1
|
Ahn J, Jang SH, Jang S, Yoon JH, Lee MG, Chi SG. XAF1 is secreted from stressed tumor cells to activate T cell-mediated tumor surveillance via Lck-ERK signaling. Neoplasia 2025; 59:101094. [PMID: 39615106 DOI: 10.1016/j.neo.2024.101094] [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: 07/16/2024] [Accepted: 11/20/2024] [Indexed: 12/20/2024]
Abstract
X-linked inhibitor of apoptosis-associated factor 1 (XAF1) is a stress-inducible tumor suppressor that is commonly inactivated in multiple types of human malignancies. Nevertheless, the molecular basis for the XAF1-mediated tumor suppression remains largely undefined. Here, we report that XAF1 is secreted from cells under various cytotoxic stress conditions and activates T cell-mediated tumor surveillance. In cancer cells exposed to interferon -γ, tumor necrosis factor -α, and etoposide, XAF1 is elevated and actively secreted through the unconventional endo-lysosomal trafficking pathway and the zinc finger 4 domain of XAF1 plays an essential for this secretion. Secreted XAF1 is internalized into nearby T cells through clathrin-mediated endocytosis and stimulates proliferation, migration, and tumor infiltration of T cells. Internalized XAF1 activates RAF-MEK-ERK signaling through the direct interaction with and phosphorylation of lymphocyte-specific protein tyrosine kinase. In response to interferon -γ injection, Xaf1+/+ tumors display significantly higher regression rate and T cell infiltration compared to Xaf1-/- tumors while Xaf1-/- tumors are markedly reduced by injection of recombinant Xaf1. XAF1 expression is associated with overall survival in T cell-enriched cancer patients and also correlates with prognosis in T cell-based immunotherapies. Together, our study identifies XAF1 as a novel secretory immune-modulatory tumor suppressor, illuminating the mechanistic consequence of its inactivation in tumorigenesis.
Collapse
Affiliation(s)
- Jieun Ahn
- Department of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Seung-Hun Jang
- Department of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Sungchan Jang
- Department of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Ji-Hye Yoon
- Department of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Min-Goo Lee
- Department of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Sung-Gil Chi
- Department of Life Sciences, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
2
|
Lopez A, Siddiqi FH, Villeneuve J, Ureshino RP, Jeon HY, Koulousakis P, Keeling S, McEwan WA, Fleming A, Rubinsztein DC. Carbonic anhydrase inhibition ameliorates tau toxicity via enhanced tau secretion. Nat Chem Biol 2024:10.1038/s41589-024-01762-7. [PMID: 39482469 DOI: 10.1038/s41589-024-01762-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 09/22/2024] [Indexed: 11/03/2024]
Abstract
Tauopathies are neurodegenerative diseases that manifest with intracellular accumulation and aggregation of tau protein. These include Pick's disease, progressive supranuclear palsy, corticobasal degeneration and argyrophilic grain disease, where tau is believed to be the primary disease driver, as well as secondary tauopathies, such as Alzheimer's disease. There is a need to develop effective pharmacological therapies. Here we tested >1,400 clinically approved compounds using transgenic zebrafish tauopathy models. This revealed that carbonic anhydrase (CA) inhibitors protected against tau toxicity. CRISPR experiments confirmed that CA depletion mimicked the effects of these drugs. CA inhibition promoted faster clearance of human tau by promoting lysosomal exocytosis. Importantly, methazolamide, a CA inhibitor used in the clinic, also reduced total and phosphorylated tau levels, increased neuronal survival and ameliorated neurodegeneration in mouse tauopathy models at concentrations similar to those seen in people. These data underscore the feasibility of in vivo drug screens using zebrafish models and suggest serious consideration of CA inhibitors for treating tauopathies.
Collapse
Affiliation(s)
- Ana Lopez
- Department of Medical Genetics, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK
| | - Farah H Siddiqi
- Department of Medical Genetics, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK
| | - Julien Villeneuve
- Department of Medical Genetics, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK
| | - Rodrigo Portes Ureshino
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Hee-Yeon Jeon
- Department of Medical Genetics, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK
| | - Philippos Koulousakis
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Sophie Keeling
- UK Dementia Research Institute, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK
| | - William A McEwan
- UK Dementia Research Institute, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK
| | - Angeleen Fleming
- Department of Medical Genetics, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK.
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
- UK Dementia Research Institute, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK.
| | - David C Rubinsztein
- Department of Medical Genetics, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK.
- UK Dementia Research Institute, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK.
| |
Collapse
|
3
|
Tebeje BM, Thiex NW, Swanson JA. Growing Macrophages Regulate High Rates of Solute Flux by Pinocytosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619691. [PMID: 39484410 PMCID: PMC11526976 DOI: 10.1101/2024.10.22.619691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
In metazoan cells, growth factors stimulate solute ingestion by pinocytosis. To examine the role of pinocytosis in cell growth, this study measured cell proliferation and the attendant rates of solute flux by pinocytosis in murine macrophages in response to the growth factor colony-stimulating factor-1 (CSF1). During CSF1-dependent growth in rich medium, macrophages internalized 72 percent of their cell volume in extracellular fluid every hour. Removal of the essential amino acid leucine from growth medium limited rates of protein synthesis and growth, but increased rates of solute accumulation by macropinocytosis. The amount of protein synthesized during leucine-dependent growth exceeded the capacity of pinocytosis to internalize enough soluble leucine to support growth and proliferation. Fluid-phase solute recycling from lysosomes secreted small molecules from the cells at high rates. Inhibitors of pinocytosis and the mechanistic target-of-rapamycin (mTOR) reduced cell growth and solute recycling, indicating roles for pinocytosis in growth and for nutrient sensing in the regulation of solute flux by pinocytosis.
Collapse
Affiliation(s)
- Biniam M Tebeje
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109-5620
| | - Natalie W Thiex
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD 57007
| | - Joel A Swanson
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109-5620
| |
Collapse
|
4
|
Ferrari V, Tedesco B, Cozzi M, Chierichetti M, Casarotto E, Pramaggiore P, Cornaggia L, Mohamed A, Patelli G, Piccolella M, Cristofani R, Crippa V, Galbiati M, Poletti A, Rusmini P. Lysosome quality control in health and neurodegenerative diseases. Cell Mol Biol Lett 2024; 29:116. [PMID: 39237893 PMCID: PMC11378602 DOI: 10.1186/s11658-024-00633-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024] Open
Abstract
Lysosomes are acidic organelles involved in crucial intracellular functions, including the degradation of organelles and protein, membrane repair, phagocytosis, endocytosis, and nutrient sensing. Given these key roles of lysosomes, maintaining their homeostasis is essential for cell viability. Thus, to preserve lysosome integrity and functionality, cells have developed a complex intracellular system, called lysosome quality control (LQC). Several stressors may affect the integrity of lysosomes, causing Lysosomal membrane permeabilization (LMP), in which membrane rupture results in the leakage of luminal hydrolase enzymes into the cytosol. After sensing the damage, LQC either activates lysosome repair, or induces the degradation of the ruptured lysosomes through autophagy. In addition, LQC stimulates the de novo biogenesis of functional lysosomes and lysosome exocytosis. Alterations in LQC give rise to deleterious consequences for cellular homeostasis. Specifically, the persistence of impaired lysosomes or the malfunctioning of lysosomal processes leads to cellular toxicity and death, thereby contributing to the pathogenesis of different disorders, including neurodegenerative diseases (NDs). Recently, several pieces of evidence have underlined the importance of the role of lysosomes in NDs. In this review, we describe the elements of the LQC system, how they cooperate to maintain lysosome homeostasis, and their implication in the pathogenesis of different NDs.
Collapse
Affiliation(s)
- Veronica Ferrari
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Barbara Tedesco
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Marta Cozzi
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Marta Chierichetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Elena Casarotto
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Paola Pramaggiore
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Laura Cornaggia
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Ali Mohamed
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Guglielmo Patelli
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Margherita Piccolella
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Riccardo Cristofani
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Valeria Crippa
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Mariarita Galbiati
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy.
| | - Paola Rusmini
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| |
Collapse
|
5
|
Néel E, Chiritoiu-Butnaru M, Fargues W, Denus M, Colladant M, Filaquier A, Stewart SE, Lehmann S, Zurzolo C, Rubinsztein DC, Marin P, Parmentier ML, Villeneuve J. The endolysosomal system in conventional and unconventional protein secretion. J Cell Biol 2024; 223:e202404152. [PMID: 39133205 PMCID: PMC11318669 DOI: 10.1083/jcb.202404152] [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: 05/02/2024] [Revised: 07/12/2024] [Accepted: 07/26/2024] [Indexed: 08/13/2024] Open
Abstract
Most secreted proteins are transported through the "conventional" endoplasmic reticulum-Golgi apparatus exocytic route for their delivery to the cell surface and release into the extracellular space. Nonetheless, formative discoveries have underscored the existence of alternative or "unconventional" secretory routes, which play a crucial role in exporting a diverse array of cytosolic proteins outside the cell in response to intrinsic demands, external cues, and environmental changes. In this context, lysosomes emerge as dynamic organelles positioned at the crossroads of multiple intracellular trafficking pathways, endowed with the capacity to fuse with the plasma membrane and recognized for their key role in both conventional and unconventional protein secretion. The recent recognition of lysosomal transport and exocytosis in the unconventional secretion of cargo proteins provides new and promising insights into our understanding of numerous physiological processes.
Collapse
Affiliation(s)
- Eloïse Néel
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | | | - William Fargues
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | - Morgane Denus
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | - Maëlle Colladant
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | - Aurore Filaquier
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | - Sarah E Stewart
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Sylvain Lehmann
- Laboratoire de Biochimie-Protéomique Clinique-Plateforme de Protéomique Clinique, Université de Montpellier, Institute for Regenerative Medicine and Biotherapy Centre Hospitalier Universitaire de Montpellier, Institute for Neurosciences of Montpellier INSERM , Montpellier, France
| | - Chiara Zurzolo
- Unité de Trafic Membranaire et Pathogenèse, Institut Pasteur, UMR3691 CNRS , Paris, France
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute , Cambridge, UK
| | - Philippe Marin
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | - Marie-Laure Parmentier
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| | - Julien Villeneuve
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM , Montpellier, France
| |
Collapse
|
6
|
Cen J, Hu N, Shen J, Gao Y, Lu H. Pathological Functions of Lysosomal Ion Channels in the Central Nervous System. Int J Mol Sci 2024; 25:6565. [PMID: 38928271 PMCID: PMC11203704 DOI: 10.3390/ijms25126565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Lysosomes are highly dynamic organelles that maintain cellular homeostasis and regulate fundamental cellular processes by integrating multiple metabolic pathways. Lysosomal ion channels such as TRPML1-3, TPC1/2, ClC6/7, CLN7, and TMEM175 mediate the flux of Ca2+, Cl-, Na+, H+, and K+ across lysosomal membranes in response to osmotic stimulus, nutrient-dependent signals, and cellular stresses. These ion channels serve as the crucial transducers of cell signals and are essential for the regulation of lysosomal biogenesis, motility, membrane contact site formation, and lysosomal homeostasis. In terms of pathophysiology, genetic variations in these channel genes have been associated with the development of lysosomal storage diseases, neurodegenerative diseases, inflammation, and cancer. This review aims to discuss the current understanding of the role of these ion channels in the central nervous system and to assess their potential as drug targets.
Collapse
Affiliation(s)
| | | | | | - Yongjing Gao
- Institute of Pain Medicine and Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China; (J.C.); (N.H.); (J.S.)
| | - Huanjun Lu
- Institute of Pain Medicine and Special Environmental Medicine, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China; (J.C.); (N.H.); (J.S.)
| |
Collapse
|
7
|
Kim M, Park JH, Go M, Lee N, Seo J, Lee H, Kim D, Ha H, Kim T, Jeong MS, Kim S, Kim T, Kim HS, Kang D, Shim H, Lee SY. RUFY4 deletion prevents pathological bone loss by blocking endo-lysosomal trafficking of osteoclasts. Bone Res 2024; 12:29. [PMID: 38744829 PMCID: PMC11094054 DOI: 10.1038/s41413-024-00326-8] [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: 08/06/2023] [Revised: 02/01/2024] [Accepted: 03/08/2024] [Indexed: 05/16/2024] Open
Abstract
Mature osteoclasts degrade bone matrix by exocytosis of active proteases from secretory lysosomes through a ruffled border. However, the molecular mechanisms underlying lysosomal trafficking and secretion in osteoclasts remain largely unknown. Here, we show with GeneChip analysis that RUN and FYVE domain-containing protein 4 (RUFY4) is strongly upregulated during osteoclastogenesis. Mice lacking Rufy4 exhibited a high trabecular bone mass phenotype with abnormalities in osteoclast function in vivo. Furthermore, deleting Rufy4 did not affect osteoclast differentiation, but inhibited bone-resorbing activity due to disruption in the acidic maturation of secondary lysosomes, their trafficking to the membrane, and their secretion of cathepsin K into the extracellular space. Mechanistically, RUFY4 promotes late endosome-lysosome fusion by acting as an adaptor protein between Rab7 on late endosomes and LAMP2 on primary lysosomes. Consequently, Rufy4-deficient mice were highly protected from lipopolysaccharide- and ovariectomy-induced bone loss. Thus, RUFY4 plays as a new regulator in osteoclast activity by mediating endo-lysosomal trafficking and have a potential to be specific target for therapies against bone-loss diseases such as osteoporosis.
Collapse
Affiliation(s)
- Minhee Kim
- Department of Life Science, Ewha Womans University, Seoul, 03760, South Korea
| | - Jin Hee Park
- Department of Life Science, Ewha Womans University, Seoul, 03760, South Korea
- The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, 03760, South Korea
| | - Miyeon Go
- Department of Life Science, Ewha Womans University, Seoul, 03760, South Korea
| | - Nawon Lee
- Department of Life Science, Ewha Womans University, Seoul, 03760, South Korea
| | - Jeongin Seo
- Department of Life Science, Ewha Womans University, Seoul, 03760, South Korea
| | - Hana Lee
- Department of Biomedical Engineering, Yonsei University, Wonju, 26493, South Korea
| | - Doyong Kim
- Department of Biomedical Engineering, Yonsei University, Wonju, 26493, South Korea
| | - Hyunil Ha
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, 34054, South Korea
| | - Taesoo Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, 34054, South Korea
| | - Myeong Seon Jeong
- Chuncheon Center, Korea Basic Science Institute, Chuncheon, 24341, South Korea
| | - Suree Kim
- Fluorescence Core Imaging Center and Bioimaging Data Curation Center, Ewha Womans University, Seoul, 03760, South Korea
| | - Taesoo Kim
- Department of Life Science, Ewha Womans University, Seoul, 03760, South Korea
- The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, 03760, South Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, 03760, South Korea
| | - Han Sung Kim
- Department of Biomedical Engineering, Yonsei University, Wonju, 26493, South Korea
| | - Dongmin Kang
- Department of Life Science, Ewha Womans University, Seoul, 03760, South Korea
- Fluorescence Core Imaging Center and Bioimaging Data Curation Center, Ewha Womans University, Seoul, 03760, South Korea
| | - Hyunbo Shim
- Department of Life Science, Ewha Womans University, Seoul, 03760, South Korea
| | - Soo Young Lee
- Department of Life Science, Ewha Womans University, Seoul, 03760, South Korea.
- The Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, 03760, South Korea.
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, 03760, South Korea.
| |
Collapse
|
8
|
Zhang S, Yin H, Zhang Y, Zhu Y, Zhu X, Zhu W, Tang L, Liu Y, Wu K, Zhao B, Tian Y, Lu H. Autophagic-lysosomal damage induced by swainsonine is protected by trehalose through activation of TFEB-regulated pathway in renal tubular epithelial cells. Chem Biol Interact 2024; 394:110990. [PMID: 38579922 DOI: 10.1016/j.cbi.2024.110990] [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: 11/10/2023] [Revised: 03/19/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
Swainsonine (SW) is the main toxic component of locoweed. Previous studies have shown that kidney damage is an early pathologic change in locoweed poisoning in animals. Trehalose induces autophagy and alleviates lysosomal damage, while its protective effect and mechanism against the toxic injury induced by SW is not clear. Based on the published literature, we hypothesize that transcription factor EB(TFEB) -regulated is targeted by SW and activating TFEB by trehalose would reverse the toxic effects. In this study, we investigate the mechanism of protective effects of trehalose using renal tubular epithelial cells. The results showed that SW induced an increase in the expression level of microtubule-associated protein light chain 3-II and p62 proteins and a decrease in the expression level of ATPase H+ transporting V1 Subunit A, Cathepsin B, Cathepsin D, lysosome-associated membrane protein 2 and TFEB proteins in renal tubular epithelial cells in a time and dose-dependent manner suggesting TFEB-regulated lysosomal pathway is adversely affected by SW. Conversely, treatment with trehalose, a known activator of TFEB promote TFEB nuclear translocation suggesting that TFEB plays an important role in protection against SW toxicity. We demonstrated in lysosome staining that SW reduced the number of lysosomes and increased the luminal pH, while trehalose could counteract these SW-induced effects. In summary, our results demonstrated for the first time that trehalose could alleviate the autophagy degradation disorder and lysosomal damage induced by SW. Our results provide an interesting method for reversion of SW-induced toxicity in farm animals and furthermore, activation of TFEB by trehalose suggesting novel mechanism of treating lysosomal storage diseases.
Collapse
Affiliation(s)
- Shuhang Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Hai Yin
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yiqingqing Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yanli Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Xueyao Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Wenting Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Lihui Tang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yiling Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Kexin Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Baoyu Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yanan Tian
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX, 77843, USA
| | - Hao Lu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
9
|
Eriksson I, Öllinger K. Lysosomes in Cancer-At the Crossroad of Good and Evil. Cells 2024; 13:459. [PMID: 38474423 DOI: 10.3390/cells13050459] [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: 12/21/2023] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
Although it has been known for decades that lysosomes are central for degradation and recycling in the cell, their pivotal role as nutrient sensing signaling hubs has recently become of central interest. Since lysosomes are highly dynamic and in constant change regarding content and intracellular position, fusion/fission events allow communication between organelles in the cell, as well as cell-to-cell communication via exocytosis of lysosomal content and release of extracellular vesicles. Lysosomes also mediate different forms of regulated cell death by permeabilization of the lysosomal membrane and release of their content to the cytosol. In cancer cells, lysosomal biogenesis and autophagy are increased to support the increased metabolism and allow growth even under nutrient- and oxygen-poor conditions. Tumor cells also induce exocytosis of lysosomal content to the extracellular space to promote invasion and metastasis. However, due to the enhanced lysosomal function, cancer cells are often more susceptible to lysosomal membrane permeabilization, providing an alternative strategy to induce cell death. This review summarizes the current knowledge of cancer-associated alterations in lysosomal structure and function and illustrates how lysosomal exocytosis and release of extracellular vesicles affect disease progression. We focus on functional differences depending on lysosomal localization and the regulation of intracellular transport, and lastly provide insight how new therapeutic strategies can exploit the power of the lysosome and improve cancer treatment.
Collapse
Affiliation(s)
- Ida Eriksson
- Division of Cell Biology, Department of Biomedical and Clinical Sciences, Linköping University, 58185 Linköping, Sweden
| | - Karin Öllinger
- Division of Cell Biology, Department of Biomedical and Clinical Sciences, Linköping University, 58185 Linköping, Sweden
| |
Collapse
|
10
|
Zaffagnini G, Cheng S, Salzer MC, Pernaute B, Duran JM, Irimia M, Schuh M, Böke E. Mouse oocytes sequester aggregated proteins in degradative super-organelles. Cell 2024; 187:1109-1126.e21. [PMID: 38382525 DOI: 10.1016/j.cell.2024.01.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 12/04/2023] [Accepted: 01/19/2024] [Indexed: 02/23/2024]
Abstract
Oocytes are among the longest-lived cells in the body and need to preserve their cytoplasm to support proper embryonic development. Protein aggregation is a major threat for intracellular homeostasis in long-lived cells. How oocytes cope with protein aggregation during their extended life is unknown. Here, we find that mouse oocytes accumulate protein aggregates in specialized compartments that we named endolysosomal vesicular assemblies (ELVAs). Combining live-cell imaging, electron microscopy, and proteomics, we found that ELVAs are non-membrane-bound compartments composed of endolysosomes, autophagosomes, and proteasomes held together by a protein matrix formed by RUFY1. Functional assays revealed that in immature oocytes, ELVAs sequester aggregated proteins, including TDP-43, and degrade them upon oocyte maturation. Inhibiting degradative activity in ELVAs leads to the accumulation of protein aggregates in the embryo and is detrimental for embryo survival. Thus, ELVAs represent a strategy to safeguard protein homeostasis in long-lived cells.
Collapse
Affiliation(s)
- Gabriele Zaffagnini
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Shiya Cheng
- Department of Meiosis, Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
| | - Marion C Salzer
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Barbara Pernaute
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Juan Manuel Duran
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Manuel Irimia
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
| | - Melina Schuh
- Department of Meiosis, Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, 37077 Göttingen, Germany
| | - Elvan Böke
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain.
| |
Collapse
|
11
|
Kim B, Kim G, Kim H, Song YS, Jung J. Modulation of Cisplatin Sensitivity through TRPML1-Mediated Lysosomal Exocytosis in Ovarian Cancer Cells: A Comprehensive Metabolomic Approach. Cells 2024; 13:115. [PMID: 38247807 PMCID: PMC10814698 DOI: 10.3390/cells13020115] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND The lysosome has emerged as a promising target for overcoming chemoresistance, owing to its role in facilitating the lysosomal sequestration of drugs. The lysosomal calcium channel TRPML1 not only influences lysosomal biogenesis but also coordinates both endocytosis and exocytosis. This study explored the modulation of cisplatin sensitivity by regulating TRPML1-mediated lysosomal exocytosis and identified the metabolomic profile altered by TRPML1 inhibition. METHODS We used four types of ovarian cancer cells: two cancer cell lines (OVCAR8 and TOV21G) and two patient-derived ovarian cancer cells. Metabolomic analyses were conducted to identify altered metabolites by TRPML1 inhibition. RESULTS Lysosomal exocytosis in response to cisplatin was observed in resistant cancer cells, whereas the phenomenon was absent in sensitive cancer cells. Through the pharmacological intervention of TRPML1, lysosomal exocytosis was interrupted, leading to the sensitization of resistant cancer cells to cisplatin treatment. To assess the impact of lysosomal exocytosis on chemoresistance, we conducted an untargeted metabolomic analysis on cisplatin-resistant ovarian cancer cells with TRPML1 inhibition. Among the 1446 differentially identified metabolites, we focused on 84 significant metabolites. Metabolite set analysis revealed their involvement in diverse pathways. CONCLUSIONS These findings collectively have the potential to enhance our understanding of the interplay between lysosomal exocytosis and chemoresistance, providing valuable insights for the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Boyun Kim
- Department of SmartBio, College of Life and Health Science, Kyungsung University, Busan 48434, Republic of Korea; (B.K.); (G.K.)
| | - Gaeun Kim
- Department of SmartBio, College of Life and Health Science, Kyungsung University, Busan 48434, Republic of Korea; (B.K.); (G.K.)
| | - Heeyeon Kim
- WCU Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea; (H.K.); (Y.S.S.)
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Yong Sang Song
- WCU Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea; (H.K.); (Y.S.S.)
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Jewon Jung
- Department of SmartBio, College of Life and Health Science, Kyungsung University, Busan 48434, Republic of Korea; (B.K.); (G.K.)
| |
Collapse
|
12
|
Shirbhate E, Singh V, Mishra A, Jahoriya V, Veerasamy R, Tiwari AK, Rajak H. Targeting Lysosomes: A Strategy Against Chemoresistance in Cancer. Mini Rev Med Chem 2024; 24:1449-1468. [PMID: 38343053 DOI: 10.2174/0113895575287242240129120002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/11/2024] [Accepted: 01/19/2024] [Indexed: 07/23/2024]
Abstract
Chemotherapy is still the major method of treatment for many types of cancer. Curative cancer therapy is hampered significantly by medication resistance. Acidic organelles like lysosomes serve as protagonists in cellular digestion. Lysosomes, however, are gaining popularity due to their speeding involvement in cancer progression and resistance. For instance, weak chemotherapeutic drugs of basic nature permeate through the lysosomal membrane and are retained in lysosomes in their cationic state, while extracellular release of lysosomal enzymes induces cancer, cytosolic escape of lysosomal hydrolases causes apoptosis, and so on. Drug availability at the sites of action is decreased due to lysosomal drug sequestration, which also enhances cancer resistance. This review looks at lysosomal drug sequestration mechanisms and how they affect cancer treatment resistance. Using lysosomes as subcellular targets to combat drug resistance and reverse drug sequestration is another method for overcoming drug resistance that is covered in this article. The present review has identified lysosomal drug sequestration as one of the reasons behind chemoresistance. The article delves deeper into specific aspects of lysosomal sequestration, providing nuanced insights, critical evaluations, or novel interpretations of different approaches that target lysosomes to defect cancer.
Collapse
Affiliation(s)
- Ekta Shirbhate
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| | - Vaibhav Singh
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| | - Aditya Mishra
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| | - Varsha Jahoriya
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| | - Ravichandran Veerasamy
- Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| | - Amit K Tiwari
- UAMS College of Pharmacy; UAMS - University of Arkansas for Medical Sciences, (AR) USA
| | - Harish Rajak
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| |
Collapse
|
13
|
Shtuhin-Rahav R, Olender A, Zlotkin-Rivkin E, Bouman EA, Danieli T, Nir-Keren Y, Weiss AM, Nandi I, Aroeti B. Enteropathogenic E. coli infection co-elicits lysosomal exocytosis and lytic host cell death. mBio 2023; 14:e0197923. [PMID: 38038448 PMCID: PMC10746156 DOI: 10.1128/mbio.01979-23] [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: 07/25/2023] [Accepted: 10/16/2023] [Indexed: 12/02/2023] Open
Abstract
IMPORTANCE Enteropathogenic Escherichia coli (EPEC) infection is a significant cause of gastroenteritis, mainly in children. Therefore, studying the mechanisms of EPEC infection is an important research theme. EPEC modulates its host cell life by injecting via a type III secretion machinery cell death modulating effector proteins. For instance, while EspF and Map promote mitochondrial cell death, EspZ antagonizes cell death. We show that these effectors also control lysosomal exocytosis, i.e., the trafficking of lysosomes to the host cell plasma membrane. Interestingly, the capacity of these effectors to induce or protect against cell death correlates completely with their ability to induce LE, suggesting that the two processes are interconnected. Modulating host cell death is critical for establishing bacterial attachment to the host and subsequent dissemination. Therefore, exploring the modes of LE involvement in host cell death is crucial for elucidating the mechanisms underlying EPEC infection and disease.
Collapse
Affiliation(s)
- Raisa Shtuhin-Rahav
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, The Edmond J. Safra Campus–Givat Ram, Jerusalem, Israel
- Department of Cell and Developmental Biology, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, The Edmond J. Safra Campus–Givat Ram, Jerusalem, Israel
| | - Aaron Olender
- Department of Cell and Developmental Biology, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, The Edmond J. Safra Campus–Givat Ram, Jerusalem, Israel
- The Alexander Grass Center for Bioengineering, The Hebrew University of Jerusalem, The Edmond J. Safra Campus–Givat Ram, Jerusalem, Israel
| | - Efrat Zlotkin-Rivkin
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, The Edmond J. Safra Campus–Givat Ram, Jerusalem, Israel
- Department of Cell and Developmental Biology, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, The Edmond J. Safra Campus–Givat Ram, Jerusalem, Israel
| | - Etan Amse Bouman
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, The Edmond J. Safra Campus–Givat Ram, Jerusalem, Israel
- Department of Cell and Developmental Biology, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, The Edmond J. Safra Campus–Givat Ram, Jerusalem, Israel
| | - Tsafi Danieli
- The Protein Production Facility, Wolfson Centre for Applied Structural Biology, The Hebrew University of Jerusalem, The Edmond J. Safra Campus–Givat Ram, Jerusalem, Israel
| | - Yael Nir-Keren
- The Protein Production Facility, Wolfson Centre for Applied Structural Biology, The Hebrew University of Jerusalem, The Edmond J. Safra Campus–Givat Ram, Jerusalem, Israel
| | - Aryeh M. Weiss
- Faculty of Engineering, Bar Ilan University, Ramat Gan, Israel
| | - Ipsita Nandi
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, The Edmond J. Safra Campus–Givat Ram, Jerusalem, Israel
- Department of Cell and Developmental Biology, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, The Edmond J. Safra Campus–Givat Ram, Jerusalem, Israel
| | - Benjamin Aroeti
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, The Edmond J. Safra Campus–Givat Ram, Jerusalem, Israel
- Department of Cell and Developmental Biology, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, The Edmond J. Safra Campus–Givat Ram, Jerusalem, Israel
| |
Collapse
|
14
|
Nagahama M, Takehara M, Seike S, Sakaguchi Y. Cellular Uptake and Cytotoxicity of Clostridium perfringens Iota-Toxin. Toxins (Basel) 2023; 15:695. [PMID: 38133199 PMCID: PMC10747272 DOI: 10.3390/toxins15120695] [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: 11/22/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 12/23/2023] Open
Abstract
Clostridium perfringens iota-toxin is composed of two separate proteins: a binding protein (Ib) that recognizes a host cell receptor and promotes the cellular uptake of a catalytic protein and (Ia) possessing ADP-ribosyltransferase activity that induces actin cytoskeleton disorganization. Ib exhibits the overall structure of bacterial pore-forming toxins (PFTs). Lipolysis-stimulated lipoprotein receptor (LSR) is defined as a host cell receptor for Ib. The binding of Ib to LSR causes an oligomer formation of Ib in lipid rafts of plasma membranes, mediating the entry of Ia into the cytoplasm. Ia induces actin cytoskeleton disruption via the ADP-ribosylation of G-actin and causes cell rounding and death. The binding protein alone disrupts the cell membrane and induces cytotoxicity in sensitive cells. Host cells permeabilized by the pore formation of Ib are repaired by a Ca2+-dependent plasma repair pathway. This review shows that the cellular uptake of iota-toxin utilizes a pathway of plasma membrane repair and that Ib alone induces cytotoxicity.
Collapse
Affiliation(s)
- Masahiro Nagahama
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan; (M.T.); (Y.S.)
| | - Masaya Takehara
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan; (M.T.); (Y.S.)
| | - Soshi Seike
- Laboratory of Molecular Microbiological Science, Faculty of Pharmaceutical Sciences, Hiroshima International University, Kure, Hiroshima 737-0112, Japan;
| | - Yoshihiko Sakaguchi
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan; (M.T.); (Y.S.)
| |
Collapse
|
15
|
Tsarouhas V, Liu D, Tsikala G, Engström Y, Strigini M, Samakovlis C. A surfactant lipid layer of endosomal membranes facilitates airway gas filling in Drosophila. Curr Biol 2023; 33:5132-5146.e5. [PMID: 37992718 DOI: 10.1016/j.cub.2023.10.058] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 09/14/2023] [Accepted: 10/26/2023] [Indexed: 11/24/2023]
Abstract
The mechanisms underlying the construction of an air-liquid interface in respiratory organs remain elusive. Here, we use live imaging and genetic analysis to describe the morphogenetic events generating an extracellular lipid lining of the Drosophila airways required for their gas filing and animal survival. We show that sequential Rab39/Syx1A/Syt1-mediated secretion of lysosomal acid sphingomyelinase (Drosophila ASM [dASM]) and Rab11/35/Syx1A/Rop-dependent exosomal secretion provides distinct components for lipid film assembly. Tracheal inactivation of Rab11 or Rab35 or loss of Rop results in intracellular accumulation of exosomal, multi-vesicular body (MVB)-derived vesicles. On the other hand, loss of dASM or Rab39 causes luminal bubble-like accumulations of exosomal membranes and liquid retention in the airways. Inactivation of the exosomal secretion in dASM mutants counteracts this phenotype, arguing that the exosomal secretion provides the lipid vesicles and that secreted lysosomal dASM organizes them into a continuous film. Our results reveal the coordinated functions of extracellular vesicle and lysosomal secretions in generating a lipid layer crucial for airway gas filling and survival.
Collapse
Affiliation(s)
- Vasilios Tsarouhas
- Stockholm University, Department of Molecular Biosciences, The Wenner-Gren Institute, 10691 Stockholm, Sweden; Science for Life Laboratory, SciLifeLab, 171 65 Stockholm, Sweden.
| | - Dan Liu
- Stockholm University, Department of Molecular Biosciences, The Wenner-Gren Institute, 10691 Stockholm, Sweden
| | - Georgia Tsikala
- Stockholm University, Department of Molecular Biosciences, The Wenner-Gren Institute, 10691 Stockholm, Sweden; IMBB, 70013 Heraklion, Crete, Greece
| | - Ylva Engström
- Stockholm University, Department of Molecular Biosciences, The Wenner-Gren Institute, 10691 Stockholm, Sweden
| | | | - Christos Samakovlis
- Stockholm University, Department of Molecular Biosciences, The Wenner-Gren Institute, 10691 Stockholm, Sweden; Science for Life Laboratory, SciLifeLab, 171 65 Stockholm, Sweden; ECCPS, Justus Liebig University of Giessen, 35390 Giessen, Germany.
| |
Collapse
|
16
|
Yeo S, Jang J, Jung HJ, Lee H, Choe Y. Primary cilia-mediated regulation of microglial secretion in Alzheimer's disease. Front Mol Biosci 2023; 10:1250335. [PMID: 37942288 PMCID: PMC10627801 DOI: 10.3389/fmolb.2023.1250335] [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: 06/30/2023] [Accepted: 09/28/2023] [Indexed: 11/10/2023] Open
Abstract
Alzheimer's disease (AD) is a brain disorder manifested by a gradual decline in cognitive function due to the accumulation of extracellular amyloid plaques, disruptions in neuronal substance transport, and the degeneration of neurons. In affected neurons, incomplete clearance of toxic proteins by neighboring microglia leads to irreversible brain inflammation, for which cellular signaling is poorly understood. Through single-cell transcriptomic analysis, we discovered distinct regional differences in the ability of microglia to clear damaged neurites. Specifically, microglia in the septal region of wild type mice exhibited a transcriptomic signature resembling disease-associated microglia (DAM). These lateral septum (LS)-enriched microglia were associated with dense axonal bundles originating from the hippocampus. Further transcriptomic and proteomic approaches revealed that primary cilia, small hair-like structures found on cells, played a role in the regulation of microglial secretory function. Notably, primary cilia were transiently observed in microglia, and their presence was significantly reduced in microglia from AD mice. We observed significant changes in the secretion and proteomic profiles of the secretome after inhibiting the primary cilia gene intraflagellar transport particle 88 (Ift88) in microglia. Intriguingly, inhibiting primary cilia in the septal microglia of AD mice resulted in the expansion of extracellular amyloid plaques and damage to adjacent neurites. These results indicate that DAM-like microglia are present in the LS, a critical target region for hippocampal nerve bundles, and that the primary ciliary signaling system regulates microglial secretion, affecting extracellular proteostasis. Age-related primary ciliopathy probably contributes to the selective sensitivity of microglia, thereby exacerbating AD. Targeting the primary ciliary signaling system could therefore be a viable strategy for modulating neuroimmune responses in AD treatments.
Collapse
Affiliation(s)
- Seungeun Yeo
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Jaemyung Jang
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Hyun Jin Jung
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Hyeyoung Lee
- Division of Applied Bioengineering, Dong-eui University, Busan, Republic of Korea
| | - Youngshik Choe
- Korea Brain Research Institute, Daegu, Republic of Korea
| |
Collapse
|
17
|
Skotland T, Llorente A, Sandvig K. Lipids in Extracellular Vesicles: What Can Be Learned about Membrane Structure and Function? Cold Spring Harb Perspect Biol 2023; 15:a041415. [PMID: 37277192 PMCID: PMC10411865 DOI: 10.1101/cshperspect.a041415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Extracellular vesicles, such as exosomes, can be used as interesting models to study the structure and function of biological membranes as these vesicles contain only one membrane (i.e., one lipid bilayer). In addition to lipids, they contain proteins, nucleic acids, and various other molecules. The lipid composition of exosomes is here compared to HIV particles and detergent-resistant membranes, which also have a high content of sphingolipids, cholesterol, and phosphatidylserine (PS). We discuss interactions between the lipids in the two bilayers, and especially those between PS 18:0/18:1 in the inner leaflet and the very-long-chain sphingolipids in the outer leaflet, and the importance of cholesterol for these interactions. We also briefly discuss the involvement of ether-linked phospholipids (PLs) in such lipid raft-like structures, and the possible involvement of these and other lipid classes in the formation of exosomes. The urgent need to improve the quality of quantitative lipidomic studies is highlighted.
Collapse
Affiliation(s)
- Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway
| | - Alicia Llorente
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway
- Department of Mechanical, Electronics and Chemical Engineering, Oslo Metropolitan University, 0167 Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, 0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway
- Department of Molecular Biosciences, University of Oslo, 0316 Oslo, Norway
| |
Collapse
|
18
|
Placci M, Giannotti MI, Muro S. Polymer-based drug delivery systems under investigation for enzyme replacement and other therapies of lysosomal storage disorders. Adv Drug Deliv Rev 2023; 197:114683. [PMID: 36657645 PMCID: PMC10629597 DOI: 10.1016/j.addr.2022.114683] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/30/2022] [Accepted: 12/25/2022] [Indexed: 01/18/2023]
Abstract
Lysosomes play a central role in cellular homeostasis and alterations in this compartment associate with many diseases. The most studied example is that of lysosomal storage disorders (LSDs), a group of 60 + maladies due to genetic mutations affecting lysosomal components, mostly enzymes. This leads to aberrant intracellular storage of macromolecules, altering normal cell function and causing multiorgan syndromes, often fatal within the first years of life. Several treatment modalities are available for a dozen LSDs, mostly consisting of enzyme replacement therapy (ERT) strategies. Yet, poor biodistribution to main targets such as the central nervous system, musculoskeletal tissue, and others, as well as generation of blocking antibodies and adverse effects hinder effective LSD treatment. Drug delivery systems are being studied to surmount these obstacles, including polymeric constructs and nanoparticles that constitute the focus of this article. We provide an overview of the formulations being tested, the diseases they aim to treat, and the results observed from respective in vitro and in vivo studies. We also discuss the advantages and disadvantages of these strategies, the remaining gaps of knowledge regarding their performance, and important items to consider for their clinical translation. Overall, polymeric nanoconstructs hold considerable promise to advance treatment for LSDs.
Collapse
Affiliation(s)
- Marina Placci
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain
| | - Marina I Giannotti
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain; CIBER-BBN, ISCIII, Barcelona, Spain; Department of Materials Science and Physical Chemistry, University of Barcelona, Barcelona 08028, Spain
| | - Silvia Muro
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain; Institute of Catalonia for Research and Advanced Studies (ICREA), Barcelona 08010, Spain; Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, USA; Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
19
|
Wagh AR, Sulakshane P, Glickman MH. Alzheimer's disease-associated mutant ubiquitin (UBB +1) is secreted through an autophagosome-like vesicle-mediated unconventional pathway. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194936. [PMID: 37075976 DOI: 10.1016/j.bbagrm.2023.194936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/28/2023] [Accepted: 04/13/2023] [Indexed: 04/21/2023]
Abstract
Misfolded protein aggregation at both intracellular and extracellular milieus is thought to be the major etiology of Alzheimer's disease (AD). UBB+1, a frameshift variant of the ubiquitin B gene (UBB) results in a folded ubiquitin domain fused to a flexible unstructured extension. Accumulation of UBB+1 in extracellular plaques in the brains of AD patients undoubtedly suggests a role of the ubiquitin-proteasome system in AD. However, the exact mechanism of extracellular secretion of UBB+1 remains unknown. In an attempt to understand the molecular mechanism of UBB+1 secretion, we performed a survey of secretory pathways and identified the involvement of unconventional autophagosome-mediated UBB+1 secretion. Expression of UBB+1 was sufficient to stimulate LC3B/Atg8 conversion from LC3B-I to LC3B-II, which indicates initiation of the autophagy pathway. Furthermore, deficiency of ATG5 - a key player in autophagosome formation - inhibited UBB+1 secretion. Based on immunofluorescence 3D structured illumination (SIM) microscopy and co-immunoprecipitation, we provide evidence that UBB+1 is associated with the secretory autophagosome marker, SEC22B, while HSP90 possibly acts as a carrier. Using LC-MS/MS and mutagenesis we found that in cells, UBB+1 is ubiquitinated on lysine 11, 29, and 48, however, this ubiquitination does not contribute to its secretion. By contrast, proteasome or lysosome inhibition slightly enhanced secretion. Taken together, this study suggests that by ridding cells of UBB+1, secretory autophagosomes may alleviate the cellular stress associated with UBB+1, yet simultaneously mediate the spreading of a mutant specie with disordered characteristics to the extracellular milieu.
Collapse
Affiliation(s)
- Ajay R Wagh
- The Faculty of Biology, Technion Israel Institute of Technology, Haifa 32000, Israel
| | - Prasad Sulakshane
- The Faculty of Biology, Technion Israel Institute of Technology, Haifa 32000, Israel
| | - Michael H Glickman
- The Faculty of Biology, Technion Israel Institute of Technology, Haifa 32000, Israel.
| |
Collapse
|
20
|
Huang Y, Liang J, Fan Z. A review: Small organic molecule dual/multi-organelle-targeted fluorescent probes. Talanta 2023; 259:124529. [PMID: 37084606 DOI: 10.1016/j.talanta.2023.124529] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/21/2023] [Accepted: 04/03/2023] [Indexed: 04/23/2023]
Abstract
In recent years, the dual/multi-organelle-targeted fluorescent probe based on small organic molecules has good biocompatibility and can visualize the interaction between different organelles, which has attracted much attention. In addition, these probes can also be used to detect small molecules in the organelle environment, such as active sulfur species (RSS), reactive oxygen species (ROS), pH, viscosity and so on. However, the review of dual/multi-organelle-targeted fluorescent probe for small organic molecules lacks a systematic summary, which may hinder the development of this field. In this review, we will focus on the design strategies and bioimaging applications of dual/multi-organelle-targeted fluorescent probe, and classify them into six classes according to different organelles targeted. The first class probe targeted mitochondria and lysosome. The second class probe targeted endoplasmic reticulum and lysosome. The third class probe targeted mitochondria and lipid droplets. The fourth class probe targeted endoplasmic reticulum and lipid droplets. The fifth class probe targeted lysosome and lipid droplets. The sixth class multi-targeted probe. The mechanism of these probes targeting organelles and the visualization of the interaction between different organelles are emphasized, and the prospect and future development direction of this research field are prospected. This will provide a systematic idea for the development and functional research of dual/multi-organelle-targeted fluorescent probe, and promote its research in related physiological and pathological medicine field in the future.
Collapse
Affiliation(s)
- Yongfei Huang
- Key Laboratory of Magnetic Molecules and Magnetic Information Materials of Ministry of Education & School of Chemistry and Materials Science of Shanxi Normal University, TaiYuan, 030032, China
| | - Junping Liang
- Key Laboratory of Magnetic Molecules and Magnetic Information Materials of Ministry of Education & School of Chemistry and Materials Science of Shanxi Normal University, TaiYuan, 030032, China
| | - Zhefeng Fan
- Key Laboratory of Magnetic Molecules and Magnetic Information Materials of Ministry of Education & School of Chemistry and Materials Science of Shanxi Normal University, TaiYuan, 030032, China.
| |
Collapse
|
21
|
Racchetti G, Meldolesi J. Four distinct cytoplasmic structures generate and release specific vesicles, thus opening the way to intercellular communication. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2023; 4:44-58. [PMID: 39698300 PMCID: PMC11648438 DOI: 10.20517/evcna.2023.03] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/26/2023] [Accepted: 03/07/2023] [Indexed: 12/20/2024]
Abstract
In all cells, generation and release of specific vesicles are the initial steps of back-and-forth intercellular communication. These processes are critical in normal physiology and pathophysiology. Vesicles have particular functions appropriate to their targets. When stimulated, they are released into the extracellular space. Four cytoplasmic membrane-bound structures generate their particular vesicles. Among these structures, multivesicular bodies (MVBs) can accumulate many small vesicles in their lumen; release occurs upon MVB exocytosis. Ectosomes are larger vesicles characterized by their responses and are generated directly and released independently from specific microdomains pre-established in the thickness of the plasma membrane. Most lysosomes do not generate vesicles. However, unique components of a minor form, the endo-lysosome, constitute the third class of structures that release a few vesicles by exocytosis with molecules and structures inducing changes in the extracellular environment. The autophagosome, the fourth structure, releases several heterogeneous vesicles by exocytosis with malformed bio-molecules, assembled structures, and damaged organelles. Interestingly, the frequent interaction of autophagosomes with MVBs and their exosomes contributes to the regulation and intensity of their action. The specificity and function of released vesicles depend on their membranes' and luminal cargoes' composition and dynamics. An ongoing investigation of the various vesicles reveals new properties regarding their generation, release, and resulting extracellular processes. The growth of information about structures and their vesicles progressively extends the knowledge base regarding cell communication and contributes to their clinical applications.
Collapse
Affiliation(s)
- Gabriella Racchetti
- San Raffaele Institute, Vita-Salute San Raffaele University, Milan 20132, Italy
| | - Jacopo Meldolesi
- San Raffaele Institute, Vita-Salute San Raffaele University, Milan 20132, Italy
- CNR Institute of Neuroscience at the Milano-Bicocca University, Vedano al Lambro, Milan 20854, Italy
| |
Collapse
|
22
|
Riederer E, Cang C, Ren D. Lysosomal Ion Channels: What Are They Good For and Are They Druggable Targets? Annu Rev Pharmacol Toxicol 2023; 63:19-41. [PMID: 36151054 DOI: 10.1146/annurev-pharmtox-051921-013755] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Lysosomes play fundamental roles in material digestion, cellular clearance, recycling, exocytosis, wound repair, Ca2+ signaling, nutrient signaling, and gene expression regulation. The organelle also serves as a hub for important signaling networks involving the mTOR and AKT kinases. Electrophysiological recording and molecular and structural studies in the past decade have uncovered several unique lysosomal ion channels and transporters, including TPCs, TMEM175, TRPMLs, CLN7, and CLC-7. They underlie the organelle's permeability to major ions, including K+, Na+, H+, Ca2+, and Cl-. The channels are regulated by numerous cellular factors, ranging from H+ in the lumen and voltage across the lysosomal membrane to ATP in the cytosol to growth factors outside the cell. Genetic variations in the channel/transporter genes are associated with diseases that include lysosomal storage diseases and neurodegenerative diseases. Recent studies with human genetics and channel activators suggest that lysosomal channels may be attractive targets for the development of therapeutics for the prevention of and intervention in human diseases.
Collapse
Affiliation(s)
- Erika Riederer
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA; ,
| | - Chunlei Cang
- CAS Key Laboratory of Innate Immunity and Chronic Disease, Neurodegenerative Disorder Research Center, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China;
| | - Dejian Ren
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA; ,
| |
Collapse
|
23
|
Abstract
Lysosomes are acidic membrane-bound organelles that use hydrolytic enzymes to break down material through pathways such as endocytosis, phagocytosis, mitophagy, and autophagy. To function properly, intralysosomal environments are strictly controlled by a set of integral membrane proteins such as ion channels and transporters. Potassium ion (K+) channels are a large and diverse family of membrane proteins that control K+ flux across both the plasma membrane and intracellular membranes. In the plasma membrane, they are essential in both excitable and non-excitable cells for the control of membrane potential and cell signaling. However, our understanding of intracellular K+ channels is very limited. In this review, we summarize the recent development in studies of K+ channels in the lysosome. We focus on their characterization, potential roles in maintaining lysosomal membrane potential and lysosomal function, and pathological implications.
Collapse
Affiliation(s)
- Peng Huang
- Collaborative Innovation Center for Biomedicine, School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Mengnan Xu
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, Halifax, NS, Canada
| | - Yi Wu
- Collaborative Innovation Center for Biomedicine, School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Alia Kazim Rizvi Syeda
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, Halifax, NS, Canada
| | - Xian-Ping Dong
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, Halifax, NS, Canada.
| |
Collapse
|
24
|
Abstract
Ca2+ is a universal second messenger that plays a wide variety of fundamental roles in cellular physiology. Thus, to warrant selective responses and to allow rapid mobilization upon specific stimuli, Ca2+ is accumulated in organelles to keep it at very low levels in the cytoplasm during resting conditions. Major Ca2+ storage organelles include the endoplasmic reticulum (ER), the mitochondria, and as recently demonstrated, the lysosome (Xu and Ren, Annu Rev Physiol 77:57-80, 2015). The importance of Ca2+ signaling deregulation in human physiology is underscored by its involvement in several human diseases, including lysosomal storage disorders, neurodegenerative disease and cancer (Shen et al., Nat Commun 3:731, 2012; Bae et al., J Neurosci 34:11485-11503, 2014). Recent evidence strongly suggests that lysosomal Ca2+ plays a major role in the regulation of lysosomal adaptation to nutrient availability through a lysosomal signaling pathway involving the lysosomal Ca2+ channel TRPML1 and the transcription factor TFEB, a master regulator for lysosomal function and autophagy (Sardiello et al., Science 325:473-477, 2009; Settembre et al., Science 332:1429-1433, 2011; Medina et al., Nat Cell Biol 17:288-299, 2015; Di Paola et al., Cell Calcium 69:112-121, 2018). Due to the tight relationship of this lysosomal Ca2+ channel and TFEB, in this chapter, we will focus on the role of the TRPML1/TFEB pathway in the regulation of lysosomal function and autophagy.
Collapse
Affiliation(s)
- Diego Luis Medina
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy.
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Naples, Italy.
| |
Collapse
|
25
|
Bao F, Zhou L, Xiao J, Liu X. Mitolysosome exocytosis: a novel mitochondrial quality control pathway linked with parkinsonism-like symptoms. Biochem Soc Trans 2022; 50:1773-1783. [PMID: 36484629 DOI: 10.1042/bst20220726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/14/2022] [Accepted: 11/25/2022] [Indexed: 06/17/2023]
Abstract
Quality control of mitochondria is essential for their homeostasis and function. Light chain 3 (LC3) associated autophagosomes-mediated mitophagy represents a canonical mitochondrial quality control pathway. Alternative quality control processes, such as mitochondrial-derived vesicles (MDVs), have been discovered, but the intact mitochondrial quality control remains unknown. We recently discovered a novel mitolysosome exocytosis mechanism for mitochondrial quality control in flunarizine (FNZ)-induced mitochondria clearance, where autophagosomes are not required, but rather mitochondria are engulfed directly by lysosomes, mediating mitochondrial secretion. As FNZ results in parkinsonism, we propose that excessive mitolysosome exocytosis is the cause.
Collapse
Affiliation(s)
- Feixiang Bao
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Lingyan Zhou
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiahui Xiao
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xingguo Liu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
| |
Collapse
|
26
|
Gaudioso Á, Silva TP, Ledesma MD. Models to study basic and applied aspects of lysosomal storage disorders. Adv Drug Deliv Rev 2022; 190:114532. [PMID: 36122863 DOI: 10.1016/j.addr.2022.114532] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 08/05/2022] [Accepted: 09/04/2022] [Indexed: 01/24/2023]
Abstract
The lack of available treatments and fatal outcome in most lysosomal storage disorders (LSDs) have spurred research on pathological mechanisms and novel therapies in recent years. In this effort, experimental methodology in cellular and animal models have been developed, with aims to address major challenges in many LSDs such as patient-to-patient variability and brain condition. These techniques and models have advanced knowledge not only of LSDs but also for other lysosomal disorders and have provided fundamental insights into the biological roles of lysosomes. They can also serve to assess the efficacy of classical therapies and modern drug delivery systems. Here, we summarize the techniques and models used in LSD research, which include both established and recently developed in vitro methods, with general utility or specifically addressing lysosomal features. We also review animal models of LSDs together with cutting-edge technology that may reduce the need for animals in the study of these devastating diseases.
Collapse
Affiliation(s)
- Ángel Gaudioso
- Centro Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Teresa P Silva
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | | |
Collapse
|
27
|
Gletten RB, Cantrell LS, Bhattacharya S, Schey KL. Lens Aquaporin-5 Inserts Into Bovine Fiber Cell Plasma Membranes Via Unconventional Protein Secretion. Invest Ophthalmol Vis Sci 2022; 63:5. [PMID: 35816045 PMCID: PMC9284464 DOI: 10.1167/iovs.63.8.5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose To spatially map aquaporin-5 (AQP5) expression in the bovine lens, molecularly characterize cytoplasmic AQP5-containing vesicles in the outer cortex, and elucidate AQP5 membrane trafficking mechanisms. Methods Immunofluorescence was performed on bovine lens cryosections using AQP5, TOMM20, COX IV, calnexin, LC3B, Sec22β, LIMP-2, and connexin 50 antibodies and the membrane dye CM-DiI. AQP5 plasma membrane insertion was defined via line expression profile analysis. Transmission electron microscopy (TEM) was performed on bovine lens sections to examine cytoplasmic organelle morphology and subcellular localization in cortical fiber cells. Bovine lenses were treated with 10-nM bafilomycin A1 or 0.1% dimethyl sulfoxide vehicle control for 24 hours in ex vivo culture to determine changes in AQP5 plasma membrane expression. Results Immunofluorescence analysis revealed cytoplasmic AQP5 expression in lens epithelial cells and differentiating fiber cells. In the lens cortex, complete AQP5 plasma membrane insertion occurs at r/a = 0.951 ± 0.005. AQP5-containing cytoplasmic vesicles are spheroidal in morphology with linear extensions, express TOMM20, and contain LC3B and LIMP-2, but not Sec22β, as fiber cells mature. TEM analysis revealed complex vesicular assemblies with congruent subcellular localization to AQP5-containing cytoplasmic vesicles. AQP5-containing cytoplasmic vesicles appear to dock with the plasma membrane. Bafilomycin A1 treatment reduced AQP5 plasma membrane expression by 27%. Conclusions AQP5 localizes to spheroidal, linear cytoplasmic vesicles in the differentiating bovine lens fiber cells. During fiber cell differentiation, these vesicles incorporate LC3B and presumably fuse with LIMP-2–positive lysosomes. Our data suggest that AQP5 to the plasma membrane through lysosome-associated unconventional protein secretion, a novel mechanism of AQP5 trafficking.
Collapse
Affiliation(s)
- Romell B Gletten
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, United States
| | - Lee S Cantrell
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, United States
| | - Sujoy Bhattacharya
- Department of Ophthalmology and Visual Sciences, Vanderbilt University, Nashville, Tennessee, United States
| | - Kevin L Schey
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, United States
| |
Collapse
|
28
|
Moon OJ, Yoon CJ, Lee BR, Lee J. An Optimally Fabricated Platform Guides Cancer-Specific Activation of Chemotherapeutic Drugs and Toxicity-free Cancer Treatment. Adv Healthc Mater 2022; 11:e2200765. [PMID: 35670274 DOI: 10.1002/adhm.202200765] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/12/2022] [Indexed: 11/08/2022]
Abstract
Cancer chemotherapeutic drugs such as doxorubicin, mitomycin C, and gemcitabine, which are mostly small synthetic molecules, are still clinically useful for cancer treatment. However, despite considerable therapeutic efficacy, severe toxicity-associated problems, which are mainly caused by the non-specific mode of action such as chromosomal DNA damage and interference in the DNA replication even in normal cells, remain unresolved and a major challenge for safer and thus more widespread adoption of chemotherapy. Here we developed an innovative platform through beneficially integrating core peptide units into highly-ordered, stable, and flexibly guest-adaptable structure of apoferritin, which simultaneously fulfills high-capacity loading of chemotherapeutic drugs compared with the case of FDA-approved antibody-drug conjugates, efficient drug targeting to cancer cells, and cancer cell-specific drug release and activation. This approach dramatically reduced drug toxicity to normal cells, significantly enhanced efficacy in in vivo cancer treatment without toxicity to normal organs of mice, and thus is expected to open up a novel clinical route to break through the limits of current cancer chemotherapy. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ok Jeong Moon
- Department of Chemical and Biological Engineering, College of Engineering, Korea University, Anam-Ro 145, Seoul, 136-713, Republic of Korea
| | - Chul Joo Yoon
- Department of Chemical and Biological Engineering, College of Engineering, Korea University, Anam-Ro 145, Seoul, 136-713, Republic of Korea
| | - Bo-Ram Lee
- Department of Chemical and Biological Engineering, College of Engineering, Korea University, Anam-Ro 145, Seoul, 136-713, Republic of Korea
| | - Jeewon Lee
- Department of Chemical and Biological Engineering, College of Engineering, Korea University, Anam-Ro 145, Seoul, 136-713, Republic of Korea
| |
Collapse
|
29
|
Lan Y, He W, Wang G, Wang Z, Chen Y, Gao F, Song D. Potential Antiviral Strategy Exploiting Dependence of SARS-CoV-2 Replication on Lysosome-Based Pathway. Int J Mol Sci 2022; 23:ijms23116188. [PMID: 35682877 PMCID: PMC9181800 DOI: 10.3390/ijms23116188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/29/2022] [Accepted: 05/30/2022] [Indexed: 12/04/2022] Open
Abstract
The recent novel coronavirus (SARS-CoV-2) disease (COVID-19) outbreak created a severe public health burden worldwide. Unfortunately, the SARS-CoV-2 variant is still spreading at an unprecedented speed in many countries and regions. There is still a lack of effective treatment for moderate and severe COVID-19 patients, due to a lack of understanding of the SARS-CoV-2 life cycle. Lysosomes, which act as “garbage disposals” for nearly all types of eukaryotic cells, were shown in numerous studies to support SARS-CoV-2 replication. Lysosome-associated pathways are required for virus entry and exit during replication. In this review, we summarize experimental evidence demonstrating a correlation between lysosomal function and SARS-CoV-2 replication, and the development of lysosomal perturbation drugs as anti-SARS-CoV-2 agents.
Collapse
Affiliation(s)
- Yungang Lan
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130022, China; (W.H.); (Z.W.); (Y.C.); (F.G.)
- Correspondence: (Y.L.); (D.S.)
| | - Wenqi He
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130022, China; (W.H.); (Z.W.); (Y.C.); (F.G.)
| | - Gaili Wang
- Jilin Academy of Animal Husbandry and Veterinary Medicine, Changchun 130022, China;
| | - Zhenzhen Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130022, China; (W.H.); (Z.W.); (Y.C.); (F.G.)
| | - Yuzhu Chen
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130022, China; (W.H.); (Z.W.); (Y.C.); (F.G.)
| | - Feng Gao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130022, China; (W.H.); (Z.W.); (Y.C.); (F.G.)
| | - Deguang Song
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130022, China; (W.H.); (Z.W.); (Y.C.); (F.G.)
- Correspondence: (Y.L.); (D.S.)
| |
Collapse
|
30
|
Barral DC, Staiano L, Guimas Almeida C, Cutler DF, Eden ER, Futter CE, Galione A, Marques ARA, Medina DL, Napolitano G, Settembre C, Vieira OV, Aerts JMFG, Atakpa‐Adaji P, Bruno G, Capuozzo A, De Leonibus E, Di Malta C, Escrevente C, Esposito A, Grumati P, Hall MJ, Teodoro RO, Lopes SS, Luzio JP, Monfregola J, Montefusco S, Platt FM, Polishchuck R, De Risi M, Sambri I, Soldati C, Seabra MC. Current methods to analyze lysosome morphology, positioning, motility and function. Traffic 2022; 23:238-269. [PMID: 35343629 PMCID: PMC9323414 DOI: 10.1111/tra.12839] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 01/09/2023]
Abstract
Since the discovery of lysosomes more than 70 years ago, much has been learned about the functions of these organelles. Lysosomes were regarded as exclusively degradative organelles, but more recent research has shown that they play essential roles in several other cellular functions, such as nutrient sensing, intracellular signalling and metabolism. Methodological advances played a key part in generating our current knowledge about the biology of this multifaceted organelle. In this review, we cover current methods used to analyze lysosome morphology, positioning, motility and function. We highlight the principles behind these methods, the methodological strategies and their advantages and limitations. To extract accurate information and avoid misinterpretations, we discuss the best strategies to identify lysosomes and assess their characteristics and functions. With this review, we aim to stimulate an increase in the quantity and quality of research on lysosomes and further ground-breaking discoveries on an organelle that continues to surprise and excite cell biologists.
Collapse
Affiliation(s)
- Duarte C. Barral
- CEDOC, NOVA Medical School, NMS, Universidade NOVA de LisboaLisbonPortugal
| | - Leopoldo Staiano
- Telethon Institute of Genetics and Medicine (TIGEM)PozzuoliItaly
- Institute for Genetic and Biomedical ResearchNational Research Council (CNR)MilanItaly
| | | | - Dan F. Cutler
- MRC Laboratory for Molecular Cell BiologyUniversity College LondonLondonUK
| | - Emily R. Eden
- University College London (UCL) Institute of OphthalmologyLondonUK
| | - Clare E. Futter
- University College London (UCL) Institute of OphthalmologyLondonUK
| | | | | | - Diego Luis Medina
- Telethon Institute of Genetics and Medicine (TIGEM)PozzuoliItaly
- Medical Genetics Unit, Department of Medical and Translational ScienceFederico II UniversityNaplesItaly
| | - Gennaro Napolitano
- Telethon Institute of Genetics and Medicine (TIGEM)PozzuoliItaly
- Medical Genetics Unit, Department of Medical and Translational ScienceFederico II UniversityNaplesItaly
| | - Carmine Settembre
- Telethon Institute of Genetics and Medicine (TIGEM)PozzuoliItaly
- Clinical Medicine and Surgery DepartmentFederico II UniversityNaplesItaly
| | - Otília V. Vieira
- CEDOC, NOVA Medical School, NMS, Universidade NOVA de LisboaLisbonPortugal
| | | | | | - Gemma Bruno
- Telethon Institute of Genetics and Medicine (TIGEM)PozzuoliItaly
| | | | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine (TIGEM)PozzuoliItaly
- Institute of Biochemistry and Cell Biology, CNRRomeItaly
| | - Chiara Di Malta
- Telethon Institute of Genetics and Medicine (TIGEM)PozzuoliItaly
- Medical Genetics Unit, Department of Medical and Translational ScienceFederico II UniversityNaplesItaly
| | | | | | - Paolo Grumati
- Telethon Institute of Genetics and Medicine (TIGEM)PozzuoliItaly
| | - Michael J. Hall
- CEDOC, NOVA Medical School, NMS, Universidade NOVA de LisboaLisbonPortugal
| | - Rita O. Teodoro
- CEDOC, NOVA Medical School, NMS, Universidade NOVA de LisboaLisbonPortugal
| | - Susana S. Lopes
- CEDOC, NOVA Medical School, NMS, Universidade NOVA de LisboaLisbonPortugal
| | - J. Paul Luzio
- Cambridge Institute for Medical ResearchUniversity of CambridgeCambridgeUK
| | | | | | | | | | - Maria De Risi
- Telethon Institute of Genetics and Medicine (TIGEM)PozzuoliItaly
| | - Irene Sambri
- Telethon Institute of Genetics and Medicine (TIGEM)PozzuoliItaly
- Medical Genetics Unit, Department of Medical and Translational ScienceFederico II UniversityNaplesItaly
| | - Chiara Soldati
- Telethon Institute of Genetics and Medicine (TIGEM)PozzuoliItaly
| | - Miguel C. Seabra
- CEDOC, NOVA Medical School, NMS, Universidade NOVA de LisboaLisbonPortugal
| |
Collapse
|
31
|
Reinheckel T, Tholen M. Low level lysosomal membrane permeabilization for limited release and sub-lethal functions of cathepsin proteases in the cytosol and nucleus. FEBS Open Bio 2022; 12:694-707. [PMID: 35203107 PMCID: PMC8972055 DOI: 10.1002/2211-5463.13385] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/21/2022] [Accepted: 02/15/2022] [Indexed: 11/12/2022] Open
Abstract
For a long time, lysosomes were purely seen as organelles in charge of garbage disposal within the cell. They destroy any cargo delivered into their lumen with a plethora of highly potent hydrolytic enzymes, including various proteases. In case of damage to their limiting membranes, the lysosomes release their soluble content with detrimental outcomes for the cell. In recent years however, this view of the lysosome changed towards acknowledging it as a platform for integration of manifold intra- and extracellular signals. Even impaired lysosomal membrane integrity is no longer considered to be a one-way street to cell death. Increasing evidence suggests that lysosomal enzymes, mainly cathepsin proteases, can be released in a spatially and temporarily restricted manner that is compatible with cellular survival. This way, cathepsins can act in the cytosol and the nucleus, where they affect important cellular processes such as cell division. Here, we review this evidence and discuss the routes and molecular mechanisms by which the cathepsins may reach their unusual destination.
Collapse
Affiliation(s)
- Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University, Freiburg, Germany.,German Cancer Consortium (DKTK) Partner Site Freiburg, Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Biological Signaling Studies BIOSS, Albert Ludwigs University, Freiburg, Germany
| | - Martina Tholen
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University, Freiburg, Germany
| |
Collapse
|
32
|
Wu Y, Xu M, Wang P, Syeda AKR, Huang P, Dong XP. Lysosomal potassium channels. Cell Calcium 2022; 102:102536. [PMID: 35016151 DOI: 10.1016/j.ceca.2022.102536] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/21/2022]
Abstract
The lysosome is an important membrane-bound acidic organelle that is regarded as the degradative center as well as multifunctional signaling hub. It digests unwanted macromolecules, damaged organelles, microbes, and other materials derived from endocytosis, autophagy, and phagocytosis. To function properly, the ionic homeostasis and membrane potential of the lysosome are strictly regulated by transporters and ion channels. As the most abundant cation inside the cell, potassium ions (K+) are vital for lysosomal membrane potential and lysosomal calcium (Ca2+) signaling. However, our understanding about how lysosomal K+homeostasis is regulated and what are the functions of K+in the lysosome is very limited. Currently, two lysosomal K+channels have been identified: large-conductance Ca2+-activated K+channel (BK) and transmembrane Protein 175 (TMEM175). In this review, we summarize recent development in our understanding of K+ homeostasis and K+channels in the lysosome. We hope to guide the readers into a more in-depth discussion of lysosomal K+ channels in lysosomal physiology and human diseases.
Collapse
Affiliation(s)
- Yi Wu
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Rd, Shanghai 201318, China; School of Pharmacy, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Rd, Shanghai 201318, China
| | - Mengnan Xu
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, Halifax, Nova NS B3H 4R2, Canada
| | - Pingping Wang
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, Halifax, Nova NS B3H 4R2, Canada
| | - Alia Kazim Rizvi Syeda
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, Halifax, Nova NS B3H 4R2, Canada
| | - Peng Huang
- Collaborative Innovation Center for Biomedicine, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Rd, Shanghai 201318, China; School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Rd, Shanghai 201318, China.
| | - Xian-Ping Dong
- Department of Physiology and Biophysics, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, Halifax, Nova NS B3H 4R2, Canada.
| |
Collapse
|
33
|
Yoshihara M, Mizutani S, Kato Y, Matsumoto K, Mizutani E, Mizutani H, Fujimoto H, Osuka S, Kajiyama H. Recent Insights into Human Endometrial Peptidases in Blastocyst Implantation via Shedding of Microvesicles. Int J Mol Sci 2021; 22:13479. [PMID: 34948276 PMCID: PMC8708926 DOI: 10.3390/ijms222413479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/02/2021] [Accepted: 12/14/2021] [Indexed: 12/13/2022] Open
Abstract
Blastocyst implantation involves multiple interactions with numerous molecules expressed in endometrial epithelial cells (EECs) during the implantation window; however, there is limited information regarding the molecular mechanism underlying the crosstalk. In blastocysts, fibronectin plays a major role in the adhesion of various types of cells by binding to extracellular matrix proteins via the Arg-Gly-Asp (RGD) motif. In EECs, RGD-recognizing integrins are important bridging receptors for fibronectin, whereas the non-RGD binding of fibronectin includes interactions with dipeptidyl peptidase IV (DPPIV)/cluster of differentiation (CD) 26. Fibronectin may also bind to aminopeptidase N (APN)/CD13, and in the endometrium, these peptidases are present in plasma membranes and lysosomal membranes. Blastocyst implantation is accompanied by lysosome exocytosis, which transports various peptidases and nutrients into the endometrial cavity to facilitate blastocyst implantation. Both DPPIV and APN are released into the uterine cavity via shedding of microvesicles (MVs) from EECs. Recently, extracellular vesicles derived from endometrial cells have been proposed to act on trophectoderm cells to promote implantation. MVs are also secreted from embryonal stem cells and may play an active role in implantation. Thus, crosstalk between the blastocyst and endometrium via extracellular vesicles is a new insight into the fundamental molecular basis of blastocyst implantation.
Collapse
Affiliation(s)
- Masato Yoshihara
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan; (H.M.); (H.F.); (S.O.); (H.K.)
| | - Shigehiko Mizutani
- Daiyabilding Lady’s Clinic, 1-1-17 Meieki, Nishi-ku, Nagoya 451-0045, Japan;
| | - Yukio Kato
- Department of Molecular Pharmacotherapeutics, Faculty of Pharmacy, Kanazawa University, Kanazawa 920-1192, Japan;
| | - Kunio Matsumoto
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kanazawa 920-1192, Japan;
| | - Eita Mizutani
- Daiyabilding Lady’s Clinic, 1-1-17 Meieki, Nishi-ku, Nagoya 451-0045, Japan;
| | - Hidesuke Mizutani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan; (H.M.); (H.F.); (S.O.); (H.K.)
| | - Hiroki Fujimoto
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan; (H.M.); (H.F.); (S.O.); (H.K.)
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Satoko Osuka
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan; (H.M.); (H.F.); (S.O.); (H.K.)
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan; (H.M.); (H.F.); (S.O.); (H.K.)
| |
Collapse
|
34
|
Costa-Verdera H, Collaud F, Riling CR, Sellier P, Nordin JML, Preston GM, Cagin U, Fabregue J, Barral S, Moya-Nilges M, Krijnse-Locker J, van Wittenberghe L, Daniele N, Gjata B, Cosette J, Abad C, Simon-Sola M, Charles S, Li M, Crosariol M, Antrilli T, Quinn WJ, Gross DA, Boyer O, Anguela XM, Armour SM, Colella P, Ronzitti G, Mingozzi F. Hepatic expression of GAA results in enhanced enzyme bioavailability in mice and non-human primates. Nat Commun 2021; 12:6393. [PMID: 34737297 PMCID: PMC8568898 DOI: 10.1038/s41467-021-26744-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 10/05/2021] [Indexed: 12/22/2022] Open
Abstract
Pompe disease (PD) is a severe neuromuscular disorder caused by deficiency of the lysosomal enzyme acid alpha-glucosidase (GAA). PD is currently treated with enzyme replacement therapy (ERT) with intravenous infusions of recombinant human GAA (rhGAA). Although the introduction of ERT represents a breakthrough in the management of PD, the approach suffers from several shortcomings. Here, we developed a mouse model of PD to compare the efficacy of hepatic gene transfer with adeno-associated virus (AAV) vectors expressing secretable GAA with long-term ERT. Liver expression of GAA results in enhanced pharmacokinetics and uptake of the enzyme in peripheral tissues compared to ERT. Combination of gene transfer with pharmacological chaperones boosts GAA bioavailability, resulting in improved rescue of the PD phenotype. Scale-up of hepatic gene transfer to non-human primates also successfully results in enzyme secretion in blood and uptake in key target tissues, supporting the ongoing clinical translation of the approach.
Collapse
Affiliation(s)
- Helena Costa-Verdera
- Genethon, 91000, Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Integrare research Unit UMR_S951, 91000, Evry, France.,Sorbonne University Paris and INSERM U974, 75013, Paris, France
| | - Fanny Collaud
- Genethon, 91000, Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Integrare research Unit UMR_S951, 91000, Evry, France
| | | | - Pauline Sellier
- Genethon, 91000, Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Integrare research Unit UMR_S951, 91000, Evry, France
| | | | | | - Umut Cagin
- Genethon, 91000, Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Integrare research Unit UMR_S951, 91000, Evry, France
| | - Julien Fabregue
- Genethon, 91000, Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Integrare research Unit UMR_S951, 91000, Evry, France
| | - Simon Barral
- Genethon, 91000, Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Integrare research Unit UMR_S951, 91000, Evry, France
| | | | | | | | | | | | | | - Catalina Abad
- Université de Rouen Normandie-IRIB, 76183, Rouen, France
| | - Marcelo Simon-Sola
- Genethon, 91000, Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Integrare research Unit UMR_S951, 91000, Evry, France
| | - Severine Charles
- Genethon, 91000, Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Integrare research Unit UMR_S951, 91000, Evry, France
| | - Mathew Li
- Spark Therapeutics, Philadelphia, PA, 19104, USA
| | | | - Tom Antrilli
- Spark Therapeutics, Philadelphia, PA, 19104, USA
| | | | - David A Gross
- Genethon, 91000, Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Integrare research Unit UMR_S951, 91000, Evry, France
| | - Olivier Boyer
- Université de Rouen Normandie-IRIB, 76183, Rouen, France
| | | | | | - Pasqualina Colella
- Genethon, 91000, Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Integrare research Unit UMR_S951, 91000, Evry, France
| | - Giuseppe Ronzitti
- Genethon, 91000, Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Integrare research Unit UMR_S951, 91000, Evry, France
| | - Federico Mingozzi
- Genethon, 91000, Evry, France. .,Université Paris-Saclay, Univ Evry, Inserm, Integrare research Unit UMR_S951, 91000, Evry, France. .,Sorbonne University Paris and INSERM U974, 75013, Paris, France. .,Spark Therapeutics, Philadelphia, PA, 19104, USA.
| |
Collapse
|
35
|
Loss of Christianson Syndrome Na +/H + Exchanger 6 (NHE6) Causes Abnormal Endosome Maturation and Trafficking Underlying Lysosome Dysfunction in Neurons. J Neurosci 2021; 41:9235-9256. [PMID: 34526390 PMCID: PMC8570832 DOI: 10.1523/jneurosci.1244-20.2021] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/05/2021] [Accepted: 08/07/2021] [Indexed: 01/08/2023] Open
Abstract
Loss-of-function mutations in endosomal Na+/H+ exchanger 6 (NHE6) cause the X-linked neurologic disorder Christianson syndrome. Patients exhibit symptoms associated with both neurodevelopmental and neurodegenerative abnormalities. While loss of NHE6 has been shown to overacidify the endosome lumen, and is associated with endolysosome neuropathology, NHE6-mediated mechanisms in endosome trafficking and lysosome function have been understudied. Here, we show that NHE6-null mouse neurons demonstrate worsening lysosome function with time in culture, likely as a result of defective endosome trafficking. NHE6-null neurons exhibit overall reduced lysosomal proteolysis despite overacidification of the endosome and lysosome lumen. Akin to Nhx1 mutants in Saccharomyces cerevisiae, we observe decreased endosome-lysosome fusion in NHE6-null neurons. Also, we find premature activation of pH-dependent cathepsin D (CatD) in endosomes. While active CatD is increased in endosomes, CatD activation and CatD protein levels are reduced in the lysosome. Protein levels of another mannose 6-phosphate receptor (M6PR)-dependent enzyme, β-N-acetylglucosaminidase, were also decreased in lysosomes of NHE6-null neurons. M6PRs accumulate in late endosomes, suggesting defective M6PR recycling and retromer function in NHE6-null neurons. Finally, coincident with decreased endosome-lysosome fusion, using total internal reflection fluorescence, we also find a prominent increase in fusion between endosomal multivesicular bodies and the plasma membrane, indicating enhanced exosome secretion from NHE6-null neurons. In summary, in addition to overacidification of endosomes and lysosomes, loss of NHE6 leads to defects in endosome maturation and trafficking, including enhanced exosome release, contributing to lysosome deficiency and potentially leading to neurodegenerative disease. SIGNIFICANCE STATEMENT Loss-of-function mutations in the endosomal Na+/H+ exchanger 6 (NHE6) cause Christianson syndrome, an X-linked neurologic disorder. Loss of NHE6 has been shown to overacidify endosomes; however, endosome trafficking mechanisms have been understudied, and the mechanisms leading to neurodegeneration are largely unknown. In NHE6-null mouse neurons in vitro, we find worsening lysosome function with days in culture. Notably, pH-dependent lysosome enzymes, such as cathepsin D, have reduced activity in lysosomes yet increased, precocious activity in endosomes in NHE6-null neurons. Further, endosomes show reduced fusion to lysosomes, and increased fusion to the plasma membrane with increased exosome release. This study identifies new mechanisms involving defective endosome maturation and trafficking that impair lysosome function in Christianson syndrome, likely contributing to neurodegeneration.
Collapse
|
36
|
Johnson JL, Ramadass M, Rahman F, Meneses-Salas E, Zgajnar NR, Carvalho Gontijo R, Zhang J, Kiosses WB, Zhu YP, Hedrick CC, Perego M, Gunton JE, Pestonjamasp K, Napolitano G, Catz SD. The atypical small GTPase GEM/Kir is a negative regulator of the NADPH oxidase and NETs production through macroautophagy. J Leukoc Biol 2021; 110:629-649. [PMID: 34085299 DOI: 10.1002/jlb.2hi0421-123r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Despite the important function of neutrophils in the eradication of infections and induction of inflammation, the molecular mechanisms regulating the activation and termination of the neutrophil immune response is not well understood. Here, the function of the small GTPase from the RGK family, Gem, is characterized as a negative regulator of the NADPH oxidase through autophagy regulation. Gem knockout (Gem KO) neutrophils show increased NADPH oxidase activation and increased production of extracellular and intracellular reactive oxygen species (ROS). Enhanced ROS production in Gem KO neutrophils was associated with increased NADPH oxidase complex-assembly as determined by quantitative super-resolution microscopy, but normal exocytosis of gelatinase and azurophilic granules. Gem-deficiency was associated with increased basal autophagosomes and autolysosome numbers but decreased autophagic flux under phorbol ester-induced conditions. Neutrophil stimulation triggered the localization of the NADPH oxidase subunits p22phox and p47phox at LC3-positive structures suggesting that the assembled NADPH oxidase complex is recruited to autophagosomes, which was significantly increased in Gem KO neutrophils. Prevention of new autophagosome formation by treatment with SAR405 increased ROS production while induction of autophagy by Torin-1 decreased ROS production in Gem KO neutrophils, and also in wild-type neutrophils, suggesting that macroautophagy contributes to the termination of NADPH oxidase activity. Autophagy inhibition decreased NETs formation independently of enhanced ROS production. NETs production, which was significantly increased in Gem-deficient neutrophils, was decreased by inhibition of both autophagy and calmodulin, a known GEM interactor. Intracellular ROS production was increased in Gem KO neutrophils challenged with live Gram-negative bacteria Pseudomonas aeruginosa or Salmonella Typhimurium, but phagocytosis was not affected in Gem-deficient cells. In vivo analysis in a model of Salmonella Typhimurium infection indicates that Gem-deficiency provides a genetic advantage manifested as a moderate increased in survival to infections. Altogether, the data suggest that Gem-deficiency leads to the enhancement of the neutrophil innate immune response by increasing NADPH oxidase assembly and NETs production and that macroautophagy differentially regulates ROS and NETs in neutrophils.
Collapse
Affiliation(s)
- Jennifer L Johnson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Mahalakshmi Ramadass
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Farhana Rahman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Elsa Meneses-Salas
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Nadia R Zgajnar
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | | | - Jinzhong Zhang
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - William B Kiosses
- Center for Diabetes, Obesity, and Endocrinology (CDOE), The Westmead Institute for Medical Research (WIMR), The University of Sydney, Sydney, NSW, Australia
| | - Yanfang Peipei Zhu
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Catherine C Hedrick
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Marta Perego
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Jenny E Gunton
- Center for Diabetes, Obesity, and Endocrinology (CDOE), The Westmead Institute for Medical Research (WIMR), The University of Sydney, Sydney, NSW, Australia
| | - Kersi Pestonjamasp
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | | | - Sergio D Catz
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
37
|
Zhu F, Miao Y, Cheng M, Ye X, Chen A, Zheng G, Tian X. The CACNA1A Mutant Disrupts Lysosome Calcium Homeostasis in Cerebellar Neurons and the Resulting Endo-Lysosomal Fusion Defect Can be Improved by Calcium Modulation. Neurochem Res 2021; 47:249-263. [PMID: 34476720 DOI: 10.1007/s11064-021-03438-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 11/24/2022]
Abstract
Mutations in P/Q type voltage gated calcium channel (VGCC) lead severe human neurological diseases such as episodic ataxia 2, familial hemiplegic migraine 1, absence epilepsy, progressive ataxia and spinocerebellar ataxia 6. The pathogenesis of these diseases remains unclear. Mice with spontaneous mutation in the Cacna1a gene encoding the pore-forming subunit of P/Q type VGCC also exhibit ataxia, epilepsy and neurodegeneration. Based on the previous work showing that the P/Q type VGCC in neurons regulates lysosomal fusion through its calcium channel activity on lysosomes, we utilized CACNA1A mutant mice to further investigate the mechanism by which P/Q-type VGCCs regulate lysosomal function and neuronal homeostasis. We found CACNA1A mutant neurons have reduced lysosomal calcium storage without changing the resting calcium concentration in cytoplasm and the acidification of lysosomes. Immunohistochemistry and transmission electron microscopy reveal axonal degeneration due to lysosome dysfunction in the CACNA1A mutant cerebella. The calcium modulating drug thapsigargin, by depleting the ER calcium store, which locally increases the calcium concentration can alleviate the defective lysosomal fusion in mutant neurons. We propose a model that in cerebellar neurons, P/Q-type VGCC maintains the integrity of the nervous system by regulating lysosomal calcium homeostasis to affect lysosomal fusion, which in turn regulates multiple important cellular processes such as autophagy and endocytosis. This study helps us to better understand the pathogenesis of P/Q-type VGCC related neurodegenerative diseases and provides a feasible direction for future pharmacological treatment.
Collapse
Affiliation(s)
- Feng Zhu
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, 310013, China.,Department of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Yunping Miao
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, 310013, China.,Department of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Min Cheng
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, 310013, China.,Department of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Xiaodi Ye
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, 310013, China.,Department of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Aiying Chen
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, 310013, China.,Department of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Gaoli Zheng
- National Zhejiang Center for Safety Evaluation of New Drugs, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, 310013, China
| | - Xuejun Tian
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Institute of Materia Medica, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, 310013, China. .,Department of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China. .,Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
38
|
Escrevente C, Bento-Lopes L, Ramalho JS, Barral DC. Rab11 is required for lysosome exocytosis through the interaction with Rab3a, Sec15 and GRAB. J Cell Sci 2021; 134:jcs246694. [PMID: 34100549 PMCID: PMC8214760 DOI: 10.1242/jcs.246694] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 04/26/2021] [Indexed: 12/19/2022] Open
Abstract
Lysosomes are dynamic organelles, capable of undergoing exocytosis. This process is crucial for several cellular functions, namely plasma membrane repair. Nevertheless, the molecular machinery involved in this process is poorly understood. Here, we identify Rab11a and Rab11b as regulators of Ca2+-induced lysosome exocytosis. Interestingly, Rab11-positive vesicles transiently interact with lysosomes at the cell periphery, indicating that this interaction is required for the last steps of lysosome exocytosis. Additionally, we found that the silencing of the exocyst subunit Sec15, a Rab11 effector, impairs lysosome exocytosis, suggesting that Sec15 acts together with Rab11 in the regulation of lysosome exocytosis. Furthermore, we show that Rab11 binds the guanine nucleotide exchange factor for Rab3a (GRAB) as well as Rab3a, which we have previously described to be a regulator of the positioning and exocytosis of lysosomes. Thus, our study identifies new players required for lysosome exocytosis and suggest the existence of a Rab11-Rab3a cascade involved in this process.
Collapse
Affiliation(s)
| | | | | | - Duarte C. Barral
- iNOVA4Health, CEDOC, NOVA Medical School, NMS, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| |
Collapse
|
39
|
Ermini L, Farrell A, Alahari S, Ausman J, Park C, Sallais J, Melland-Smith M, Porter T, Edson M, Nevo O, Litvack M, Post M, Caniggia I. Ceramide-Induced Lysosomal Biogenesis and Exocytosis in Early-Onset Preeclampsia Promotes Exosomal Release of SMPD1 Causing Endothelial Dysfunction. Front Cell Dev Biol 2021; 9:652651. [PMID: 34017832 PMCID: PMC8130675 DOI: 10.3389/fcell.2021.652651] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/06/2021] [Indexed: 11/13/2022] Open
Abstract
Aberrant ceramide build-up in preeclampsia, a serious disorder of pregnancy, causes exuberant autophagy-mediated trophoblast cell death. The significance of ceramide accumulation for lysosomal biogenesis in preeclampsia is unknown. Here we report that lysosome formation is markedly increased in trophoblast cells of early-onset preeclamptic placentae, in particular in syncytiotrophoblasts. This is accompanied by augmented levels of transcription factor EB (TFEB). In vitro and in vivo experiments demonstrate that ceramide increases TFEB expression and nuclear translocation and induces lysosomal formation and exocytosis. Further, we show that TFEB directly regulates the expression of lysosomal sphingomyelin phosphodiesterase (L-SMPD1) that degrades sphingomyelin to ceramide. In early-onset preeclampsia, ceramide-induced lysosomal exocytosis carries L-SMPD1 to the apical membrane of the syncytial epithelium, resulting in ceramide accumulation in lipid rafts and release of active L-SMPD1 via ceramide-enriched exosomes into the maternal circulation. The SMPD1-containing exosomes promote endothelial activation and impair endothelial tubule formation in vitro. Both exosome-induced processes are attenuated by SMPD1 inhibitors. These findings suggest that ceramide-induced lysosomal biogenesis and exocytosis in preeclamptic placentae contributes to maternal endothelial dysfunction, characteristic of this pathology.
Collapse
Affiliation(s)
- Leonardo Ermini
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Abby Farrell
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Sruthi Alahari
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Jonathan Ausman
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Chanho Park
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Julien Sallais
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Megan Melland-Smith
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Tyler Porter
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Michael Edson
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Ori Nevo
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Michael Litvack
- Translational Medicine Program, Peter Gilgan Center, The Hospital for Sick Children, Toronto, ON, Canada
| | - Martin Post
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Translational Medicine Program, Peter Gilgan Center, The Hospital for Sick Children, Toronto, ON, Canada
| | - Isabella Caniggia
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
40
|
Plasma membrane integrity in health and disease: significance and therapeutic potential. Cell Discov 2021; 7:4. [PMID: 33462191 PMCID: PMC7813858 DOI: 10.1038/s41421-020-00233-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/23/2020] [Indexed: 12/13/2022] Open
Abstract
Maintenance of plasma membrane integrity is essential for normal cell viability and function. Thus, robust membrane repair mechanisms have evolved to counteract the eminent threat of a torn plasma membrane. Different repair mechanisms and the bio-physical parameters required for efficient repair are now emerging from different research groups. However, less is known about when these mechanisms come into play. This review focuses on the existence of membrane disruptions and repair mechanisms in both physiological and pathological conditions, and across multiple cell types, albeit to different degrees. Fundamentally, irrespective of the source of membrane disruption, aberrant calcium influx is the common stimulus that activates the membrane repair response. Inadequate repair responses can tip the balance between physiology and pathology, highlighting the significance of plasma membrane integrity. For example, an over-activated repair response can promote cancer invasion, while the inability to efficiently repair membrane can drive neurodegeneration and muscular dystrophies. The interdisciplinary view explored here emphasises the widespread potential of targeting plasma membrane repair mechanisms for therapeutic purposes.
Collapse
|
41
|
Tancini B, Buratta S, Delo F, Sagini K, Chiaradia E, Pellegrino RM, Emiliani C, Urbanelli L. Lysosomal Exocytosis: The Extracellular Role of an Intracellular Organelle. MEMBRANES 2020; 10:E406. [PMID: 33316913 PMCID: PMC7764620 DOI: 10.3390/membranes10120406] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/01/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022]
Abstract
Lysosomes are acidic cell compartments containing a large set of hydrolytic enzymes. These lysosomal hydrolases degrade proteins, lipids, polysaccharides, and nucleic acids into their constituents. Materials to be degraded can reach lysosomes either from inside the cell, by autophagy, or from outside the cell, by different forms of endocytosis. In addition to their degradative functions, lysosomes are also able to extracellularly release their contents by lysosomal exocytosis. These organelles move from the perinuclear region along microtubules towards the proximity of the plasma membrane, then the lysosomal and plasma membrane fuse together via a Ca2+-dependent process. The fusion of the lysosomal membrane with plasma membrane plays an important role in plasma membrane repair, while the secretion of lysosomal content is relevant for the remodelling of extracellular matrix and release of functional substrates. Lysosomal storage disorders (LSDs) and age-related neurodegenerative disorders, such as Parkinson's and Alzheimer's diseases, share as a pathological feature the accumulation of undigested material within organelles of the endolysosomal system. Recent studies suggest that lysosomal exocytosis stimulation may have beneficial effects on the accumulation of these unprocessed aggregates, leading to their extracellular elimination. However, many details of the molecular machinery required for lysosomal exocytosis are only beginning to be unravelled. Here, we are going to review the current literature on molecular mechanisms and biological functions underlying lysosomal exocytosis, to shed light on the potential of lysosomal exocytosis stimulation as a therapeutic approach.
Collapse
Affiliation(s)
- Brunella Tancini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (B.T.); (S.B.); (F.D.); (K.S.); (R.M.P.)
| | - Sandra Buratta
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (B.T.); (S.B.); (F.D.); (K.S.); (R.M.P.)
| | - Federica Delo
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (B.T.); (S.B.); (F.D.); (K.S.); (R.M.P.)
| | - Krizia Sagini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (B.T.); (S.B.); (F.D.); (K.S.); (R.M.P.)
| | - Elisabetta Chiaradia
- Department of Veterinary Medicine, University of Perugia, Via S. Costanzo 4, 06126 Perugia, Italy;
| | - Roberto Maria Pellegrino
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (B.T.); (S.B.); (F.D.); (K.S.); (R.M.P.)
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (B.T.); (S.B.); (F.D.); (K.S.); (R.M.P.)
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Lorena Urbanelli
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (B.T.); (S.B.); (F.D.); (K.S.); (R.M.P.)
| |
Collapse
|
42
|
Wu PH, Onodera Y, Giaccia AJ, Le QT, Shimizu S, Shirato H, Nam JM. Lysosomal trafficking mediated by Arl8b and BORC promotes invasion of cancer cells that survive radiation. Commun Biol 2020; 3:620. [PMID: 33110168 PMCID: PMC7591908 DOI: 10.1038/s42003-020-01339-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 09/02/2020] [Indexed: 12/18/2022] Open
Abstract
Enhanced invasiveness, a critical determinant of metastasis and poor prognosis, has been observed in cancer cells that survive cancer therapy, including radiotherapy. Here, we show that invasiveness in radiation-surviving cancer cells is associated with alterations in lysosomal exocytosis caused by the enhanced activation of Arl8b, a small GTPase that regulates lysosomal trafficking. The binding of Arl8b with its effector, SKIP, is increased after radiation through regulation of BORC-subunits. Knockdown of Arl8b or BORC-subunits decreases lysosomal exocytosis and the invasiveness of radiation-surviving cells. Notably, high expression of ARL8B and BORC-subunit genes is significantly correlated with poor prognosis in breast cancer patients. Sp1, an ATM-regulated transcription factor, is found to increase BORC-subunit genes expression after radiation. In vivo experiments show that ablation of Arl8b decreases IR-induced invasive tumor growth and distant metastasis. These findings suggest that BORC-Arl8b-mediated lysosomal trafficking is a target for improving radiotherapy by inhibiting invasive tumor growth and metastasis.
Collapse
Affiliation(s)
- Ping-Hsiu Wu
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, 060-8638, Sapporo, Hokkaido, Japan
| | - Yasuhito Onodera
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, 060-8638, Sapporo, Hokkaido, Japan.
- Department of Molecular Biology, Faculty of Medicine, Hokkaido University, 060-8638, Sapporo, Hokkaido, Japan.
| | - Amato J Giaccia
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Quynh-Thu Le
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Shinichi Shimizu
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, 060-8638, Sapporo, Hokkaido, Japan
- Department of Radiation Medical Science and Engineering, Faculty of Medicine, Hokkaido University, 060-8638, Sapporo, Hokkaido, Japan
| | - Hiroki Shirato
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, 060-8638, Sapporo, Hokkaido, Japan
| | - Jin-Min Nam
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, 060-8638, Sapporo, Hokkaido, Japan.
| |
Collapse
|
43
|
Two-pore and TRPML cation channels: Regulators of phagocytosis, autophagy and lysosomal exocytosis. Pharmacol Ther 2020; 220:107713. [PMID: 33141027 DOI: 10.1016/j.pharmthera.2020.107713] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023]
Abstract
The old Greek saying "Panta Rhei" ("everything flows") is true for all life and all living things in general. It also becomes nicely evident when looking closely into cells. There, material from the extracellular space is taken up by endocytic processes and transported to endosomes where it is sorted either for recycling or degradation. Cargo is also packaged for export through exocytosis involving the Golgi network, lysosomes and other organelles. Everything in this system is in constant motion and many proteins are necessary to coordinate transport along the different intracellular pathways to avoid chaos. Among these proteins are ion channels., in particular TRPML channels (mucolipins) and two-pore channels (TPCs) which reside on endosomal and lysosomal membranes to speed up movement between organelles, e.g. by regulating fusion and fission; they help readjust pH and osmolarity changes due to such processes, or they promote exocytosis of export material. Pathophysiologically, these channels are involved in neurodegenerative, metabolic, retinal and infectious diseases, cancer, pigmentation defects, and immune cell function, and thus have been proposed as novel pharmacological targets, e.g. for the treatment of lysosomal storage disorders, Duchenne muscular dystrophy, or different types of cancer. Here, we discuss the similarities but also differences of TPCs and TRPMLs in regulating phagocytosis, autophagy and lysosomal exocytosis, and we address the contradictions and open questions in the field relating to the roles TPCs and TRPMLs play in these different processes.
Collapse
|
44
|
Bowman SL, Bi-Karchin J, Le L, Marks MS. The road to lysosome-related organelles: Insights from Hermansky-Pudlak syndrome and other rare diseases. Traffic 2020; 20:404-435. [PMID: 30945407 DOI: 10.1111/tra.12646] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/02/2019] [Accepted: 04/02/2019] [Indexed: 12/11/2022]
Abstract
Lysosome-related organelles (LROs) comprise a diverse group of cell type-specific, membrane-bound subcellular organelles that derive at least in part from the endolysosomal system but that have unique contents, morphologies and functions to support specific physiological roles. They include: melanosomes that provide pigment to our eyes and skin; alpha and dense granules in platelets, and lytic granules in cytotoxic T cells and natural killer cells, which release effectors to regulate hemostasis and immunity; and distinct classes of lamellar bodies in lung epithelial cells and keratinocytes that support lung plasticity and skin lubrication. The formation, maturation and/or secretion of subsets of LROs are dysfunctional or entirely absent in a number of hereditary syndromic disorders, including in particular the Hermansky-Pudlak syndromes. This review provides a comprehensive overview of LROs in humans and model organisms and presents our current understanding of how the products of genes that are defective in heritable diseases impact their formation, motility and ultimate secretion.
Collapse
Affiliation(s)
- Shanna L Bowman
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jing Bi-Karchin
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Linh Le
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael S Marks
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
45
|
Cagin U, Puzzo F, Gomez MJ, Moya-Nilges M, Sellier P, Abad C, Van Wittenberghe L, Daniele N, Guerchet N, Gjata B, Collaud F, Charles S, Sola MS, Boyer O, Krijnse-Locker J, Ronzitti G, Colella P, Mingozzi F. Rescue of Advanced Pompe Disease in Mice with Hepatic Expression of Secretable Acid α-Glucosidase. Mol Ther 2020; 28:2056-2072. [PMID: 32526204 PMCID: PMC7474269 DOI: 10.1016/j.ymthe.2020.05.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/15/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022] Open
Abstract
Pompe disease is a neuromuscular disorder caused by disease-associated variants in the gene encoding for the lysosomal enzyme acid α-glucosidase (GAA), which converts lysosomal glycogen to glucose. We previously reported full rescue of Pompe disease in symptomatic 4-month-old Gaa knockout (Gaa−/−) mice by adeno-associated virus (AAV) vector-mediated liver gene transfer of an engineered secretable form of GAA (secGAA). Here, we showed that hepatic expression of secGAA rescues the phenotype of 4-month-old Gaa−/− mice at vector doses at which the native form of GAA has little to no therapeutic effect. Based on these results, we then treated severely affected 9-month-old Gaa−/− mice with an AAV vector expressing secGAA and followed the animals for 9 months thereafter. AAV-treated Gaa−/− mice showed complete reversal of the Pompe phenotype, with rescue of glycogen accumulation in most tissues, including the central nervous system, and normalization of muscle strength. Transcriptomic profiling of skeletal muscle showed rescue of most altered pathways, including those involved in mitochondrial defects, a finding supported by structural and biochemical analyses, which also showed restoration of lysosomal function. Together, these results provide insight into the reversibility of advanced Pompe disease in the Gaa−/− mouse model via liver gene transfer of secGAA.
Collapse
Affiliation(s)
- Umut Cagin
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Francesco Puzzo
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France; Sorbonne Université, Paris, France
| | - Manuel Jose Gomez
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain
| | | | - Pauline Sellier
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Catalina Abad
- Université de Rouen Normandie-IRIB, 76183 Rouen, France
| | | | - Nathalie Daniele
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Nicolas Guerchet
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Bernard Gjata
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Fanny Collaud
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Severine Charles
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Marcelo Simon Sola
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Olivier Boyer
- Université de Rouen Normandie-IRIB, 76183 Rouen, France
| | | | - Giuseppe Ronzitti
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Pasqualina Colella
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Federico Mingozzi
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France; Sorbonne Université, Paris, France; Spark Therapeutics, Philadelphia, PA 19103, USA.
| |
Collapse
|
46
|
Florentin A, Cobb DW, Kudyba HM, Muralidharan V. Directing traffic: Chaperone-mediated protein transport in malaria parasites. Cell Microbiol 2020; 22:e13215. [PMID: 32388921 PMCID: PMC7282954 DOI: 10.1111/cmi.13215] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/12/2020] [Accepted: 04/14/2020] [Indexed: 12/16/2022]
Abstract
The ability of eukaryotic parasites from the phylum Apicomplexa to cause devastating diseases is predicated upon their ability to maintain faithful and precise protein trafficking mechanisms. Their parasitic life cycle depends on the trafficking of effector proteins to the infected host cell, transport of proteins to several critical organelles required for survival, as well as transport of parasite and host proteins to the digestive organelles to generate the building blocks for parasite growth. Several recent studies have shed light on the molecular mechanisms parasites utilise to transform the infected host cells, transport proteins to essential metabolic organelles and for biogenesis of organelles required for continuation of their life cycle. Here, we review key pathways of protein transport originating and branching from the endoplasmic reticulum, focusing on the essential roles of chaperones in these processes. Further, we highlight key gaps in our knowledge that prevents us from building a holistic view of protein trafficking in these deadly human pathogens.
Collapse
Affiliation(s)
- Anat Florentin
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA.,Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| | - David W Cobb
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA.,Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| | - Heather M Kudyba
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA.,Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| | - Vasant Muralidharan
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA.,Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
47
|
Gomez-Auli A, Hillebrand LE, Christen D, Günther SC, Biniossek ML, Peters C, Schilling O, Reinheckel T. The secreted inhibitor of invasive cell growth CREG1 is negatively regulated by cathepsin proteases. Cell Mol Life Sci 2020; 78:733-755. [PMID: 32385587 PMCID: PMC7873128 DOI: 10.1007/s00018-020-03528-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 03/31/2020] [Accepted: 04/13/2020] [Indexed: 01/15/2023]
Abstract
Previous clinical and experimental evidence strongly supports a breast cancer-promoting function of the lysosomal protease cathepsin B. However, the cathepsin B-dependent molecular pathways are not completely understood. Here, we studied the cathepsin-mediated secretome changes in the context of the MMTV-PyMT breast cancer mouse model. Employing the cell-conditioned media from tumor-macrophage co-cultures, as well as tumor interstitial fluid obtained by a novel strategy from PyMT mice with differential cathepsin B expression, we identified an important proteolytic and lysosomal signature, highlighting the importance of this organelle and these enzymes in the tumor micro-environment. The Cellular Repressor of E1A Stimulated Genes 1 (CREG1), a secreted endolysosomal glycoprotein, displayed reduced abundance upon over-expression of cathepsin B as well as increased abundance upon cathepsin B deletion or inhibition. Moreover, it was cleaved by cathepsin B in vitro. CREG1 reportedly could act as tumor suppressor. We show that treatment of PyMT tumor cells with recombinant CREG1 reduced proliferation, migration, and invasion; whereas, the opposite was observed with reduced CREG1 expression. This was further validated in vivo by orthotopic transplantation. Our study highlights CREG1 as a key player in tumor–stroma interaction and suggests that cathepsin B sustains malignant cell behavior by reducing the levels of the growth suppressor CREG1 in the tumor microenvironment.
Collapse
Affiliation(s)
- Alejandro Gomez-Auli
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Larissa Elisabeth Hillebrand
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Daniel Christen
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Sira Carolin Günther
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Martin Lothar Biniossek
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Christoph Peters
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany.,German Cancer Research Center (DKFZ) Heidelberg, and German Cancer Consortium (DKTK), Partner Site Freiburg, 79104, Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany
| | - Oliver Schilling
- Institute of Surgical Pathology, University Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany.,German Cancer Research Center (DKFZ) Heidelberg, and German Cancer Consortium (DKTK), Partner Site Freiburg, 79104, Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany. .,German Cancer Research Center (DKFZ) Heidelberg, and German Cancer Consortium (DKTK), Partner Site Freiburg, 79104, Freiburg, Germany. .,BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany.
| |
Collapse
|
48
|
Lysosomal Exocytosis, Exosome Release and Secretory Autophagy: The Autophagic- and Endo-Lysosomal Systems Go Extracellular. Int J Mol Sci 2020; 21:ijms21072576. [PMID: 32276321 PMCID: PMC7178086 DOI: 10.3390/ijms21072576] [Citation(s) in RCA: 248] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/04/2020] [Accepted: 04/06/2020] [Indexed: 12/13/2022] Open
Abstract
Beyond the consolidated role in degrading and recycling cellular waste, the autophagic- and endo-lysosomal systems play a crucial role in extracellular release pathways. Lysosomal exocytosis is a process leading to the secretion of lysosomal content upon lysosome fusion with plasma membrane and is an important mechanism of cellular clearance, necessary to maintain cell fitness. Exosomes are a class of extracellular vesicles originating from the inward budding of the membrane of late endosomes, which may not fuse with lysosomes but be released extracellularly upon exocytosis. In addition to garbage disposal tools, they are now considered a cell-to-cell communication mechanism. Autophagy is a cellular process leading to sequestration of cytosolic cargoes for their degradation within lysosomes. However, the autophagic machinery is also involved in unconventional protein secretion and autophagy-dependent secretion, which are fundamental mechanisms for toxic protein disposal, immune signalling and pathogen surveillance. These cellular processes underline the crosstalk between the autophagic and the endosomal system and indicate an intersection between degradative and secretory functions. Further, they suggest that the molecular mechanisms underlying fusion, either with lysosomes or plasma membrane, are key determinants to maintain cell homeostasis upon stressing stimuli. When they fail, the accumulation of undigested substrates leads to pathological consequences, as indicated by the involvement of autophagic and lysosomal alteration in human diseases, namely lysosomal storage disorders, age-related neurodegenerative diseases and cancer. In this paper, we reviewed the current knowledge on the functional role of extracellular release pathways involving lysosomes and the autophagic- and endo-lysosomal systems, evaluating their implication in health and disease.
Collapse
|
49
|
Kudriaeva AA, Sokolov AV, Belogurov AAJ. Stochastics of Degradation: The Autophagic-Lysosomal System of the Cell. Acta Naturae 2020; 12:18-32. [PMID: 32477595 PMCID: PMC7245954 DOI: 10.32607/actanaturae.10936] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Autophagy is a conservative and evolutionarily ancient process that enables the
transfer of various cellular compounds, organelles, and potentially dangerous
cellular components to the lysosome for their degradation. This process is
crucial for the recycling of energy and substrates, which are required for
cellular biosynthesis. Autophagy not only plays a major role in the survival of
cells under stress conditions, but is also actively involved in maintaining
cellular homeostasis. It has multiple effects on the immune system and cellular
remodeling during organism development. The effectiveness of autophagy is
ensured by a controlled interaction between two organelles – the
autophagosome and the lysosome. Despite significant progress in the description
of the molecular mechanisms underlying autophagic-lysosomal system (ALS)
functioning, many fundamental questions remain. Namely, the specialized
functions of lysosomes and the role of ALS in the pathogenesis of human
diseases are still enigmatic. Understanding of the mechanisms that are
triggered at all stages of autophagic- lysosomal degradation, from the
initiation of autophagy to the terminal stage of substrate destruction in the
lysosome, may result in new approaches that could help better uderstand ALS
and, therefore, selectively control cellular proteostasis.
Collapse
Affiliation(s)
- A. A. Kudriaeva
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russia
| | - A. V. Sokolov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russia
| | - A. A. Jr. Belogurov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997 Russia
- Lomonosov Moscow State University, Moscow, 119991 Russia
| |
Collapse
|
50
|
Dai Y, Li K, Wu W, Wu K, Yi H, Li W, Xiao Y, Zhong Y, Cao Y, Tian L. Steroid hormone 20-hydroxyecdysone induces the transcription and complex assembly of V-ATPases to facilitate autophagy in Bombyx mori. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2020; 116:103255. [PMID: 31654713 DOI: 10.1016/j.ibmb.2019.103255] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 06/10/2023]
Abstract
Vacuolar-type H + -adenosine triphosphatases (V-ATPases) are indispensable for lysosome acidification and participate in autophagic processes. The steroid hormone 20-hydroxyecdysone (20E) predominantly induces autophagy and regulates insect larval molting and metamorphosis; however, the specific mechanism of lysosome acidification regulation by 20E remains unclear. Here, we showed that the developmental profiles of Bombyx V-ATPases were in accordance with autophagy occurrence and lysosome acidification in the fat body during larval-pupal metamorphosis. BmV-ATPase-A and BmV-ATPase-B were required for lysosome acidification and autophagic flux. Both 20E treatment and starvation were able to induce lysosome acidification. Furthermore, BmV-ATPase transcription was induced by 20E treatment and reduced by RNAi targeting the 20E receptor BmUsp. On the one hand, 20E upregulated the transcription of BmV-ATPases through inducing Bombyx transcription factor EB (TFEB) and its nuclear translocation; on the other hand, 20E inhibited mTOR signaling to induce the transcription and assembly of BmV-ATPase subunits. Overall, 20E induces lysosome acidification by upregulating the transcription and assembly of V-ATPase subunits via activating BmTFEB and cooperating with nutrient signaling. These findings improve our understanding of the regulatory mechanisms underlying lysosome acidification and autophagic flux in Bombyx mori.
Collapse
Affiliation(s)
- Yichen Dai
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding/Guangdong Provincial Sericulture and Mulberry Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Kang Li
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Wenmei Wu
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding/Guangdong Provincial Sericulture and Mulberry Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Kunzhong Wu
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding/Guangdong Provincial Sericulture and Mulberry Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Huiyu Yi
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding/Guangdong Provincial Sericulture and Mulberry Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Wangyu Li
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding/Guangdong Provincial Sericulture and Mulberry Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yang Xiao
- The Sericultural and Agri-Food Research Institute of the Guangdong Academy of Agricultural Sciences, Guangzhou, 510610, China
| | - Yangjin Zhong
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding/Guangdong Provincial Sericulture and Mulberry Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yang Cao
- Biological Science Research Center/Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing, 400716, China; Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding/Guangdong Provincial Sericulture and Mulberry Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Ling Tian
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding/Guangdong Provincial Sericulture and Mulberry Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, China.
| |
Collapse
|