51
|
Meneses-Salas E, García-Melero A, Blanco-Muñoz P, Jose J, Brenner MS, Lu A, Tebar F, Grewal T, Rentero C, Enrich C. Selective Degradation Permits a Feedback Loop Controlling Annexin A6 and Cholesterol Levels in Endolysosomes of NPC1 Mutant Cells. Cells 2020; 9:cells9051152. [PMID: 32392809 PMCID: PMC7291204 DOI: 10.3390/cells9051152] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/23/2020] [Accepted: 05/05/2020] [Indexed: 12/22/2022] Open
Abstract
We recently identified elevated annexin A6 (AnxA6) protein levels in Niemann–Pick-type C1 (NPC1) mutant cells. In these cells, AnxA6 depletion rescued the cholesterol accumulation associated with NPC1 deficiency. Here, we demonstrate that elevated AnxA6 protein levels in NPC1 mutants or upon pharmacological NPC1 inhibition, using U18666A, were not due to upregulated AnxA6 mRNA expression, but caused by defects in AnxA6 protein degradation. Two KFERQ-motifs are believed to target AnxA6 to lysosomes for chaperone-mediated autophagy (CMA), and we hypothesized that the cholesterol accumulation in endolysosomes (LE/Lys) triggered by the NPC1 inhibition could interfere with the CMA pathway. Therefore, AnxA6 protein amounts and cholesterol levels in the LE/Lys (LE-Chol) compartment were analyzed in NPC1 mutant cells ectopically expressing lysosome-associated membrane protein 2A (Lamp2A), which is well known to induce the CMA pathway. Strikingly, AnxA6 protein amounts were strongly decreased and coincided with significantly reduced LE-Chol levels in NPC1 mutant cells upon Lamp2A overexpression. Therefore, these findings suggest Lamp2A-mediated restoration of CMA in NPC1 mutant cells to lower LE-Chol levels with concomitant lysosomal AnxA6 degradation. Collectively, we propose CMA to permit a feedback loop between AnxA6 and cholesterol levels in LE/Lys, encompassing a novel mechanism for regulating cholesterol homeostasis in NPC1 disease.
Collapse
Affiliation(s)
- Elsa Meneses-Salas
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (E.M.-S.); (A.G.-M.); (P.B.-M.); (F.T.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036-Barcelona, Spain
| | - Ana García-Melero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (E.M.-S.); (A.G.-M.); (P.B.-M.); (F.T.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036-Barcelona, Spain
| | - Patricia Blanco-Muñoz
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (E.M.-S.); (A.G.-M.); (P.B.-M.); (F.T.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036-Barcelona, Spain
| | - Jaimy Jose
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney 2006, NSW, Australia; (J.J.); (M.-S.B.)
| | - Marie-Sophie Brenner
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney 2006, NSW, Australia; (J.J.); (M.-S.B.)
| | - Albert Lu
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Francesc Tebar
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (E.M.-S.); (A.G.-M.); (P.B.-M.); (F.T.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036-Barcelona, Spain
| | - Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney 2006, NSW, Australia; (J.J.); (M.-S.B.)
- Correspondence: (T.G.); (C.R.); (C.E.); Tel.: +34-934021908 (C.R.)
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (E.M.-S.); (A.G.-M.); (P.B.-M.); (F.T.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036-Barcelona, Spain
- Correspondence: (T.G.); (C.R.); (C.E.); Tel.: +34-934021908 (C.R.)
| | - Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (E.M.-S.); (A.G.-M.); (P.B.-M.); (F.T.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036-Barcelona, Spain
- Correspondence: (T.G.); (C.R.); (C.E.); Tel.: +34-934021908 (C.R.)
| |
Collapse
|
52
|
Ballout RA, Sviridov D, Bukrinsky MI, Remaley AT. The lysosome: A potential juncture between SARS-CoV-2 infectivity and Niemann-Pick disease type C, with therapeutic implications. FASEB J 2020; 34:7253-7264. [PMID: 32367579 PMCID: PMC7383733 DOI: 10.1096/fj.202000654r] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/21/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023]
Abstract
Drug repurposing is potentially the fastest available option in the race to identify safe and efficacious drugs that can be used to prevent and/or treat COVID‐19. By describing the life cycle of the newly emergent coronavirus, SARS‐CoV‐2, in light of emerging data on the therapeutic efficacy of various repurposed antimicrobials undergoing testing against the virus, we highlight in this review a possible mechanistic convergence between some of these tested compounds. Specifically, we propose that the lysosomotropic effects of hydroxychloroquine and several other drugs undergoing testing may be responsible for their demonstrated in vitro antiviral activities against COVID‐19. Moreover, we propose that Niemann‐Pick disease type C (NPC), a lysosomal storage disorder, may provide new insights into potential future therapeutic targets for SARS‐CoV‐2, by highlighting key established features of the disorder that together result in an “unfavorable” host cellular environment that may interfere with viral propagation. Our reasoning evolves from previous biochemical and cell biology findings related to NPC, coupled with the rapidly evolving data on COVID‐19. Our overall aim is to suggest that pharmacological interventions targeting lysosomal function in general, and those particularly capable of reversibly inducing transient NPC‐like cellular and biochemical phenotypes, constitute plausible mechanisms that could be used to therapeutically target COVID‐19.
Collapse
Affiliation(s)
- Rami A Ballout
- Lipoprotein Metabolism Section, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dmitri Sviridov
- Lipoproteins and Atherosclerosis Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Michael I Bukrinsky
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Alan T Remaley
- Lipoprotein Metabolism Section, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
53
|
Stamatakou E, Wróbel L, Hill SM, Puri C, Son SM, Fujimaki M, Zhu Y, Siddiqi F, Fernandez-Estevez M, Manni MM, Park SJ, Villeneuve J, Rubinsztein DC. Mendelian neurodegenerative disease genes involved in autophagy. Cell Discov 2020; 6:24. [PMID: 32377374 PMCID: PMC7198619 DOI: 10.1038/s41421-020-0158-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 03/11/2020] [Indexed: 12/13/2022] Open
Abstract
The lysosomal degradation pathway of macroautophagy (herein referred to as autophagy) plays a crucial role in cellular physiology by regulating the removal of unwanted cargoes such as protein aggregates and damaged organelles. Over the last five decades, significant progress has been made in understanding the molecular mechanisms that regulate autophagy and its roles in human physiology and diseases. These advances, together with discoveries in human genetics linking autophagy-related gene mutations to specific diseases, provide a better understanding of the mechanisms by which autophagy-dependent pathways can be potentially targeted for treating human diseases. Here, we review mutations that have been identified in genes involved in autophagy and their associations with neurodegenerative diseases.
Collapse
Affiliation(s)
- Eleanna Stamatakou
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, CB2 0XY UK
- UK Dementia Research Institute, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY UK
| | - Lidia Wróbel
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, CB2 0XY UK
- UK Dementia Research Institute, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY UK
| | - Sandra Malmgren Hill
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, CB2 0XY UK
- UK Dementia Research Institute, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY UK
| | - Claudia Puri
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, CB2 0XY UK
- UK Dementia Research Institute, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY UK
| | - Sung Min Son
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, CB2 0XY UK
- UK Dementia Research Institute, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY UK
| | - Motoki Fujimaki
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, CB2 0XY UK
- UK Dementia Research Institute, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY UK
| | - Ye Zhu
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, CB2 0XY UK
- UK Dementia Research Institute, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY UK
| | - Farah Siddiqi
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, CB2 0XY UK
- UK Dementia Research Institute, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY UK
| | - Marian Fernandez-Estevez
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, CB2 0XY UK
- UK Dementia Research Institute, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY UK
| | - Marco M. Manni
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, CB2 0XY UK
- UK Dementia Research Institute, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY UK
| | - So Jung Park
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, CB2 0XY UK
- UK Dementia Research Institute, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY UK
| | - Julien Villeneuve
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, CB2 0XY UK
- UK Dementia Research Institute, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY UK
| | - David Chaim Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, CB2 0XY UK
- UK Dementia Research Institute, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY UK
| |
Collapse
|
54
|
Pathogenesis of Mucopolysaccharidoses, an Update. Int J Mol Sci 2020; 21:ijms21072515. [PMID: 32260444 PMCID: PMC7178160 DOI: 10.3390/ijms21072515] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/29/2020] [Accepted: 04/02/2020] [Indexed: 01/08/2023] Open
Abstract
The recent advancements in the knowledge of lysosomal biology and function have translated into an improved understanding of the pathophysiology of mucopolysaccharidoses (MPSs). The concept that MPS manifestations are direct consequences of lysosomal engorgement with undegraded glycosaminoglycans (GAGs) has been challenged by new information on the multiple biological roles of GAGs and by a new vision of the lysosome as a signaling hub involved in many critical cellular functions. MPS pathophysiology is now seen as the result of a complex cascade of secondary events that lead to dysfunction of several cellular processes and pathways, such as abnormal composition of membranes and its impact on vesicle fusion and trafficking; secondary storage of substrates; impairment of autophagy; impaired mitochondrial function and oxidative stress; dysregulation of signaling pathways. The characterization of this cascade of secondary cellular events is critical to better understand the pathophysiology of MPS clinical manifestations. In addition, some of these pathways may represent novel therapeutic targets and allow for the development of new therapies for these disorders.
Collapse
|
55
|
Lakkaraju A, Umapathy A, Tan LX, Daniele L, Philp NJ, Boesze-Battaglia K, Williams DS. The cell biology of the retinal pigment epithelium. Prog Retin Eye Res 2020; 78:100846. [PMID: 32105772 PMCID: PMC8941496 DOI: 10.1016/j.preteyeres.2020.100846] [Citation(s) in RCA: 245] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/19/2020] [Accepted: 02/23/2020] [Indexed: 02/07/2023]
Abstract
The retinal pigment epithelium (RPE), a monolayer of post-mitotic polarized epithelial cells, strategically situated between the photoreceptors and the choroid, is the primary caretaker of photoreceptor health and function. Dysfunction of the RPE underlies many inherited and acquired diseases that cause permanent blindness. Decades of research have yielded valuable insight into the cell biology of the RPE. In recent years, new technologies such as live-cell imaging have resulted in major advancement in our understanding of areas such as the daily phagocytosis and clearance of photoreceptor outer segment tips, autophagy, endolysosome function, and the metabolic interplay between the RPE and photoreceptors. In this review, we aim to integrate these studies with an emphasis on appropriate models and techniques to investigate RPE cell biology and metabolism, and discuss how RPE cell biology informs our understanding of retinal disease.
Collapse
Affiliation(s)
- Aparna Lakkaraju
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Ankita Umapathy
- Department of Ophthalmology and Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Li Xuan Tan
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Lauren Daniele
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nancy J Philp
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kathleen Boesze-Battaglia
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David S Williams
- Department of Ophthalmology and Stein Eye Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
56
|
Suresh SN, Chakravorty A, Giridharan M, Garimella L, Manjithaya R. Pharmacological Tools to Modulate Autophagy in Neurodegenerative Diseases. J Mol Biol 2020; 432:2822-2842. [PMID: 32105729 DOI: 10.1016/j.jmb.2020.02.023] [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: 11/05/2019] [Revised: 02/03/2020] [Accepted: 02/12/2020] [Indexed: 12/11/2022]
Abstract
Considerable evidences suggest a link between autophagy dysfunction, protein aggregation, and neurodegenerative diseases. Given that autophagy is a conserved intracellular housekeeping process, modulation of autophagy flux in various model organisms have highlighted its importance for maintaining proteostasis. In postmitotic cells such as neurons, compromised autophagy is sufficient to cause accumulation of ubiquitinated aggregates, neuronal dysfunction, degeneration, and loss of motor coordination-all hallmarks of neurodegenerative diseases. Reciprocally, enhanced autophagy flux augments cellular and organismal health, in addition to extending life span. These genetic studies not-withstanding a plethora of small molecule modulators of autophagy flux have been reported that alleviate disease symptoms in models of neurodegenerative diseases. This review summarizes the potential of such molecules to be, perhaps, one of the first autophagy drugs for treating these currently incurable diseases.
Collapse
Affiliation(s)
- S N Suresh
- Centre for Brain Research, Indian Institute of Science, Bangalore, 560012, Karnataka, India
| | - Anushka Chakravorty
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, 560064, Karnataka, India
| | - Mridhula Giridharan
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, 560064, Karnataka, India
| | - Lakshmi Garimella
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, 560064, Karnataka, India
| | - Ravi Manjithaya
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, 560064, Karnataka, India; Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bangalore, 560064, Karnataka, India.
| |
Collapse
|
57
|
Sawamoto K, Álvarez González JV, Piechnik M, Otero FJ, Couce ML, Suzuki Y, Tomatsu S. Mucopolysaccharidosis IVA: Diagnosis, Treatment, and Management. Int J Mol Sci 2020; 21:E1517. [PMID: 32102177 PMCID: PMC7073202 DOI: 10.3390/ijms21041517] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/15/2020] [Accepted: 02/19/2020] [Indexed: 12/16/2022] Open
Abstract
Mucopolysaccharidosis type IVA (MPS IVA, or Morquio syndrome type A) is an inherited metabolic lysosomal disease caused by the deficiency of the N-acetylglucosamine-6-sulfate sulfatase enzyme. The deficiency of this enzyme accumulates the specific glycosaminoglycans (GAG), keratan sulfate, and chondroitin-6-sulfate mainly in bone, cartilage, and its extracellular matrix. GAG accumulation in these lesions leads to unique skeletal dysplasia in MPS IVA patients. Clinical, radiographic, and biochemical tests are needed to complete the diagnosis of MPS IVA since some clinical characteristics in MPS IVA are overlapped with other disorders. Early and accurate diagnosis is vital to optimizing patient management, which provides a better quality of life and prolonged life-time in MPS IVA patients. Currently, enzyme replacement therapy (ERT) and hematopoietic stem cell transplantation (HSCT) are available for patients with MPS IVA. However, ERT and HSCT do not have enough impact on bone and cartilage lesions in patients with MPS IVA. Penetrating the deficient enzyme into an avascular lesion remains an unmet challenge, and several innovative therapies are under development in a preclinical study. In this review article, we comprehensively describe the current diagnosis, treatment, and management for MPS IVA. We also illustrate developing future therapies focused on the improvement of skeletal dysplasia in MPS IVA.
Collapse
Affiliation(s)
- Kazuki Sawamoto
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; (K.S.); (J.V.Á.G.); (M.P.)
| | | | - Matthew Piechnik
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; (K.S.); (J.V.Á.G.); (M.P.)
- University of Delaware, Newark, DE 19716, USA
| | - Francisco J. Otero
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, 15872 Santiago de Compostela, Spain;
| | - Maria L. Couce
- Department of Forensic Sciences, Pathology, Gynecology and Obstetrics and Pediatrics Neonatology Service, Metabolic Unit, IDIS, CIBERER, MetabERN, University Clinic Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain;
| | - Yasuyuki Suzuki
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu 501-1193, Japan;
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; (K.S.); (J.V.Á.G.); (M.P.)
- University of Delaware, Newark, DE 19716, USA
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu 501-1193, Japan;
- Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
58
|
Rontogianni S, Iskit S, van Doorn S, Peeper DS, Altelaar M. Combined EGFR and ROCK Inhibition in Triple-negative Breast Cancer Leads to Cell Death Via Impaired Autophagic Flux. Mol Cell Proteomics 2020; 19:261-277. [PMID: 31772060 PMCID: PMC7000121 DOI: 10.1074/mcp.ra119.001800] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with very limited therapeutic options. We have recently shown that the combined inhibition of EGFR and ROCK in TNBC cells results in cell death, however, the underlying mechanisms remain unclear. To investigate this, here we applied a mass spectrometry-based proteomic approach to identify proteins altered on single and combination treatments. Our proteomic data revealed autophagy as the major molecular mechanism implicated in the cells' response to combinatorial treatment. We here show that EGFR inhibition by gefitinib treatment alone induces autophagy, a cellular recycling process that acts as a cytoprotective response for TNBC cells. However, combined inhibition of EGFR and ROCK leads to autophagy blockade and accumulation of autophagic vacuoles. Our data show impaired autophagosome clearance as a likely cause of antitumor activity. We propose that the inhibition of the autophagic flux on combinatorial treatment is attributed to the major cytoskeletal changes induced on ROCK inhibition, given the essential role the cytoskeleton plays throughout the various steps of the autophagy process.
Collapse
Affiliation(s)
- Stamatia Rontogianni
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands; Netherlands Proteomics Center, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Sedef Iskit
- Division of Molecular Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Sander van Doorn
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands; Netherlands Proteomics Center, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Daniel S Peeper
- Division of Molecular Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands; Netherlands Proteomics Center, Padualaan 8, 3584 CH Utrecht, The Netherlands; Mass Spectrometry and Proteomics Facility, The Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands.
| |
Collapse
|
59
|
Ren H, Wang G. Autophagy and Lysosome Storage Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1207:87-102. [DOI: 10.1007/978-981-15-4272-5_5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
60
|
Anderson A, Campo A, Fulton E, Corwin A, Jerome WG, O'Connor MS. 7-Ketocholesterol in disease and aging. Redox Biol 2020; 29:101380. [PMID: 31926618 PMCID: PMC6926354 DOI: 10.1016/j.redox.2019.101380] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/06/2019] [Accepted: 11/10/2019] [Indexed: 02/08/2023] Open
Abstract
7-Ketocholesterol (7KC) is a toxic oxysterol that is associated with many diseases and disabilities of aging, as well as several orphan diseases. 7KC is the most common product of a reaction between cholesterol and oxygen radicals and is the most concentrated oxysterol found in the blood and arterial plaques of coronary artery disease patients as well as various other disease tissues and cell types. Unlike cholesterol, 7KC consistently shows cytotoxicity to cells and its physiological function in humans or other complex organisms is unknown. Oxysterols, particularly 7KC, have also been shown to diffuse through membranes where they affect receptor and enzymatic function. Here, we will explore the known and proposed mechanisms of pathologies that are associated with 7KC, as well speculate about the future of 7KC as a diagnostic and therapeutic target in medicine.
Collapse
|
61
|
de Araujo MEG, Liebscher G, Hess MW, Huber LA. Lysosomal size matters. Traffic 2019; 21:60-75. [PMID: 31808235 PMCID: PMC6972631 DOI: 10.1111/tra.12714] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 12/25/2022]
Abstract
Lysosomes are key cellular catabolic centers that also perform fundamental metabolic, signaling and quality control functions. Lysosomes are not static and they respond dynamically to intra‐ and extracellular stimuli triggering changes in organelle numbers, size and position. Such physical changes have a strong impact on lysosomal activity ultimately influencing cellular homeostasis. In this review, we summarize the current knowledge on lysosomal size regulation, on its physiological role(s) and association to several disease conditions.
Collapse
Affiliation(s)
- Mariana E G de Araujo
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Gudrun Liebscher
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael W Hess
- Institute of Histology and Embryology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas A Huber
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria.,Austrian Drug Screening Institute, ADSI, Innsbruck, Austria
| |
Collapse
|
62
|
Lysosomes as dynamic regulators of cell and organismal homeostasis. Nat Rev Mol Cell Biol 2019; 21:101-118. [DOI: 10.1038/s41580-019-0185-4] [Citation(s) in RCA: 408] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2019] [Indexed: 12/11/2022]
|
63
|
Wheeler S, Sillence DJ. Niemann-Pick type C disease: cellular pathology and pharmacotherapy. J Neurochem 2019; 153:674-692. [PMID: 31608980 DOI: 10.1111/jnc.14895] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/10/2019] [Accepted: 09/15/2019] [Indexed: 12/22/2022]
Abstract
Niemann-Pick type C disease (NPCD) was first described in 1914 and affects approximately 1 in 150 000 live births. It is characterized clinically by diverse symptoms affecting liver, spleen, motor control, and brain; premature death invariably results. Its molecular origins were traced, as late as 1997, to a protein of late endosomes and lysosomes which was named NPC1. Mutation or absence of this protein leads to accumulation of cholesterol in these organelles. In this review, we focus on the intracellular events that drive the pathology of this disease. We first introduce endocytosis, a much-studied area of dysfunction in NPCD cells, and survey the various ways in which this process malfunctions. We briefly consider autophagy before attempting to map the more complex pathways by which lysosomal cholesterol storage leads to protein misregulation, mitochondrial dysfunction, and cell death. We then briefly introduce the metabolic pathways of sphingolipids (as these emerge as key species for treatment) and critically examine the various treatment approaches that have been attempted to date.
Collapse
Affiliation(s)
- Simon Wheeler
- School of Pharmacy, De Montfort University, The Gateway, Leicester, UK
| | - Dan J Sillence
- School of Pharmacy, De Montfort University, The Gateway, Leicester, UK
| |
Collapse
|
64
|
ER-lysosome contacts enable cholesterol sensing by mTORC1 and drive aberrant growth signalling in Niemann-Pick type C. Nat Cell Biol 2019; 21:1206-1218. [PMID: 31548609 PMCID: PMC6936960 DOI: 10.1038/s41556-019-0391-5] [Citation(s) in RCA: 219] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 08/15/2019] [Indexed: 12/24/2022]
Abstract
Cholesterol activates the master growth regulator, mTORC1 kinase, by promoting its recruitment to the surface of lysosomes via the Rag guanosine triphosphatases (GTPases). The mechanisms that regulate lysosomal cholesterol content to enable mTORC1 signaling are unknown. We show that Oxysterol Binding Protein (OSBP) and its anchors at the endoplasmic reticulum (ER), VAPA/B, deliver cholesterol across ER-lysosome contacts to activate mTORC1. In cells lacking OSBP, but not other VAP-interacting cholesterol carriers, mTORC1 recruitment by the Rag GTPases is inhibited due to impaired cholesterol transport to lysosomes. Conversely, OSBP-mediated cholesterol trafficking drives constitutive mTORC1 activation in a disease model caused by loss of the lysosomal cholesterol transporter, Niemann-Pick C1 (NPC1). Chemical and genetic inactivation of OSBP suppresses aberrant mTORC1 signaling and restores autophagic function in cellular models of NPC. Thus, ER-lysosome contacts are signaling hubs that enable cholesterol sensing by mTORC1, and targeting their sterol-transfer activity could be beneficial in NPC.
Collapse
|
65
|
Álvarez JV, Bravo SB, García-Vence M, De Castro MJ, Luzardo A, Colón C, Tomatsu S, Otero-Espinar FJ, Couce ML. Proteomic Analysis in Morquio A Cells Treated with Immobilized Enzymatic Replacement Therapy on Nanostructured Lipid Systems. Int J Mol Sci 2019; 20:ijms20184610. [PMID: 31540344 PMCID: PMC6769449 DOI: 10.3390/ijms20184610] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/07/2019] [Accepted: 09/13/2019] [Indexed: 12/14/2022] Open
Abstract
Morquio A syndrome, or mucopolysaccharidosis type IVA (MPS IVA), is a lysosomal storage disease due to mutations in the N-acetylgalactosamine-6-sulfatase (GALNS) gene. Systemic skeletal dysplasia and the related clinical features of MPS IVA are due to disruption of cartilage and its extracellular matrix, leading to an imbalance of growth. Enzyme replacement therapy (ERT) with recombinant human GALNS, alpha elosulfase, provides a systemic treatment. However, this therapy has a limited impact on skeletal dysplasia because the infused enzyme cannot penetrate cartilage and bone. Therefore, an alternative therapeutic approach to reach the cartilage is an unmet challenge. We have developed a new drug delivery system based on a nanostructure lipid carrier with the capacity to immobilize enzymes used for ERT and to target the lysosomes. This study aimed to assess the effect of the encapsulated enzyme in this new delivery system, using in vitro proteomic technology. We found a greater internalization of the enzyme carried by nanoparticles inside the cells and an improvement of cellular protein routes previously impaired by the disease, compared with conventional ERT. This is the first qualitative and quantitative proteomic assay that demonstrates the advantages of a new delivery system to improve the MPS IVA ERT.
Collapse
Affiliation(s)
- J Víctor Álvarez
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Campus Vida, University of Santiago de Compostela, 15872 Santiago de Compostela, Spain.
- Department of Forensic Sciences, Pathology, Gynecology and Obstetrics, Pediatrics, Neonatology Service, Department of Paediatrics, Hospital Clínico Universitario de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), CIBERER, MetabERN, 15706 Santiago de Compostela, Spain.
- Skeletal Dysplasia Lab Nemours Biomedical Research Nemours/Alfred I. duPont Hospital for Children, 1600 Rockland Road, Wilmington, DE 19803, USA.
| | - Susana B Bravo
- Proteomic Platform, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, 15706 Santiago de Compostea, Spain.
| | - María García-Vence
- Proteomic Platform, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, 15706 Santiago de Compostea, Spain.
| | - María J De Castro
- Department of Forensic Sciences, Pathology, Gynecology and Obstetrics, Pediatrics, Neonatology Service, Department of Paediatrics, Hospital Clínico Universitario de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), CIBERER, MetabERN, 15706 Santiago de Compostela, Spain.
| | - Asteria Luzardo
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Sciences, Campus de Lugo, University of Santiago de Compostela, 27002 Lugo, Spain.
- Paraquasil Platform, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, 15706 Santiago de Compostela, Spain.
| | - Cristóbal Colón
- Department of Forensic Sciences, Pathology, Gynecology and Obstetrics, Pediatrics, Neonatology Service, Department of Paediatrics, Hospital Clínico Universitario de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), CIBERER, MetabERN, 15706 Santiago de Compostela, Spain.
| | - Shunji Tomatsu
- Skeletal Dysplasia Lab Nemours Biomedical Research Nemours/Alfred I. duPont Hospital for Children, 1600 Rockland Road, Wilmington, DE 19803, USA.
| | - Francisco J Otero-Espinar
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Campus Vida, University of Santiago de Compostela, 15872 Santiago de Compostela, Spain.
- Paraquasil Platform, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, 15706 Santiago de Compostela, Spain.
| | - María L Couce
- Department of Forensic Sciences, Pathology, Gynecology and Obstetrics, Pediatrics, Neonatology Service, Department of Paediatrics, Hospital Clínico Universitario de Santiago de Compostela, Health Research Institute of Santiago de Compostela (IDIS), CIBERER, MetabERN, 15706 Santiago de Compostela, Spain.
| |
Collapse
|
66
|
Palhegyi AM, Seranova E, Dimova S, Hoque S, Sarkar S. Biomedical Implications of Autophagy in Macromolecule Storage Disorders. Front Cell Dev Biol 2019; 7:179. [PMID: 31555645 PMCID: PMC6742707 DOI: 10.3389/fcell.2019.00179] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 08/19/2019] [Indexed: 12/20/2022] Open
Abstract
An imbalance between the production and clearance of macromolecules such as proteins, lipids and carbohydrates can lead to a category of diseases broadly known as macromolecule storage disorders. These include, but not limited to, neurodegenerative diseases such as Alzheimer’s, Parkinson’s and Huntington’s disease associated with accumulation of aggregation-prone proteins, Lafora and Pompe disease associated with glycogen accumulation, whilst lipid accumulation is characteristic to Niemann-Pick disease and Gaucher disease. One of the underlying factors contributing to the build-up of macromolecules in these storage disorders is the intracellular degradation pathway called autophagy. This process is the primary clearance route for unwanted macromolecules, either via bulk non-selective degradation, or selectively via aggrephagy, glycophagy and lipophagy. Since autophagy plays a vital role in maintaining cellular homeostasis, cell viability and human health, malfunction of this process could be detrimental. Indeed, defective autophagy has been reported in a number of macromolecule storage disorders where autophagy is impaired at distinct stages, such as at the level of autophagosome formation, autophagosome maturation or improper lysosomal degradation of the autophagic cargo. Of biomedical relevance, autophagy is regulated by multiple signaling pathways that are amenable to chemical perturbations by small molecules. Induction of autophagy has been shown to improve cell viability and exert beneficial effects in experimental models of various macromolecule storage disorders where the lysosomal functionality is not overtly compromised. In this review, we will discuss the role of autophagy in certain macromolecule storage disorders and highlight the potential therapeutic benefits of autophagy enhancers in these pathological conditions.
Collapse
Affiliation(s)
- Adina Maria Palhegyi
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom
| | - Elena Seranova
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom
| | - Simona Dimova
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom
| | - Sheabul Hoque
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom
| | - Sovan Sarkar
- College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
67
|
Safary A, Akbarzadeh Khiavi M, Omidi Y, Rafi MA. Targeted enzyme delivery systems in lysosomal disorders: an innovative form of therapy for mucopolysaccharidosis. Cell Mol Life Sci 2019; 76:3363-3381. [PMID: 31101939 PMCID: PMC11105648 DOI: 10.1007/s00018-019-03135-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 04/19/2019] [Accepted: 05/06/2019] [Indexed: 12/27/2022]
Abstract
Mucopolysaccharidoses (MPSs), which are inherited lysosomal storage disorders caused by the accumulation of undegraded glycosaminoglycans, can affect the central nervous system (CNS) and elicit cognitive and behavioral issues. Currently used enzyme replacement therapy methodologies often fail to adequately treat the manifestations of the disease in the CNS and other organs such as bone, cartilage, cornea, and heart. Targeted enzyme delivery systems (EDSs) can efficiently cross biological barriers such as blood-brain barrier and provide maximal therapeutic effects with minimal side effects, and hence, offer great clinical benefits over the currently used conventional enzyme replacement therapies. In this review, we provide comprehensive insights into MPSs and explore the clinical impacts of multimodal targeted EDSs.
Collapse
Affiliation(s)
- Azam Safary
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, 51656-65811, Iran
| | - Mostafa Akbarzadeh Khiavi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, 51656-65811, Iran
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, 51656-65811, Iran.
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad A Rafi
- Department of Neurology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| |
Collapse
|
68
|
Manthe RL, Rappaport JA, Long Y, Solomon M, Veluvolu V, Hildreth M, Gugutkov D, Marugan J, Zheng W, Muro S. δ-Tocopherol Effect on Endocytosis and Its Combination with Enzyme Replacement Therapy for Lysosomal Disorders: A New Type of Drug Interaction? J Pharmacol Exp Ther 2019; 370:823-833. [PMID: 31101681 PMCID: PMC6806345 DOI: 10.1124/jpet.119.257345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/15/2019] [Indexed: 12/27/2022] Open
Abstract
Induction of lysosomal exocytosis alleviates lysosomal storage of undigested metabolites in cell models of lysosomal disorders (LDs). However, whether this strategy affects other vesicular compartments, e.g., those involved in endocytosis, is unknown. This is important both to predict side effects and to use this strategy in combination with therapies that require endocytosis for intracellular delivery, such as lysosomal enzyme replacement therapy (ERT). We investigated this using δ-tocopherol as a model previously shown to induce lysosomal exocytosis and cell models of type A Niemann-Pick disease, a LD characterized by acid sphingomyelinase (ASM) deficiency and sphingomyelin storage. δ-Tocopherol and derivative CF3-T reduced net accumulation of fluid phase, ligands, and polymer particles via phagocytic, caveolae-, clathrin-, and cell adhesion molecule (CAM)-mediated pathways, yet the latter route was less affected due to receptor overexpression. In agreement, δ-tocopherol lowered uptake of recombinant ASM by deficient cells (known to occur via the clathrin pathway) and via targeting intercellular adhesion molecule-1 (associated to the CAM pathway). However, the net enzyme activity delivered and lysosomal storage attenuation were greater via the latter route. Data suggest stimulation of exocytosis by tocopherols is not specific of lysosomes and affects endocytic cargo. However, this effect was transient and became unnoticeable several hours after tocopherol removal. Therefore, induction of exocytosis in combination with therapies requiring endocytic uptake, such as ERT, may represent a new type of drug interaction, yet this strategy could be valuable if properly timed for minimal interference.
Collapse
Affiliation(s)
- Rachel L Manthe
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Jeffrey A Rappaport
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Yan Long
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Melani Solomon
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Vinay Veluvolu
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Michael Hildreth
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Dencho Gugutkov
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Juan Marugan
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Wei Zheng
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| | - Silvia Muro
- Fischell Department of Bioengineering (R.L.M., J.A.R., V.V., M.H.) and Institute for Bioscience and Biotechnology Research (M.S., S.M.), University of Maryland, College Park, Maryland; National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland (Y.L., J.M., W.Z.); Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Barcelona, Spain (D.G., S.M.); and Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain (S.M.)
| |
Collapse
|
69
|
Wheeler S, Haberkant P, Bhardwaj M, Tongue P, Ferraz MJ, Halter D, Sprong H, Schmid R, Aerts JM, Sullo N, Sillence DJ. Cytosolic glucosylceramide regulates endolysosomal function in Niemann-Pick type C disease. Neurobiol Dis 2019; 127:242-252. [DOI: 10.1016/j.nbd.2019.03.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 03/06/2019] [Accepted: 03/09/2019] [Indexed: 12/22/2022] Open
|
70
|
Wheeler S, Schmid R, Sillence DJ. Lipid⁻Protein Interactions in Niemann⁻Pick Type C Disease: Insights from Molecular Modeling. Int J Mol Sci 2019; 20:E717. [PMID: 30736449 PMCID: PMC6387118 DOI: 10.3390/ijms20030717] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 01/31/2019] [Accepted: 02/03/2019] [Indexed: 12/19/2022] Open
Abstract
The accumulation of lipids in the late endosomes and lysosomes of Niemann⁻Pick type C disease (NPCD) cells is a consequence of the dysfunction of one protein (usually NPC1) but induces dysfunction in many proteins. We used molecular docking to propose (a) that NPC1 exports not just cholesterol, but also sphingosine, (b) that the cholesterol sensitivity of big potassium channel (BK) can be traced to a previously unappreciated site on the channel's voltage sensor, (c) that transient receptor potential mucolipin 1 (TRPML1) inhibition by sphingomyelin is likely an indirect effect, and (d) that phosphoinositides are responsible for both the mislocalization of annexin A2 (AnxA2) and a soluble NSF (N-ethylmaleimide Sensitive Fusion) protein attachment receptor (SNARE) recycling defect. These results are set in the context of existing knowledge of NPCD to sketch an account of the endolysosomal pathology key to this disease.
Collapse
Affiliation(s)
- Simon Wheeler
- School of Pharmacy, De Montfort University, The Gateway, Leicester LE1 9BH, UK.
| | - Ralf Schmid
- Leicester Institute of Structural and Chemical Biology, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 7RH, UK.
| | - Dan J Sillence
- School of Pharmacy, De Montfort University, The Gateway, Leicester LE1 9BH, UK.
| |
Collapse
|
71
|
Tsugawa H, Mori H, Matsuzaki J, Sato A, Saito Y, Imoto M, Suematsu M, Suzuki H. CAPZA1 determines the risk of gastric carcinogenesis by inhibiting Helicobacter pylori CagA-degraded autophagy. Autophagy 2019; 15:242-258. [PMID: 30176157 PMCID: PMC6333452 DOI: 10.1080/15548627.2018.1515530] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 06/18/2018] [Accepted: 08/21/2018] [Indexed: 01/04/2023] Open
Abstract
Helicobacter pylori-derived CagA, a type IV secretion system effector, plays a role as an oncogenic driver in gastric epithelial cells. However, upon delivery into gastric epithelial cells, CagA is usually degraded by macroautophagy/autophagy. Hence, the induction of autophagy in H. pylori-infected epithelial cells is an important host-protective ability against gastric carcinogenesis. However, the mechanisms by which autophagosome-lysosome fusion is regulated, are unknown. Here, we report that enhancement of LAMP1 (lysosomal associated membrane protein 1) expression is necessary for autolysosome formation. LAMP1 expression is induced by nuclear translocated LRP1 (LDL receptor related protein 1) intracellular domain (LRP1-ICD) binding to the proximal LAMP1 promoter region. Nuclear translocation of LRP1-ICD is enhanced by H. pylori infection. In contrast, CAPZA1 (capping actin protein of muscle Z-line alpha subunit 1) inhibits LAMP1 expression via binding to LRP1-ICD in the nuclei. The binding of CAPZA1 to LRP1-ICD prevents LRP1-ICD binding to the LAMP1 proximal promoter. Thus, in CAPZA1-overexpressing gastric epithelial cells infected with H. pylori, autolysosome formation is inhibited and CagA escapes autophagic degradation. These findings identify CAPZA1 as a novel negative regulator of autolysosome formation and suggest that deregulation of CAPZA1 expression leads to increased risk of gastric carcinogenesis. Abbreviations: CagA: cytotoxin-associated gene A; CAPZA1: capping actin protein of muscle Z-line alpha subunit 1; ChIP: chromatin immunoprecipitation; GTF2I: general transcription factor IIi; HDAC: histone deacetylase; LAMP1: lysosomal associated membrane protein 1; LRP1: LDL receptor related protein 1; LRP1-ICD: CagA intracellular domain; qPCR: quantitative polymerase chain reaction; VacA: vacuolating cytotoxin.
Collapse
Affiliation(s)
- Hitoshi Tsugawa
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Hideki Mori
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Juntaro Matsuzaki
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan
| | - Akira Sato
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Yoshimasa Saito
- Division of Pharmacotherapeutics, Keio University Faculty of Pharmacy, Tokyo, Japan
| | - Masaya Imoto
- Department of Biosciences and Informatics, Faculty of Science and Technology, Keio University, Kohoku-ku, Yokohama, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Hidekazu Suzuki
- Fellowship Training Center and Medical Education Center, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
72
|
Zhao YG, Zhang H. Autophagosome maturation: An epic journey from the ER to lysosomes. J Cell Biol 2018; 218:757-770. [PMID: 30578282 PMCID: PMC6400552 DOI: 10.1083/jcb.201810099] [Citation(s) in RCA: 258] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/05/2018] [Accepted: 12/05/2018] [Indexed: 12/23/2022] Open
Abstract
Macroautophagy involves the sequestration of cytoplasmic contents in a double-membrane autophagosome and their delivery to lysosomes for degradation. In multicellular organisms, nascent autophagosomes fuse with vesicles originating from endolysosomal compartments before forming degradative autolysosomes, a process known as autophagosome maturation. ATG8 family members, tethering factors, Rab GTPases, and SNARE proteins act coordinately to mediate fusion of autophagosomes with endolysosomal vesicles. The machinery mediating autophagosome maturation is under spatiotemporal control and provides regulatory nodes to integrate nutrient availability with autophagy activity. Dysfunction of autophagosome maturation is associated with various human diseases, including neurodegenerative diseases, Vici syndrome, cancer, and lysosomal storage disorders. Understanding the molecular mechanisms underlying autophagosome maturation will provide new insights into the pathogenesis and treatment of these diseases.
Collapse
Affiliation(s)
- Yan G Zhao
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA
| | - Hong Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China .,National Laboratory of Biomacromolecules, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
73
|
Clayton EL, Milioto C, Muralidharan B, Norona FE, Edgar JR, Soriano A, Jafar-nejad P, Rigo F, Collinge J, Isaacs AM. Frontotemporal dementia causative CHMP2B impairs neuronal endolysosomal traffic-rescue by TMEM106B knockdown. Brain 2018; 141:3428-3442. [PMID: 30496365 PMCID: PMC6262218 DOI: 10.1093/brain/awy284] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 09/19/2018] [Accepted: 09/23/2018] [Indexed: 12/12/2022] Open
Abstract
Mutations in the endosome-associated protein CHMP2B cause frontotemporal dementia and lead to lysosomal storage pathology in neurons. We here report that physiological levels of mutant CHMP2B causes reduced numbers and significantly impaired trafficking of endolysosomes within neuronal dendrites, accompanied by increased dendritic branching. Mechanistically, this is due to the stable incorporation of mutant CHMP2B onto neuronal endolysosomes, which we show renders them unable to traffic within dendrites. This defect is due to the inability of mutant CHMP2B to recruit the ATPase VPS4, which is required for release of CHMP2B from endosomal membranes. Strikingly, both impaired trafficking and the increased dendritic branching were rescued by treatment with antisense oligonucleotides targeting the well validated frontotemporal dementia risk factor TMEM106B, which encodes an endolysosomal protein. This indicates that reducing TMEM106B levels can restore endosomal health in frontotemporal dementia. As TMEM106B is a risk factor for frontotemporal dementia caused by both C9orf72 and progranulin mutations, and antisense oligonucleotides are showing promise as therapeutics for neurodegenerative diseases, our data suggests a potential new strategy for treating the wide range of frontotemporal dementias associated with endolysosomal dysfunction.
Collapse
Affiliation(s)
- Emma L Clayton
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, Queen Square, London, UK
| | - Carmelo Milioto
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, UCL Institute of Neurology, Queen Square, London, UK
| | - Bhavana Muralidharan
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, UCL Institute of Neurology, Queen Square, London, UK
| | - Frances E Norona
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - James R Edgar
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | | | | | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, California, USA
| | - John Collinge
- MRC Prion Unit at UCL, Institute of Prion Diseases, Queen Square, London, UK
| | - Adrian M Isaacs
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, UCL Institute of Neurology, Queen Square, London, UK
| |
Collapse
|
74
|
Settembre C, Cinque L, Bartolomeo R, Di Malta C, De Leonibus C, Forrester A. Defective collagen proteostasis and matrix formation in the pathogenesis of lysosomal storage disorders. Matrix Biol 2018; 71-72:283-293. [DOI: 10.1016/j.matbio.2018.06.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 12/14/2022]
|
75
|
Niemann-Pick type C disease: The atypical sphingolipidosis. Adv Biol Regul 2018; 70:82-88. [PMID: 30205942 DOI: 10.1016/j.jbior.2018.08.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 08/27/2018] [Indexed: 12/29/2022]
Abstract
Niemann-Pick type C (NPC) disease is a lysosomal storage disorder resulting from mutations in either the NPC1 (95%) or NPC2 (5%) genes. NPC typically presents in childhood with visceral lipid accumulation and complex progressive neurodegeneration characterized by cerebellar ataxia, dysphagia, and dementia, resulting in a shortened lifespan. While cholesterol is widely acknowledged as the principal storage lipid in NPC, multiple species of sphingolipids accumulate as well. This accumulation of sphingolipids led to the initial assumption that NPC disease was caused by a deficiency in a sphingolipid catabolism enzyme, similar to sphingomyelinase deficiencies with which it shares a family name. It took about half a century to determine that NPC was in fact caused by a cholesterol trafficking defect, and still as we approach a century after the initial identification of the disease, the mechanisms by which sphingolipids accumulate remain poorly understood. Here we focus on the defects of sphingolipid catabolism in the endolysosomal compartment and how they contribute to the biology and pathology observed in NPC disease. This review highlights the need for further work on understanding and possibly developing treatments to correct the accumulation of sphingolipids in addition to cholesterol in this currently untreatable disease.
Collapse
|
76
|
Muro S. Alterations in Cellular Processes Involving Vesicular Trafficking and Implications in Drug Delivery. Biomimetics (Basel) 2018; 3:biomimetics3030019. [PMID: 31105241 PMCID: PMC6352689 DOI: 10.3390/biomimetics3030019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 07/09/2018] [Accepted: 07/10/2018] [Indexed: 12/31/2022] Open
Abstract
Endocytosis and vesicular trafficking are cellular processes that regulate numerous functions required to sustain life. From a translational perspective, they offer avenues to improve the access of therapeutic drugs across cellular barriers that separate body compartments and into diseased cells. However, the fact that many factors have the potential to alter these routes, impacting our ability to effectively exploit them, is often overlooked. Altered vesicular transport may arise from the molecular defects underlying the pathological syndrome which we aim to treat, the activity of the drugs being used, or side effects derived from the drug carriers employed. In addition, most cellular models currently available do not properly reflect key physiological parameters of the biological environment in the body, hindering translational progress. This article offers a critical overview of these topics, discussing current achievements, limitations and future perspectives on the use of vesicular transport for drug delivery applications.
Collapse
Affiliation(s)
- Silvia Muro
- Institute for Bioscience and Biotechnology Research and Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA.
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain.
- Institute for Bioengineering of Catalonia (IBEC) of the Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain.
| |
Collapse
|
77
|
Barbero-Camps E, Roca-Agujetas V, Bartolessis I, de Dios C, Fernández-Checa JC, Marí M, Morales A, Hartmann T, Colell A. Cholesterol impairs autophagy-mediated clearance of amyloid beta while promoting its secretion. Autophagy 2018; 14:1129-1154. [PMID: 29862881 PMCID: PMC6103708 DOI: 10.1080/15548627.2018.1438807] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Macroautophagy/autophagy failure with the accumulation of autophagosomes is an early neuropathological feature of Alzheimer disease (AD) that directly affects amyloid beta (Aβ) metabolism. Although loss of presenilin 1 function has been reported to impair lysosomal function and prevent autophagy flux, the detailed mechanism leading to autophagy dysfunction in AD remains to be elucidated. The resemblance between pathological hallmarks of AD and Niemann-Pick Type C disease, including endosome-lysosome abnormalities and impaired autophagy, suggests cholesterol accumulation as a common link. Using a mouse model of AD (APP-PSEN1-SREBF2 mice), expressing chimeric mouse-human amyloid precursor protein with the familial Alzheimer Swedish mutation (APP695swe) and mutant presenilin 1 (PSEN1-dE9), together with a dominant-positive, truncated and active form of SREBF2/SREBP2 (sterol regulatory element binding factor 2), we demonstrated that high brain cholesterol enhanced autophagosome formation, but disrupted its fusion with endosomal-lysosomal vesicles. The combination of these alterations resulted in impaired degradation of Aβ and endogenous MAPT (microtubule associated protein tau), and stimulated autophagy-dependent Aβ secretion. Exacerbated Aβ-induced oxidative stress in APP-PSEN1-SREBF2 mice, due to cholesterol-mediated depletion of mitochondrial glutathione/mGSH, is critical for autophagy induction. In agreement, in vivo mitochondrial GSH recovery with GSH ethyl ester, inhibited autophagosome synthesis by preventing the oxidative inhibition of ATG4B deconjugation activity exerted by Aβ. Moreover, cholesterol-enrichment within the endosomes-lysosomes modified the levels and membrane distribution of RAB7A and SNAP receptors (SNAREs), which affected its fusogenic ability. Accordingly, in vivo treatment with 2-hydroxypropyl-β-cyclodextrin completely rescued these alterations, making it a potential therapeutic tool for AD.
Collapse
Affiliation(s)
- Elisabet Barbero-Camps
- a Department of Cell Death and Proliferation , Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , Barcelona , Spain
| | - Vicente Roca-Agujetas
- a Department of Cell Death and Proliferation , Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , Barcelona , Spain
| | - Isabel Bartolessis
- a Department of Cell Death and Proliferation , Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , Barcelona , Spain
| | - Cristina de Dios
- a Department of Cell Death and Proliferation , Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , Barcelona , Spain.,c Departament de Biomedicina, Facultat de Medicina , Universitat de Barcelona , Barcelona , Spain
| | - Jose C Fernández-Checa
- a Department of Cell Death and Proliferation , Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , Barcelona , Spain.,d Liver Unit , Hospital Clinic, CIBEREHD , Barcelona , Spain , Research Center for Alcoholic Liver and Pancreatic Diseases , Keck School of Medicine of the University of Southern California , Los Angeles , CA , USA
| | - Montserrat Marí
- a Department of Cell Death and Proliferation , Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , Barcelona , Spain
| | - Albert Morales
- a Department of Cell Death and Proliferation , Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , Barcelona , Spain
| | - Tobias Hartmann
- e Experimental Neurology , Saarland University , Homburg/Saar , Germany
| | - Anna Colell
- a Department of Cell Death and Proliferation , Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , Barcelona , Spain.,b Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) , Spain
| |
Collapse
|
78
|
Abstract
Macroautophagy (herein referred to as autophagy) is a highly conserved stress response that engulfs damaged proteins, lipids, and/or organelles within double-membrane vesicles called autophagosomes for lysosomal degradation. Dysregulated autophagy is a hallmark of cancer; and thus, there is great interest in modulating autophagy for cancer therapy. Sphingolipids regulate each step of autophagosome biogenesis with roles for sphingolipid metabolites and enzymes spanning from the initial step of de novo ceramide synthesis to the sphingosine-1-phosphate lyase 1-mediated exit from the sphingolipid pathway. Notably, sphingolipid metabolism occurs at several of the organelles that contribute to autophagosome biogenesis to suggest that local changes in sphingolipids may regulate autophagy. As sphingolipid metabolism is frequently dysregulated in cancer, a molecular understanding of sphingolipids in stress-induced autophagy may provide insight into the mechanisms driving tumor development and progression. On the contrary, modulation of sphingolipid metabolites and/or enzymes can induce autophagy-dependent cell death for cancer therapy. This chapter will overview the major steps in mammalian autophagy, discuss the regulation of each step by sphingolipid metabolites, and describe the functions of sphingolipid-mediated autophagy in cancer. While our understanding of the signaling and biophysical properties of sphingolipids in autophagy remains in its infancy, the unique cross talk between the two pathways is an exciting area for further development, particularly in the context of cancer therapy.
Collapse
Affiliation(s)
- Megan M Young
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Hong-Gang Wang
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
79
|
Abstract
Actin cytoskeleton dynamics play vital roles in most forms of intracellular trafficking by promoting the biogenesis and transport of vesicular cargoes. Mounting evidence indicates that actin dynamics and membrane-cytoskeleton scaffolds also have essential roles in macroautophagy, the process by which cellular waste is isolated inside specialized vesicles called autophagosomes for recycling and degradation. Branched actin polymerization is necessary for the biogenesis of autophagosomes from the endoplasmic reticulum (ER) membrane. Actomyosin-based transport is then used to feed the growing phagophore with pre-selected cargoes and debris derived from different membranous organelles inside the cell. Finally, mature autophagosomes detach from the ER membrane by an as yet unknown mechanism, undergo intracellular transport and then fuse with lysosomes, endosomes and multivesicular bodies through mechanisms that involve actin- and microtubule-mediated motility, cytoskeleton-membrane scaffolds and signaling proteins. In this review, we highlight the considerable progress made recently towards understanding the diverse roles of the cytoskeleton in autophagy.
Collapse
|
80
|
Yan P, Sun X, Chen X, Chen Y, Wang X, Su D, Zhou H, Gao L, Lu L, Wang J, Zhang J. The Polyphenolic Compound Curcumin Conjugation with an Alkyne Moiety in the Process of Autophagy. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2018; 46:673-687. [PMID: 29614882 DOI: 10.1142/s0192415x18500350] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Curcumin is a hydrophobic polyphenol derived from turmeric: the rhizome of the herb Curcumalonga. Autophagy is an evolutionarily conserved process, in which cellular proteins and organelles are engulfed in autophagosome and then fuses with lysosome for degradation. Our previous study showed that Curcumin activates lysosome and induce autophagy through inhibition of AKT (protein kinase K, PKB)-mammalian target of rapamycin (mTOR) pathway. But whether Curucmin affects the fusion of autophagosome-lysosome is still not clear. Here, we used Curcumin-probe conjugation with an alkyne moiety to label mouse embryonic fibroblasts (MEFs) and found that Curcumin targets autophagy-related proteins, enhances autophagic flux and activates lysosome in cells. Moreover, Curcumin treatment promotes the fusion of autophasosome-lysosome in MEFs. Second, the enhanced fusion of autophagosome-lysosome is attributed to mTOR suppression. Third, blockage of the autophagosome-lysosome fusion leads to cell growth inhibition by Curcumin. Taken together, data from our study indicates the importance of the fusion of autophagosome-lysosome in Curcumin-induced autophagy, which may facilitate the development of Curcumin as a potential therapeutic agent for oxidative stress-related diseases.
Collapse
Affiliation(s)
- Peiyi Yan
- Department of Clinical Laboratory, Shanghai Putuo District People’s Hospital, Shanghai, China
| | - Xin Sun
- Department of Oncology, Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Xiaochen Chen
- Department of Oncology, Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Yun Chen
- Department of Oncology, Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Xiao Wang
- Department of Oncology, Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Dan Su
- Department of Oncology, Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Hongying Zhou
- Department of Oncology, Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Liang Gao
- Department of Oncology, Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Liqin Lu
- Department of Oncology, Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Jigang Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jianbin Zhang
- Department of Oncology, Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
81
|
Gurda BL, Bagel JH, Fisher SJ, Schultz ML, Lieberman AP, Hand P, Vite CH, Swain GP. LC3 Immunostaining in the Inferior Olivary Nuclei of Cats With Niemann-Pick Disease Type C1 Is Associated With Patterned Purkinje Cell Loss. J Neuropathol Exp Neurol 2018; 77:229-245. [PMID: 29346563 PMCID: PMC5989620 DOI: 10.1093/jnen/nlx119] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The feline model of Niemann-Pick disease, type C1 (NPC1) recapitulates the clinical, neuropathological, and biochemical abnormalities present in children with NPC1. The hallmarks of disease are the lysosomal storage of unesterified cholesterol and multiple sphingolipids in neurons, and the spatial and temporal distribution of Purkinje cell death. In feline NPC1 brain, microtubule-associated protein 1 light chain 3 (LC3) accumulations, indicating autophagosomes, were found within axons and presynaptic terminals. High densities of accumulated LC3 were seen in subdivisions of the inferior olive, which project to cerebellar regions that show the most Purkinje cell loss, suggesting that autophagic abnormalities in specific climbing fibers may contribute to the spatial pattern of Purkinje cell loss seen. Biweekly intrathecal administration of 2-hydroxypropyl-beta cyclodextrin (HPβCD) ameliorated neurological dysfunction, reduced cholesterol and sphingolipid accumulation, and increased lifespan in NPC1 cats. LC3 pathology was reduced in treated animals suggesting that HPβCD administration also ameliorates autophagic abnormalities. This study is the first to (i) identify specific brain regions exhibiting autophagic abnormalities in any species with NPC1, (ii) provide evidence of differential vulnerability among discrete brain nuclei and pathways, and (iii) show the amelioration of these abnormalities in NPC1 cats treated with HPβCD.
Collapse
Affiliation(s)
- Brittney L Gurda
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jessica H Bagel
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Samantha J Fisher
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mark L Schultz
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Andrew P Lieberman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Peter Hand
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Charles H Vite
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gary P Swain
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
82
|
Gegg ME, Schapira AHV. The role of glucocerebrosidase in Parkinson disease pathogenesis. FEBS J 2018; 285:3591-3603. [DOI: 10.1111/febs.14393] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/17/2018] [Accepted: 01/25/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Matthew E. Gegg
- Department of Clinical Neuroscience; Institute of Neurology; University College London; UK
| | - Anthony H. V. Schapira
- Department of Clinical Neuroscience; Institute of Neurology; University College London; UK
| |
Collapse
|
83
|
Hesketh GG, Wartosch L, Davis LJ, Bright NA, Luzio JP. The Lysosome and Intracellular Signalling. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2018; 57:151-180. [PMID: 30097775 DOI: 10.1007/978-3-319-96704-2_6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In addition to being the terminal degradative compartment of the cell's endocytic and autophagic pathways, the lysosome is a multifunctional signalling hub integrating the cell's response to nutrient status and growth factor/hormone signalling. The cytosolic surface of the limiting membrane of the lysosome is the site of activation of the multiprotein complex mammalian target of rapamycin complex 1 (mTORC1), which phosphorylates numerous cell growth-related substrates, including transcription factor EB (TFEB). Under conditions in which mTORC1 is inhibited including starvation, TFEB becomes dephosphorylated and translocates to the nucleus where it functions as a master regulator of lysosome biogenesis. The signalling role of lysosomes is not limited to this pathway. They act as an intracellular Ca2+ store, which can release Ca2+ into the cytosol for both local effects on membrane fusion and pleiotropic effects within the cell. The relationship and crosstalk between the lysosomal and endoplasmic reticulum (ER) Ca2+ stores play a role in shaping intracellular Ca2+ signalling. Lysosomes also perform other signalling functions, which are discussed. Current views of the lysosomal compartment recognize its dynamic nature. It includes endolysosomes, autolysosome and storage lysosomes that are constantly engaged in fusion/fission events and lysosome regeneration. How signalling is affected by individual lysosomal organelles being at different stages of these processes and/or at different sites within the cell is poorly understood, but is discussed.
Collapse
Affiliation(s)
- Geoffrey G Hesketh
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, M5G 1X5, Canada
| | - Lena Wartosch
- Department of Clinical Biochemistry and Cambridge Institute for Medical Research, School of Clinical Medicine, Wellcome Trust/MRC Building, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY, UK
| | - Luther J Davis
- Department of Clinical Biochemistry and Cambridge Institute for Medical Research, School of Clinical Medicine, Wellcome Trust/MRC Building, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY, UK
| | - Nicholas A Bright
- Department of Clinical Biochemistry and Cambridge Institute for Medical Research, School of Clinical Medicine, Wellcome Trust/MRC Building, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY, UK
| | - J Paul Luzio
- Department of Clinical Biochemistry and Cambridge Institute for Medical Research, School of Clinical Medicine, Wellcome Trust/MRC Building, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY, UK.
| |
Collapse
|
84
|
Dysregulation of autophagy as a common mechanism in lysosomal storage diseases. Essays Biochem 2017; 61:733-749. [PMID: 29233882 PMCID: PMC5869865 DOI: 10.1042/ebc20170055] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 10/08/2017] [Accepted: 10/12/2017] [Indexed: 12/19/2022]
Abstract
The lysosome plays a pivotal role between catabolic and anabolic processes as the nexus for signalling pathways responsive to a variety of factors, such as growth, nutrient availability, energetic status and cellular stressors. Lysosomes are also the terminal degradative organelles for autophagy through which macromolecules and damaged cellular components and organelles are degraded. Autophagy acts as a cellular homeostatic pathway that is essential for organismal physiology. Decline in autophagy during ageing or in many diseases, including late-onset forms of neurodegeneration is considered a major contributing factor to the pathology. Multiple lines of evidence indicate that impairment in autophagy is also a central mechanism underlying several lysosomal storage disorders (LSDs). LSDs are a class of rare, inherited disorders whose histopathological hallmark is the accumulation of undegraded materials in the lysosomes due to abnormal lysosomal function. Inefficient degradative capability of the lysosomes has negative impact on the flux through the autophagic pathway, and therefore dysregulated autophagy in LSDs is emerging as a relevant disease mechanism. Pathology in the LSDs is generally early-onset, severe and life-limiting but current therapies are limited or absent; recognizing common autophagy defects in the LSDs raises new possibilities for therapy. In this review, we describe the mechanisms by which LSDs occur, focusing on perturbations in the autophagy pathway and present the latest data supporting the development of novel therapeutic approaches related to the modulation of autophagy.
Collapse
|
85
|
Menzies FM, Fleming A, Caricasole A, Bento CF, Andrews SP, Ashkenazi A, Füllgrabe J, Jackson A, Jimenez Sanchez M, Karabiyik C, Licitra F, Lopez Ramirez A, Pavel M, Puri C, Renna M, Ricketts T, Schlotawa L, Vicinanza M, Won H, Zhu Y, Skidmore J, Rubinsztein DC. Autophagy and Neurodegeneration: Pathogenic Mechanisms and Therapeutic Opportunities. Neuron 2017; 93:1015-1034. [PMID: 28279350 DOI: 10.1016/j.neuron.2017.01.022] [Citation(s) in RCA: 839] [Impact Index Per Article: 104.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 12/11/2022]
Abstract
Autophagy is a conserved pathway that delivers cytoplasmic contents to the lysosome for degradation. Here we consider its roles in neuronal health and disease. We review evidence from mouse knockout studies demonstrating the normal functions of autophagy as a protective factor against neurodegeneration associated with intracytoplasmic aggregate-prone protein accumulation as well as other roles, including in neuronal stem cell differentiation. We then describe how autophagy may be affected in a range of neurodegenerative diseases. Finally, we describe how autophagy upregulation may be a therapeutic strategy in a wide range of neurodegenerative conditions and consider possible pathways and druggable targets that may be suitable for this objective.
Collapse
Affiliation(s)
- Fiona M Menzies
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Angeleen Fleming
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Andrea Caricasole
- Alzheimer's Research UK Cambridge Drug Discovery Institute, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, UK
| | - Carla F Bento
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Stephen P Andrews
- Alzheimer's Research UK Cambridge Drug Discovery Institute, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, UK
| | - Avraham Ashkenazi
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Jens Füllgrabe
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Anne Jackson
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Maria Jimenez Sanchez
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Cansu Karabiyik
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Floriana Licitra
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Ana Lopez Ramirez
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Mariana Pavel
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Claudia Puri
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Maurizio Renna
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Thomas Ricketts
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Lars Schlotawa
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Mariella Vicinanza
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Hyeran Won
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Ye Zhu
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - John Skidmore
- Alzheimer's Research UK Cambridge Drug Discovery Institute, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, UK
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK.
| |
Collapse
|
86
|
Grasso G, Santoro AM, Lanza V, Sbardella D, Tundo GR, Ciaccio C, Marini S, Coletta M, Milardi D. The double faced role of copper in Aβ homeostasis: A survey on the interrelationship between metal dyshomeostasis, UPS functioning and autophagy in neurodegeneration. Coord Chem Rev 2017. [DOI: 10.1016/j.ccr.2017.06.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
87
|
Solomon M, Muro S. Lysosomal enzyme replacement therapies: Historical development, clinical outcomes, and future perspectives. Adv Drug Deliv Rev 2017; 118:109-134. [PMID: 28502768 PMCID: PMC5828774 DOI: 10.1016/j.addr.2017.05.004] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/26/2017] [Accepted: 05/08/2017] [Indexed: 01/06/2023]
Abstract
Lysosomes and lysosomal enzymes play a central role in numerous cellular processes, including cellular nutrition, recycling, signaling, defense, and cell death. Genetic deficiencies of lysosomal components, most commonly enzymes, are known as "lysosomal storage disorders" or "lysosomal diseases" (LDs) and lead to lysosomal dysfunction. LDs broadly affect peripheral organs and the central nervous system (CNS), debilitating patients and frequently causing fatality. Among other approaches, enzyme replacement therapy (ERT) has advanced to the clinic and represents a beneficial strategy for 8 out of the 50-60 known LDs. However, despite its value, current ERT suffers from several shortcomings, including various side effects, development of "resistance", and suboptimal delivery throughout the body, particularly to the CNS, lowering the therapeutic outcome and precluding the use of this strategy for a majority of LDs. This review offers an overview of the biomedical causes of LDs, their socio-medical relevance, treatment modalities and caveats, experimental alternatives, and future treatment perspectives.
Collapse
Affiliation(s)
- Melani Solomon
- Institute for Bioscience and Biotechnology Research, University Maryland, College Park, MD 20742, USA
| | - Silvia Muro
- Institute for Bioscience and Biotechnology Research, University Maryland, College Park, MD 20742, USA; Fischell Department of Bioengineering, University Maryland, College Park, MD 20742, USA.
| |
Collapse
|
88
|
Thelen AM, Zoncu R. Emerging Roles for the Lysosome in Lipid Metabolism. Trends Cell Biol 2017; 27:833-850. [PMID: 28838620 DOI: 10.1016/j.tcb.2017.07.006] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 07/28/2017] [Accepted: 07/31/2017] [Indexed: 12/20/2022]
Abstract
Precise regulation of lipid biosynthesis, transport, and storage is key to the homeostasis of cells and organisms. Cells rely on a sophisticated but poorly understood network of vesicular and nonvesicular transport mechanisms to ensure efficient delivery of lipids to target organelles. The lysosome stands at the crossroads of this network due to its ability to process and sort exogenous and endogenous lipids. The lipid-sorting function of the lysosome is intimately connected to its recently discovered role as a metabolic command-and-control center, which relays multiple nutrient cues to the master growth regulator, mechanistic target of rapamycin complex (mTORC)1 kinase. In turn, mTORC1 potently drives anabolic processes, including de novo lipid synthesis, while inhibiting lipid catabolism. Here, we describe the dual role of the lysosome in lipid transport and biogenesis, and we discuss how integration of these two processes may play important roles both in normal physiology and in disease.
Collapse
Affiliation(s)
- Ashley M Thelen
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA; The Paul F. Glenn Center for Aging Research at the University of California, Berkeley, Berkeley, CA 94720, USA
| | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA; The Paul F. Glenn Center for Aging Research at the University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
89
|
Mohamed FE, Al-Gazali L, Al-Jasmi F, Ali BR. Pharmaceutical Chaperones and Proteostasis Regulators in the Therapy of Lysosomal Storage Disorders: Current Perspective and Future Promises. Front Pharmacol 2017; 8:448. [PMID: 28736525 PMCID: PMC5500627 DOI: 10.3389/fphar.2017.00448] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 06/22/2017] [Indexed: 02/05/2023] Open
Abstract
Different approaches have been utilized or proposed for the treatment of lysosomal storage disorders (LSDs) including enzyme replacement and hematopoietic stem cell transplant therapies, both aiming to compensate for the enzymatic loss of the underlying mutated lysosomal enzymes. However, these approaches have their own limitations and therefore the vast majority of LSDs are either still untreatable or their treatments are inadequate. Missense mutations affecting enzyme stability, folding and cellular trafficking are common in LSDs resulting often in low protein half-life, premature degradation, aggregation and retention of the mutant proteins in the endoplasmic reticulum. Small molecular weight compounds such as pharmaceutical chaperones (PCs) and proteostasis regulators have been in recent years to be promising approaches for overcoming some of these protein processing defects. These compounds are thought to enhance lysosomal enzyme activity by specific binding to the mutated enzyme or by manipulating components of the proteostasis pathways promoting protein stability, folding and trafficking and thus enhancing and restoring some of the enzymatic activity of the mutated protein in lysosomes. Multiple compounds have already been approved for clinical use to treat multiple LSDs like migalastat in the treatment of Fabry disease and others are currently under research or in clinical trials such as Ambroxol hydrochloride and Pyrimethamine. In this review, we are presenting a general overview of LSDs, their molecular and cellular bases, and focusing on recent advances on targeting and manipulation proteostasis, including the use of PCs and proteostasis regulators, as therapeutic targets for some LSDs. In addition, we present the successes, limitations and future perspectives in this field.
Collapse
Affiliation(s)
- Fedah E. Mohamed
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates UniversityAl Ain, United Arab Emirates
| | - Lihadh Al-Gazali
- Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates UniversityAl Ain, United Arab Emirates
| | - Fatma Al-Jasmi
- Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates UniversityAl Ain, United Arab Emirates
| | - Bassam R. Ali
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates UniversityAl Ain, United Arab Emirates
- Zayed Bin Sultan Center for Health Sciences, United Arab Emirates UniversityAl-Ain, United Arab Emirates
| |
Collapse
|
90
|
Dai S, Dulcey AE, Hu X, Wassif CA, Porter FD, Austin CP, Ory DS, Marugan J, Zheng W. Methyl-β-cyclodextrin restores impaired autophagy flux in Niemann-Pick C1-deficient cells through activation of AMPK. Autophagy 2017; 13:1435-1451. [PMID: 28613987 PMCID: PMC5584846 DOI: 10.1080/15548627.2017.1329081] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The drug 2-hydroxypropyl-β-cyclodextrin (HPβCD) reduces lysosomal cholesterol accumulation in Niemann-Pick disease, type C (NPC) and has been advanced to human clinical trials. However, its mechanism of action for reducing cholesterol accumulation in NPC cells is uncertain and its molecular target is unknown. We found that methyl-β-cyclodextrin (MβCD), a potent analog of HPβCD, restored impaired macroautophagy/autophagy flux in Niemann-Pick disease, type C1 (NPC1) cells. This effect was mediated by a direct activation of AMP-activated protein kinase (AMPK), an upstream kinase in the autophagy pathway, through MβCD binding to its β-subunits. Knockdown of PRKAB1 or PRKAB2 (encoding the AMPK β1 or β2 subunit) expression and an AMPK inhibitor abolished MβCD-mediated reduction of cholesterol storage in NPC1 cells. The results demonstrate that AMPK is the molecular target of MβCD and its activation enhances autophagy flux, thereby mitigating cholesterol accumulation in NPC1 cells. The results identify AMPK as an attractive target for drug development to treat NPC.
Collapse
Affiliation(s)
- Sheng Dai
- a National Center for Advancing Translational Sciences (NCATS), NIH , Bethesda , MD , USA.,b Sir Run Run Shaw Hospital , Zhejiang University School of Medicine , Hangzhou , China
| | - Andrés E Dulcey
- a National Center for Advancing Translational Sciences (NCATS), NIH , Bethesda , MD , USA
| | - Xin Hu
- a National Center for Advancing Translational Sciences (NCATS), NIH , Bethesda , MD , USA
| | - Christopher A Wassif
- c National Institute of Child Health and Human Development, NIH , Bethesda , MD , USA
| | - Forbes D Porter
- c National Institute of Child Health and Human Development, NIH , Bethesda , MD , USA
| | - Christopher P Austin
- a National Center for Advancing Translational Sciences (NCATS), NIH , Bethesda , MD , USA
| | - Daniel S Ory
- d Diabetic Cardiovascular Disease Center , Washington University School of Medicine , St. Louis , MO USA
| | - Juan Marugan
- a National Center for Advancing Translational Sciences (NCATS), NIH , Bethesda , MD , USA
| | - Wei Zheng
- a National Center for Advancing Translational Sciences (NCATS), NIH , Bethesda , MD , USA
| |
Collapse
|
91
|
Beard H, Hassiotis S, Gai WP, Parkinson-Lawrence E, Hopwood JJ, Hemsley KM. Axonal dystrophy in the brain of mice with Sanfilippo syndrome. Exp Neurol 2017; 295:243-255. [PMID: 28601604 DOI: 10.1016/j.expneurol.2017.06.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 04/16/2017] [Accepted: 06/06/2017] [Indexed: 01/29/2023]
Abstract
Axonal dystrophy has been described as an early pathological feature of neurodegenerative disorders including Alzheimer's disease and amyotrophic lateral sclerosis. Axonal inclusions have also been reported to occur in several neurodegenerative lysosomal storage disorders including Mucopolysaccharidosis type IIIA (MPS IIIA; Sanfilippo syndrome). This disorder results from a mutation in the gene encoding the lysosomal sulphatase sulphamidase, and as a consequence heparan sulphate accumulates, accompanied by secondarily-stored gangliosides. The precise basis of symptom generation in MPS IIIA has not been elucidated, however axonal dystrophy may conceivably lead to impaired vesicular trafficking, neuronal dysfunction and/or death. We have utilised a faithful murine model of MPS IIIA to determine the spatio-temporal profile of neuronal inclusion formation and determine the effect of restoring normal lysosomal function. Dopaminergic (tyrosine hydroxylase-positive), cholinergic (choline acetyltransferase-positive) and GABAergic (glutamic acid decarboxylase65/67-positive) neurons were found to exhibit axonal dystrophy in MPS IIIA mouse brain. Axonal lesions present by ~seven weeks of age were Rab5-positive but lysosomal integral membrane protein-2 negative, suggesting early endosomal involvement. By 9-12-weeks of age, immunoreactivity for the autophagosome-related proteins LC3 and p62 and the proteasomal subunit 19S was noted in the spheroidal structures, together with wildtype α-synuclein, phosphorylated Thr-181 Tau and amyloid precursor protein, indicative of impaired axonal trafficking. Sulphamidase replacement reduced but did not abrogate the axonal lesions. Therefore, if axonal dystrophy impairs neuronal activity and ultimately, neuronal function, its incomplete resolution warrants further investigation.
Collapse
Affiliation(s)
- Helen Beard
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5001, Australia
| | - Sofia Hassiotis
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5001, Australia
| | - Wei-Ping Gai
- Dept Human Physiology, The Flinders University of SA, Bedford Park, SA 5052, Australia
| | - Emma Parkinson-Lawrence
- School of Pharmacy & Medical Sciences, University of South Australia, Adelaide, SA 5001, Australia
| | - John J Hopwood
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5001, Australia
| | - Kim M Hemsley
- Lysosomal Diseases Research Unit, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5001, Australia.
| |
Collapse
|
92
|
Budini M, Buratti E, Morselli E, Criollo A. Autophagy and Its Impact on Neurodegenerative Diseases: New Roles for TDP-43 and C9orf72. Front Mol Neurosci 2017; 10:170. [PMID: 28611593 PMCID: PMC5447761 DOI: 10.3389/fnmol.2017.00170] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 05/15/2017] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a catabolic mechanism where intracellular material is degraded by vesicular structures called autophagolysosomes. Autophagy is necessary to maintain the normal function of the central nervous system (CNS), avoiding the accumulation of misfolded and aggregated proteins. Consistently, impaired autophagy has been associated with the pathogenesis of various neurodegenerative diseases. The proteins TAR DNA-binding protein-43 (TDP-43), which regulates RNA processing at different levels, and chromosome 9 open reading frame 72 (C9orf72), probably involved in membrane trafficking, are crucial in the development of neurodegenerative diseases such as Amyotrophic lateral sclerosis (ALS) and Frontotemporal Lobar Degeneration (FTLD). Additionally, recent studies have identified a role for these proteins in the control of autophagy. In this manuscript, we review what is known regarding the autophagic mechanism and discuss the involvement of TDP-43 and C9orf72 in autophagy and their impact on neurodegenerative diseases.
Collapse
Affiliation(s)
- Mauricio Budini
- Dentistry Faculty, Institute in Dentistry Sciences, University of ChileSantiago, Chile
| | - Emanuele Buratti
- International Centre for Genetic Engineering and BiotechnologyTrieste, Italy
| | - Eugenia Morselli
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de ChileSantiago, Chile
| | - Alfredo Criollo
- Dentistry Faculty, Institute in Dentistry Sciences, University of ChileSantiago, Chile.,Advanced Center for Chronic DiseasesSantiago, Chile
| |
Collapse
|
93
|
Yang DS, Stavrides P, Kumar A, Jiang Y, Mohan PS, Ohno M, Dobrenis K, Davidson CD, Saito M, Pawlik M, Huo C, Walkley SU, Nixon RA. Cyclodextrin has conflicting actions on autophagy flux in vivo in brains of normal and Alzheimer model mice. Hum Mol Genet 2017; 26:843-859. [PMID: 28062666 DOI: 10.1093/hmg/ddx001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 01/03/2016] [Indexed: 12/13/2022] Open
Abstract
2-hydroxypropyl-β-cyclodextrin (CYCLO), a modifier of cholesterol efflux from cellular membrane and endo-lysosomal compartments, reduces lysosomal lipid accumulations and has therapeutic effects in animal models of Niemann-Pick disease type C and several other neurodegenerative states. Here, we investigated CYCLO effects on autophagy in wild-type mice and TgCRND8 mice-an Alzheimer's Disease (AD) model exhibiting β-amyloidosis, neuronal autophagy deficits leading to protein and lipid accumulation within greatly enlarged autolysosomes. A 14-day intracerebroventricular administration of CYCLO to 8-month-old TgCRND8 mice that exhibit moderately advanced neuropathology markedly diminished the sizes of enlarged autolysosomes and lowered their content of GM2 ganglioside and Aβ-immunoreactivity without detectably altering amyloid precursor protein processing or extracellular Aβ/β-amyloid burden. We identified two major actions of CYCLO on autophagy underlying amelioration of lysosomal pathology. First, CYCLO stimulated lysosomal proteolytic activity by increasing cathepsin D activity, levels of cathepsins B and D and two proteins known to interact with cathepsin D, NPC1 and ABCA1. Second, CYCLO impeded autophagosome-lysosome fusion as evidenced by the accumulation of LC3, SQSTM1/p62, and ubiquitinated substrates in an expanded population of autophagosomes in the absence of greater autophagy induction. By slowing substrate delivery to lysosomes, autophagosome maturational delay, as further confirmed by our in vitro studies, may relieve lysosomal stress due to accumulated substrates. These findings provide in vivo evidence for lysosomal enhancing properties of CYCLO, but caution that prolonged interference with cellular membrane fusion/autophagosome maturation could have unfavorable consequences, which might require careful optimization of dosage and dosing schedules.
Collapse
Affiliation(s)
- Dun-Sheng Yang
- Nathan Kline Institute, Orangeburg, NY, USA.,Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA
| | | | - Asok Kumar
- Nathan Kline Institute, Orangeburg, NY, USA.,Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA
| | - Ying Jiang
- Nathan Kline Institute, Orangeburg, NY, USA.,Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA
| | - Panaiyur S Mohan
- Nathan Kline Institute, Orangeburg, NY, USA.,Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA
| | - Masuo Ohno
- Nathan Kline Institute, Orangeburg, NY, USA.,Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA
| | - Kostantin Dobrenis
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Cristin D Davidson
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mitsuo Saito
- Nathan Kline Institute, Orangeburg, NY, USA.,Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA
| | | | | | - Steven U Walkley
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ralph A Nixon
- Nathan Kline Institute, Orangeburg, NY, USA.,Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA.,Cell Biology, New York University Langone Medical Center, New York, NY, USA
| |
Collapse
|
94
|
Tamura A, Yui N. Rational design of stimuli-cleavable polyrotaxanes for therapeutic applications. Polym J 2017. [DOI: 10.1038/pj.2017.17] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
95
|
Ríos-Marco P, Marco C, Gálvez X, Jiménez-López JM, Carrasco MP. Alkylphospholipids: An update on molecular mechanisms and clinical relevance. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:1657-1667. [PMID: 28238819 DOI: 10.1016/j.bbamem.2017.02.016] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 02/20/2017] [Accepted: 02/21/2017] [Indexed: 11/16/2022]
Abstract
Alkylphospholipids (APLs) represent a new class of drugs which do not interact directly with DNA but act on the cell membrane where they accumulate and interfere with lipid metabolism and signalling pathways. This review summarizes the mode of action at the molecular level of these compounds. In this sense, a diversity of mechanisms has been suggested to explain the actions of clinically-relevant APLs, in particular, in cancer treatment. One consistently reported finding is that APLs reduce the biosynthesis of phosphatidylcholine (PC) by inhibiting the rate-limiting enzyme CTP:phosphocholine cytidylyltransferase (CT). APLs also alter intracellular cholesterol traffic and metabolism in human tumour-cell lines, leading to an accumulation of cholesterol inside the cell. An increase in cholesterol biosynthesis associated with a decrease in the synthesis of choline-containing phospholipids and cholesterol esterification leads to a change in the free-cholesterol:PC ratio in cells exposed to APLs. Akt phosphorylation status after APL exposure shows that this critical regulator for cell survival is modulated by changes in cholesterol levels induced in the plasma membrane by these lipid analogues. Furthermore, APLs produce cell ultrastructural alterations with an abundant autophagic vesicles and autolysosomes in treated cells, indicating an interference of autophagy process after APL exposure. Thus, antitumoural APLs interfere with the proliferation of tumour cells via a complex mechanism involving phospholipid and cholesterol metabolism, interfere with lipid-dependent survival-signalling pathways and autophagy. Although APLs also exert antiparasitic, antibacterial, and antifungal effects, in this review we provide a summary of the antileishmanial activity of these lipid analogues. This article is part of a Special Issue entitled: Membrane Lipid Therapy: Drugs Targeting Biomembranes edited by Pablo V. Escribá.
Collapse
Affiliation(s)
- Pablo Ríos-Marco
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Av. Fuentenueva s/n, Granada 18001, Spain
| | - Carmen Marco
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Av. Fuentenueva s/n, Granada 18001, Spain
| | - Xiomara Gálvez
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Av. Fuentenueva s/n, Granada 18001, Spain
| | - José M Jiménez-López
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Av. Fuentenueva s/n, Granada 18001, Spain.
| | - María P Carrasco
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Av. Fuentenueva s/n, Granada 18001, Spain.
| |
Collapse
|
96
|
Borland H, Vilhardt F. Prelysosomal Compartments in the Unconventional Secretion of Amyloidogenic Seeds. Int J Mol Sci 2017; 18:ijms18010227. [PMID: 28124989 PMCID: PMC5297856 DOI: 10.3390/ijms18010227] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/09/2017] [Accepted: 01/16/2017] [Indexed: 12/18/2022] Open
Abstract
A mechanistic link between neuron-to-neuron transmission of secreted amyloid and propagation of protein malconformation cytopathology and disease has recently been uncovered in animal models. An enormous interest in the unconventional secretion of amyloids from neurons has followed. Amphisomes and late endosomes are the penultimate maturation products of the autophagosomal and endosomal pathways, respectively, and normally fuse with lysosomes for degradation. However, under conditions of perturbed membrane trafficking and/or lysosomal deficiency, prelysosomal compartments may instead fuse with the plasma membrane to release any contained amyloid. After a brief introduction to the endosomal and autophagosomal pathways, we discuss the evidence for autophagosomal secretion (exophagy) of amyloids, with a comparative emphasis on Aβ1-42 and α-synuclein, as luminal and cytosolic amyloids, respectively. The ESCRT-mediated import of cytosolic amyloid into late endosomal exosomes, a known vehicle of transmission of macromolecules between cells, is also reviewed. Finally, mechanisms of lysosomal dysfunction, deficiency, and exocytosis are exemplified in the context of genetically identified risk factors, mainly for Parkinson's disease. Exocytosis of prelysosomal or lysosomal organelles is a last resort for clearance of cytotoxic material and alleviates cytopathy. However, they also represent a vehicle for the concentration, posttranslational modification, and secretion of amyloid seeds.
Collapse
Affiliation(s)
- Helena Borland
- Department of Neurodegeneration In Vitro, H. Lundbeck A/S, 2500 Valby, Denmark.
| | - Frederik Vilhardt
- Department of Cellular and Molecular Medicine, Panum Institute, University of Copenhagen, 2200N Copenhagen, Denmark.
| |
Collapse
|
97
|
Bouché V, Espinosa AP, Leone L, Sardiello M, Ballabio A, Botas J. Drosophila Mitf regulates the V-ATPase and the lysosomal-autophagic pathway. Autophagy 2016; 12:484-98. [PMID: 26761346 DOI: 10.1080/15548627.2015.1134081] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
An evolutionarily conserved gene network regulates the expression of genes involved in lysosome biogenesis, autophagy, and lipid metabolism. In mammals, TFEB and other members of the MiTF-TFE family of transcription factors control this network. Here we report that the lysosomal-autophagy pathway is controlled by Mitf gene in Drosophila melanogaster. Mitf is the single MiTF-TFE family member in Drosophila and prior to this work was known only for its function in eye development. We show that Mitf regulates the expression of genes encoding V-ATPase subunits as well as many additional genes involved in the lysosomal-autophagy pathway. Reduction of Mitf function leads to abnormal lysosomes and impairs autophagosome fusion and lipid breakdown during the response to starvation. In contrast, elevated Mitf levels increase the number of lysosomes, autophagosomes and autolysosomes, and decrease the size of lipid droplets. Inhibition of Drosophila MTORC1 induces Mitf translocation to the nucleus, underscoring conserved regulatory mechanisms between Drosophila and mammalian systems. Furthermore, we show Mitf-mediated clearance of cytosolic and nuclear expanded ATXN1 (ataxin 1) in a cellular model of spinocerebellar ataxia type 1 (SCA1). This remarkable observation illustrates the potential of the lysosomal-autophagy system to prevent toxic protein aggregation in both the cytoplasmic and nuclear compartments. We anticipate that the genetics of the Drosophila model and the absence of redundant MIT transcription factors will be exploited to investigate the regulation and function of the lysosomal-autophagy gene network.
Collapse
Affiliation(s)
- Valentina Bouché
- a Department of Molecular and Human Genetics , Baylor College of Medicine , Houston , TX , USA.,b Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital , Houston , TX , USA.,c Telethon Institute of Genetics and Medicine (TIGEM) , Naples , Italy
| | - Alma Perez Espinosa
- a Department of Molecular and Human Genetics , Baylor College of Medicine , Houston , TX , USA.,b Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital , Houston , TX , USA
| | - Luigi Leone
- a Department of Molecular and Human Genetics , Baylor College of Medicine , Houston , TX , USA.,b Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital , Houston , TX , USA.,d Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche , Pozzuoli , Italy
| | - Marco Sardiello
- a Department of Molecular and Human Genetics , Baylor College of Medicine , Houston , TX , USA.,b Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital , Houston , TX , USA
| | - Andrea Ballabio
- a Department of Molecular and Human Genetics , Baylor College of Medicine , Houston , TX , USA.,b Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital , Houston , TX , USA.,c Telethon Institute of Genetics and Medicine (TIGEM) , Naples , Italy.,e Medical Genetics, Department of Translational Medicine, Federico II University , Naples , Italy
| | - Juan Botas
- a Department of Molecular and Human Genetics , Baylor College of Medicine , Houston , TX , USA.,b Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital , Houston , TX , USA
| |
Collapse
|
98
|
Shi B, Huang QQ, Birkett R, Doyle R, Dorfleutner A, Stehlik C, He C, Pope RM. SNAPIN is critical for lysosomal acidification and autophagosome maturation in macrophages. Autophagy 2016; 13:285-301. [PMID: 27929705 DOI: 10.1080/15548627.2016.1261238] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
We previously observed that SNAPIN, which is an adaptor protein in the SNARE core complex, was highly expressed in rheumatoid arthritis synovial tissue macrophages, but its role in macrophages and autoimmunity is unknown. To identify SNAPIN's role in these cells, we employed siRNA to silence the expression of SNAPIN in primary human macrophages. Silencing SNAPIN resulted in swollen lysosomes with impaired CTSD (cathepsin D) activation, although total CTSD was not reduced. Neither endosome cargo delivery nor lysosomal fusion with endosomes or autophagosomes was inhibited following the forced silencing of SNAPIN. The acidification of lysosomes and accumulation of autolysosomes in SNAPIN-silenced cells was inhibited, resulting in incomplete lysosomal hydrolysis and impaired macroautophagy/autophagy flux. Mechanistic studies employing ratiometric color fluorescence on living cells demonstrated that the reduction of SNAPIN resulted in a modest reduction of H+ pump activity; however, the more critical mechanism was a lysosomal proton leak. Overall, our results demonstrate that SNAPIN is critical in the maintenance of healthy lysosomes and autophagy through its role in lysosome acidification and autophagosome maturation in macrophages largely through preventing proton leak. These observations suggest an important role for SNAPIN and autophagy in the homeostasis of macrophages, particularly long-lived tissue resident macrophages.
Collapse
Affiliation(s)
- Bo Shi
- a Division of Rheumatology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Qi-Quan Huang
- a Division of Rheumatology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Robert Birkett
- a Division of Rheumatology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Renee Doyle
- a Division of Rheumatology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Andrea Dorfleutner
- a Division of Rheumatology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Christian Stehlik
- a Division of Rheumatology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Congcong He
- b Department of Cell and Molecular Biology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| | - Richard M Pope
- a Division of Rheumatology , Feinberg School of Medicine, Northwestern University , Chicago , IL , USA
| |
Collapse
|
99
|
Gómez-Sintes R, Ledesma MD, Boya P. Lysosomal cell death mechanisms in aging. Ageing Res Rev 2016; 32:150-168. [PMID: 26947122 DOI: 10.1016/j.arr.2016.02.009] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 02/22/2016] [Accepted: 02/29/2016] [Indexed: 12/14/2022]
Abstract
Lysosomes are degradative organelles essential for cell homeostasis that regulate a variety of processes, from calcium signaling and nutrient responses to autophagic degradation of intracellular components. Lysosomal cell death is mediated by the lethal effects of cathepsins, which are released into the cytoplasm following lysosomal damage. This process of lysosomal membrane permeabilization and cathepsin release is observed in several physiopathological conditions and plays a role in tissue remodeling, the immune response to intracellular pathogens and neurodegenerative diseases. Many evidences indicate that aging strongly influences lysosomal activity by altering the physical and chemical properties of these organelles, rendering them more sensitive to stress. In this review we focus on how aging alters lysosomal function and increases cell sensitivity to lysosomal membrane permeabilization and lysosomal cell death, both in physiological conditions and age-related pathologies.
Collapse
Affiliation(s)
- Raquel Gómez-Sintes
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biologicas, CIB-CSIC, C/Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - María Dolores Ledesma
- Department of Molecular Neurobiology, Centro Biologia Molecular Severo Ochoa, CSIC-UAM, C/Nicolás Cabrera 1, 28049 Madrid, Spain
| | - Patricia Boya
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biologicas, CIB-CSIC, C/Ramiro de Maeztu 9, 28040 Madrid, Spain.
| |
Collapse
|
100
|
One-Step Selective Exoenzymatic Labeling (SEEL) Strategy for the Biotinylation and Identification of Glycoproteins of Living Cells. J Am Chem Soc 2016; 138:11575-11582. [PMID: 27541995 DOI: 10.1021/jacs.6b04049] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Technologies that can visualize, capture, and identify subsets of biomolecules that are not encoded by the genome in the context of healthy and diseased cells will offer unique opportunities to uncover the molecular mechanism of a multitude of physiological and disease processes. We describe here a chemical reporter strategy for labeling of cell surface glycoconjugates that takes advantage of recombinant glycosyltransferases and a corresponding sugar nucleotide functionalized by biotin. The exceptional efficiency of this method, termed one-step selective exoenzymatic labeling, or SEEL, greatly improved the ability to enrich and identify large numbers of tagged glycoproteins by LC-MS/MS. We further demonstrated that this labeling method resulted in far superior enrichment and detection of glycoproteins at the plasma membrane compared to a sulfo-NHS-activated biotinylation or two-step SEEL. This new methodology will make it possible to profile cell surface glycoproteomes with unprecedented sensitivity in the context of physiological and disease states.
Collapse
|