1
|
Marques ARA, Ferreira IS, Ribeiro Q, Ferraz MJ, Lopes E, Pinto D, Hall M, Ramalho J, Artola M, Almeida MS, Rodrigues G, Gonçalves PA, Ferreira J, Borbinha C, Marto JP, Viana-Baptista M, Gouveia E Melo R, Pedro LM, Soares MIL, Vaz WLC, Vieira OV, Aerts JMFG. Glucosylated cholesterol accumulates in atherosclerotic lesions and impacts macrophage immune response. J Lipid Res 2025:100825. [PMID: 40381699 DOI: 10.1016/j.jlr.2025.100825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 05/09/2025] [Accepted: 05/10/2025] [Indexed: 05/20/2025] Open
Abstract
Atherosclerosis can be described as a local acquired lysosomal storage disorder (LSD), resulting from the build-up of undegraded material in lysosomes. Atherosclerotic foam cells accumulate cholesterol (Chol) and glycosphingolipids (GSLs) within lysosomes. This constitutes the ideal milieu for the formation of a side product of lysosomal storage: glucosylated cholesterol (GlcChol), previously found in several LSDs. Using LC-MS/MS, we demonstrated that GlcChol is abundant in atherosclerotic lesions. Patients suffering from cardiovascular diseases presented unaltered plasma GlcChol levels but slightly elevated GlcChol/Chol ratios. Furthermore, we mimicked GlcChol formation in vitro by exposing macrophages (Mφ) to a pro-atherogenic oxidized cholesteryl ester, an atherosclerosis foam cell model. Additionally, Mφ exposed to GlcChol exhibited an enlarged and multinucleated phenotype. These Mφ present signs of decreased proliferation and reduced pro-inflammatory capacity. Mechanistically the process seems to be associated with the activation of the AMPK signalling pathway and the cyclin-dependent kinase inhibitor 1 (CDKN1A/p21), in response to DNA damage inflicted by reactive oxygen species (ROS). At the organelle level, exposure to GlcChol impacted the lysosomal compartment, resulting in the activation of the mTOR signalling pathway and lysosomal biogenesis mediated by the transcription factor EB (TFEB). This suggests that high concentrations of GlcChol impact cellular homeostasis. In contrast, under this threshold GlcChol formation most likely represents a relatively innocuous compensatory mechanism to cope with Chol and GSL build-up within lesions. Our findings demonstrate that glycosidase-mediated lipid modifications may play a role in the aetiology of genetic and acquired LSDs, warranting further investigation.
Collapse
Affiliation(s)
- André R A Marques
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal.
| | - Inês S Ferreira
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Quélia Ribeiro
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Maria J Ferraz
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Elizeth Lopes
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Daniela Pinto
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Michael Hall
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - José Ramalho
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Marta Artola
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Manuel S Almeida
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; Hospital Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Av. Prof. Dr. Reinaldo dos Santos, 2790-134 Carnaxide, Portugal
| | - Gustavo Rodrigues
- Hospital Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Av. Prof. Dr. Reinaldo dos Santos, 2790-134 Carnaxide, Portugal
| | - Pedro Araújo Gonçalves
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; Hospital Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Av. Prof. Dr. Reinaldo dos Santos, 2790-134 Carnaxide, Portugal
| | - Jorge Ferreira
- Hospital Santa Cruz, Centro Hospitalar de Lisboa Ocidental, Av. Prof. Dr. Reinaldo dos Santos, 2790-134 Carnaxide, Portugal
| | - Cláudia Borbinha
- Department of Neurology, Hospital de Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Rua da Junqueira 126 1349-019 Lisboa, Portugal
| | - João Pedro Marto
- Department of Neurology, Hospital de Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Rua da Junqueira 126 1349-019 Lisboa, Portugal
| | - Miguel Viana-Baptista
- Department of Neurology, Hospital de Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Rua da Junqueira 126 1349-019 Lisboa, Portugal
| | - Ryan Gouveia E Melo
- Department of Vascular Surgery, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisboa, Portugal
| | - Luís Mendes Pedro
- Department of Vascular Surgery, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisboa, Portugal
| | - Maria I L Soares
- University of Coimbra, Coimbra Chemistry Centre - Institute of Molecular Sciences (CQC-IMS), Department of Chemistry, 3004-535 Coimbra, Portugal
| | - Winchil L C Vaz
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Otília V Vieira
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Johannes M F G Aerts
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| |
Collapse
|
2
|
Appu AP, Bagh MB, Plavelil N, Mondal A, Sadhukhan T, Singh SP, Perkins NJ, Liu A, Mukherjee AB. Niemann Pick C1 mistargeting disrupts lysosomal cholesterol homeostasis contributing to neurodegeneration in a Batten disease model. SCIENCE ADVANCES 2025; 11:eadr5703. [PMID: 40333988 PMCID: PMC12057685 DOI: 10.1126/sciadv.adr5703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 04/01/2025] [Indexed: 05/09/2025]
Abstract
Neurodegeneration is a devastating manifestation in most lysosomal storage disorders (LSDs). Loss-of-function mutations in CLN1, encoding palmitoyl-protein thioesterase-1 (PPT1), cause CLN1 disease, a devastating neurodegenerative LSD that has no curative treatment. Numerous proteins in the brain require dynamic S-palmitoylation (palmitoylation-depalmitoylation) for trafficking to their destination. Although PPT1 depalmitoylates S-palmitoylated proteins and its deficiency causes CLN1 disease, the underlying pathogenic mechanism has remained elusive. We report that Niemann-Pick C1 (NPC1), a polytopic membrane protein mediating lysosomal cholesterol egress, requires dynamic S-palmitoylation for trafficking to the lysosome. In Cln1-/- mice, Ppt1 deficiency misroutes NPC1-dysregulating lysosomal cholesterol homeostasis. Along with this defect, increased oxysterol-binding protein (OSBP) promotes cholesterol-mediated activation of mechanistic target of rapamycin C1 (mTORC1), which inhibits autophagy contributing to neurodegeneration. Pharmacological inhibition of OSBP suppresses mTORC1 activation, rescues autophagy, and ameliorates neuropathology in Cln1-/- mice. Our findings reveal a previously unrecognized role of CLN1/PPT1 in lysosomal cholesterol homeostasis and suggest that suppression of mTORC1 activation may be beneficial for CLN1 disease.
Collapse
Affiliation(s)
- Abhilash P. Appu
- Section on Developmental Genetics, Division of Translational Medicine, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1830, USA
| | - Maria B. Bagh
- Section on Developmental Genetics, Division of Translational Medicine, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1830, USA
| | - Nisha Plavelil
- Section on Developmental Genetics, Division of Translational Medicine, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1830, USA
| | - Avisek Mondal
- Section on Developmental Genetics, Division of Translational Medicine, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1830, USA
| | - Tamal Sadhukhan
- Section on Developmental Genetics, Division of Translational Medicine, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1830, USA
| | - Satya P. Singh
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1830, USA
| | - Neil J. Perkins
- Biostatistics and Bioinformatics Branch (HNT72), Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1830, USA
| | - Aiyi Liu
- Biostatistics and Bioinformatics Branch (HNT72), Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1830, USA
| | - Anil B. Mukherjee
- Section on Developmental Genetics, Division of Translational Medicine, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1830, USA
| |
Collapse
|
3
|
İnci A, Dökmeci S. Extracellular chaperones in lysosomal storage diseases. Mol Genet Metab 2025; 145:109086. [PMID: 40106871 DOI: 10.1016/j.ymgme.2025.109086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/23/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Lysosomal storage disorders (LSDs) are a diverse group of inherited metabolic disorders characterized by the accumulation of undegraded substrates within lysosomes due to defective lysosomal function. Recent research has highlighted the pivotal role of extracellular chaperones in the pathophysiology of LSDs, revealing their crucial involvement in modulating disease progression. These chaperones aid in stabilizing and refolding misfolded lysosomal enzymes, enhancing their proper trafficking and function, which in turn reduces substrate accumulation. Furthermore, extracellular chaperones have emerged as promising biomarkers, with their levels in bodily fluids offering potential for disease diagnosis and monitoring. This review explores the current understanding of extracellular chaperones in the context of LSDs, examining their mechanisms of action, biomarker and therapeutic potential, and future directions in clinical application of LSDs.
Collapse
Affiliation(s)
- Aslı İnci
- Gazi University School of Medicine, Department of Pediatric Metabolism, Ankara, Turkey; Hacettepe University School of Medicine, Department of Medical Biology, Ankara, Turkey.
| | - Serap Dökmeci
- Hacettepe University School of Medicine, Department of Medical Biology, Ankara, Turkey
| |
Collapse
|
4
|
Alvarez-Valadez K, Sauvat A, Diharce J, Leduc M, Stoll G, Guittat L, Lambertucci F, Paillet J, Motiño O, Ferret L, Muller A, Forveille S, Maiuri MC, Kepp O, de Brevern AG, Wodrich H, Pol JG, Kroemer G, Djavaheri-Mergny M. Lysosomal damage due to cholesterol accumulation triggers immunogenic cell death. Autophagy 2025; 21:934-956. [PMID: 39663580 PMCID: PMC12013445 DOI: 10.1080/15548627.2024.2440842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024] Open
Abstract
Cholesterol serves as a vital lipid that regulates numerous physiological processes. Nonetheless, its role in regulating cell death processes remains incompletely understood. In this study, we investigated the role of cholesterol trafficking in immunogenic cell death. Through cell-based drug screening, we identified two antidepressants, sertraline and indatraline, as potent inducers of the nuclear translocation of TFEB (transcription factor EB). Activation of TFEB was mediated through the autophagy-independent lipidation of MAP1LC3/LC3 (microtubule associated protein 1 light chain 3). Both compounds promoted cholesterol accumulation within lysosomes, resulting in lysosomal membrane permeabilization, disruption of autophagy and cell death that could be reversed by cholesterol depletion. Molecular docking analysis indicated that sertraline and indatraline have the potential to inhibit cholesterol binding to the lysosomal cholesterol transporters, NPC1 (NPC intracellular cholesterol transporter 1) and NPC2. This inhibitory effect might be further enhanced by the upregulation of NPC1 and NPC2 expression by TFEB. Both antidepressants also upregulated PLA2G15 (phospholipase A2 group XV), an enzyme that elevates lysosomal cholesterol. In cancer cells, sertraline and indatraline elicited immunogenic cell death, converting dying cells into prophylactic vaccines that were able to confer protection against tumor growth in mice. In a therapeutic setting, a single dose of each compound was sufficient to significantly reduce the outgrowth of established tumors in a T-cell-dependent manner. These results identify sertraline and indatraline as immunostimulatory agents for cancer treatment. More generally, this research shed light on novel therapeutic avenues harnessing lysosomal cholesterol transport to regulate immunogenic cell death.Abbreviation: ATG5: autophagy related 5; ATG13: autophagy related 13; DKO: double knockout; ICD: immunogenic cell death; KO: knockout; LAMP1: lysosomal associated membrane protein 1; LAMP2: lysosomal associated membrane protein 2; LGALS3: galectin 3; LDL: low-density lipoprotein; LMP: lysosomal membrane permeabilization; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTX: mitoxantrone; NPC1: NPC intracellular cholesterol transporter 1; NPC2: NPC intracellular cholesterol transporter 2; TFE3: transcription factor E3; TFEB: transcription factor EB; ULK1: unc-51 like autophagy activating kinase 1.
Collapse
Affiliation(s)
- Karla Alvarez-Valadez
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Équipe labellisée par la Ligue contre le Cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
- Faculté de Médecine, Université Paris Saclay, Paris, France
| | - Allan Sauvat
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Équipe labellisée par la Ligue contre le Cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Julien Diharce
- Université Paris Cité and Université de la Réunion, INSERM UMRS 1134, BIGR, DSIMB Bioinformatics team, Paris, France
| | - Marion Leduc
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Équipe labellisée par la Ligue contre le Cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Gautier Stoll
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Équipe labellisée par la Ligue contre le Cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Lionel Guittat
- Laboratoire d’Optique et Biosciences, École Polytechnique, CNRS UMR7645, INSERM U1182, Institut Polytechnique de Paris, Palaiseau, France
- Santé, Médecine, Biologie Humaine (SMBH), Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Flavia Lambertucci
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Équipe labellisée par la Ligue contre le Cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Juliette Paillet
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Équipe labellisée par la Ligue contre le Cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Omar Motiño
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Équipe labellisée par la Ligue contre le Cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Lucille Ferret
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Équipe labellisée par la Ligue contre le Cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
- Faculté de Médecine, Université Paris Saclay, Paris, France
| | - Alexandra Muller
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Équipe labellisée par la Ligue contre le Cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Sabrina Forveille
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Équipe labellisée par la Ligue contre le Cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Maria Chiara Maiuri
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Équipe labellisée par la Ligue contre le Cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Oliver Kepp
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Équipe labellisée par la Ligue contre le Cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Alexandre G de Brevern
- Université Paris Cité and Université de la Réunion, INSERM UMRS 1134, BIGR, DSIMB Bioinformatics team, Paris, France
| | - Harald Wodrich
- CNRS UMR 5234, Fundamental Microbiology and Pathogenicity, Université de Bordeaux, Bordeaux, France
| | - Jonathan G Pol
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Équipe labellisée par la Ligue contre le Cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Équipe labellisée par la Ligue contre le Cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
- Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Institut du Cancer Paris CARPEM, Paris, France
| | - Mojgan Djavaheri-Mergny
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Équipe labellisée par la Ligue contre le Cancer, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Villejuif, France
| |
Collapse
|
5
|
Marcó S, Muñoz S, Bosch F, Jimenez V. Rat models of musculoskeletal lysosomal storage disorders and their role in pre-clinical evaluation of gene therapy approaches. Mamm Genome 2025:10.1007/s00335-025-10121-3. [PMID: 40100425 DOI: 10.1007/s00335-025-10121-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/05/2025] [Indexed: 03/20/2025]
Abstract
Mice have been a cornerstone of biomedical research for decades for studying a wide range of biological processes, disease mechanisms, and the assessment of therapies. Moreover, mice present several practical advantages such as small size, low cost and ease of genetic manipulation. While mice offer numerous benefits, for certain disease areas, rat models provide a closer representation of human disease progression, offering better insights for translational research and therapeutic development. This closer resemblance is particularly important for research focusing on diseases involving the cardiovascular and musculoskeletal system. In rats, the pathophysiology of these diseases mirrors the clinical alterations observed in humans. This review focuses on the key phenotypic differences between mouse and rat models of lysosomal storage disorders that specifically manifest with cardiac, skeletal muscle, and bone and joint involvement (Pompe and Danon diseases, and Maroteaux-Lamy and Morquio A syndromes). Furthermore, we discuss the therapeutic potential of various adeno-associated viral vector-mediated gene therapies that have been evaluated in these rat models, highlighting their contributions to advancing treatment options for these debilitating conditions.
Collapse
Affiliation(s)
- Sara Marcó
- Center of Animal Biotechnology and Gene Therapy, Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Sergio Muñoz
- Center of Animal Biotechnology and Gene Therapy, Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Fatima Bosch
- Center of Animal Biotechnology and Gene Therapy, Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Veronica Jimenez
- Center of Animal Biotechnology and Gene Therapy, Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| |
Collapse
|
6
|
Bonacina F, Zhang X, Manel N, Yvan-Charvet L, Razani B, Norata GD. Lysosomes in the immunometabolic reprogramming of immune cells in atherosclerosis. Nat Rev Cardiol 2025; 22:149-164. [PMID: 39304748 PMCID: PMC11835540 DOI: 10.1038/s41569-024-01072-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/08/2024] [Indexed: 09/22/2024]
Abstract
Lysosomes have a central role in the disposal of extracellular and intracellular cargo and also function as metabolic sensors and signalling platforms in the immunometabolic reprogramming of macrophages and other immune cells in atherosclerosis. Lysosomes can rapidly sense the presence of nutrients within immune cells, thereby switching from catabolism of extracellular material to the recycling of intracellular cargo. Such a fine-tuned degradative response supports the generation of metabolic building blocks through effectors such as mTORC1 or TFEB. By coupling nutrients to downstream signalling and metabolism, lysosomes serve as a crucial hub for cellular function in innate and adaptive immune cells. Lysosomal dysfunction is now recognized to be a hallmark of atherogenesis. Perturbations in nutrient-sensing and signalling have profound effects on the capacity of immune cells to handle cholesterol, perform phagocytosis and efferocytosis, and limit the activation of the inflammasome and other inflammatory pathways. Strategies to improve lysosomal function hold promise as novel modulators of the immunoinflammatory response associated with atherosclerosis. In this Review, we describe the crosstalk between lysosomal biology and immune cell function and polarization, with a particular focus on cellular immunometabolic reprogramming in the context of atherosclerosis.
Collapse
Affiliation(s)
- Fabrizia Bonacina
- Department of Excellence of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| | - Xiangyu Zhang
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA
- Pittsburgh VA Medical Center, Pittsburgh, PA, USA
| | - Nicolas Manel
- Immunity and Cancer Department, Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU), Oncoage, Nice, France
| | - Babak Razani
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA
- Pittsburgh VA Medical Center, Pittsburgh, PA, USA
| | - Giuseppe D Norata
- Department of Excellence of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
7
|
Canini G, Mazzinelli E, Nocca G, Lattanzi W, Arcovito A. Targeting Glucosylceramide Synthase: Innovative Drug Repurposing Strategies for Lysosomal Diseases. Int J Mol Sci 2025; 26:2195. [PMID: 40076817 PMCID: PMC11900012 DOI: 10.3390/ijms26052195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Sphingolipidoses, a subgroup of lysosomal storage diseases (LSDs), are rare and debilitating disorders caused by defects in sphingolipid metabolism. Despite advancements in treatment, therapeutic options remain limited. Miglustat, a glucosylceramide synthase EC 2.4.1.80 (GCS) inhibitor, is one of the few available pharmacological treatments; however, it is associated with significant adverse effects that impact patients' quality of life. Drug repurposing offers a promising strategy to identify new therapeutic agents from approved drugs, expanding treatment options for rare diseases with limited therapeutic alternatives. This study aims to identify potential alternative inhibitors of GCS through a drug-repurposing approach, using computational and experimental methods to assess their therapeutic potential for sphingolipidoses. A library of approved drugs was screened using advanced computational techniques, including molecular docking, molecular dynamics simulations, and metadynamics, to identify potential GCS inhibitors. Promising candidates were selected for further in vitro validation to evaluate their inhibitory activity and potential as therapeutic alternatives to Miglustat. Computational screening identified several potential GCS inhibitors, with Dapagliflozin emerging as the most promising candidate. Experimental validation confirmed its efficacy, revealing a complementary mechanism of action to Miglustat while potentially offering a more favorable side effect profile. This study underscores the utility of computational and experimental methodologies in drug repurposing for rare diseases. The identification of Dapagliflozin as a potential GCS inhibitor provides a foundation for further preclinical and clinical evaluation, supporting its potential application in the treatment of sphingolipidoses.
Collapse
Affiliation(s)
- Giorgia Canini
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (G.C.); (E.M.); (G.N.)
| | - Elena Mazzinelli
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (G.C.); (E.M.); (G.N.)
| | - Giuseppina Nocca
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (G.C.); (E.M.); (G.N.)
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy;
| | - Wanda Lattanzi
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy;
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Alessandro Arcovito
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (G.C.); (E.M.); (G.N.)
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy;
| |
Collapse
|
8
|
Zwi-Dantsis L, Mohamed S, Massaro G, Moeendarbary E. Adeno-Associated Virus Vectors: Principles, Practices, and Prospects in Gene Therapy. Viruses 2025; 17:239. [PMID: 40006994 PMCID: PMC11861813 DOI: 10.3390/v17020239] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/27/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Gene therapy offers promising potential as an efficacious and long-lasting therapeutic option for genetic conditions, by correcting defective mutations using engineered vectors to deliver genetic material to host cells. Among these vectors, adeno-associated viruses (AAVs) stand out for their efficiency, versatility, and safety, making them one of the leading platforms in gene therapy. The enormous potential of AAVs has been demonstrated through their use in over 225 clinical trials and the FDA's approval of six AAV-based gene therapy products, positioning these vectors at the forefront of the field. This review highlights the evolution and current applications of AAVs in gene therapy, focusing on their clinical successes, ongoing developments, and the manufacturing processes required for the rapid commercial growth anticipated in the AAV therapy market. It also discusses the broader implications of these advancements for future therapeutic strategies targeting more complex and multi-systemic conditions and biological processes such as aging. Finally, we explore some of the major challenges currently confronting the field.
Collapse
Affiliation(s)
- Limor Zwi-Dantsis
- Department of Mechanical Engineering, Roberts Building, University College London, London WC1E 6BT, UK
| | - Saira Mohamed
- Department of Mechanical Engineering, Roberts Building, University College London, London WC1E 6BT, UK
| | - Giulia Massaro
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Emad Moeendarbary
- Department of Mechanical Engineering, Roberts Building, University College London, London WC1E 6BT, UK
| |
Collapse
|
9
|
Biddeci G, Spinelli G, Colomba P, Duro G, Anania M, Francofonte D, Di Blasi F. Fabry Disease and Inflammation: Potential Role of p65 iso5, an Isoform of the NF-κB Complex. Cells 2025; 14:230. [PMID: 39937021 PMCID: PMC11817417 DOI: 10.3390/cells14030230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/24/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Fabry disease (FD) is an X-linked lysosomal storage disease, caused by mutations in the GLA gene on the X chromosome, resulting in a deficiency of the lysosomal enzyme α-GAL. This leads to the progressive accumulation of Gb3 in cells, causing multi-systemic effects. FD has been classified as a subgroup of autoinflammatory diseases. NF-κB is a family of ubiquitous and inducible transcription factors that play critical roles in inflammation, in which the p65/p50 heterodimer is the most abundant. The glucocorticoid receptor (GR) represents the physiological antagonists in the inflammation process. A novel spliced variant of p65, named p65 iso5, which can bind the dexamethasone, enhancing GR activity, has been found. This study investigates the potential role of p65 iso5 in the inflammation of subjects with FD. We evaluated in peripheral blood mononuclear cells (PBMCs), from over 100 FD patients, the p65 iso5 mRNA level, and the protein expression. The results showed significantly lower p65 iso5 mRNA and protein expression levels compared to controls. These findings, along with the ability of p65 iso5 to bind dexamethasone and the regulation of the glucocorticoid response in the opposite way of p65, strongly suggest the involvement of p65 iso5 in the inflammatory response in FD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Francesco Di Blasi
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Via Ugo La Malfa 153, 90146 Palermo, Italy; (G.B.); (G.S.); (P.C.); (G.D.); (M.A.); (D.F.)
| |
Collapse
|
10
|
Parvatam S, Pistollato F, Marshall LJ, Furtmann F, Jahagirdar D, Chakraborty Choudhury M, Mohanty S, Mittal H, Meganathan S, Mishra R. Human-based complex in vitro models: their promise and potential for rare disease therapeutics. Front Cell Dev Biol 2025; 13:1526306. [PMID: 39931243 PMCID: PMC11807990 DOI: 10.3389/fcell.2025.1526306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025] Open
Abstract
Rare diseases affect a small percentage of an individual country's population; however, with over 7,000 in total, rare diseases represent a significant disease burden impacting up to 10% of the world's population. Despite this, there are no approved treatments for almost 95% of rare diseases, and the existing treatments are cost-intensive for the patients. More than 70% of rare diseases are genetic in nature, with patient-specific mutations. This calls for the need to have personalised and patient-specific preclinical models that can lead to effective, speedy, and affordable therapeutic options. Complex in vitro models (CIVMs), including those using induced pluripotent stem cells (iPSCs), organoids, and organs-on-chips are emerging as powerful human-based pre-clinical systems with the capacity to provide efficacy data enabling drugs to move into clinical trials. In this narrative review, we discuss how CIVMs are providing insights into biomedical research on rare diseases. We also discuss how these systems are being used in clinical trials to develop efficacy models for rare diseases. Finally, we propose recommendations on how human relevant CIVMs could be leveraged to increase translatability of basic, applied and nonclinical research outcomes in the field of rare disease therapeutics in developed as well as middle-and low-income countries.
Collapse
Affiliation(s)
- Surat Parvatam
- Department of Research and Toxicology, Humane Society International/India, Hyderabad, India
| | - Francesca Pistollato
- Department of Research and Toxicology, Humane Society International/Europe, Brussels, Belgium
| | - Lindsay J. Marshall
- Animal Research Issues, The Humane Society of the United States, Washington DC, DC, United States
| | - Fabia Furtmann
- Department of Research and Toxicology, Humane Society International/Europe, Brussels, Belgium
| | - Devashree Jahagirdar
- Department of Chemical Engineering, Indian Institute of Technology (IIT), Mumbai, Maharashtra, India
| | | | - Sujata Mohanty
- Stem Cell Facility (DBT-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, New Delhi, India
| | - Harshita Mittal
- Department of Research and Toxicology, Humane Society International/India, Hyderabad, India
| | - Saveetha Meganathan
- Community Engagement and Policy Stewardship, Tata Institute for Genetics and Society, Bangalore, India
| | - Rakesh Mishra
- Tata Institute for Genetics and Society, Bangalore, India
| |
Collapse
|
11
|
Talebi SF, Seify M, Bhandari RK, Shoorei H, Oskuei SD. Fluoride-induced testicular and ovarian toxicity: evidence from animal studies. Biol Res 2025; 58:6. [PMID: 39863878 PMCID: PMC11762501 DOI: 10.1186/s40659-025-00586-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Fluoride (F), as a natural element found in a wide range of sources such as water and certain foods, has been proven to be beneficial in preventing dental caries, but concerns have been raised regarding its potential deleterious effects on overall health. Sodium fluoride (NaF), another form of F, has the ability to accumulate in reproductive organs and interfere with hormonal regulation and oxidative stress pathways, contributing to reproductive toxicity. While the exact mechanisms of F-induced reproductive toxicity are not fully understood, this review aims to elucidate the mechanisms involved in testicular and ovarian injury. In males, F exposure at different doses has been associated with reduced testis weight, reduced sperm quality in terms of count, motility, and viability, as well as abnormal sperm morphology and disruption of seminiferous tubules by altering hormone levels (especially testosterone), impairing spermatogenesis, and inducing oxidative stress and zinc deficiency. Similarly, administration of F can impact female reproductive health by affecting ovarian function, hormone levels, oocyte quality, and the regularity of the estrous cycle. However, the impact of F exposure on LH, FSH, and GnRH levels is controversial between males and females. In both males and females, F exerts its adverse effects by triggering apoptosis, autophagy, inflammation, mitochondrial dysfunction, reduction in ATP synthesis, and modulation of important genes involved in steroidogenesis. Furthermore, genetic susceptibility and individual variations in F metabolism may contribute to different responses to fluoride exposure.
Collapse
Affiliation(s)
| | - Mohammad Seify
- Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ramji Kumar Bhandari
- Division of Biological Sciences, University of Missouri, Columbia, MO, 65211, USA.
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Shahram Dabiri Oskuei
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
12
|
Wang Z, Li X, Moura AK, Hu JZ, Wang YT, Zhang Y. Lysosome Functions in Atherosclerosis: A Potential Therapeutic Target. Cells 2025; 14:183. [PMID: 39936975 PMCID: PMC11816498 DOI: 10.3390/cells14030183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 02/13/2025] Open
Abstract
Lysosomes in mammalian cells are recognized as key digestive organelles, containing a variety of hydrolytic enzymes that enable the processing of both endogenous and exogenous substrates. These organelles digest various macromolecules and recycle them through the autophagy-lysosomal system. Recent research has expanded our understanding of lysosomes, identifying them not only as centers of degradation but also as crucial regulators of nutrient sensing, immunity, secretion, and other vital cellular functions. The lysosomal pathway plays a significant role in vascular regulation and is implicated in diseases such as atherosclerosis. During atherosclerotic plaque formation, macrophages initially engulf large quantities of lipoproteins, triggering pathogenic responses that include lysosomal dysfunction, foam cell formation, and subsequent atherosclerosis development. Lysosomal dysfunction, along with the inefficient degradation of apoptotic cells and the accumulation of modified low-density lipoproteins, negatively impacts atherosclerotic lesion progression. Recent studies have highlighted that lysosomal dysfunction contributes critically to atherosclerosis in a cell- and stage-specific manner. In this review, we discuss the mechanisms of lysosomal biogenesis and its regulatory role in atherosclerotic lesions. Based on these lysosomal functions, we propose that targeting lysosomes could offer a novel therapeutic approach for atherosclerosis, shedding light on the connection between lysosomal dysfunction and disease progression while offering new insights into potential anti-atherosclerotic strategies.
Collapse
Affiliation(s)
- Zhengchao Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (Z.W.); (A.K.M.); (J.Z.H.); (Y.-T.W.)
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou 350007, China
| | - Xiang Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (Z.W.); (A.K.M.); (J.Z.H.); (Y.-T.W.)
| | - Alexandra K. Moura
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (Z.W.); (A.K.M.); (J.Z.H.); (Y.-T.W.)
| | - Jenny Z. Hu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (Z.W.); (A.K.M.); (J.Z.H.); (Y.-T.W.)
| | - Yun-Ting Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (Z.W.); (A.K.M.); (J.Z.H.); (Y.-T.W.)
| | - Yang Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (Z.W.); (A.K.M.); (J.Z.H.); (Y.-T.W.)
| |
Collapse
|
13
|
Lerussi G, Villagrasa-Araya V, Moltó-Abad M, del Toro M, Pintos-Morell G, Seras-Franzoso J, Abasolo I. Extracellular Vesicles as Tools for Crossing the Blood-Brain Barrier to Treat Lysosomal Storage Diseases. Life (Basel) 2025; 15:70. [PMID: 39860010 PMCID: PMC11766495 DOI: 10.3390/life15010070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/03/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Extracellular vesicles (EVs) are nanosized, membrane-bound structures that have emerged as promising tools for drug delivery, especially in the treatment of lysosomal storage disorders (LSDs) with central nervous system (CNS) involvement. This review highlights the unique properties of EVs, such as their biocompatibility, capacity to cross the blood-brain barrier (BBB), and potential for therapeutic cargo loading, including that of enzymes and genetic material. Current therapies for LSDs, like enzyme replacement therapy (ERT), often fail to address neurological symptoms due to their inability to cross the BBB. EVs offer a viable alternative, allowing for targeted delivery to the CNS and improving therapeutic outcomes. We discuss recent advancements in the engineering and modification of EVs to enhance targeting, circulation time and cargo stability, and provide a detailed overview of their application in LSDs, such as Gaucher and Fabry diseases, and Sanfilippo syndrome. Despite their potential, challenges remain in scaling production, ensuring isolation purity, and meeting regulatory requirements. Future developments will focus on overcoming these barriers, paving the way for the clinical translation of EV-based therapies in LSDs and other CNS disorders.
Collapse
Affiliation(s)
- Giovanni Lerussi
- Clinical Biochemistry, Drug Delivery & Therapy (CB-DDT), Vall d’Hebron Institute of Research (VHIR), 08035 Barcelona, Spain; (G.L.); (V.V.-A.); (M.M.-A.); (G.P.-M.); (J.S.-F.)
| | - Verónica Villagrasa-Araya
- Clinical Biochemistry, Drug Delivery & Therapy (CB-DDT), Vall d’Hebron Institute of Research (VHIR), 08035 Barcelona, Spain; (G.L.); (V.V.-A.); (M.M.-A.); (G.P.-M.); (J.S.-F.)
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08035 Barcelona, Spain
- Institute of Advanced Chemistry of Catalonia (IQAC), Centro Superior de Investigaciones Científicas (CSIC), 08034 Barcelona, Spain
| | - Marc Moltó-Abad
- Clinical Biochemistry, Drug Delivery & Therapy (CB-DDT), Vall d’Hebron Institute of Research (VHIR), 08035 Barcelona, Spain; (G.L.); (V.V.-A.); (M.M.-A.); (G.P.-M.); (J.S.-F.)
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08035 Barcelona, Spain
| | - Mireia del Toro
- Pediatric Neurology Unit, Hospital Universitari Vall d’Hebron and MetabERN, 08035 Barcelona, Spain;
- Networking Research Center on Rare Diseases (CIBERER), 08035 Barcelona, Spain
| | - Guillem Pintos-Morell
- Clinical Biochemistry, Drug Delivery & Therapy (CB-DDT), Vall d’Hebron Institute of Research (VHIR), 08035 Barcelona, Spain; (G.L.); (V.V.-A.); (M.M.-A.); (G.P.-M.); (J.S.-F.)
| | - Joaquin Seras-Franzoso
- Clinical Biochemistry, Drug Delivery & Therapy (CB-DDT), Vall d’Hebron Institute of Research (VHIR), 08035 Barcelona, Spain; (G.L.); (V.V.-A.); (M.M.-A.); (G.P.-M.); (J.S.-F.)
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08035 Barcelona, Spain
| | - Ibane Abasolo
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 08035 Barcelona, Spain
- Institute of Advanced Chemistry of Catalonia (IQAC), Centro Superior de Investigaciones Científicas (CSIC), 08034 Barcelona, Spain
| |
Collapse
|
14
|
Zhang H, Wang Y, Wang R, Zhang X, Chen H. TRPML1 agonist ML-SA5 mitigates uranium-induced nephrotoxicity via promoting lysosomal exocytosis. Biomed Pharmacother 2024; 181:117728. [PMID: 39647321 DOI: 10.1016/j.biopha.2024.117728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/24/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024] Open
Abstract
Uranium (U) released from U mining and spent nuclear fuel reprocessing in the nuclear industry, nuclear accidents and military activities as a primary environmental pollutant (e.g., drinking water pollution) is a threat to human health. Kidney is one of the main target organs for U accumulation, leading to nephrotoxicity mainly associated with the injuries in proximal tubular epithelial cells (PTECs). Transient receptor potential mucolipin 1 (TRPML1) is a novel therapeutic target for nephrotoxicity caused by acute or chronic U poisoning. We herein investigate the therapeutic efficacy of ML-SA5, a small molecule agonist of TRPML1, in U-induced nephrotoxicity in acute U intoxicated mice. We demonstrate that delayed treatment with ML-SA5 enhances U clearance from the kidneys via urine excretion by activating lysosomal exocytosis, and thereby attenuates U-induced kidney dysfunction and cell death/apoptosis of renal PTECs in acute U intoxicated mice. In addition, ML-SA5 promotes the nuclear translocation of transcription factor EB (TFEB) in renal PTECs in acute U intoxicated mice. Mechanistically, ML-SA5 triggers the TRPML1-mediated lysosomal calcium release and consequently induces TFEB activation in U-loaded renal PTECs-derived HK-2 cells. Moreover, knockdown of TRPML1 or TFEB abolishes the effects of ML-SA5 on the removal of intracellular U and reduction of the cellular injury/death in U-loaded HK-2 cells. Our findings indicate that pharmacological activation of TRPML1 is a promising therapeutic approach for the delayed treatment of U-induced nephrotoxicity via the activation of the positive feedback loop of TRPML1 and TFEB and consequent the induction of lysosomal exocytosis.
Collapse
Affiliation(s)
- Hongjing Zhang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, No. 2094, Xie-Tu Road, Shanghai 200032, PR China
| | - Yifei Wang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, No. 2094, Xie-Tu Road, Shanghai 200032, PR China
| | - Ruiyun Wang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, No. 2094, Xie-Tu Road, Shanghai 200032, PR China
| | - Xuxia Zhang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, No. 2094, Xie-Tu Road, Shanghai 200032, PR China
| | - Honghong Chen
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, No. 2094, Xie-Tu Road, Shanghai 200032, PR China.
| |
Collapse
|
15
|
Yang S, Chen K, Yu J, Jin Z, Zhang M, Li Z, Yu Y, Xuan N, Tian B, Li N, Mao Z, Wang W, Chen T, Wu Y, Zhao Y, Zhang M, Fei X, Ying S, Li W, Yan F, Zhang X, Zhang G, Shen H, Chen Z. Inhibition of cathepsin L ameliorates inflammation through the A20/NF-κB pathway in endotoxin-induced acute lung injury. iScience 2024; 27:111024. [PMID: 39559762 PMCID: PMC11570319 DOI: 10.1016/j.isci.2024.111024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 08/07/2024] [Accepted: 09/20/2024] [Indexed: 11/20/2024] Open
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a severe inflammatory condition that remains refractory; however, its molecular mechanisms are largely unknown. Previous studies have shown numerous compounds containing 4-indolyl-2-aminopyrimidine that display strong anti-inflammatory properties. In our research, we identified that a 4-Indole-2-Arylaminopyrimidine derivative named "IAAP" suppressed lipopolysaccharide (LPS)-induced inflammation. Immunoprecipitation and liquid chromatography-tandem mass spectrometry (LC-MS/MS) identified that IAAP interacts with a lysosomal cysteine protease, cathepsin L (CTSL), and restrains its activity. The nuclear factor kappa B (NF-κB) family plays a central role in controlling innate immunity. Canonical NF-κB activation, such as stimulation with lipopolysaccharide (LPS), typically involves the degradation of A20. We observed that IAAP suppression of CTSL prevented the LPS-induced degradation of A20, thereby ameliorating NF-κB activation. This study identifies CTSL as a crucial regulator of A20/NF-κB signaling and suggests IAAP as a potential lead compound for developing drugs to treat ALI/ARDS.
Collapse
Affiliation(s)
- Shiyi Yang
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Kaijun Chen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Jinkang Yu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Zhangchu Jin
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Min Zhang
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Zhouyang Li
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Yang Yu
- Department of Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Nanxia Xuan
- Department of Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Baoping Tian
- Department of Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Na Li
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Zhengtong Mao
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Wenbing Wang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Tianpeng Chen
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yinfang Wu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Yun Zhao
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Min Zhang
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Xia Fei
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Songmin Ying
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu 322000, China
- Department of Pharmacology & Department of Respiratory and Critical Care Medicine of the Second Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Respiratory Disease of Zhejiang Province, Hangzhou 310009, China
| | - Wen Li
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Fugui Yan
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Xingxian Zhang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Gensheng Zhang
- Department of Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Huahao Shen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
- State Key Lab of Respiratory Disease, Key Cite of National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Zhihua Chen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| |
Collapse
|
16
|
Mysior MM, Simpson JC. An automated high-content screening and assay platform for the analysis of spheroids at subcellular resolution. PLoS One 2024; 19:e0311963. [PMID: 39531451 PMCID: PMC11556727 DOI: 10.1371/journal.pone.0311963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024] Open
Abstract
The endomembrane system is essential for healthy cell function, with the various compartments carrying out a large number of specific biochemical reactions. To date, almost all of our understanding of the endomembrane system has come from the study of cultured cells growing as monolayers. However, monolayer-grown cells only poorly represent the environment encountered by cells in the human body. As a first step to address this disparity, we have developed a platform that allows us to investigate and quantify changes to the endomembrane system in three-dimensional (3D) cell models, in an automated and highly systematic manner. HeLa Kyoto cells were grown on custom-designed micropatterned 96-well plates to facilitate spheroid assembly in the form of highly uniform arrays. Fully automated high-content confocal imaging and analysis were then carried out, allowing us to measure various spheroid-, cellular- and subcellular-level parameters relating to size and morphology. Using two drugs known to perturb endomembrane function, we demonstrate that cell-based assays can be carried out in these spheroids, and that changes to the Golgi apparatus and endosomes can be quantified from individual cells within the spheroids. We also show that image texture measurements are useful tools to discriminate cellular phenotypes. The automated platform that we show here has the potential to be scaled up, thereby allowing large-scale robust screening to be carried out in 3D cell models.
Collapse
Affiliation(s)
- Margaritha M. Mysior
- Cell Screening Laboratory, UCD School of Biology & Environmental Science, University College Dublin, Dublin, Ireland
| | - Jeremy C. Simpson
- Cell Screening Laboratory, UCD School of Biology & Environmental Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
17
|
Ghoochani A, Heiby JC, Rawat ES, Medoh UN, Di Fraia D, Dong W, Gastou M, Nyame K, Laqtom NN, Gomez-Ospina N, Ori A, Abu-Remaileh M. Cell-Type Resolved Protein Atlas of Brain Lysosomes Identifies SLC45A1-Associated Disease as a Lysosomal Disorder. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618295. [PMID: 39464040 PMCID: PMC11507716 DOI: 10.1101/2024.10.14.618295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Mutations in lysosomal genes cause neurodegeneration and neurological lysosomal storage disorders (LSDs). Despite their essential role in brain homeostasis, the cell-type-specific composition and function of lysosomes remain poorly understood. Here, we report a quantitative protein atlas of the lysosome from mouse neurons, astrocytes, oligodendrocytes, and microglia. We identify dozens of novel lysosomal proteins and reveal the diversity of the lysosomal composition across brain cell types. Notably, we discovered SLC45A1, mutations in which cause a monogenic neurological disease, as a neuron-specific lysosomal protein. Loss of SLC45A1 causes lysosomal dysfunction in vitro and in vivo. Mechanistically, SLC45A1 plays a dual role in lysosomal sugar transport and stabilization of V1 subunits of the V-ATPase. SLC45A1 deficiency depletes the V1 subunits, elevates lysosomal pH, and disrupts iron homeostasis causing mitochondrial dysfunction. Altogether, our work redefines SLC45A1-associated disease as a LSD and establishes a comprehensive map to study lysosome biology at cell-type resolution in the brain and its implications for neurodegeneration.
Collapse
Affiliation(s)
- Ali Ghoochani
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network; Chevy Chase, MD, 20815, USA
- These authors contributed equally
| | - Julia C. Heiby
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI) e.V., Jena, Germany
- These authors contributed equally
| | - Eshaan S. Rawat
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network; Chevy Chase, MD, 20815, USA
| | - Uche N. Medoh
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network; Chevy Chase, MD, 20815, USA
- Current affiliation: Arc Institute, Palo Alto, CA 94304, USA
| | - Domenico Di Fraia
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI) e.V., Jena, Germany
- Current affiliation: Department of Biology, University of Rochester, Rochester, NY, USA
| | - Wentao Dong
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network; Chevy Chase, MD, 20815, USA
| | - Marc Gastou
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Kwamina Nyame
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network; Chevy Chase, MD, 20815, USA
| | - Nouf N. Laqtom
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network; Chevy Chase, MD, 20815, USA
| | - Natalia Gomez-Ospina
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Alessandro Ori
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI) e.V., Jena, Germany
- Co-senior authors
| | - Monther Abu-Remaileh
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network; Chevy Chase, MD, 20815, USA
- The Phil & Penny Knight Initiative for Brain Resilience at the Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
- Co-senior authors
- Lead author
| |
Collapse
|
18
|
Chen J, Wen P, Tang YH, Li H, Wang Z, Wang X, Zhou X, Gao XD, Fujita M, Yang G. Proteome and Glycoproteome Analyses Reveal Regulation of Protein Glycosylation Site-Specific Occupancy and Lysosomal Hydrolase Maturation by N-Glycan-Dependent ER-Quality Control. J Proteome Res 2024; 23:4409-4421. [PMID: 39235835 DOI: 10.1021/acs.jproteome.4c00378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
N-Glycan-dependent endoplasmic reticulum quality control (ERQC) primarily mediates protein folding, which determines the fate of the polypeptide. Monoglucose residues on N-glycans determine whether the nascent N-glycosylated proteins enter into and escape from the calnexin (CANX)/calreticulin (CALR) cycle, which is a central system of the ERQC. To reveal the impact of ERQC on glycosylation and protein fate, we performed comprehensive quantitative proteomic and glycoproteomic analyses using cells defective in N-glycan-dependent ERQC. Deficiency of MOGS encoding the ER α-glucosidase I, CANX, or/and CALR broadly affected protein expression and glycosylation. Among the altered glycoproteins, the occupancy of oligomannosidic N-glycans was significantly affected. Besides the expected ER stress, proteins and glycoproteins involved in pathways for lysosome and viral infection are differentially changed in those deficient cells. We demonstrated that lysosomal hydrolases were not correctly modified with mannose-6-phosphates on the N-glycans and were directly secreted to the culture medium in N-glycan-dependent ERQC mutant cells. Overall, the CANX/CALR cycle promotes the correct folding of glycosylated peptides and influences the transport of lysosomal hydrolases.
Collapse
Affiliation(s)
- Jingru Chen
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China
| | - Piaopiao Wen
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yu-He Tang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hanjie Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China
| | - Zibo Wang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiuyuan Wang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaoman Zhou
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xiao-Dong Gao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China
| | - Morihisa Fujita
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu 501-1193, Japan
| | - Ganglong Yang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
19
|
Ferrari V, Tedesco B, Cozzi M, Chierichetti M, Casarotto E, Pramaggiore P, Cornaggia L, Mohamed A, Patelli G, Piccolella M, Cristofani R, Crippa V, Galbiati M, Poletti A, Rusmini P. Lysosome quality control in health and neurodegenerative diseases. Cell Mol Biol Lett 2024; 29:116. [PMID: 39237893 PMCID: PMC11378602 DOI: 10.1186/s11658-024-00633-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024] Open
Abstract
Lysosomes are acidic organelles involved in crucial intracellular functions, including the degradation of organelles and protein, membrane repair, phagocytosis, endocytosis, and nutrient sensing. Given these key roles of lysosomes, maintaining their homeostasis is essential for cell viability. Thus, to preserve lysosome integrity and functionality, cells have developed a complex intracellular system, called lysosome quality control (LQC). Several stressors may affect the integrity of lysosomes, causing Lysosomal membrane permeabilization (LMP), in which membrane rupture results in the leakage of luminal hydrolase enzymes into the cytosol. After sensing the damage, LQC either activates lysosome repair, or induces the degradation of the ruptured lysosomes through autophagy. In addition, LQC stimulates the de novo biogenesis of functional lysosomes and lysosome exocytosis. Alterations in LQC give rise to deleterious consequences for cellular homeostasis. Specifically, the persistence of impaired lysosomes or the malfunctioning of lysosomal processes leads to cellular toxicity and death, thereby contributing to the pathogenesis of different disorders, including neurodegenerative diseases (NDs). Recently, several pieces of evidence have underlined the importance of the role of lysosomes in NDs. In this review, we describe the elements of the LQC system, how they cooperate to maintain lysosome homeostasis, and their implication in the pathogenesis of different NDs.
Collapse
Affiliation(s)
- Veronica Ferrari
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Barbara Tedesco
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Marta Cozzi
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Marta Chierichetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Elena Casarotto
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Paola Pramaggiore
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Laura Cornaggia
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Ali Mohamed
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Guglielmo Patelli
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Margherita Piccolella
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Riccardo Cristofani
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Valeria Crippa
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Mariarita Galbiati
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy.
| | - Paola Rusmini
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università degli Studi di Milano, Dipartimento Di Eccellenza, 2018-2027, Milan, Italy
| |
Collapse
|
20
|
Bandyopadhyay S, Adebayo D, Obaseki E, Hariri H. Lysosomal membrane contact sites: Integrative hubs for cellular communication and homeostasis. CURRENT TOPICS IN MEMBRANES 2024; 93:85-116. [PMID: 39181579 DOI: 10.1016/bs.ctm.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Lysosomes are more than just cellular recycling bins; they play a crucial role in regulating key cellular functions. Proper lysosomal function is essential for growth pathway regulation, cell proliferation, and metabolic homeostasis. Impaired lysosomal function is associated with lipid storage disorders and neurodegenerative diseases. Lysosomes form extensive and dynamic close contacts with the membranes of other organelles, including the endoplasmic reticulum, mitochondria, peroxisomes, and lipid droplets. These membrane contacts sites (MCSs) are vital for many lysosomal functions. In this chapter, we will explore lysosomal MCSs focusing on the machinery that mediates these contacts, how they are regulated, and their functional implications on physiology and pathology.
Collapse
Affiliation(s)
- Sumit Bandyopadhyay
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Daniel Adebayo
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Eseiwi Obaseki
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Hanaa Hariri
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States.
| |
Collapse
|
21
|
Madhana Priya N, Sidharth Kumar N, Udhaya Kumar S, Mohanraj G, Magesh R, Zayed H, Vasudevan K, C GPD. Exploring the effect of disease causing mutations in metal binding sites of human ARSA in metachromatic leukodystrophy. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 141:203-221. [PMID: 38960474 DOI: 10.1016/bs.apcsb.2023.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
The arylsulfatase A (ARSA) gene is observed to be deficient in patients with metachromatic leukodystrophy (MLD), a type of lysosomal storage disease. MLD is a severe neurodegenerative disorder characterized by an autosomal recessive inheritance pattern. This study aimed to map the most deleterious mutations at the metal binding sites of ARSA and the amino acids in proximity to the mutated positions. We utilized an array of computational tools, including PredictSNP, MAPP, PhD-SNP, PolyPhen-1, PolyPhen-2, SIFT, SNAP, and ConSurf, to identify the most detrimental mutations potentially implicated in MLD collected from UniProt, ClinVar, and HGMD. Two mutations, D29N and D30H, as being extremely deleterious based on assessments of pathogenicity, conservation, biophysical characteristics, and stability analysis. The D29 and D30 are located at the metal-interacting regions of ARSA and found to undergo post-translational modification, specifically phosphorylation. Henceforth, the in-depth effect of metal binding upon mutation was examined using molecular dynamics simulations (MDS) before and after phosphorylation. The MDS results exhibited high deviation for the D29N and D30H mutations in comparison to the native, and the same was confirmed by significant residue fluctuation and reduced compactness. These structural alterations suggest that such mutations may influence protein functionality, offering potential avenues for personalized therapeutic and providing a basis for potential mutation-specific treatments for severe MLD patients.
Collapse
Affiliation(s)
- N Madhana Priya
- Department of Biotechnology, Sri Ramachandra Institute of Higher Education and Research (DU), Chennai, Tamil Nadu, India
| | - N Sidharth Kumar
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - S Udhaya Kumar
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India; Department of Medicine, Division Endocrinology, Diabetes and Metabolism, Baylor College of Medicine, Houston, TX, United States
| | - G Mohanraj
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - R Magesh
- Department of Biotechnology, Sri Ramachandra Institute of Higher Education and Research (DU), Chennai, Tamil Nadu, India
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar
| | - Karthick Vasudevan
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, Karnataka, India
| | - George Priya Doss C
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India.
| |
Collapse
|
22
|
Jacob JR, Palanichamy K, Chakravarti A. Antipsychotics possess anti-glioblastoma activity by disrupting lysosomal function and inhibiting oncogenic signaling by stabilizing PTEN. Cell Death Dis 2024; 15:414. [PMID: 38871731 PMCID: PMC11176297 DOI: 10.1038/s41419-024-06779-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 05/22/2024] [Accepted: 05/28/2024] [Indexed: 06/15/2024]
Abstract
The repurposing of medications developed for central nervous system (CNS) disorders, possessing favorable safety profiles and blood-brain barrier permeability, represents a promising strategy for identifying new therapies to combat glioblastoma (GBM). In this study, we investigated the anti-GBM activity of specific antipsychotics and antidepressants in vitro and in vivo. Our results demonstrate that these compounds share a common mechanism of action in GBM, disrupting lysosomal function and subsequently inducing lysosomal membrane rupture and cell death. Notably, PTEN intact GBMs possess an increased sensitivity to these compounds. The inhibition of lysosomal function synergized with inhibitors targeting the EGFR-PI3K-Akt pathway, leading to an energetic and antioxidant collapse. These findings provide a foundation for the potential clinical application of CNS drugs in GBM treatment. Additionally, this work offers critical insights into the mechanisms and determinants of cytotoxicity for drugs currently undergoing clinical trials as repurposing agents for various cancers, including Fluoxetine, Sertraline, Thioridazine, Chlorpromazine, and Fluphenazine.
Collapse
Affiliation(s)
- John Ryan Jacob
- Department of Radiation Oncology, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Kamalakannan Palanichamy
- Department of Radiation Oncology, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, OH, 43210, USA.
| | - Arnab Chakravarti
- Department of Radiation Oncology, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, OH, 43210, USA
| |
Collapse
|
23
|
Lee JJ, Wang T, Wiggins K, Lu PN, Underwood C, Ochenkowska K, Samarut E, Pollard LM, Flanagan-Steet H, Steet R. Dysregulated lysosomal exocytosis drives protease-mediated cartilage pathogenesis in multiple lysosomal disorders. iScience 2024; 27:109293. [PMID: 38495824 PMCID: PMC10940929 DOI: 10.1016/j.isci.2024.109293] [Citation(s) in RCA: 5] [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: 06/21/2023] [Revised: 12/20/2023] [Accepted: 02/16/2024] [Indexed: 03/19/2024] Open
Abstract
The classic view of the lysosome as a static recycling center has been replaced with one of a dynamic and mobile hub of metabolic regulation. This revised view raises new questions about how dysfunction of this organelle causes pathology in inherited lysosomal disorders. Here we provide evidence for increased lysosomal exocytosis in the developing cartilage of three lysosomal disease zebrafish models with distinct etiologies. Dysregulated exocytosis was linked to altered cartilage development, increased activity of multiple cathepsin proteases, and cathepsin- and TGFβ-mediated pathogenesis in these models. Moreover, inhibition of cathepsin activity or direct blockade of exocytosis with small molecule modulators improved the cartilage phenotypes, reinforcing a connection between excessive extracellular protease activity and cartilage pathogenesis. This study highlights the pathogenic consequences in early cartilage development arising from uncontrolled release of lysosomal enzymes via exocytosis, and suggests that pharmacological enhancement of this process could be detrimental during tissue development.
Collapse
Affiliation(s)
- Jen-Jie Lee
- JC Self Research Institute, Greenwood Genetic Center, Greenwood, SC 29646, USA
| | - Tong Wang
- JC Self Research Institute, Greenwood Genetic Center, Greenwood, SC 29646, USA
| | - Kali Wiggins
- JC Self Research Institute, Greenwood Genetic Center, Greenwood, SC 29646, USA
| | - Po Nien Lu
- JC Self Research Institute, Greenwood Genetic Center, Greenwood, SC 29646, USA
| | - Christina Underwood
- Biochemical Genetics Laboratory, Greenwood Genetic Center, Greenwood, SC 29646, USA
| | - Katarzyna Ochenkowska
- Research Center, Centre hospitalier de l’Université de Montréal (CHUM), Montreal, Canada
- Department of Neuroscience, Université de Montréal, Montréal, Canada
| | - Eric Samarut
- Research Center, Centre hospitalier de l’Université de Montréal (CHUM), Montreal, Canada
- Department of Neuroscience, Université de Montréal, Montréal, Canada
| | - Laura M. Pollard
- Biochemical Genetics Laboratory, Greenwood Genetic Center, Greenwood, SC 29646, USA
| | | | - Richard Steet
- JC Self Research Institute, Greenwood Genetic Center, Greenwood, SC 29646, USA
| |
Collapse
|
24
|
Coelho-Ribeiro B, Silva HG, Sampaio-Marques B, Fraga AG, Azevedo O, Pedrosa J, Ludovico P. Inflammation and Exosomes in Fabry Disease Pathogenesis. Cells 2024; 13:654. [PMID: 38667269 PMCID: PMC11049543 DOI: 10.3390/cells13080654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/01/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024] Open
Abstract
Fabry Disease (FD) is one of the most prevalent lysosomal storage disorders, resulting from mutations in the GLA gene located on the X chromosome. This genetic mutation triggers glo-botriaosylceramide (Gb-3) buildup within lysosomes, ultimately impairing cellular functions. Given the role of lysosomes in immune cell physiology, FD has been suggested to have a profound impact on immunological responses. During the past years, research has been focusing on this topic, and pooled evidence strengthens the hypothesis that Gb-3 accumulation potentiates the production of pro-inflammatory mediators, revealing the existence of an acute inflammatory process in FD that possibly develops to a chronic state due to stimulus persistency. In parallel, extracellular vesicles (EVs) have gained attention due to their function as intercellular communicators. Considering EVs' capacity to convey cargo from parent to distant cells, they emerge as potential inflammatory intermediaries capable of transporting cytokines and other immunomodulatory molecules. In this review, we revisit the evidence underlying the association between FD and altered immune responses and explore the potential of EVs to function as inflammatory vehicles.
Collapse
Affiliation(s)
- Bruna Coelho-Ribeiro
- Life and Health Sciences Research Institute (ICVS), 4710-057 Braga, Portugal; (B.C.-R.); (H.G.S.); (B.S.-M.); (A.G.F.); (J.P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Helena G. Silva
- Life and Health Sciences Research Institute (ICVS), 4710-057 Braga, Portugal; (B.C.-R.); (H.G.S.); (B.S.-M.); (A.G.F.); (J.P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Belém Sampaio-Marques
- Life and Health Sciences Research Institute (ICVS), 4710-057 Braga, Portugal; (B.C.-R.); (H.G.S.); (B.S.-M.); (A.G.F.); (J.P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Alexandra G. Fraga
- Life and Health Sciences Research Institute (ICVS), 4710-057 Braga, Portugal; (B.C.-R.); (H.G.S.); (B.S.-M.); (A.G.F.); (J.P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Olga Azevedo
- Reference Center on Lysosomal Storage Disorders, Hospital Senhora da Oliveira, 4835-044 Guimarães, Portugal;
| | - Jorge Pedrosa
- Life and Health Sciences Research Institute (ICVS), 4710-057 Braga, Portugal; (B.C.-R.); (H.G.S.); (B.S.-M.); (A.G.F.); (J.P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Paula Ludovico
- Life and Health Sciences Research Institute (ICVS), 4710-057 Braga, Portugal; (B.C.-R.); (H.G.S.); (B.S.-M.); (A.G.F.); (J.P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| |
Collapse
|
25
|
Boytsov D, Madej GM, Horn G, Blaha N, Köcher T, Sitte HH, Siekhaus D, Ziegler C, Sandtner W, Roblek M. Orphan lysosomal solute carrier MFSD1 facilitates highly selective dipeptide transport. Proc Natl Acad Sci U S A 2024; 121:e2319686121. [PMID: 38507452 PMCID: PMC10990142 DOI: 10.1073/pnas.2319686121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/27/2024] [Indexed: 03/22/2024] Open
Abstract
Orphan solute carrier (SLC) represents a group of membrane transporters whose exact functions and substrate specificities are not known. Elucidating the function and regulation of orphan SLC transporters is not only crucial for advancing our knowledge of cellular and molecular biology but can potentially lead to the development of new therapeutic strategies. Here, we provide evidence for the biological function of a ubiquitous orphan lysosomal SLC, the Major Facilitator Superfamily Domain-containing Protein 1 (MFSD1), which has remained phylogenetically unassigned. Targeted metabolomics revealed that dipeptides containing either lysine or arginine residues accumulate in lysosomes of cells lacking MFSD1. Whole-cell patch-clamp electrophysiological recordings of HEK293-cells expressing MFSD1 on the cell surface displayed transport affinities for positively charged dipeptides in the lower mM range, while dipeptides that carry a negative net charge were not transported. This was also true for single amino acids and tripeptides, which MFSD1 failed to transport. Our results identify MFSD1 as a highly selective lysosomal lysine/arginine/histidine-containing dipeptide exporter, which functions as a uniporter.
Collapse
Affiliation(s)
- Danila Boytsov
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, ViennaAT-1090, Austria
| | - Gregor M. Madej
- Department of Biophysics II/Structural Biology, University of Regensburg, RegensburgDE-93053, Germany
| | - Georg Horn
- Department of Biophysics II/Structural Biology, University of Regensburg, RegensburgDE-93053, Germany
| | - Nadine Blaha
- Vienna BioCenter Core Facilities, Metabolomics, Vienna BioCenter, ViennaAT-1030, Austria
| | - Thomas Köcher
- Vienna BioCenter Core Facilities, Metabolomics, Vienna BioCenter, ViennaAT-1030, Austria
| | - Harald H. Sitte
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, ViennaAT-1090, Austria
- Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, AmmanJO-19328, Jordan
- Center for Addiction Research and Science, Medical University of Vienna, ViennaAT-1090, Austria
| | - Daria Siekhaus
- Institute of Science and Technology Austria, KlosterneuburgAT-3400, Austria
| | - Christine Ziegler
- Department of Biophysics II/Structural Biology, University of Regensburg, RegensburgDE-93053, Germany
| | - Walter Sandtner
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, ViennaAT-1090, Austria
| | - Marko Roblek
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, ViennaAT-1090, Austria
- Institute of Science and Technology Austria, KlosterneuburgAT-3400, Austria
| |
Collapse
|
26
|
Li X, Xiang C, Zhu S, Guo J, Liu C, Wang A, Cao J, Lu Y, Neculai D, Xu P, Feng XH. SNX8 enables lysosome reformation and reverses lysosomal storage disorder. Nat Commun 2024; 15:2553. [PMID: 38519472 PMCID: PMC10959956 DOI: 10.1038/s41467-024-46705-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/06/2024] [Indexed: 03/25/2024] Open
Abstract
Lysosomal Storage Disorders (LSDs), which share common phenotypes, including enlarged lysosomes and defective lysosomal storage, are caused by mutations in lysosome-related genes. Although gene therapies and enzyme replacement therapies have been explored, there are currently no effective routine therapies against LSDs. During lysosome reformation, which occurs when the functional lysosome pool is reduced, lysosomal lipids and proteins are recycled to restore lysosome functions. Here we report that the sorting nexin protein SNX8 promotes lysosome tubulation, a process that is required for lysosome reformation, and that loss of SNX8 leads to phenotypes characteristic of LSDs in human cells. SNX8 overexpression rescued features of LSDs in cells, and AAV-based delivery of SNX8 to the brain rescued LSD phenotypes in mice. Importantly, by screening a natural compound library, we identified three small molecules that enhanced SNX8-lysosome binding and reversed LSD phenotypes in human cells and in mice. Altogether, our results provide a potential solution for the treatment of LSDs.
Collapse
Affiliation(s)
- Xinran Li
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, 321000, Shaoxing, Zhejiang, China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, 311200, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Cong Xiang
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, 321000, Shaoxing, Zhejiang, China
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Shilei Zhu
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, 321000, Shaoxing, Zhejiang, China
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Jiansheng Guo
- Center of Cryo-Electron Microscopy, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chang Liu
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, 321000, Shaoxing, Zhejiang, China
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Ailian Wang
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, 321000, Shaoxing, Zhejiang, China
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Jin Cao
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, 321000, Shaoxing, Zhejiang, China
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Yan Lu
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
- Department of Cell Biology, and Department of General Surgery of Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dante Neculai
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
- Department of Cell Biology, and Department of General Surgery of Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pinglong Xu
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, 310058, Hangzhou, Zhejiang, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, 321000, Shaoxing, Zhejiang, China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, 311200, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Xin-Hua Feng
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, 310058, Hangzhou, Zhejiang, China.
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, 321000, Shaoxing, Zhejiang, China.
- Cancer Center, Zhejiang University, 310058, Hangzhou, Zhejiang, China.
- The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
27
|
van der Beek J, de Heus C, Sanza P, Liv N, Klumperman J. Loss of the HOPS complex disrupts early-to-late endosome transition, impairs endosomal recycling and induces accumulation of amphisomes. Mol Biol Cell 2024; 35:ar40. [PMID: 38198575 PMCID: PMC10916860 DOI: 10.1091/mbc.e23-08-0328] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/22/2023] [Accepted: 01/05/2024] [Indexed: 01/12/2024] Open
Abstract
The multisubunit HOPS tethering complex is a well-established regulator of lysosome fusion with late endosomes and autophagosomes. However, the role of the HOPS complex in other stages of endo-lysosomal trafficking is not well understood. To address this, we made HeLa cells knocked out for the HOPS-specific subunits Vps39 or Vps41, or the HOPS-CORVET-core subunits Vps18 or Vps11. In all four knockout cells, we found that endocytosed cargos were trapped in enlarged endosomes that clustered in the perinuclear area. By correlative light-electron microscopy, these endosomes showed a complex ultrastructure and hybrid molecular composition, displaying markers for early (Rab5, PtdIns3P, EEA1) as well as late (Rab7, CD63, LAMP1) endosomes. These "HOPS bodies" were not acidified, contained enzymatically inactive cathepsins and accumulated endocytosed cargo and cation-independent mannose-6-phosphate receptor (CI-MPR). Consequently, CI-MPR was depleted from the TGN, and secretion of lysosomal enzymes to the extracellular space was enhanced. Strikingly, HOPS bodies also contained the autophagy proteins p62 and LC3, defining them as amphisomes. Together, these findings show that depletion of the lysosomal HOPS complex has a profound impact on the functional organization of the entire endosomal system and suggest the existence of a HOPS-independent mechanism for amphisome formation.
Collapse
Affiliation(s)
- Jan van der Beek
- Center for Molecular Medicine, University Medical Center Utrecht, Institute of Biomembranes, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Cecilia de Heus
- Center for Molecular Medicine, University Medical Center Utrecht, Institute of Biomembranes, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Paolo Sanza
- Center for Molecular Medicine, University Medical Center Utrecht, Institute of Biomembranes, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Nalan Liv
- Center for Molecular Medicine, University Medical Center Utrecht, Institute of Biomembranes, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Judith Klumperman
- Center for Molecular Medicine, University Medical Center Utrecht, Institute of Biomembranes, Utrecht University, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
28
|
Abstract
Dystonia is a clinically and genetically highly heterogeneous neurological disorder characterized by abnormal movements and postures caused by involuntary sustained or intermittent muscle contractions. A number of groundbreaking genetic and molecular insights have recently been gained. While they enable genetic testing and counseling, their translation into new therapies is still limited. However, we are beginning to understand shared pathophysiological pathways and molecular mechanisms. It has become clear that dystonia results from a dysfunctional network involving the basal ganglia, cerebellum, thalamus, and cortex. On the molecular level, more than a handful of, often intertwined, pathways have been linked to pathogenic variants in dystonia genes, including gene transcription during neurodevelopment (e.g., KMT2B, THAP1), calcium homeostasis (e.g., ANO3, HPCA), striatal dopamine signaling (e.g., GNAL), endoplasmic reticulum stress response (e.g., EIF2AK2, PRKRA, TOR1A), autophagy (e.g., VPS16), and others. Thus, different forms of dystonia can be molecularly grouped, which may facilitate treatment development in the future.
Collapse
Affiliation(s)
- Mirja Thomsen
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany;
| | - Lara M Lange
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany;
| | - Michael Zech
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Katja Lohmann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany;
| |
Collapse
|
29
|
Gauthier C, El Cheikh K, Basile I, Daurat M, Morère E, Garcia M, Maynadier M, Morère A, Gary-Bobo M. Cation-independent mannose 6-phosphate receptor: From roles and functions to targeted therapies. J Control Release 2024; 365:759-772. [PMID: 38086445 DOI: 10.1016/j.jconrel.2023.12.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
The cation-independent mannose 6-phosphate receptor (CI-M6PR) is a ubiquitous transmembrane receptor whose main intracellular role is to direct enzymes carrying mannose 6-phosphate moieties to lysosomal compartments. Recently, the small membrane-bound portion of this receptor has appeared to be implicated in numerous pathophysiological processes. This review presents an overview of the main ligand partners and the roles of CI-M6PR in lysosomal storage diseases, neurology, immunology and cancer fields. Moreover, this membrane receptor has already been noted for its strong potential in therapeutic applications thanks to its cellular internalization activity and its ability to address pathogenic factors to lysosomes for degradation. A number of therapeutic delivery approaches using CI-M6PR, in particular with enzymes, antibodies or nanoparticles, are currently being proposed.
Collapse
Affiliation(s)
- Corentin Gauthier
- NanoMedSyn, Montpellier, France; IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | | | | | | | - Elodie Morère
- NanoMedSyn, Montpellier, France; IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | | | | | - Alain Morère
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | | |
Collapse
|
30
|
Wang H, Cheng L, Yu L, Guo Z. Targeting the mammalian target of rapamycin pathway in neurological manifestations of Covid-19. Rev Med Virol 2024; 34:e2503. [PMID: 38282397 DOI: 10.1002/rmv.2503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/09/2023] [Accepted: 12/12/2023] [Indexed: 01/30/2024]
Abstract
The diverse and severe nature of neurological manifestations associated with coronavirus disease 2019 (Covid-19) has garnered increasing attention. Exploring the potential to decrease neurological complications in Covid-19 patients involves targeting the mammalian target of rapamycin (mTOR) pathway as a therapeutic strategy. The mTOR pathway, widely recognised for its central role in essential cellular processes like synthesising proteins, facilitating autophagy, and modulating immune responses, has implications in various neurological disorders. Drawing parallels between these disorders and the observed neurological complications in Covid-19, we present a comprehensive review on the current understanding of mTOR signalling in the context of severe acute respiratory syndrome coronavirus 2 infection and neuroinflammation.
Collapse
Affiliation(s)
- Han Wang
- Department of Clinical Laboratory, The Affiliated Hospital to Changchun, University of Chinese Medicine, Changchun, China
| | - Li Cheng
- Department of Clinical Laboratory, The Affiliated Hospital to Changchun, University of Chinese Medicine, Changchun, China
| | - Lanlan Yu
- Department of Clinical Laboratory, The Affiliated Hospital to Changchun, University of Chinese Medicine, Changchun, China
| | - Zhigang Guo
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
31
|
Buco F, Matassini C, Vanni C, Clemente F, Paoli P, Carozzini C, Beni A, Cardona F, Goti A, Moya SE, Ortore MG, Andreozzi P, Morrone A, Marradi M. Gold nanoparticles decorated with monosaccharides and sulfated ligands as potential modulators of the lysosomal enzyme N-acetylgalactosamine-6-sulfatase (GALNS). Org Biomol Chem 2023; 21:9362-9371. [PMID: 37975191 DOI: 10.1039/d3ob01466e] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
N-Acetylgalactosamine-6-sulfatase (GALNS) is an enzyme whose deficiency is related to the lysosomal storage disease Morquio A. For the development of effective therapeutic approaches against this disease, the design of suitable enzyme enhancers (i.e. pharmacological chaperones) is fundamental. The natural substrates of GALNS are the glycosaminoglycans keratan sulfate and chondroitin 6-sulfate, which mainly display repeating units of sulfated carbohydrates. With a biomimetic approach, gold nanoparticles (AuNPs) decorated with simple monosaccharides, sulfated ligands (homoligand AuNPs), or both monosaccharides and sulfated ligands (mixed-ligand AuNPs) were designed here as multivalent inhibitors of GALNS. Among the homoligand AuNPs, the most effective inhibitors of GALNS activity are the β-D-galactoside-coated AuNPs. In the case of mixed-ligand AuNPs, β-D-galactosides/sulfated ligands do not show better inhibition than the β-D-galactoside-coated AuNPs. However, a synergistic effect is observed for α-D-mannosides in a mixed-ligand coating with sulfated ligands that reduced IC50 by one order of magnitude with respect to the homoligand α-D-mannoside-coated AuNPs. SAXS experiments corroborated the association of GALNS with β-D-galactoside AuNPs. These AuNPs are able to restore the enzyme activity by almost 2-fold after thermal denaturation, indicating a potential chaperoning activity towards GALNS. This information could be exploited for future development of nanomedicines for Morquio A. The recent implications of GALNS in cancer and neuropathic pain make these kinds of multivalent bionanomaterials of great interest towards multiple therapies.
Collapse
Affiliation(s)
- Francesca Buco
- Department of Chemistry 'Ugo Schiff', University of Firenze, via della Lastruccia 13, Sesto Fiorentino, FI, Italy.
| | - Camilla Matassini
- Department of Chemistry 'Ugo Schiff', University of Firenze, via della Lastruccia 13, Sesto Fiorentino, FI, Italy.
| | - Costanza Vanni
- Department of Chemistry 'Ugo Schiff', University of Firenze, via della Lastruccia 13, Sesto Fiorentino, FI, Italy.
| | - Francesca Clemente
- Department of Chemistry 'Ugo Schiff', University of Firenze, via della Lastruccia 13, Sesto Fiorentino, FI, Italy.
| | - Paolo Paoli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Firenze, Italy
| | - Cosimo Carozzini
- Department of Chemistry 'Ugo Schiff', University of Firenze, via della Lastruccia 13, Sesto Fiorentino, FI, Italy.
| | - Alice Beni
- Department of Chemistry 'Ugo Schiff', University of Firenze, via della Lastruccia 13, Sesto Fiorentino, FI, Italy.
| | - Francesca Cardona
- Department of Chemistry 'Ugo Schiff', University of Firenze, via della Lastruccia 13, Sesto Fiorentino, FI, Italy.
| | - Andrea Goti
- Department of Chemistry 'Ugo Schiff', University of Firenze, via della Lastruccia 13, Sesto Fiorentino, FI, Italy.
| | - Sergio Enrique Moya
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramon 182 C, Donostia-San Sebastián 20014, Spain
| | - Maria Grazia Ortore
- Department of Life and Environmental Sciences, Marche Polytechnic University, Via Brecce Bianche, Ancona, I-60130, Italy
| | - Patrizia Andreozzi
- Department of Chemistry 'Ugo Schiff', University of Firenze, via della Lastruccia 13, Sesto Fiorentino, FI, Italy.
| | - Amelia Morrone
- Laboratory of Molecular Biology of Neurometabolic Diseases, Meyer Children's Hospital, IRCCS, Viale Pieraccini 24, 50139, Firenze, Italy
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Viale Pieraccini 24, 50139 Firenze, Italy
| | - Marco Marradi
- Department of Chemistry 'Ugo Schiff', University of Firenze, via della Lastruccia 13, Sesto Fiorentino, FI, Italy.
| |
Collapse
|
32
|
Zheng H, Li G, Min J, Xu X, Huang W. Lysosome and related protein degradation technologies. Drug Discov Today 2023; 28:103767. [PMID: 37708931 DOI: 10.1016/j.drudis.2023.103767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 08/31/2023] [Accepted: 09/07/2023] [Indexed: 09/16/2023]
Abstract
Recently, targeted protein degradation technologies based on lysosomal pathways have been developed. Lysosome-based targeted protein degradation technology has a broad range of substrates and the potential to degrade intracellular and extracellular proteins, protein aggregates, damaged organelles and non-protein molecules. Thus, they hold great promise for drug R&D. This study has focused on the biogenesis of lysosomes, their basic functions, lysosome-associated diseases and targeted protein degradation technologies through the lysosomal pathway. In addition, we thoroughly examine the potential applications and limitations of this technology and engage in insightful discussions on potential avenues for future research. Our primary objective is to foster preclinical research on this technology and facilitate its successful clinical implementation.
Collapse
Affiliation(s)
- Hongmei Zheng
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China
| | - Gangjian Li
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China
| | - Jingli Min
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China
| | - Xiangwei Xu
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Wenhai Huang
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, School of Pharmacy, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Key Discipline of Zhejiang Province in Public Health and Preventive Medicine (First Class, Category A), Hangzhou Medical College, China.
| |
Collapse
|
33
|
Bianco V, D'Agostino M, Pirone D, Giugliano G, Mosca N, Di Summa M, Scerra G, Memmolo P, Miccio L, Russo T, Stella E, Ferraro P. Label-Free Intracellular Multi-Specificity in Yeast Cells by Phase-Contrast Tomographic Flow Cytometry. SMALL METHODS 2023; 7:e2300447. [PMID: 37670547 DOI: 10.1002/smtd.202300447] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 08/14/2023] [Indexed: 09/07/2023]
Abstract
In-flow phase-contrast tomography provides a 3D refractive index of label-free cells in cytometry systems. Its major limitation, as with any quantitative phase imaging approach, is the lack of specificity compared to fluorescence microscopy, thus restraining its huge potentialities in single-cell analysis and diagnostics. Remarkable results in introducing specificity are obtained through artificial intelligence (AI), but only for adherent cells. However, accessing the 3D fluorescence ground truth and obtaining accurate voxel-level co-registration of image pairs for AI training is not viable for high-throughput cytometry. The recent statistical inference approach is a significant step forward for label-free specificity but remains limited to cells' nuclei. Here, a generalized computational strategy based on a self-consistent statistical inference to achieve intracellular multi-specificity is shown. Various subcellular compartments (i.e., nuclei, cytoplasmic vacuoles, the peri-vacuolar membrane area, cytoplasm, vacuole-nucleus contact site) can be identified and characterized quantitatively at different phases of the cells life cycle by using yeast cells as a biological model. Moreover, for the first time, virtual reality is introduced for handling the information content of multi-specificity in single cells. Full fruition is proofed for exploring and interacting with 3D quantitative biophysical parameters of the identified compartments on demand, thus opening the route to a metaverse for 3D microscopy.
Collapse
Affiliation(s)
- Vittorio Bianco
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems "E. Caianiello", Via Campi Flegrei 34, Pozzuoli, Napoli, 80078, Italy
| | - Massimo D'Agostino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via S. Pansini 5, Naples, 80131, Italy
| | - Daniele Pirone
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems "E. Caianiello", Via Campi Flegrei 34, Pozzuoli, Napoli, 80078, Italy
| | - Giusy Giugliano
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems "E. Caianiello", Via Campi Flegrei 34, Pozzuoli, Napoli, 80078, Italy
| | - Nicola Mosca
- Institute of Intelligent Industrial Technologies and Systems for Advanced Manufacturing, National Research Council of Italy, Via Amendola 122/D-O, Bari, 70125, Italy
| | - Maria Di Summa
- Institute of Intelligent Industrial Technologies and Systems for Advanced Manufacturing, National Research Council of Italy, Via Amendola 122/D-O, Bari, 70125, Italy
| | - Gianluca Scerra
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via S. Pansini 5, Naples, 80131, Italy
| | - Pasquale Memmolo
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems "E. Caianiello", Via Campi Flegrei 34, Pozzuoli, Napoli, 80078, Italy
| | - Lisa Miccio
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems "E. Caianiello", Via Campi Flegrei 34, Pozzuoli, Napoli, 80078, Italy
| | - Tommaso Russo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via S. Pansini 5, Naples, 80131, Italy
| | - Ettore Stella
- Institute of Intelligent Industrial Technologies and Systems for Advanced Manufacturing, National Research Council of Italy, Via Amendola 122/D-O, Bari, 70125, Italy
| | - Pietro Ferraro
- CNR-ISASI, Institute of Applied Sciences and Intelligent Systems "E. Caianiello", Via Campi Flegrei 34, Pozzuoli, Napoli, 80078, Italy
| |
Collapse
|
34
|
Yang C, Tian F, Hu M, Kang C, Ping M, Liu Y, Hu M, Xu H, Yu Y, Gao Z, Li P. Characterization of the role of TMEM175 in an in vitro lysosomal H + fluxes model. FEBS J 2023; 290:4641-4659. [PMID: 37165739 DOI: 10.1111/febs.16814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/06/2023] [Accepted: 05/09/2023] [Indexed: 05/12/2023]
Abstract
Lysosome acidification is a dynamic equilibrium of H+ influx and efflux across the membrane, which is crucial for cell physiology. The vacuolar H+ ATPase (V-ATPase) is responsible for the H+ influx or refilling of lysosomes. TMEM175 was identified as a novel H+ permeable channel on lysosomal membranes, and it plays a critical role in lysosome acidification. However, how TMEM175 participates in lysosomal acidification remains unknown. Here, we present evidence that TMEM175 regulates lysosomal H+ influx and efflux in enlarged lysosomes isolated from COS1 treated with vacuolin-1. By utilizing the whole-endolysosome patch-clamp recording technique, a series of integrated lysosomal H+ influx and efflux signals in a ten-of-second time scale under the physiological pH gradient (luminal pH 4.60, and cytosolic pH 7.20) was recorded from this in vitro system. Lysosomal H+ fluxes constitute both the lysosomal H+ refilling and releasing, and they are asymmetrical processes with distinct featured kinetics for each of the H+ fluxes. Lysosomal H+ fluxes are entirely abolished when TMEM175 losses of function in the F39V mutant and is blocked by the antagonist (2-GBI). Meanwhile, lysosomal H+ fluxes are modulated by the pH-buffering capacity of the lumen and the lysosomal glycosylated membrane proteins, lysosome-associated membrane protein 1 (LAMP1). We propose that the TMEM175-mediated lysosomal H+ fluxes model would provide novel thoughts for studying the pathology of Parkinson's disease and lysosome storage disorders.
Collapse
Affiliation(s)
- Chuanyan Yang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Fuyun Tian
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China
| | - Mei Hu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China
- Pharmacology Laboratory, Zhongshan Hospital, Guangzhou University of Chinese Medicine, China
| | - Chunlan Kang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Meixuan Ping
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yiyao Liu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China
| | - Meiqin Hu
- Department of Molecular, Cellular, and Developmental Biology, The University of Michigan, Ann Arbor, Michigan, USA
| | - Haoxing Xu
- Department of Molecular, Cellular, and Developmental Biology, The University of Michigan, Ann Arbor, Michigan, USA
| | - Ye Yu
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhaobing Gao
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ping Li
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
35
|
Curado F, Rösner S, Zielke S, Westphal G, Grittner U, Skrahina V, Alasel M, Malik AM, Beetz C, Böttcher T, Barel G, Sah AP, Dinur T, Anjum N, Ichraf Q, Kriouile Y, Hadipour Z, Hadipour F, Revel-Vilk S, Cozma C, Hartkamp J, Cheema H, Zimran A, Bauer P, Rolfs A. Insights into the Value of Lyso-Gb1 as a Predictive Biomarker in Treatment-Naïve Patients with Gaucher Disease Type 1 in the LYSO-PROOF Study. Diagnostics (Basel) 2023; 13:2812. [PMID: 37685353 PMCID: PMC10487050 DOI: 10.3390/diagnostics13172812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/04/2023] [Accepted: 08/10/2023] [Indexed: 09/10/2023] Open
Abstract
Gaucher disease (GD) is a rare autosomal recessive disorder arising from bi-allelic variants in the GBA1 gene, encoding glucocerebrosidase. Deficiency of this enzyme leads to progressive accumulation of the sphingolipid glucosylsphingosine (lyso-Gb1). The international, multicenter, observational "Lyso-Gb1 as a Long-term Prognostic Biomarker in Gaucher Disease"-LYSO-PROOF study succeeded in enrolling a cohort of 160 treatment-naïve GD patients from diverse geographic regions and evaluated the potential of lyso-Gb1 as a specific biomarker for GD. Using genotypes based on established classifications for clinical presentation, patients were stratified into type 1 GD (n = 114) and further subdivided into mild (n = 66) and severe type 1 GD (n = 48). Due to having previously unreported genotypes, 46 patients could not be classified. Though lyso-Gb1 values at enrollment were widely distributed, they displayed a moderate and statistically highly significant correlation with disease severity measured by the GD-DS3 scoring system in all GD patients (r = 0.602, p < 0.0001). These findings support the utility of lyso-Gb1 as a sensitive biomarker for GD and indicate that it could help to predict the clinical course of patients with undescribed genotypes to improve personalized care in the future.
Collapse
Affiliation(s)
- Filipa Curado
- CENTOGENE GmbH, 18055 Rostock, Germany; (F.C.); (S.R.); (S.Z.); (G.W.); (M.A.); (A.M.M.); (C.B.); (T.B.); (A.P.S.); (C.C.); (J.H.)
| | - Sabine Rösner
- CENTOGENE GmbH, 18055 Rostock, Germany; (F.C.); (S.R.); (S.Z.); (G.W.); (M.A.); (A.M.M.); (C.B.); (T.B.); (A.P.S.); (C.C.); (J.H.)
| | - Susanne Zielke
- CENTOGENE GmbH, 18055 Rostock, Germany; (F.C.); (S.R.); (S.Z.); (G.W.); (M.A.); (A.M.M.); (C.B.); (T.B.); (A.P.S.); (C.C.); (J.H.)
| | - Gina Westphal
- CENTOGENE GmbH, 18055 Rostock, Germany; (F.C.); (S.R.); (S.Z.); (G.W.); (M.A.); (A.M.M.); (C.B.); (T.B.); (A.P.S.); (C.C.); (J.H.)
| | - Ulrike Grittner
- Berlin Institute of Health, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany;
- Institute of Biometry and Clinical Epidemiology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | | | - Mohammed Alasel
- CENTOGENE GmbH, 18055 Rostock, Germany; (F.C.); (S.R.); (S.Z.); (G.W.); (M.A.); (A.M.M.); (C.B.); (T.B.); (A.P.S.); (C.C.); (J.H.)
| | - Ahmad Mehmood Malik
- CENTOGENE GmbH, 18055 Rostock, Germany; (F.C.); (S.R.); (S.Z.); (G.W.); (M.A.); (A.M.M.); (C.B.); (T.B.); (A.P.S.); (C.C.); (J.H.)
| | - Christian Beetz
- CENTOGENE GmbH, 18055 Rostock, Germany; (F.C.); (S.R.); (S.Z.); (G.W.); (M.A.); (A.M.M.); (C.B.); (T.B.); (A.P.S.); (C.C.); (J.H.)
| | - Tobias Böttcher
- CENTOGENE GmbH, 18055 Rostock, Germany; (F.C.); (S.R.); (S.Z.); (G.W.); (M.A.); (A.M.M.); (C.B.); (T.B.); (A.P.S.); (C.C.); (J.H.)
| | - Gal Barel
- CENTOGENE GmbH, 18055 Rostock, Germany; (F.C.); (S.R.); (S.Z.); (G.W.); (M.A.); (A.M.M.); (C.B.); (T.B.); (A.P.S.); (C.C.); (J.H.)
| | - Ashish Prasad Sah
- CENTOGENE GmbH, 18055 Rostock, Germany; (F.C.); (S.R.); (S.Z.); (G.W.); (M.A.); (A.M.M.); (C.B.); (T.B.); (A.P.S.); (C.C.); (J.H.)
| | - Tama Dinur
- Gaucher Unit, Shaare Zedek Medical Center, Jerusalem 9103102, Israel; (T.D.); (S.R.-V.); (A.Z.)
| | - Nadeem Anjum
- The Children’s Hospital and University of Child Health Sciences, Lahore 54600, Pakistan; (N.A.); (H.C.)
| | - Quidad Ichraf
- Children Hospital’s Rabat, Neuropediatric-Metabolic, Rabat 6527, Morocco; (Q.I.); (Y.K.)
| | - Yamna Kriouile
- Children Hospital’s Rabat, Neuropediatric-Metabolic, Rabat 6527, Morocco; (Q.I.); (Y.K.)
| | - Zahra Hadipour
- Soodbakhash Poly Clinic, Atiyeh Hospital, Tehran 1416753955, Iran; (Z.H.); (F.H.)
- Medical Genetics Department, Pars Research Center & Hospital, Tehran 1416753955, Iran
| | - Fatemeh Hadipour
- Soodbakhash Poly Clinic, Atiyeh Hospital, Tehran 1416753955, Iran; (Z.H.); (F.H.)
- Medical Genetics Department, Pars Research Center & Hospital, Tehran 1416753955, Iran
| | - Shoshana Revel-Vilk
- Gaucher Unit, Shaare Zedek Medical Center, Jerusalem 9103102, Israel; (T.D.); (S.R.-V.); (A.Z.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel
| | - Claudia Cozma
- CENTOGENE GmbH, 18055 Rostock, Germany; (F.C.); (S.R.); (S.Z.); (G.W.); (M.A.); (A.M.M.); (C.B.); (T.B.); (A.P.S.); (C.C.); (J.H.)
| | - Jörg Hartkamp
- CENTOGENE GmbH, 18055 Rostock, Germany; (F.C.); (S.R.); (S.Z.); (G.W.); (M.A.); (A.M.M.); (C.B.); (T.B.); (A.P.S.); (C.C.); (J.H.)
| | - Huma Cheema
- The Children’s Hospital and University of Child Health Sciences, Lahore 54600, Pakistan; (N.A.); (H.C.)
| | - Ari Zimran
- Gaucher Unit, Shaare Zedek Medical Center, Jerusalem 9103102, Israel; (T.D.); (S.R.-V.); (A.Z.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel
| | - Peter Bauer
- CENTOGENE GmbH, 18055 Rostock, Germany; (F.C.); (S.R.); (S.Z.); (G.W.); (M.A.); (A.M.M.); (C.B.); (T.B.); (A.P.S.); (C.C.); (J.H.)
- Medical Faculty, University of Rostock, 18057 Rostock, Germany;
| | - Arndt Rolfs
- Medical Faculty, University of Rostock, 18057 Rostock, Germany;
- Agyany Pharmaceutics Ltd., Jerusalem 9103102, Israel
- RCV Rare Disease GmbH, 10115 Berlin, Germany
| |
Collapse
|
36
|
Dong L, Xiao J, Liu S, Deng G, Liao Y, Chu B, Zhao X, Song BL, Luo J. Lysosomal cholesterol accumulation is commonly found in most peroxisomal disorders and reversed by 2-hydroxypropyl-β-cyclodextrin. SCIENCE CHINA. LIFE SCIENCES 2023; 66:1786-1799. [PMID: 36971991 DOI: 10.1007/s11427-022-2260-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 12/10/2022] [Indexed: 03/29/2023]
Abstract
Peroxisomal disorders (PDs) are a heterogenous group of diseases caused by defects in peroxisome biogenesis or functions. X-linked adrenoleukodystrophy is the most prevalent form of PDs and results from mutations in the ABCD1 gene, which encodes a transporter mediating the uptake of very long-chain fatty acids (VLCFAs). The curative approaches for PDs are very limited. Here, we investigated whether cholesterol accumulation in the lysosomes is a biochemical feature shared by a broad spectrum of PDs. We individually knocked down fifteen PD-associated genes in cultured cells and found ten induced cholesterol accumulation in the lysosome. 2-Hydroxypropyl-β-cyclodextrin (HPCD) effectively alleviated the cholesterol accumulation phenotype in PD-mimicking cells through reducing intracellular cholesterol content as well as promoting cholesterol redistribution to other cellular membranes. In ABCD1 knockdown cells, HPCD treatment lowered reactive oxygen species and VLCFA to normal levels. In Abcd1 knockout mice, HPCD injections reduced cholesterol and VLCFA sequestration in the brain and adrenal cortex. The plasma levels of adrenocortical hormones were increased and the behavioral abnormalities were greatly ameliorated upon HPCD administration. Together, our results suggest that defective cholesterol transport underlies most, if not all, PDs, and that HPCD can serve as a novel and effective strategy for the treatment of PDs.
Collapse
Affiliation(s)
- Lewei Dong
- College of Life Sciences, Taikang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, Wuhan University, Wuhan, 430072, China
| | - Jian Xiao
- College of Life Sciences, Taikang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, Wuhan University, Wuhan, 430072, China
| | - Shuai Liu
- College of Life Sciences, Taikang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, Wuhan University, Wuhan, 430072, China
| | - Gang Deng
- College of Life Sciences, Taikang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, Wuhan University, Wuhan, 430072, China
| | - Yacheng Liao
- College of Life Sciences, Taikang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, Wuhan University, Wuhan, 430072, China
| | - Beibei Chu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, China
| | - Xiaolu Zhao
- College of Life Sciences, Taikang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, Wuhan University, Wuhan, 430072, China
| | - Bao-Liang Song
- College of Life Sciences, Taikang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, Wuhan University, Wuhan, 430072, China
| | - Jie Luo
- College of Life Sciences, Taikang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
37
|
Qiao W, Richards CM, Jabs S. LYSET/TMEM251- a novel key component of the mannose 6-phosphate pathway. Autophagy 2023; 19:2143-2145. [PMID: 36633450 PMCID: PMC10283412 DOI: 10.1080/15548627.2023.2167376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/06/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
Degradation of macromolecules delivered to lysosomes by processes such as autophagy or endocytosis is crucial for cellular function. Lysosomes require more than 60 soluble hydrolases in order to catabolize such macromolecules. These soluble hydrolases are tagged with mannose6-phosphate (M6P) moieties in sequential reactions by the Golgi-resident GlcNAc-1-phosphotransferase complex and NAGPA/UCE/uncovering enzyme (N-acetylglucosamine-1-phosphodiester alpha-N-acetylglucosaminidase), which allows their delivery to endosomal/lysosomal compartments through trafficking mediated by cation-dependent and -independent mannose 6-phosphate receptors (MPRs). We and others recently identified TMEM251 as a novel regulator of the M6P pathway via independent genome-wide genetic screening strategies. We renamed TMEM251 to LYSET (lysosomal enzyme trafficking factor) to establish nomenclature reflective to this gene's function. LYSET is a Golgi-localized transmembrane protein important for the retention of the GlcNAc-1-phosphotransferase complex in the Golgi-apparatus. The current understanding of LYSET's importance regarding human biology is 3-fold: 1) highly pathogenic viruses that depend on lysosomal hydrolase activity require LYSET for infection. 2) The presence of LYSET is critical for cancer cell proliferation in nutrient-deprived environments in which extracellular proteins must be catabolized. 3) Inherited pathogenic alleles of LYSET can cause a severe inherited disease which resembles GlcNAc-1-phosphotransferase deficiency (i.e., mucolipidosis type II).Abbreviations: GlcNAc-1-PT: GlcNAc-1-phosphotransferase; KO: knockout; LSD: lysosomal storage disorder; LYSET: lysosomal enzyme trafficking factor; M6P: mannose 6-phosphate; MPRs: mannose-6-phosphate receptors, cation-dependent or -independent; MBTPS1/site-1 protease: membrane bound transcription factor peptidase, site 1; MLII: mucolipidosis type II; WT: wild-type.
Collapse
Affiliation(s)
- Wenjie Qiao
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Christopher M. Richards
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sabrina Jabs
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Schleswig-Holstein, Germany
| |
Collapse
|
38
|
Gul R, Firasat S, Schubert M, Ullah A, Peña E, Thuesen ACB, Hussain M, Staeger FF, Gjesing AP, Albrechtsen A, Hansen T. Identifying the genetic causes of phenotypically diagnosed Pakistani mucopolysaccharidoses patients by whole genome sequencing. Front Genet 2023; 14:1128850. [PMID: 37091798 PMCID: PMC10113632 DOI: 10.3389/fgene.2023.1128850] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/13/2023] [Indexed: 04/07/2023] Open
Abstract
Background: Lysosomal storage disorders (LSDs) are a group of inherited metabolic diseases, which encompass more than 50 different subtypes of pathologies. These disorders are caused by defects in lysosomal enzymes, transporters, and other non-lysosomal proteins. Mucopolysaccharidosis (MPS) is the most common subgroup of lysosomal storage disorders in which the body is unable to properly breakdown mucopolysaccharides. The aim of the present study was to identify novel genes and pathogenic variants in families from diverse regions of Pakistan with clinically diagnosed mucopolysaccharidosis type I and mucopolysaccharidosis type II.Methods: Clinical diagnosis identified 12 with mucopolysaccharidosis I and 2 with mucopolysaccharidosis II in 14 families and whole genome sequencing (WGS) was performed to identify the causative variations in 15 affected individuals. Twenty-two unaffected individuals including parents or normal siblings of patients were also sequenced. Putative causal variants were identified by co-segregation and functional annotation.Results: Analysis of whole genome sequencing data revealed ten novel and six previously reported variants in lysosomal storage disorders-associated genes (IDUA, GALNS, SGSH, GAA, IDS, ALDOB, TRAPPC4, MASP1, SMARCAL, KIAA1109, HERC1, RRAS2) and a novel candidate gene (ABCA5) for lysosomal storage disorder-like phenotypes, which has previously been associated with symptoms strongly related with lysosomal storage disorder in animal models.Conclusion: Multigenic inheritance was found in several families highlighting the importance of searching for homozygous pathogenic variants in several genes also in families with a high degree of consanguinity.
Collapse
Affiliation(s)
- Rutaba Gul
- Department of Zoology, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, Pakistan
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sabika Firasat
- Department of Zoology, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, Pakistan
- *Correspondence: Sabika Firasat, ; Torben Hansen,
| | - Mikkel Schubert
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Asmat Ullah
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Elionora Peña
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne C. B. Thuesen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mulazim Hussain
- The Children Hospital, Pakistan Institute of Medical Sciences (PIMS), Islamabad, Pakistan
| | - Frederik F. Staeger
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Anette P. Gjesing
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders Albrechtsen
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Sabika Firasat, ; Torben Hansen,
| |
Collapse
|
39
|
Pandey MK. Exploring Pro-Inflammatory Immunological Mediators: Unraveling the Mechanisms of Neuroinflammation in Lysosomal Storage Diseases. Biomedicines 2023; 11:biomedicines11041067. [PMID: 37189685 DOI: 10.3390/biomedicines11041067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 04/05/2023] Open
Abstract
Lysosomal storage diseases are a group of rare and ultra-rare genetic disorders caused by defects in specific genes that result in the accumulation of toxic substances in the lysosome. This excess accumulation of such cellular materials stimulates the activation of immune and neurological cells, leading to neuroinflammation and neurodegeneration in the central and peripheral nervous systems. Examples of lysosomal storage diseases include Gaucher, Fabry, Tay–Sachs, Sandhoff, and Wolman diseases. These diseases are characterized by the accumulation of various substrates, such as glucosylceramide, globotriaosylceramide, ganglioside GM2, sphingomyelin, ceramide, and triglycerides, in the affected cells. The resulting pro-inflammatory environment leads to the generation of pro-inflammatory cytokines, chemokines, growth factors, and several components of complement cascades, which contribute to the progressive neurodegeneration seen in these diseases. In this study, we provide an overview of the genetic defects associated with lysosomal storage diseases and their impact on the induction of neuro-immune inflammation. By understanding the underlying mechanisms behind these diseases, we aim to provide new insights into potential biomarkers and therapeutic targets for monitoring and managing the severity of these diseases. In conclusion, lysosomal storage diseases present a complex challenge for patients and clinicians, but this study offers a comprehensive overview of the impact of these diseases on the central and peripheral nervous systems and provides a foundation for further research into potential treatments.
Collapse
Affiliation(s)
- Manoj Kumar Pandey
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, Cincinnati, OH 45229-3026, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0515, USA
| |
Collapse
|
40
|
Zhang X, Misra SK, Moitra P, Zhang X, Jeong SJ, Stitham J, Rodriguez-Velez A, Park A, Yeh YS, Gillanders WE, Fan D, Diwan A, Cho J, Epelman S, Lodhi IJ, Pan D, Razani B. Use of acidic nanoparticles to rescue macrophage lysosomal dysfunction in atherosclerosis. Autophagy 2023; 19:886-903. [PMID: 35982578 PMCID: PMC9980706 DOI: 10.1080/15548627.2022.2108252] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 12/19/2022] Open
Abstract
Dysfunction in the macrophage lysosomal system including reduced acidity and diminished degradative capacity is a hallmark of atherosclerosis, leading to blunted clearance of excess cellular debris and lipids in plaques and contributing to lesion progression. Devising strategies to rescue this macrophage lysosomal dysfunction is a novel therapeutic measure. Nanoparticles have emerged as an effective platform to both target specific tissues and serve as drug delivery vehicles. In most cases, administered nanoparticles are taken up non-selectively by the mononuclear phagocyte system including monocytes/macrophages leading to the undesirable degradation of cargo in lysosomes. We took advantage of this default route to target macrophage lysosomes to rectify their acidity in disease states such as atherosclerosis. Herein, we develop and test two commonly used acidic nanoparticles, poly-lactide-co-glycolic acid (PLGA) and polylactic acid (PLA), both in vitro and in vivo. Our results in cultured macrophages indicate that the PLGA-based nanoparticles are the most effective at trafficking to and enhancing acidification of lysosomes. PLGA nanoparticles also provide functional benefits including enhanced lysosomal degradation, promotion of macroautophagy/autophagy and protein aggregate removal, and reduced apoptosis and inflammasome activation. We demonstrate the utility of this system in vivo, showing nanoparticle accumulation in, and lysosomal acidification of, macrophages in atherosclerotic plaques. Long-term administration of PLGA nanoparticles results in significant reductions in surrogates of plaque complexity with reduced apoptosis, necrotic core formation, and cytotoxic protein aggregates and increased fibrous cap formation. Taken together, our data support the use of acidic nanoparticles to rescue macrophage lysosomal dysfunction in the treatment of atherosclerosis.Abbreviations: BCA: brachiocephalic arteries; FACS: fluorescence activated cell sorting; FITC: fluorescein-5-isothiocyanatel; IL1B: interleukin 1 beta; LAMP: lysosomal associated membrane protein; LIPA/LAL: lipase A, lysosomal acid type; LSDs: lysosomal storage disorders; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MFI: mean fluorescence intensity; MPS: mononuclear phagocyte system; PEGHDE: polyethylene glycol hexadecyl ether; PLA: polylactic acid; PLGA: poly-lactide-co-glycolic acid; SQSTM1/p62: sequestosome 1.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Cardiovascular Division, Washington University, St. Louis, MO, USA
| | - Santosh Kumar Misra
- Department of Bioengineering, University of Illinois at Urbana Champaign, IL, USA
| | - Parikshit Moitra
- Departments of Diagnostic Radiology and Nuclear Medicine and Pediatrics, Baltimore, Maryland, USA
- Department of Nuclear Engineering, The Pennsylvania State University, University Park, Pennsylvania16802, USA
| | - Xiuli Zhang
- Department of Surgery, Washington University, St. Louis, MO, USA
| | - Se-Jin Jeong
- Cardiovascular Division, Washington University, St. Louis, MO, USA
| | - Jeremiah Stitham
- Cardiovascular Division, Washington University, St. Louis, MO, USA
- Division of Endocrinology, Metabolism, and Lipid Research, St. Louis, MO, USA
| | | | - Arick Park
- Cardiovascular Division, Washington University, St. Louis, MO, USA
| | - Yu-Sheng Yeh
- Cardiovascular Division, Washington University, St. Louis, MO, USA
| | | | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Abhinav Diwan
- Cardiovascular Division, Washington University, St. Louis, MO, USA
- John Cochran Division, VA Medical Center, St. Louis, MO, USA
| | - Jaehyung Cho
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Slava Epelman
- Peter Munk Cardiac Center, Toronto General Hospital Research Institute, University Health Network, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
| | - Irfan J. Lodhi
- Division of Endocrinology, Metabolism, and Lipid Research, St. Louis, MO, USA
| | - Dipanjan Pan
- Department of Bioengineering, University of Illinois at Urbana Champaign, IL, USA
- Departments of Diagnostic Radiology and Nuclear Medicine and Pediatrics, Baltimore, Maryland, USA
- Department of Nuclear Engineering, The Pennsylvania State University, University Park, Pennsylvania16802, USA
| | - Babak Razani
- Cardiovascular Division, Washington University, St. Louis, MO, USA
- John Cochran Division, VA Medical Center, St. Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
41
|
Shaimardanova AA, Solovyeva VV, Issa SS, Rizvanov AA. Gene Therapy of Sphingolipid Metabolic Disorders. Int J Mol Sci 2023; 24:3627. [PMID: 36835039 PMCID: PMC9964151 DOI: 10.3390/ijms24043627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Sphingolipidoses are defined as a group of rare hereditary diseases resulting from mutations in the genes encoding lysosomal enzymes. This group of lysosomal storage diseases includes more than 10 genetic disorders, including GM1-gangliosidosis, Tay-Sachs disease, Sandhoff disease, the AB variant of GM2-gangliosidosis, Fabry disease, Gaucher disease, metachromatic leukodystrophy, Krabbe disease, Niemann-Pick disease, Farber disease, etc. Enzyme deficiency results in accumulation of sphingolipids in various cell types, and the nervous system is also usually affected. There are currently no known effective methods for the treatment of sphingolipidoses; however, gene therapy seems to be a promising therapeutic variant for this group of diseases. In this review, we discuss gene therapy approaches for sphingolipidoses that are currently being investigated in clinical trials, among which adeno-associated viral vector-based approaches and transplantation of hematopoietic stem cells genetically modified with lentiviral vectors seem to be the most effective.
Collapse
Affiliation(s)
- Alisa A. Shaimardanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Valeriya V. Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Shaza S. Issa
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| |
Collapse
|
42
|
Lu G, Shen HM. CLN3 clinches lysosomes in clearance of glycerophospholipids. LIFE METABOLISM 2023; 2:loac029. [PMID: 39872513 PMCID: PMC11749349 DOI: 10.1093/lifemeta/loac029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 10/17/2022] [Indexed: 01/30/2025]
Affiliation(s)
- Guang Lu
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Han-Ming Shen
- Department of Biomedical Sciences, Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macao 999078, China
| |
Collapse
|
43
|
De Pasquale V, Esposito A, Scerra G, Scarcella M, Ciampa M, Luongo A, D’Alonzo D, Guaragna A, D’Agostino M, Pavone LM. N-Substituted l-Iminosugars for the Treatment of Sanfilippo Type B Syndrome. J Med Chem 2023; 66:1790-1808. [PMID: 36696678 PMCID: PMC9923752 DOI: 10.1021/acs.jmedchem.2c01617] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Sanfilippo syndrome comprises a group of four genetic diseases due to the lack or decreased activity of enzymes involved in heparan sulfate (HS) catabolism. HS accumulation in lysosomes and other cellular compartments results in tissue and organ dysfunctions, leading to a wide range of clinical symptoms including severe neurodegeneration. To date, no approved treatments for Sanfilippo disease exist. Here, we report the ability of N-substituted l-iminosugars to significantly reduce substrate storage and lysosomal dysfunctions in Sanfilippo fibroblasts and in a neuronal cellular model of Sanfilippo B subtype. Particularly, we found that they increase the levels of defective α-N-acetylglucosaminidase and correct its proper sorting toward the lysosomal compartment. Furthermore, l-iminosugars reduce HS accumulation by downregulating protein levels of exostosin glycosyltransferases. These results highlight an interesting pharmacological potential of these glycomimetics in Sanfilippo syndrome, paving the way for the development of novel therapeutic approaches for the treatment of such incurable disease.
Collapse
Affiliation(s)
- Valeria De Pasquale
- Department
of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via F. Delpino 1, 80137 Naples, Italy
| | - Anna Esposito
- Department
of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale V. Tecchio 80, 80125 Naples, Italy
| | - Gianluca Scerra
- Department
of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Melania Scarcella
- Department
of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Mariangela Ciampa
- Department
of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Antonietta Luongo
- AORN
Sant’Anna e San Sebastiano, Via F. Palasciano, 81100 Caserta, Italy
| | - Daniele D’Alonzo
- Department
of Chemical Sciences, University of Naples
Federico II, Via Cintia, 80126 Napoli, Italy
| | - Annalisa Guaragna
- Department
of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale V. Tecchio 80, 80125 Naples, Italy,
| | - Massimo D’Agostino
- Department
of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy,
| | - Luigi Michele Pavone
- Department
of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy,
| |
Collapse
|
44
|
Amaral O, Martins M, Oliveira AR, Duarte AJ, Mondragão-Rodrigues I, Macedo MF. The Biology of Lysosomes: From Order to Disorder. Biomedicines 2023; 11:213. [PMID: 36672721 PMCID: PMC9856021 DOI: 10.3390/biomedicines11010213] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/30/2022] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
Since its discovery in 1955, the understanding of the lysosome has continuously increased. Once considered a mere waste removal system, the lysosome is now recognised as a highly crucial cellular component for signalling and energy metabolism. This notable evolution raises the need for a summarized review of the lysosome's biology. As such, throughout this article, we will be compiling the current knowledge regarding the lysosome's biogenesis and functions. The comprehension of this organelle's inner mechanisms is crucial to perceive how its impairment can give rise to lysosomal disease (LD). In this review, we highlight some examples of LD fine-tuned mechanisms that are already established, as well as others, which are still under investigation. Even though the understanding of the lysosome and its pathologies has expanded through the years, some of its intrinsic molecular aspects remain unknown. In order to illustrate the complexity of the lysosomal diseases we provide a few examples that have challenged the established single gene-single genetic disorder model. As such, we believe there is a strong need for further investigation of the exact abnormalities in the pathological pathways in lysosomal disease.
Collapse
Affiliation(s)
- Olga Amaral
- Departamento de Genética Humana, Unidade de Investigação e Desenvolvimento, Instituto Nacional de Saúde Ricardo Jorge (INSA), 4000-055 Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA, ICETA), Universidade do Porto, 4485-661 Porto, Portugal
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Mariana Martins
- Departamento de Ciências Médicas, Universidade de Aveiro, Campus Universitário de Santiago, Agra do Crasto, Edifício 30, 3810-193 Aveiro, Portugal
| | - Ana Rita Oliveira
- Departamento de Ciências Médicas, Universidade de Aveiro, Campus Universitário de Santiago, Agra do Crasto, Edifício 30, 3810-193 Aveiro, Portugal
| | - Ana Joana Duarte
- Departamento de Genética Humana, Unidade de Investigação e Desenvolvimento, Instituto Nacional de Saúde Ricardo Jorge (INSA), 4000-055 Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA, ICETA), Universidade do Porto, 4485-661 Porto, Portugal
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), 1300-477 Lisboa, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, 4050-313 Porto, Portugal
| | - Inês Mondragão-Rodrigues
- Departamento de Ciências Médicas, Universidade de Aveiro, Campus Universitário de Santiago, Agra do Crasto, Edifício 30, 3810-193 Aveiro, Portugal
- CAGE, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - M. Fátima Macedo
- Departamento de Ciências Médicas, Universidade de Aveiro, Campus Universitário de Santiago, Agra do Crasto, Edifício 30, 3810-193 Aveiro, Portugal
- CAGE, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| |
Collapse
|
45
|
Hegeman CV, de Jong OG, Lorenowicz MJ. A kaleidoscopic view of extracellular vesicles in lysosomal storage disorders. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2022; 3:393-421. [PMID: 39697359 PMCID: PMC11651879 DOI: 10.20517/evcna.2022.41] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/16/2022] [Accepted: 12/26/2022] [Indexed: 12/20/2024]
Abstract
Extracellular vesicles (EVs) are a heterogeneous population of stable lipid membrane particles that play a critical role in the regulation of numerous physiological and pathological processes. EV cargo, which includes lipids, proteins, and RNAs including miRNAs, is affected by the metabolic status of the parental cell. Concordantly, abnormalities in the autophagic-endolysosomal pathway, as seen in lysosomal storage disorders (LSDs), can affect EV release as well as EV cargo. LSDs are a group of over 70 inheritable diseases, characterized by lysosomal dysfunction and gradual accumulation of undigested molecules. LSDs are caused by single gene mutations that lead to a deficiency of a lysosomal protein or lipid. Lysosomal dysfunction sets off a cascade of alterations in the endolysosomal pathway that can affect autophagy and alter calcium homeostasis, leading to energy imbalance, oxidative stress, and apoptosis. The pathophysiology of these diseases is very heterogenous, complex, and currently incompletely understood. LSDs lead to progressive multisystemic symptoms that often include neurological deficits. In this review, a kaleidoscopic overview will be given on the roles of EVs in LSDs, from their contribution to pathology and diagnostics to their role as drug delivery vehicles. Furthermore, EV cargo and surface engineering strategies will be discussed to show the potential of EVs in future LSD treatment, both in the context of enzyme replacement therapy, as well as future gene editing strategies like CRISPR/Cas. The use of engineered EVs as drug delivery vehicles may mask therapeutic cargo from the immune system and protect it from degradation, improving circulation time and targeted delivery.
Collapse
Affiliation(s)
- Charlotte V. Hegeman
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht 3584 CG, The Netherlands
| | - Olivier G. de Jong
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht 3584 CG, The Netherlands
- Authors contributed equally
| | - Magdalena J. Lorenowicz
- Regenerative Medicine Center, Uppsalalaan 8, Utrecht 3584 CT, The Netherlands
- Biomedical Primate Research Centre, Lange Kleinweg 161, Rijswijk 2288 GJ, The Netherlands
- Authors contributed equally
| |
Collapse
|
46
|
Chen H, Jin X, Li T, Ye Z. Brain organoids: Establishment and application. Front Cell Dev Biol 2022; 10:1029873. [PMID: 36506083 PMCID: PMC9726712 DOI: 10.3389/fcell.2022.1029873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 11/10/2022] [Indexed: 11/24/2022] Open
Abstract
Brain organoids are produced by the differentiation of pluripotent stem cells under three-dimensional culture conditions by adding neurodevelopment-related regulatory signals. They are similar to the cell composition and anatomical structure of the brain, and can reflect the developmental process of the brain, as well as their physiology, pathology, and pharmacology. Brain organoids are good models to study human brain development and brain-related diseases in vitro. Here, we mainly focus on the construction of brain organoids and review the application of brain organoids in disease modelingand drug screening.
Collapse
Affiliation(s)
- Hao Chen
- Department of Neurovascular Surgery, First Hospital, Jilin University, Changchun, China
| | - Xin Jin
- Department of Oncology and Hematology, Second Hospital, Jilin University, Changchun, China
| | - Tie Li
- Department of Rheumatology, First Hospital, Jilin University, Changchun, China
| | - Zhuang Ye
- Department of Rheumatology, First Hospital, Jilin University, Changchun, China,*Correspondence: Zhuang Ye,
| |
Collapse
|
47
|
Scerra G, De Pasquale V, Scarcella M, Caporaso MG, Pavone LM, D'Agostino M. Lysosomal positioning diseases: beyond substrate storage. Open Biol 2022; 12:220155. [PMID: 36285443 PMCID: PMC9597170 DOI: 10.1098/rsob.220155] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Lysosomal storage diseases (LSDs) comprise a group of inherited monogenic disorders characterized by lysosomal dysfunctions due to undegraded substrate accumulation. They are caused by a deficiency in specific lysosomal hydrolases involved in cellular catabolism, or non-enzymatic proteins essential for normal lysosomal functions. In LSDs, the lack of degradation of the accumulated substrate and its lysosomal storage impairs lysosome functions resulting in the perturbation of cellular homeostasis and, in turn, the damage of multiple organ systems. A substantial number of studies on the pathogenesis of LSDs has highlighted how the accumulation of lysosomal substrates is only the first event of a cascade of processes including the accumulation of secondary metabolites and the impairment of cellular trafficking, cell signalling, autophagic flux, mitochondria functionality and calcium homeostasis, that significantly contribute to the onset and progression of these diseases. Emerging studies on lysosomal biology have described the fundamental roles of these organelles in a variety of physiological functions and pathological conditions beyond their canonical activity in cellular waste clearance. Here, we discuss recent advances in the knowledge of cellular and molecular mechanisms linking lysosomal positioning and trafficking to LSDs.
Collapse
Affiliation(s)
- Gianluca Scerra
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Valeria De Pasquale
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Via Federico Delpino 1, 80137 Naples, Italy
| | - Melania Scarcella
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Maria Gabriella Caporaso
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Luigi Michele Pavone
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Massimo D'Agostino
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy
| |
Collapse
|
48
|
Zhao P, Zhang N, An Z. Engineering antibody and protein therapeutics to cross the blood-brain barrier. Antib Ther 2022; 5:311-331. [PMID: 36540309 PMCID: PMC9759110 DOI: 10.1093/abt/tbac028] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/10/2022] [Accepted: 11/01/2022] [Indexed: 08/17/2023] Open
Abstract
Diseases in the central nervous system (CNS) are often difficult to treat. Antibody- and protein-based therapeutics hold huge promises in CNS disease treatment. However, proteins are restricted from entering the CNS by the blood-brain barrier (BBB). To achieve enhanced BBB crossing, antibody-based carriers have been developed by utilizing the endogenous macromolecule transportation pathway, known as receptor-mediated transcytosis. In this report, we first provided an overall review on key CNS diseases and the most promising antibody- or protein-based therapeutics approved or in clinical trials. We then reviewed the platforms that are being explored to increase the macromolecule brain entry to combat CNS diseases. Finally, we have analyzed the lessons learned from past experiences and have provided a perspective on the future engineering of novel delivery vehicles for antibody- and protein-based therapies for CNS diseases.
Collapse
Affiliation(s)
- Peng Zhao
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, Texas, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, Texas, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler Street, Houston, Texas, USA
| |
Collapse
|
49
|
Laqtom NN, Dong W, Medoh UN, Cangelosi AL, Dharamdasani V, Chan SH, Kunchok T, Lewis CA, Heinze I, Tang R, Grimm C, Dang Do AN, Porter FD, Ori A, Sabatini DM, Abu-Remaileh M. CLN3 is required for the clearance of glycerophosphodiesters from lysosomes. Nature 2022; 609:1005-1011. [PMID: 36131016 PMCID: PMC10510443 DOI: 10.1038/s41586-022-05221-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 08/10/2022] [Indexed: 11/09/2022]
Abstract
Lysosomes have many roles, including degrading macromolecules and signalling to the nucleus1. Lysosomal dysfunction occurs in various human conditions, such as common neurodegenerative diseases and monogenic lysosomal storage disorders (LSDs)2-4. For most LSDs, the causal genes have been identified but, in some, the function of the implicated gene is unknown, in part because lysosomes occupy a small fraction of the cellular volume so that changes in lysosomal contents are difficult to detect. Here we develop the LysoTag mouse for the tissue-specific isolation of intact lysosomes that are compatible with the multimodal profiling of their contents. We used the LysoTag mouse to study CLN3, a lysosomal transmembrane protein with an unknown function. In children, the loss of CLN3 causes juvenile neuronal ceroid lipofuscinosis (Batten disease), a lethal neurodegenerative LSD. Untargeted metabolite profiling of lysosomes from the brains of mice lacking CLN3 revealed a massive accumulation of glycerophosphodiesters (GPDs)-the end products of glycerophospholipid catabolism. GPDs also accumulate in the lysosomes of CLN3-deficient cultured cells and we show that CLN3 is required for their lysosomal egress. Loss of CLN3 also disrupts glycerophospholipid catabolism in the lysosome. Finally, we found elevated levels of glycerophosphoinositol in the cerebrospinal fluid of patients with Batten disease, suggesting the potential use of glycerophosphoinositol as a disease biomarker. Our results show that CLN3 is required for the lysosomal clearance of GPDs and reveal Batten disease as a neurodegenerative LSD with a defect in glycerophospholipid metabolism.
Collapse
Affiliation(s)
- Nouf N Laqtom
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- The Institute for Chemistry, Engineering & Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
| | - Wentao Dong
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- The Institute for Chemistry, Engineering & Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
| | - Uche N Medoh
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- The Institute for Chemistry, Engineering & Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Andrew L Cangelosi
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | | | - Sze Ham Chan
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Tenzin Kunchok
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | | | - Ivonne Heinze
- Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Rachel Tang
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Christian Grimm
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - An N Dang Do
- Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Forbes D Porter
- Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Alessandro Ori
- Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | | | - Monther Abu-Remaileh
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA.
- Department of Genetics, Stanford University, Stanford, CA, USA.
- The Institute for Chemistry, Engineering & Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA.
| |
Collapse
|
50
|
McCluskey G, Donaghy C, Morrison KE, McConville J, Duddy W, Duguez S. The Role of Sphingomyelin and Ceramide in Motor Neuron Diseases. J Pers Med 2022; 12:jpm12091418. [PMID: 36143200 PMCID: PMC9501626 DOI: 10.3390/jpm12091418] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS), Spinal Bulbar Muscular Atrophy (SBMA), and Spinal Muscular Atrophy (SMA) are motor neuron diseases (MNDs) characterised by progressive motor neuron degeneration, weakness and muscular atrophy. Lipid dysregulation is well recognised in each of these conditions and occurs prior to neurodegeneration. Several lipid markers have been shown to predict prognosis in ALS. Sphingolipids are complex lipids enriched in the central nervous system and are integral to key cellular functions including membrane stability and signalling pathways, as well as being mediators of neuroinflammation and neurodegeneration. This review highlights the metabolism of sphingomyelin (SM), the most abundant sphingolipid, and of its metabolite ceramide, and its role in the pathophysiology of neurodegeneration, focusing on MNDs. We also review published lipidomic studies in MNDs. In the 13 studies of patients with ALS, 12 demonstrated upregulation of multiple SM species and 6 demonstrated upregulation of ceramides. SM species also correlated with markers of clinical progression in five of six studies. These data highlight the potential use of SM and ceramide as biomarkers in ALS. Finally, we review potential therapeutic strategies for targeting sphingolipid metabolism in neurodegeneration.
Collapse
Affiliation(s)
- Gavin McCluskey
- Personalised Medicine Center, School of Medicine, Ulster University, Derry BT47 6SB, UK
- Department of Neurology, Altnagelvin Hospital, Derry, BT47 6SB, UK
- Department of Neurology, Royal Victoria Hospital, Belfast BT12 6BA, UK
| | - Colette Donaghy
- Department of Neurology, Altnagelvin Hospital, Derry, BT47 6SB, UK
| | - Karen E. Morrison
- Department of Neurology, Royal Victoria Hospital, Belfast BT12 6BA, UK
- Faculty of Medicine, Health & Life Sciences, Queen’s University, Belfast BT9 6AG, UK
| | - John McConville
- Department of Neurology, Royal Victoria Hospital, Belfast BT12 6BA, UK
- Department of Neurology, Ulster Hospital, Dundonald, Belfast BT16 1RH, UK
| | - William Duddy
- Personalised Medicine Center, School of Medicine, Ulster University, Derry BT47 6SB, UK
| | - Stephanie Duguez
- Personalised Medicine Center, School of Medicine, Ulster University, Derry BT47 6SB, UK
- Correspondence:
| |
Collapse
|