA Nanocarrier Trafficking in Cells. Bioconjug Chem 2021; 32:2245-2256. [PMID: 34543006 PMCID: PMC9014481 DOI: 10.1021/acs.bioconjchem.1c00405] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The use of nucleic acids to regulate gene expression is a rapidly developing field with immense clinical potential. Nanomaterials are frequently used to deliver nucleic acids into cells as they can overcome the poor cellular uptake and endo/lysosomal degradation of bare nucleic acids. For these nanocarriers to be effective, they must escape endo/lysosomal compartments to deliver their nucleic acid cargo into the cytosol (for ribonucleic acid (RNA)) or nucleus (for deoxyribonucleic acid (DNA)). This process is poorly understood and remains an area of active research toward the goal of developing effective delivery strategies. Fluorescent endo/lysosomal markers are among the most widely employed tools used to evaluate the endosomal escape of nucleic acid nanocarriers. However, the endo/lysosomal labeling method may alter the extent of and route of nanocarrier uptake by cells. The impact of these markers on cellular function and cell-nanocarrier interactions has not been probed in a systematic manner. To investigate this, we compared the effects of several common lysosomal labeling methods, namely, LysoTracker Red (LT Red), transient lysosomal-associated membrane protein 1-mutant green fluorescent protein (LAMP1-mGFP) transfection (Transient GFP), and stable lentiviral LAMP1-mGFP transfection (Stable GFP), on cellular metabolic activity, nanocarrier uptake, nanocarrier/lysosomal label colocalization, and gene silencing potency in U87 glioblastoma and MDA-MB-231 breast cancer cells using polyethyleneimine (PEI)/ribonucleic acid (RNA) polyplexes as a model nanocarrier. In both U87s and MDA-MB-231s, Transient GFP and LT Red labeling reduced metabolic activity relative to untransfected (Parental) cells, while Stable GFP labeling increased metabolic activity. Congruently, flow cytometry indicates Stable GFP cells have greater polyplex uptake than LT Red-labeled cells in both cell lines. Despite these similar trends in uptake, polyplex intracellular trafficking differs in the two cell lines, as confocal imaging revealed greater polyplex/lysosome colocalization in Stable GFP U87 cells than LT Red-labeled U87 cells, while the trend was reversed in MBA-MB-231s. The level of RNA-mediated gene silencing achieved in Parental versus Stable GFP U87 and MDA-MB-231 cells agreed with the observed levels of polyplex/lysosome colocalization, supporting the established concept that endosomal escape is the rate-limiting step for RNA interference. These findings indicate that lysosomal labels can profoundly alter cellular function and cell-nanocarrier interactions, presenting critical new considerations for researchers investigating nanoparticle trafficking.
Collapse
Affiliation(s)
- Shoaib Iqbal
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, Delaware 19716, United States
| | - Benjamin Luo
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, Delaware 19716, United States
| | - Jilian R Melamed
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, Delaware 19716, United States
| | - Emily S Day
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, Delaware 19716, United States
- Department of Materials Science & Engineering, University of Delaware, 201 DuPont Hall, Newark, Delaware 19716, United States
- Helen F. Graham Cancer Center & Research Institute, 4701 Ogletown Stanton Road, Newark, Delaware 19713, United States
| |
Collapse
|
35
|
Dilna A, Deepak KV, Damodaran N, Kielkopf CS, Kagedal K, Ollinger K, Nath S. Amyloid-β induced membrane damage instigates tunneling nanotube-like conduits by p21-activated kinase dependent actin remodulation. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166246. [PMID: 34403739 DOI: 10.1016/j.bbadis.2021.166246] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/19/2021] [Accepted: 08/09/2021] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) pathology progresses gradually via anatomically connected brain regions. Direct transfer of amyloid-β1-42 oligomers (oAβ) between connected neurons has been shown, however, the mechanism is not fully revealed. We observed formation of oAβ induced tunneling nanotubes (TNTs)-like nanoscaled f-actin containing membrane conduits, in differentially differentiated SH-SY5Y neuronal models. Time-lapse images showed that oAβ propagate from one cell to another via TNT-like structures. Preceding the formation of TNT-like conduits, we detected oAβ-induced plasma membrane (PM) damage and calcium-dependent repair through lysosomal-exocytosis, followed by massive endocytosis to re-establish the PM. Massive endocytosis was monitored by an influx of the membrane-staining dye TMA-DPH and PM damage was quantified by propidium iodide influx in the absence of Ca2+. The massive endocytosis eventually caused accumulation of internalized oAβ in Lamp1 positive multivesicular bodies/lysosomes via the actin cytoskeleton remodulating p21-activated kinase1 (PAK1) dependent endocytic pathway. Three-dimensional quantitative confocal imaging, structured illumination superresolution microscopy, and flowcytometry quantifications revealed that oAβ induces activation of phospho-PAK1, which modulates the formation of long stretched f-actin extensions between cells. Moreover, the formation of TNT-like conduits was inhibited by preventing PAK1-dependent internalization of oAβ using the small-molecule inhibitor IPA-3, a highly selective cell-permeable auto-regulatory inhibitor of PAK1. The present study reveals that the TNT-like conduits are probably instigated as a consequence of oAβ induced PM damage and repair process, followed by PAK1 dependent endocytosis and actin remodeling, probably to maintain cell surface expansion and/or membrane tension in equilibrium.
Collapse
Affiliation(s)
- Aysha Dilna
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Bangalore 560065, India
| | - K V Deepak
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Bangalore 560065, India
| | - Nandini Damodaran
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Bangalore 560065, India
| | - Claudia S Kielkopf
- Experimental Pathology, Department of Biomedical and Clinical Sciences Linköping University, 581 85 Linköping, Sweden
| | - Katarina Kagedal
- Experimental Pathology, Department of Biomedical and Clinical Sciences Linköping University, 581 85 Linköping, Sweden
| | - Karin Ollinger
- Experimental Pathology, Department of Biomedical and Clinical Sciences Linköping University, 581 85 Linköping, Sweden
| | - Sangeeta Nath
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Bangalore 560065, India.
| |
Collapse
|
36
|
Matos ALL, Keller F, Wegner T, del Castillo CEC, Grill D, Kudruk S, Spang A, Glorius F, Heuer A, Gerke V. CHIMs are versatile cholesterol analogs mimicking and visualizing cholesterol behavior in lipid bilayers and cells. Commun Biol 2021; 4:720. [PMID: 34117357 PMCID: PMC8196198 DOI: 10.1038/s42003-021-02252-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 05/20/2021] [Indexed: 02/05/2023] Open
Abstract
Cholesterol is an essential component of cellular membranes regulating the structural integrity and fluidity of biological bilayers and cellular processes such as signal transduction and membrane trafficking. However, tools to investigate the role and dynamics of cholesterol in live cells are still scarce and often show limited applicability. To address this, we previously developed a class of imidazolium-based cholesterol analogs, CHIMs. Here we confirm that CHIM membrane integration characteristics largely mimic those of cholesterol. Computational studies in simulated phospholipid bilayers and biophysical analyses of model membranes reveal that in biologically relevant systems CHIMs behave similarly to natural cholesterol. Importantly, the analogs can functionally replace cholesterol in membranes, can be readily labeled by click chemistry and follow trafficking pathways of cholesterol in live cells. Thus, CHIMs represent chemically versatile cholesterol analogs that can serve as a flexible toolbox to study cholesterol behavior and function in live cells and organisms.
Collapse
Affiliation(s)
- Anna L. L. Matos
- grid.5949.10000 0001 2172 9288Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - Fabian Keller
- grid.5949.10000 0001 2172 9288Physical Chemistry Institute, University of Münster, Münster, Germany ,Center for Multiscale Theory and Computation (CMTC), Münster, Germany
| | - Tristan Wegner
- grid.5949.10000 0001 2172 9288Institute of Organic Chemistry, University of Münster, Münster, Germany
| | | | - David Grill
- grid.5949.10000 0001 2172 9288Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - Sergej Kudruk
- grid.5949.10000 0001 2172 9288Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - Anne Spang
- grid.6612.30000 0004 1937 0642Biozentrum, University of Basel, Basel, Switzerland
| | - Frank Glorius
- grid.5949.10000 0001 2172 9288Institute of Organic Chemistry, University of Münster, Münster, Germany
| | - Andreas Heuer
- grid.5949.10000 0001 2172 9288Physical Chemistry Institute, University of Münster, Münster, Germany ,Center for Multiscale Theory and Computation (CMTC), Münster, Germany
| | - Volker Gerke
- grid.5949.10000 0001 2172 9288Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| |
Collapse
|
37
|
Early onset effects of single substrate accumulation recapitulate major features of LSD in patient-derived lysosomes. iScience 2021; 24:102707. [PMID: 34258549 PMCID: PMC8253970 DOI: 10.1016/j.isci.2021.102707] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 04/06/2021] [Accepted: 06/08/2021] [Indexed: 12/27/2022] Open
Abstract
Lysosome functions mainly rely on their ability to either degrade substrates or release them into the extracellular space. Lysosomal storage disorders (LSDs) are commonly characterized by a chronic lysosomal accumulation of different substrates, thereby causing lysosomal dysfunctions and secretion defects. However, the early effects of substrate accumulation on lysosomal homeostasis have not been analyzed so far. Here, we describe how the acute accumulation of a single substrate determines a rapid centripetal redistribution of the lysosomes, triggering their expansion and reducing their secretion, by limiting the motility of these organelles toward the plasma membrane. Moreover, we provide evidence that such defects could be explained by a trapping mechanism exerted by the extensive contacts between the enlarged lysosomes and the highly intertwined membrane structures of the endoplasmic reticulum which might represent a crucial biological cue ultimately leading to the clinically relevant secondary defects observed in the LSD experimental models and patients. LEU-ME triggers a rapid expansion of the lysosomal compartment Expanded lysosomes display motility and secretion defects Enlarged lysosomes display extended endoplasmic reticulum membrane contact sites
Collapse
|
38
|
Fragment-linking peptide design yields a high-affinity ligand for microtubule-based transport. Cell Chem Biol 2021; 28:1347-1355.e5. [PMID: 33838110 DOI: 10.1016/j.chembiol.2021.03.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/08/2021] [Accepted: 03/17/2021] [Indexed: 01/05/2023]
Abstract
Synthetic peptides are attractive candidates to manipulate protein-protein interactions inside the cell as they mimic natural interactions to compete for binding. However, protein-peptide interactions are often dynamic and weak. A challenge is to design peptides that make improved interactions with the target. Here, we devise a fragment-linking strategy-"mash-up" design-to deliver a high-affinity ligand, KinTag, for the kinesin-1 motor. Using structural insights from natural micromolar-affinity cargo-adaptor ligands, we have identified and combined key binding features in a single, high-affinity ligand. An X-ray crystal structure demonstrates interactions as designed and reveals only a modest increase in interface area. Moreover, when genetically encoded, KinTag promotes transport of lysosomes with higher efficiency than natural sequences, revealing a direct link between motor-adaptor binding affinity and organelle transport. Together, these data demonstrate a fragment-linking strategy for peptide design and its application in a synthetic motor ligand to direct cellular cargo transport.
Collapse
|
39
|
Baek M, Choe YJ, Bannwarth S, Kim J, Maitra S, Dorn GW, Taylor JP, Paquis-Flucklinger V, Kim NC. TDP-43 and PINK1 mediate CHCHD10 S59L mutation-induced defects in Drosophila and in vitro. Nat Commun 2021; 12:1924. [PMID: 33772006 PMCID: PMC7997989 DOI: 10.1038/s41467-021-22145-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/03/2021] [Indexed: 02/01/2023] Open
Abstract
Mutations in coiled-coil-helix-coiled-coil-helix domain containing 10 (CHCHD10) can cause amyotrophic lateral sclerosis and frontotemporal dementia (ALS-FTD). However, the underlying mechanisms are unclear. Here, we generate CHCH10S59L-mutant Drosophila melanogaster and HeLa cell lines to model CHCHD10-associated ALS-FTD. The CHCHD10S59L mutation results in cell toxicity in several tissues and mitochondrial defects. CHCHD10S59L independently affects the TDP-43 and PINK1 pathways. CHCHD10S59L expression increases TDP-43 insolubility and mitochondrial translocation. Blocking TDP-43 mitochondrial translocation with a peptide inhibitor reduced CHCHD10S59L-mediated toxicity. While genetic and pharmacological modulation of PINK1 expression and activity of its substrates rescues and mitigates the CHCHD10S59L-induced phenotypes and mitochondrial defects, respectively, in both Drosophila and HeLa cells. Our findings suggest that CHCHD10S59L-induced TDP-43 mitochondrial translocation and chronic activation of PINK1-mediated pathways result in dominant toxicity, providing a mechanistic insight into the CHCHD10 mutations associated with ALS-FTD.
Collapse
Affiliation(s)
- Minwoo Baek
- grid.17635.360000000419368657Department of Pharmacy Practice and Pharmaceutical Sciences, College of Pharmacy, University of Minnesota, Duluth, MN USA
| | - Yun-Jeong Choe
- grid.17635.360000000419368657Department of Pharmacy Practice and Pharmaceutical Sciences, College of Pharmacy, University of Minnesota, Duluth, MN USA
| | - Sylvie Bannwarth
- grid.410528.a0000 0001 2322 4179Inserm U1081, CNRS UMR7284, IRCAN, Université Côte d’Azur, CHU de Nice, Nice, France
| | - JiHye Kim
- grid.17635.360000000419368657Department of Pharmacy Practice and Pharmaceutical Sciences, College of Pharmacy, University of Minnesota, Duluth, MN USA
| | - Swati Maitra
- grid.17635.360000000419368657Department of Pharmacy Practice and Pharmaceutical Sciences, College of Pharmacy, University of Minnesota, Duluth, MN USA
| | - Gerald W. Dorn
- grid.4367.60000 0001 2355 7002Center for Pharmacogenomics, Washington University School of Medicine, St. Louis, MO USA
| | - J. Paul Taylor
- grid.240871.80000 0001 0224 711XHoward Hughes Medical Institute and Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN USA
| | - Veronique Paquis-Flucklinger
- grid.410528.a0000 0001 2322 4179Inserm U1081, CNRS UMR7284, IRCAN, Université Côte d’Azur, CHU de Nice, Nice, France
| | - Nam Chul Kim
- grid.17635.360000000419368657Department of Pharmacy Practice and Pharmaceutical Sciences, College of Pharmacy, University of Minnesota, Duluth, MN USA
| |
Collapse
|
40
|
Khundadze M, Ribaudo F, Hussain A, Stahlberg H, Brocke-Ahmadinejad N, Franzka P, Varga RE, Zarkovic M, Pungsrinont T, Kokal M, Ganley IG, Beetz C, Sylvester M, Hübner CA. Mouse models for hereditary spastic paraplegia uncover a role of PI4K2A in autophagic lysosome reformation. Autophagy 2021; 17:3690-3706. [PMID: 33618608 PMCID: PMC8632344 DOI: 10.1080/15548627.2021.1891848] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hereditary spastic paraplegia (HSP) denotes genetically heterogeneous disorders characterized by leg spasticity due to degeneration of corticospinal axons. SPG11 and SPG15 have a similar clinical course and together are the most prevalent autosomal recessive HSP entity. The respective proteins play a role for macroautophagy/autophagy and autophagic lysosome reformation (ALR). Here, we report that spg11 and zfyve26 KO mice developed motor impairments within the same course of time. This correlated with enhanced accumulation of autofluorescent material in neurons and progressive neuron loss. In agreement with defective ALR, tubulation events were diminished in starved KO mouse embryonic fibroblasts (MEFs) and lysosomes decreased in neurons of KO brain sections. Confirming that both proteins act in the same molecular pathway, the pathologies were not aggravated upon simultaneous disruption of both. We further show that PI4K2A (phosphatidylinositol 4-kinase type 2 alpha), which phosphorylates phosphatidylinositol to phosphatidylinositol-4-phosphate (PtdIns4P), accumulated in autofluorescent deposits isolated from KO but not WT brains. Elevated PI4K2A abundance was already found at autolysosomes of neurons of presymptomatic KO mice. Immunolabelings further suggested higher levels of PtdIns4P at LAMP1-positive structures in starved KO MEFs. An increased association with LAMP1-positive structures was also observed for clathrin and DNM2/dynamin 2, which are important effectors of ALR recruited by phospholipids. Because PI4K2A overexpression impaired ALR, while its knockdown increased tubulation, we conclude that PI4K2A modulates phosphoinositide levels at autolysosomes and thus the recruitment of downstream effectors of ALR. Therefore, PI4K2A may play an important role in the pathogenesis of SPG11 and SPG15. Abbreviations: ALR: autophagic lysosome reformation; AP-5: adaptor protein complex 5; BFP: blue fluorescent protein; dKO: double knockout; EBSS: Earle’s balanced salt solution; FBA: foot base angle; GFP: green fluorescent protein; HSP: hereditary spastic paraplegia; KO: knockout; LAMP1: lysosomal-associated membrane protein 1; MAP1LC3B/LC3: microtubule-associated protein 1 light chain 3 beta; MEF: mouse embryonic fibroblast; SQSTM1/p62: sequestosome 1; PI4K2A: phosphatidylinositol 4-kinase type 2 alpha; PtdIns3P: phosphatidylinositol-3-phosphate; PtdIns4P: phosphatidylinositol-4-phosphate; RFP: red fluorescent protein; SPG: spastic paraplegia gene; TGN: trans-Golgi network; WT: wild type
Collapse
Affiliation(s)
- Mukhran Khundadze
- Institute of Human Genetics, University Hospital Jena, Friedrich-Schiller-University Jena, Jena, Germany
| | - Federico Ribaudo
- Institute of Human Genetics, University Hospital Jena, Friedrich-Schiller-University Jena, Jena, Germany
| | - Adeela Hussain
- Institute of Human Genetics, University Hospital Jena, Friedrich-Schiller-University Jena, Jena, Germany
| | - Henry Stahlberg
- Institute of Human Genetics, University Hospital Jena, Friedrich-Schiller-University Jena, Jena, Germany
| | - Nahal Brocke-Ahmadinejad
- Core Facility Mass Spectrometry, Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Patricia Franzka
- Institute of Human Genetics, University Hospital Jena, Friedrich-Schiller-University Jena, Jena, Germany
| | - Rita-Eva Varga
- Institute of Human Genetics, University Hospital Jena, Friedrich-Schiller-University Jena, Jena, Germany
| | - Milena Zarkovic
- Institute of Human Genetics, University Hospital Jena, Friedrich-Schiller-University Jena, Jena, Germany
| | - Thanakorn Pungsrinont
- Institute of Human Genetics, University Hospital Jena, Friedrich-Schiller-University Jena, Jena, Germany
| | - Miriam Kokal
- Institute of Human Genetics, University Hospital Jena, Friedrich-Schiller-University Jena, Jena, Germany
| | - Ian G Ganley
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, Scotland
| | - Christian Beetz
- Institute of Clinical Chemistry, University Hospital Jena, Friedrich-Schiller-University Jena, Germany; Current Affiliation: Centogene GmbH, Rostock, Germany
| | - Marc Sylvester
- Core Facility Mass Spectrometry, Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Christian A Hübner
- Institute of Human Genetics, University Hospital Jena, Friedrich-Schiller-University Jena, Jena, Germany
| |
Collapse
|
41
|
Kesisova IA, Robinson BP, Spiliotis ET. A septin GTPase scaffold of dynein-dynactin motors triggers retrograde lysosome transport. J Cell Biol 2021; 220:211663. [PMID: 33416861 PMCID: PMC7802366 DOI: 10.1083/jcb.202005219] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 11/22/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022] Open
Abstract
The metabolic and signaling functions of lysosomes depend on their intracellular positioning and trafficking, but the underlying mechanisms are little understood. Here, we have discovered a novel septin GTPase-based mechanism for retrograde lysosome transport. We found that septin 9 (SEPT9) associates with lysosomes, promoting the perinuclear localization of lysosomes in a Rab7-independent manner. SEPT9 targeting to mitochondria and peroxisomes is sufficient to recruit dynein and cause perinuclear clustering. We show that SEPT9 interacts with both dynein and dynactin through its GTPase domain and N-terminal extension, respectively. Strikingly, SEPT9 associates preferentially with the dynein intermediate chain (DIC) in its GDP-bound state, which favors dimerization and assembly into septin multimers. In response to oxidative cell stress induced by arsenite, SEPT9 localization to lysosomes is enhanced, promoting the perinuclear clustering of lysosomes. We posit that septins function as GDP-activated scaffolds for the cooperative assembly of dynein-dynactin, providing an alternative mechanism of retrograde lysosome transport at steady state and during cellular adaptation to stress.
Collapse
|
42
|
Sotoma S, Zhong C, Kah JCY, Yamashita H, Plakhotnik T, Harada Y, Suzuki M. In situ measurements of intracellular thermal conductivity using heater-thermometer hybrid diamond nanosensors. SCIENCE ADVANCES 2021; 7:7/3/eabd7888. [PMID: 33523906 PMCID: PMC7810374 DOI: 10.1126/sciadv.abd7888] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/30/2020] [Indexed: 05/21/2023]
Abstract
Understanding heat dissipation processes at nanoscale during cellular thermogenesis is essential to clarify the relationships between the heat and biological processes in cells and organisms. A key parameter determining the heat flux inside a cell is the local thermal conductivity, a factor poorly investigated both experimentally and theoretically. Here, using a nanoheater/nanothermometer hybrid made of a polydopamine encapsulating a fluorescent nanodiamond, we measured the intracellular thermal conductivities of HeLa and MCF-7 cells with a spatial resolution of about 200 nm. The mean values determined in these two cell lines are both 0.11 ± 0.04 W m-1 K-1, which is significantly smaller than that of water. Bayesian analysis of the data suggests there is a variation of the thermal conductivity within a cell. These results make the biological impact of transient temperature spikes in a cell much more feasible, and suggest that cells may use heat flux for short-distance thermal signaling.
Collapse
Affiliation(s)
- Shingo Sotoma
- Institute for Protein Research, Osaka University, Osaka, Japan
- Japan Society for the Promotion of Science, Tokyo, Japan
| | - Chongxia Zhong
- Institute for Protein Research, Osaka University, Osaka, Japan
| | - James Chen Yong Kah
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Hayato Yamashita
- Graduate School of Engineering Science, Osaka University, Osaka, Japan
- PRESTO, Japan Science and Technology Agency, Saitama, Japan
| | - Taras Plakhotnik
- School of Mathematics and Physics, The University of Queensland, QLD, Australia.
| | - Yoshie Harada
- Institute for Protein Research, Osaka University, Osaka, Japan.
- Quantum Information and Quantum Biology Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| | - Madoka Suzuki
- Institute for Protein Research, Osaka University, Osaka, Japan.
- PRESTO, Japan Science and Technology Agency, Saitama, Japan
| |
Collapse
|
43
|
Vaillant-Beuchot L, Mary A, Pardossi-Piquard R, Bourgeois A, Lauritzen I, Eysert F, Kinoshita PF, Cazareth J, Badot C, Fragaki K, Bussiere R, Martin C, Mary R, Bauer C, Pagnotta S, Paquis-Flucklinger V, Buée-Scherrer V, Buée L, Lacas-Gervais S, Checler F, Chami M. Accumulation of amyloid precursor protein C-terminal fragments triggers mitochondrial structure, function, and mitophagy defects in Alzheimer's disease models and human brains. Acta Neuropathol 2021; 141:39-65. [PMID: 33079262 PMCID: PMC7785558 DOI: 10.1007/s00401-020-02234-7] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/03/2020] [Accepted: 10/01/2020] [Indexed: 02/07/2023]
Abstract
Several lines of recent evidence indicate that the amyloid precursor protein-derived C-terminal fragments (APP-CTFs) could correspond to an etiological trigger of Alzheimer's disease (AD) pathology. Altered mitochondrial homeostasis is considered an early event in AD development. However, the specific contribution of APP-CTFs to mitochondrial structure, function, and mitophagy defects remains to be established. Here, we demonstrate in neuroblastoma SH-SY5Y cells expressing either APP Swedish mutations, or the β-secretase-derived APP-CTF fragment (C99) combined with β- and γ-secretase inhibition, that APP-CTFs accumulation independently of Aβ triggers excessive mitochondrial morphology alteration (i.e., size alteration and cristae disorganization) associated with enhanced mitochondrial reactive oxygen species production. APP-CTFs accumulation also elicit basal mitophagy failure illustrated by enhanced conversion of LC3, accumulation of LC3-I and/or LC3-II, non-degradation of SQSTM1/p62, inconsistent Parkin and PINK1 recruitment to mitochondria, enhanced levels of membrane and matrix mitochondrial proteins, and deficient fusion of mitochondria with lysosomes. We confirm the contribution of APP-CTFs accumulation to morphological mitochondria alteration and impaired basal mitophagy in vivo in young 3xTgAD transgenic mice treated with γ-secretase inhibitor as well as in adeno-associated-virus-C99 injected mice. Comparison of aged 2xTgAD and 3xTgAD mice indicates that, besides APP-CTFs, an additional contribution of Aβ to late-stage mitophagy activation occurs. Importantly, we report on mitochondrial accumulation of APP-CTFs in human post-mortem sporadic AD brains correlating with mitophagy failure molecular signature. Since defective mitochondria homeostasis plays a pivotal role in AD pathogenesis, targeting mitochondrial dysfunctions and/or mitophagy by counteracting early APP-CTFs accumulation may represent relevant therapeutic interventions in AD.
Collapse
Affiliation(s)
- Loan Vaillant-Beuchot
- Institut of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
| | - Arnaud Mary
- Institut of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
| | - Raphaëlle Pardossi-Piquard
- Institut of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
| | - Alexandre Bourgeois
- Institut of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
| | - Inger Lauritzen
- Institut of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
| | - Fanny Eysert
- Institut of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
| | - Paula Fernanda Kinoshita
- Institut of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
- Department of Pharmacology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Julie Cazareth
- Institut of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
| | - Céline Badot
- Institut of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
| | | | - Renaud Bussiere
- Institut of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
- Department of Medicine, Burlington Danes Building, Hammersmith Hospital Campus, Imperial College London, UK Dementia Research Institute, Du Cane Road, London, W12 0NN, UK
| | - Cécile Martin
- Institut of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
| | - Rosanna Mary
- Institut of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
| | - Charlotte Bauer
- Institut of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
| | - Sophie Pagnotta
- Université Côte d'Azur, Centre Commun de Microscopie Appliquée (CCMA), Parc Valrose, 06108, Nice, France
| | | | - Valérie Buée-Scherrer
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience and Cognition, Place de Verdun, 59045, Lille, France
- Inserm UMR-S 1172, Laboratory of Excellence DistALZ, 'Alzheimer and Tauopathies', Bâtiment Biserte, rue Polonovski, 59045, Lille Cedex, France
| | - Luc Buée
- Univ. Lille, Inserm, CHU-Lille, Lille Neuroscience and Cognition, Place de Verdun, 59045, Lille, France
- Inserm UMR-S 1172, Laboratory of Excellence DistALZ, 'Alzheimer and Tauopathies', Bâtiment Biserte, rue Polonovski, 59045, Lille Cedex, France
| | - Sandra Lacas-Gervais
- Université Côte d'Azur, Centre Commun de Microscopie Appliquée (CCMA), Parc Valrose, 06108, Nice, France
| | - Frédéric Checler
- Institut of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
| | - Mounia Chami
- Institut of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France.
| |
Collapse
|
44
|
Nijenhuis W, van Grinsven MMP, Kapitein LC. An optimized toolbox for the optogenetic control of intracellular transport. J Cell Biol 2020; 219:133834. [PMID: 32328628 PMCID: PMC7147098 DOI: 10.1083/jcb.201907149] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 12/18/2019] [Accepted: 01/16/2020] [Indexed: 12/31/2022] Open
Abstract
Cellular functioning relies on active transport of organelles by molecular motors. To explore how intracellular organelle distributions affect cellular functions, several optogenetic approaches enable organelle repositioning through light-inducible recruitment of motors to specific organelles. Nonetheless, robust application of these methods in cellular populations without side effects has remained challenging. Here, we introduce an improved toolbox for optogenetic control of intracellular transport that optimizes cellular responsiveness and limits adverse effects. To improve dynamic range, we employed improved optogenetic heterodimerization modules and engineered a photosensitive kinesin-3, which is activated upon blue light–sensitive homodimerization. This opto-kinesin prevented motor activation before experimental onset, limited dark-state activation, and improved responsiveness. In addition, we adopted moss kinesin-14 for efficient retrograde transport with minimal adverse effects on endogenous transport. Using this optimized toolbox, we demonstrate robust reversible repositioning of (endogenously tagged) organelles within cellular populations. More robust control over organelle motility will aid in dissecting spatial cell biology and transport-related diseases.
Collapse
Affiliation(s)
- Wilco Nijenhuis
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Mariëlle M P van Grinsven
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Lukas C Kapitein
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
45
|
Lu M, van Tartwijk FW, Lin JQ, Nijenhuis W, Parutto P, Fantham M, Christensen CN, Avezov E, Holt CE, Tunnacliffe A, Holcman D, Kapitein L, Schierle GSK, Kaminski CF. The structure and global distribution of the endoplasmic reticulum network are actively regulated by lysosomes. SCIENCE ADVANCES 2020; 6:eabc7209. [PMID: 33328230 PMCID: PMC7744115 DOI: 10.1126/sciadv.abc7209] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 10/27/2020] [Indexed: 06/12/2023]
Abstract
The endoplasmic reticulum (ER) comprises morphologically and functionally distinct domains: sheets and interconnected tubules. These domains undergo dynamic reshaping in response to changes in the cellular environment. However, the mechanisms behind this rapid remodeling are largely unknown. Here, we report that ER remodeling is actively driven by lysosomes, following lysosome repositioning in response to changes in nutritional status: The anchorage of lysosomes to ER growth tips is critical for ER tubule elongation and connection. We validate this causal link via the chemo- and optogenetically driven repositioning of lysosomes, which leads to both a redistribution of the ER tubules and a change of its global morphology. Therefore, lysosomes sense metabolic change in the cell and regulate ER tubule distribution accordingly. Dysfunction in this mechanism during axonal extension may lead to axonal growth defects. Our results demonstrate a critical role of lysosome-regulated ER dynamics and reshaping in nutrient responses and neuronal development.
Collapse
Affiliation(s)
- Meng Lu
- Cambridge Infinitus Research Centre, Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK
| | - Francesca W van Tartwijk
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK
| | - Julie Qiaojin Lin
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK
- UK Dementia Research Institute at the University of Cambridge and Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0AH, UK
| | - Wilco Nijenhuis
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, Netherlands
| | - Pierre Parutto
- Group of Computational Biology and Applied Mathematics, Institut de Biologie de l'École Normale Supérieure-PSL, 46 rue d'Ulm, 75005 Paris, France
| | - Marcus Fantham
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK
| | - Charles N Christensen
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK
| | - Edward Avezov
- UK Dementia Research Institute at the University of Cambridge and Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0AH, UK
| | - Christine E Holt
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Alan Tunnacliffe
- Cambridge Infinitus Research Centre, Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK
| | - David Holcman
- Group of Computational Biology and Applied Mathematics, Institut de Biologie de l'École Normale Supérieure-PSL, 46 rue d'Ulm, 75005 Paris, France
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge CB3 0WA, UK
| | - Lukas Kapitein
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, Netherlands
| | - Gabriele S Kaminski Schierle
- Cambridge Infinitus Research Centre, Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK
| | - Clemens F Kaminski
- Cambridge Infinitus Research Centre, Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK.
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK
- UK Dementia Research Institute at the University of Cambridge and Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0AH, UK
| |
Collapse
|
46
|
Houston R, Sekine S, Calderon MJ, Seifuddin F, Wang G, Kawagishi H, Malide DA, Li Y, Gucek M, Pirooznia M, Nelson AJ, Stokes MP, Stewart-Ornstein J, Mullett SJ, Wendell SG, Watkins SC, Finkel T, Sekine Y. Acetylation-mediated remodeling of the nucleolus regulates cellular acetyl-CoA responses. PLoS Biol 2020; 18:e3000981. [PMID: 33253182 PMCID: PMC7728262 DOI: 10.1371/journal.pbio.3000981] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 12/10/2020] [Accepted: 10/29/2020] [Indexed: 12/14/2022] Open
Abstract
The metabolite acetyl-coenzyme A (acetyl-CoA) serves as an essential element for a wide range of cellular functions including adenosine triphosphate (ATP) production, lipid synthesis, and protein acetylation. Intracellular acetyl-CoA concentrations are associated with nutrient availability, but the mechanisms by which a cell responds to fluctuations in acetyl-CoA levels remain elusive. Here, we generate a cell system to selectively manipulate the nucleo-cytoplasmic levels of acetyl-CoA using clustered regularly interspaced short palindromic repeat (CRISPR)-mediated gene editing and acetate supplementation of the culture media. Using this system and quantitative omics analyses, we demonstrate that acetyl-CoA depletion alters the integrity of the nucleolus, impairing ribosomal RNA synthesis and evoking the ribosomal protein-dependent activation of p53. This nucleolar remodeling appears to be mediated through the class IIa histone deacetylases (HDACs). Our findings highlight acetylation-mediated control of the nucleolus as an important hub linking acetyl-CoA fluctuations to cellular stress responses.
Collapse
Affiliation(s)
- Ryan Houston
- Aging Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Shiori Sekine
- Aging Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Michael J. Calderon
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Fayaz Seifuddin
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, United States of America
| | - Guanghui Wang
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, United States of America
| | - Hiroyuki Kawagishi
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, United States of America
| | - Daniela A. Malide
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, United States of America
| | - Yuesheng Li
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, United States of America
| | - Marjan Gucek
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, United States of America
| | - Mehdi Pirooznia
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, United States of America
| | - Alissa J. Nelson
- Cell Signaling Technology, INC., Danvers, Massachusetts, United States of America
| | - Matthew P. Stokes
- Cell Signaling Technology, INC., Danvers, Massachusetts, United States of America
| | - Jacob Stewart-Ornstein
- Department of Computational and Systems Biology, University of Pittsburgh and Hillman Cancer Center, Pittsburgh, Pennsylvania, United States of America
| | - Steven J. Mullett
- Department of Pharmacology and Chemical Biology, the Health Sciences Metabolomics and Lipidomics Core, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Stacy G. Wendell
- Department of Pharmacology and Chemical Biology, the Health Sciences Metabolomics and Lipidomics Core, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Simon C. Watkins
- Department of Cell Biology, Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Toren Finkel
- Aging Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, United States of America
| | - Yusuke Sekine
- Aging Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, United States of America
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
47
|
Schormann W, Hariharan S, Andrews DW. A reference library for assigning protein subcellular localizations by image-based machine learning. J Cell Biol 2020; 219:133635. [PMID: 31968357 PMCID: PMC7055006 DOI: 10.1083/jcb.201904090] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 09/30/2019] [Accepted: 12/15/2019] [Indexed: 12/11/2022] Open
Abstract
Confocal micrographs of EGFP fusion proteins localized at key cell organelles in murine and human cells were acquired for use as subcellular localization landmarks. For each of the respective 789,011 and 523,319 optically validated cell images, morphology and statistical features were measured. Machine learning algorithms using these features permit automated assignment of the localization of other proteins and dyes in both cell types with very high accuracy. Automated assignment of subcellular localizations for model tail-anchored proteins with randomly mutated C-terminal targeting sequences allowed the discovery of motifs responsible for targeting to mitochondria, endoplasmic reticulum, and the late secretory pathway. Analysis of directed mutants enabled refinement of these motifs and characterization of protein distributions in within cellular subcompartments.
Collapse
Affiliation(s)
- Wiebke Schormann
- Biological Sciences, Sunnybrook Research Institute, Toronto, Canada
| | | | - David W Andrews
- Biological Sciences, Sunnybrook Research Institute, Toronto, Canada.,Department of Biochemistry, University of Toronto, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| |
Collapse
|
48
|
Lee C, Lamech L, Johns E, Overholtzer M. Selective Lysosome Membrane Turnover Is Induced by Nutrient Starvation. Dev Cell 2020; 55:289-297.e4. [PMID: 32916093 DOI: 10.1016/j.devcel.2020.08.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/02/2020] [Accepted: 08/17/2020] [Indexed: 12/20/2022]
Abstract
Lysosome function is essential for cellular homeostasis, but quality-control mechanisms that maintain healthy lysosomes remain poorly characterized. Here, we developed a method to measure lysosome turnover and use this to identify a selective mechanism of membrane degradation that involves lipidation of the autophagy protein LC3 onto lysosomal membranes and the formation of intraluminal vesicles through microautophagy. This mechanism is induced in response to metabolic stress resulting from glucose starvation or by treatment with pharmacological agents that induce osmotic stress on lysosomes. Cells lacking ATG5, an essential component of the LC3 lipidation machinery, show reduced ability to regulate lysosome size and degradative capacity in response to activation of this mechanism. These findings identify a selective mechanism of lysosome membrane turnover that is induced by stress and uncover a function for LC3 lipidation in regulating lysosome size and activity through microautophagy.
Collapse
Affiliation(s)
- Chan Lee
- Cell Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA; BCMB Allied Program, Weill Cornell Medical College, New York, NY 10065, USA
| | - Lilian Lamech
- Cell Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA
| | - Eleanor Johns
- Cell Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA; Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Michael Overholtzer
- Cell Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY 10065, USA; BCMB Allied Program, Weill Cornell Medical College, New York, NY 10065, USA; Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
49
|
Scheefhals N, Catsburg LAE, Westerveld ML, Blanpied TA, Hoogenraad CC, MacGillavry HD. Shank Proteins Couple the Endocytic Zone to the Postsynaptic Density to Control Trafficking and Signaling of Metabotropic Glutamate Receptor 5. Cell Rep 2020; 29:258-269.e8. [PMID: 31597090 PMCID: PMC6815225 DOI: 10.1016/j.celrep.2019.08.102] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 07/25/2019] [Accepted: 08/29/2019] [Indexed: 12/17/2022] Open
Abstract
Activation of postsynaptic metabotropic glutamate receptors (mGluRs) modulates neuronal excitability and synaptic plasticity, while deregulation of mGluR signaling has been implicated in neurodevelopmental disorders. Overstimulation of mGluRs is restricted by the rapid endocytosis of receptors after activation. However, how membrane trafficking of mGluRs at synapses is controlled remains poorly defined. We find that in hippocampal neurons, the agonist-induced receptor internalization of synaptic mGluR5 is significantly reduced in Shank knockdown neurons. This is rescued by the re-expression of wild-type Shanks, but not by mutants unable to bind Homer1b/c, Dynamin2, or Cortactin. These effects are paralleled by a reduction in synapses associated with an endocytic zone. Moreover, a mutation in SHANK2 found in autism spectrum disorders (ASDs) similarly disrupts these processes. On the basis of these findings, we propose that synaptic Shank scaffolds anchor the endocytic machinery to govern the efficient trafficking of mGluR5 and to balance the surface expression of mGluRs to efficiently modulate neuronal functioning. Receptor activation triggers efficient internalization of mGluR5 in spines Shank proteins control mGluR5 trafficking and signaling Shanks link essential components of the endocytic zone to the postsynaptic density Mutation in SHANK2 found in ASD disrupt these processes
Collapse
Affiliation(s)
- Nicky Scheefhals
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Lisa A E Catsburg
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Margriet L Westerveld
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Thomas A Blanpied
- Department of Physiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Casper C Hoogenraad
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Harold D MacGillavry
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, the Netherlands.
| |
Collapse
|
50
|
He H, Guo J, Lin X, Xu B. Enzyme-Instructed Assemblies Enable Mitochondria Localization of Histone H2B in Cancer Cells. Angew Chem Int Ed Engl 2020; 59:9330-9334. [PMID: 32119754 PMCID: PMC7269854 DOI: 10.1002/anie.202000983] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Indexed: 01/11/2023]
Abstract
Presently, little is known of how the inter-organelle crosstalk impacts cancer cells owing to the lack of approaches that can manipulate inter-organelle communication in cancer cells. We found that a negatively charged, enzyme cleavable peptide (MitoFlag) enables the trafficking of histone protein H2B, a nuclear protein, to the mitochondria in cancer cells. MitoFlag interacts with the nuclear location sequence of H2B to block it from entering the nucleus. A protease on the mitochondria cleaves the Flag from the MitoFlag/H2B complex to form assemblies that retain H2B on the mitochondria and facilitate H2B entering the mitochondria. Adding NLS, replacing aspartic acid by glutamic acid residues, or changing the l- to d-aspartic acid residue on MitoFlag abolishes the trafficking of H2B into mitochondria of HeLa cells. As the first example of the enzyme-instructed self-assembly of a synthetic peptide for trafficking endogenous proteins, this work provides insights for understanding and manipulating inter-organelle communication in cells.
Collapse
Affiliation(s)
- Hongjian He
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Jiaqi Guo
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Xingyi Lin
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| |
Collapse
|