1
|
Gospodaryov DV. Alternative NADH dehydrogenase: A complex I backup, a drug target, and a tool for mitochondrial gene therapy. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2025; 1866:149529. [PMID: 39615731 DOI: 10.1016/j.bbabio.2024.149529] [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: 03/28/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024]
Abstract
Alternative NADH dehydrogenase, also known as type II NADH dehydrogenase (NDH-2), catalyzes the same redox reaction as mitochondrial respiratory chain complex I. Specifically, it oxidizes reduced nicotinamide adenine dinucleotide (NADH) while simultaneously reducing ubiquinone to ubiquinol. However, unlike complex I, this enzyme is non-proton pumping, comprises of a single subunit, and is resistant to rotenone. Initially identified in bacteria, fungi and plants, NDH-2 was subsequently discovered in protists and certain animal taxa including sea squirts. The gene coding for NDH-2 is also present in the genomes of some annelids, tardigrades, and crustaceans. For over two decades, NDH-2 has been investigated as a potential substitute for defective complex I. In model organisms, NDH-2 has been shown to ameliorate a broad spectrum of conditions associated with complex I malfunction, including symptoms of Parkinson's disease. Recently, lifespan extension has been observed in animals expressing NDH-2 in a heterologous manner. A variety of mechanisms have been put forward by which NDH-2 may extend lifespan. Such mechanisms include the activation of pro-longevity pathways through modulation of the NAD+/NADH ratio, decreasing production of reactive oxygen species (ROS) in mitochondria, or then through moderate increases in ROS production followed by activation of defense pathways (mitohormesis). This review gives an overview of the latest research on NDH-2, including the structural peculiarities of NDH-2, its inhibitors, its role in the pathogenicity of mycobacteria and apicomplexan parasites, and its function in bacteria, fungi, and animals.
Collapse
Affiliation(s)
- Dmytro V Gospodaryov
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka, 76018, Ivano-Frankivsk, Ukraine.
| |
Collapse
|
2
|
Bondy SC, Wu M. The Critical Role of Autophagy and Phagocytosis in the Aging Brain. Int J Mol Sci 2024; 26:57. [PMID: 39795916 PMCID: PMC11720579 DOI: 10.3390/ijms26010057] [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: 10/31/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025] Open
Abstract
As the organism ages, there is a decline in effective energy supply, and this retards the ability to elaborate new proteins. The consequences of this are especially marked in the gradual decline in brain function. The senescence of cells and their constituent organelles is ultimately the cause of aging of the entire nervous system. What is less immediately obvious is that brain aging is also accompanied by the failure of catabolic events that lead to the removal of non-functional cells and ineffective subcellular components. The removal of non-working cellular and subcellular elements within the brain is essential in order to allow the appearance of fresh cells and organelles with a full range of capacities. Thus, the maintenance of operative mechanisms for the dispersal of failed tissue components is important, and its diminished capacity with aging is a significant contributory factor to the onset and progression of age-related neurological disorder. This report discusses the mechanisms underlying autophagy and phagocytosis and how these can be adversely modulated as aging proceeds. The means by which the effective recycling of cellular components may be reinstated in the aged brain are considered.
Collapse
Affiliation(s)
- Stephen C. Bondy
- Department of Occupational and Environmental Health and Department of Medicine, University of California, Irvine, CA 92697, USA
| | - Meixia Wu
- Evergreen World ADHC, Westminster, CA 92844, USA;
| |
Collapse
|
3
|
Sun B, Zhang J, Wang N, Zhang Z, Wu Y, Xie M, Peng Y, Ye Y, Jiang Z, Wei S. The bioinformatics analysis and experimental validation of the carcinogenic role of EXO1 in lung adenocarcinoma. Front Oncol 2024; 14:1492725. [PMID: 39777332 PMCID: PMC11703735 DOI: 10.3389/fonc.2024.1492725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Background Exonuclease 1 (EXO1), a protein involved in mismatch repair and recombination processes, has been identified as a prognostic biomarker in lung adenocarcinoma (LUAD). Nevertheless, its role in LUAD progression remains elusive. This study seeks to elucidate the functional significance of EXO1 in LUAD and evaluate its potential as a therapeutic target. Materials and methods Patient RNA-seq and clinical data were acquired from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Subsequently, a protein-protein interaction (PPI) network was constructed using differentially expressed genes (DEGs) to identify pivotal genes. Validation of the expression of signature genes was carried out through quantitative real-time PCR (qRT-PCR). Additionally, the association between EXO1 expression and clinical data was investigated. Immunohistochemistry was utilized to assess EXO1 expression in 93 cases of invasive pulmonary adenocarcinoma. Finally, cellular functional assays were conducted to investigate the impact of EXO1 on LUAD cells. Results Ten key molecules (PBK, ASPM, NCAPG, EXO1, MKI67, RRM2, AURKA, DLGAP5, UBE2C, and CDC6) exhibited significantly elevated expression levels in LUAD tissues. Moreover, elevated levels of EXO1 gene expression correlated strongly with advanced T, N, and M stages and were significantly associated with immune cell infiltration in LUAD. Furthermore, marked increases in EXO1 protein expression were observed in patients diagnosed with invasive pulmonary adenocarcinoma. Notably, patients diagnosed with invasive pulmonary adenocarcinoma who exhibited elevated EXO1 expression levels exhibited increased lymph node metastasis, pleural invasion, poor tumor differentiation, and advanced clinical stage. Additionally, this study employed wound healing assay and CCK-8 cell proliferation assays to investigate the significant role of EXO1 in promoting the growth and migration of lung adenocarcinoma cells. Conclusions This study identified ten hub genes associated with the initiation and progression of LUAD. Additionally, EXO1 may serve as a prognostic marker for LUAD patients, offering new perspectives for clinical treatments.
Collapse
Affiliation(s)
- Bohao Sun
- Department of Pathology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jing Zhang
- Department of Pathology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Nan Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhirong Zhang
- Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Yichen Wu
- Department of Pathology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Mengzhen Xie
- Department of Pathology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yanmei Peng
- Department of Pathology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yifan Ye
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhaochang Jiang
- Department of Pathology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shumei Wei
- Department of Pathology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Hofer SJ, Daskalaki I, Bergmann M, Friščić J, Zimmermann A, Mueller MI, Abdellatif M, Nicastro R, Masser S, Durand S, Nartey A, Waltenstorfer M, Enzenhofer S, Faimann I, Gschiel V, Bajaj T, Niemeyer C, Gkikas I, Pein L, Cerrato G, Pan H, Liang Y, Tadic J, Jerkovic A, Aprahamian F, Robbins CE, Nirmalathasan N, Habisch H, Annerer E, Dethloff F, Stumpe M, Grundler F, Wilhelmi de Toledo F, Heinz DE, Koppold DA, Rajput Khokhar A, Michalsen A, Tripolt NJ, Sourij H, Pieber TR, de Cabo R, McCormick MA, Magnes C, Kepp O, Dengjel J, Sigrist SJ, Gassen NC, Sedej S, Madl T, De Virgilio C, Stelzl U, Hoffmann MH, Eisenberg T, Tavernarakis N, Kroemer G, Madeo F. Spermidine is essential for fasting-mediated autophagy and longevity. Nat Cell Biol 2024; 26:1571-1584. [PMID: 39117797 PMCID: PMC11392816 DOI: 10.1038/s41556-024-01468-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: 04/04/2024] [Accepted: 07/02/2024] [Indexed: 08/10/2024]
Abstract
Caloric restriction and intermittent fasting prolong the lifespan and healthspan of model organisms and improve human health. The natural polyamine spermidine has been similarly linked to autophagy enhancement, geroprotection and reduced incidence of cardiovascular and neurodegenerative diseases across species borders. Here, we asked whether the cellular and physiological consequences of caloric restriction and fasting depend on polyamine metabolism. We report that spermidine levels increased upon distinct regimens of fasting or caloric restriction in yeast, flies, mice and human volunteers. Genetic or pharmacological blockade of endogenous spermidine synthesis reduced fasting-induced autophagy in yeast, nematodes and human cells. Furthermore, perturbing the polyamine pathway in vivo abrogated the lifespan- and healthspan-extending effects, as well as the cardioprotective and anti-arthritic consequences of fasting. Mechanistically, spermidine mediated these effects via autophagy induction and hypusination of the translation regulator eIF5A. In summary, the polyamine-hypusination axis emerges as a phylogenetically conserved metabolic control hub for fasting-mediated autophagy enhancement and longevity.
Collapse
Affiliation(s)
- Sebastian J Hofer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
| | - Ioanna Daskalaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Heraklion, Greece
- Department of Biology, School of Sciences and Engineering, University of Crete, Heraklion, Greece
| | - Martina Bergmann
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Jasna Friščić
- Department of Dermatology, Allergy and Venerology, University of Lübeck, Lübeck, Germany
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Andreas Zimmermann
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Melanie I Mueller
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Mahmoud Abdellatif
- BioTechMed Graz, Graz, Austria
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Raffaele Nicastro
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Sarah Masser
- BioTechMed Graz, Graz, Austria
- Institute of Pharmaceutical Sciences, Pharmaceutical Chemistry, University of Graz, Graz, Austria
| | - Sylvère Durand
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
| | - Alexander Nartey
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Mara Waltenstorfer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Sarah Enzenhofer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Isabella Faimann
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Verena Gschiel
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Thomas Bajaj
- Neurohomeostasis Research Group, Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Christine Niemeyer
- Neurohomeostasis Research Group, Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Ilias Gkikas
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Heraklion, Greece
- Department of Biology, School of Sciences and Engineering, University of Crete, Heraklion, Greece
| | - Lukas Pein
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Giulia Cerrato
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
| | - Hui Pan
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
| | - YongTian Liang
- Institute for Biology and Genetics, Freie Universität Berlin, Berlin, Germany
- Cluster of Excellence, NeuroCure, Berlin, Germany
| | - Jelena Tadic
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Andrea Jerkovic
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Fanny Aprahamian
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
| | - Christine E Robbins
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Nitharsshini Nirmalathasan
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
| | - Hansjörg Habisch
- Research Unit Integrative Structural Biology, Otto Loewi Research Center, Medicinal Chemistry, Medical University of Graz, Graz, Austria
| | - Elisabeth Annerer
- Institute of Pharmaceutical Sciences, Pharmaceutical Chemistry, University of Graz, Graz, Austria
| | | | - Michael Stumpe
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | | | | | - Daniel E Heinz
- Neurohomeostasis Research Group, Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Daniela A Koppold
- Institute of Social Medicine, Epidemiology and Health Economics, corporate member of Freie Universität Berlin and Humboldt-Universität, Charité-Universitätsmedizin, Berlin, Germany
- Department of Pediatrics, Division of Oncology and Hematology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Internal Medicine and Nature-based Therapies, Immanuel Hospital Berlin, Berlin, Germany
| | - Anika Rajput Khokhar
- Institute of Social Medicine, Epidemiology and Health Economics, corporate member of Freie Universität Berlin and Humboldt-Universität, Charité-Universitätsmedizin, Berlin, Germany
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andreas Michalsen
- Institute of Social Medicine, Epidemiology and Health Economics, corporate member of Freie Universität Berlin and Humboldt-Universität, Charité-Universitätsmedizin, Berlin, Germany
- Department of Internal Medicine and Nature-based Therapies, Immanuel Hospital Berlin, Berlin, Germany
| | - Norbert J Tripolt
- Interdisciplinary Metabolic Medicine Trials Unit, Division of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Harald Sourij
- Interdisciplinary Metabolic Medicine Trials Unit, Division of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Thomas R Pieber
- BioTechMed Graz, Graz, Austria
- Interdisciplinary Metabolic Medicine Trials Unit, Division of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- HEALTH - Institute for Biomedical Research and Technologies, Joanneum Research Forschungsgesellschaft, Graz, Austria
| | - Rafael de Cabo
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Mark A McCormick
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Christoph Magnes
- HEALTH - Institute for Biomedical Research and Technologies, Joanneum Research Forschungsgesellschaft, Graz, Austria
| | - Oliver Kepp
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
| | - Joern Dengjel
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Stephan J Sigrist
- Institute for Biology and Genetics, Freie Universität Berlin, Berlin, Germany
- Cluster of Excellence, NeuroCure, Berlin, Germany
| | - Nils C Gassen
- Neurohomeostasis Research Group, Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Simon Sedej
- BioTechMed Graz, Graz, Austria
- Division of Cardiology, Medical University of Graz, Graz, Austria
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Tobias Madl
- BioTechMed Graz, Graz, Austria
- Research Unit Integrative Structural Biology, Otto Loewi Research Center, Medicinal Chemistry, Medical University of Graz, Graz, Austria
| | | | - Ulrich Stelzl
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- Institute of Pharmaceutical Sciences, Pharmaceutical Chemistry, University of Graz, Graz, Austria
| | - Markus H Hoffmann
- Department of Dermatology, Allergy and Venerology, University of Lübeck, Lübeck, Germany
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Tobias Eisenberg
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Heraklion, Greece.
- Division of Basic Sciences, School of Medicine, University of Crete, Heraklion, Greece.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France.
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria.
- Field of Excellence BioHealth, University of Graz, Graz, Austria.
- BioTechMed Graz, Graz, Austria.
| |
Collapse
|
5
|
Zhang P, Catterson JH, Grönke S, Partridge L. Inhibition of S6K lowers age-related inflammation and increases lifespan through the endolysosomal system. NATURE AGING 2024; 4:491-509. [PMID: 38413780 PMCID: PMC11031405 DOI: 10.1038/s43587-024-00578-3] [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] [Received: 09/09/2022] [Accepted: 01/24/2024] [Indexed: 02/29/2024]
Abstract
Suppression of target of rapamycin complex 1 (TORC1) by rapamycin ameliorates aging in diverse species. S6 kinase (S6K) is an essential mediator, but the mechanisms involved are unclear. Here we show that activation of S6K specifically in Drosophila fat-body blocked extension of lifespan by rapamycin, induced accumulation of multilamellar lysosomes and blocked age-associated hyperactivation of the NF-κB-like immune deficiency (IMD) pathway, indicative of reduced inflammaging. Syntaxin 13 mediated the effects of TORC1-S6K signaling on lysosome morphology and inflammaging, suggesting they may be linked. Inflammaging depended on the IMD receptor regulatory isoform PGRP-LC, and repression of the IMD pathway from midlife extended lifespan. Age-related inflammaging was higher in females than in males and was not lowered in males by rapamycin treatment or lowered S6K. Rapamycin treatment also elevated Syntaxin 12/13 levels in mouse liver and prevented age-related increase in noncanonical NF-κB signaling, suggesting that the effect of TORC1 on inflammaging is conserved from flies to mammals.
Collapse
Affiliation(s)
- Pingze Zhang
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - James H Catterson
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
- Centre for Discovery Brain Sciences, UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | | | - Linda Partridge
- Max Planck Institute for Biology of Ageing, Cologne, Germany.
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK.
| |
Collapse
|
6
|
Melicher D, Torson AS, Yocum GD, Bosch J, Kemp WP, Bowsher JH, Rinehart JP. Metabolic and transcriptomic characterization of summer and winter dormancy in the solitary bee, Osmia lignaria. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2024; 166:104074. [PMID: 38228213 DOI: 10.1016/j.ibmb.2024.104074] [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: 09/20/2023] [Revised: 01/08/2024] [Accepted: 01/13/2024] [Indexed: 01/18/2024]
Abstract
The solitary bee Osmia lignaria is a native pollinator in North America with growing economic importance. The life cycle of O. lignaria provides a unique opportunity to compare the physiological and molecular mechanisms underlying two ecologically contrasting dormancies within the same species. O. lignaria prepupae become dormant during the summer to avoid high temperatures. Shortly after adult eclosion, they enter a second dormancy and overwinter as diapausing adults. To compare these two dormancies, we measured metabolic rates and gene expression across development as bees initiate, maintain, and terminate both prepupal (summer) and adult (overwintering) dormancies. We observed a moderate temperature-independent decrease in gas exchange during both the prepupal dormancy after cocoon spinning (45 %) and during adult diapause after eclosion (60 %). We sequenced and assembled a high-quality reference genome from a single haploid male bee with a contiguous n50 of 5.5 Mbp to facilitate our transcriptomic analysis. The transcriptomes of dormant prepupae and diapausing adults clustered into distinct groups more closely associated with life stage than dormancy status. Membrane transport, membrane-bound cellular components, oxidoreductase activity, glutathione metabolism, and transcription factor activity increased during adult diapause, relative to prepupal dormancy. Further, the transcriptomes of adults in diapause clustered into two groups, supporting multiple phases of diapause during winter. Late adult diapause was associated with gene expression profiles supporting increased insulin/IGF, juvenile hormone, and ecdysone signaling.
Collapse
Affiliation(s)
- Dacotah Melicher
- Edward T. Schafer Agricultural Research Center, U.S. Department of Agriculture/Agricultural Research Service, 1616 Albrecht Boulevard North, Fargo, ND, 58102, USA
| | - Alex S Torson
- Edward T. Schafer Agricultural Research Center, U.S. Department of Agriculture/Agricultural Research Service, 1616 Albrecht Boulevard North, Fargo, ND, 58102, USA.
| | - George D Yocum
- Edward T. Schafer Agricultural Research Center, U.S. Department of Agriculture/Agricultural Research Service, 1616 Albrecht Boulevard North, Fargo, ND, 58102, USA
| | - Jordi Bosch
- CREAF, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - William P Kemp
- Edward T. Schafer Agricultural Research Center, U.S. Department of Agriculture/Agricultural Research Service, 1616 Albrecht Boulevard North, Fargo, ND, 58102, USA
| | - Julia H Bowsher
- Department of Biological Sciences, North Dakota State University, 1340 Bolley Drive, 218 Stevens Hall, Fargo, ND, 58102, USA
| | - Joseph P Rinehart
- Edward T. Schafer Agricultural Research Center, U.S. Department of Agriculture/Agricultural Research Service, 1616 Albrecht Boulevard North, Fargo, ND, 58102, USA
| |
Collapse
|
7
|
Qian Q, Niwa R. Endocrine Regulation of Aging in the Fruit Fly Drosophila melanogaster. Zoolog Sci 2024; 41:4-13. [PMID: 38587512 DOI: 10.2108/zs230056] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 08/16/2023] [Indexed: 04/09/2024]
Abstract
The past few decades have witnessed increasing research clarifying the role of endocrine signaling in the regulation of aging in both vertebrates and invertebrates. Studies using the model organism fruit fly Drosophila melanogaster have largely advanced our understanding of evolutionarily conserved mechanisms in the endocrinology of aging and anti-aging. Mutations in single genes involved in endocrine signaling modify lifespan, as do alterations of endocrine signaling in a tissue- or cell-specific manner, highlighting a central role of endocrine signaling in coordinating the crosstalk between tissues and cells to determine the pace of aging. Here, we review the current landscape of research in D. melanogaster that offers valuable insights into the endocrine-governed mechanisms which influence lifespan and age-related physiology.
Collapse
Affiliation(s)
- Qingyin Qian
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan
| | - Ryusuke Niwa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan,
| |
Collapse
|
8
|
de Magalhães JP. Distinguishing between driver and passenger mechanisms of aging. Nat Genet 2024; 56:204-211. [PMID: 38242993 DOI: 10.1038/s41588-023-01627-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 11/28/2023] [Indexed: 01/21/2024]
Abstract
Understanding why we age is a long-standing question, and many mechanistic theories of aging have been proposed. Owing to limitations in studying the aging process, including a lack of adequate quantitative measurements, its mechanistic basis remains a subject of debate. Here, I explore theories of aging from the perspective of causal relationships. Many aging-related changes have been observed and touted as drivers of aging, including molecular changes in the genome, telomeres, mitochondria, epigenome and proteins and cellular changes affecting stem cells, the immune system and senescent cell buildup. Determining which changes are drivers and not passengers of aging remains a challenge, however, and I discuss how animal models and human genetic studies have been used empirically to infer causality. Overall, our understanding of the drivers of human aging is still inadequate; yet with a global aging population, elucidating the causes of aging has the potential to revolutionize biomedical research.
Collapse
Affiliation(s)
- João Pedro de Magalhães
- Genomics of Ageing and Rejuvenation Lab, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.
| |
Collapse
|
9
|
Marzoog BA. Autophagy Behavior in Endothelial Cell Regeneration. Curr Aging Sci 2024; 17:58-67. [PMID: 37861048 DOI: 10.2174/0118746098260689231002044435] [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: 06/28/2023] [Revised: 08/16/2023] [Accepted: 08/25/2023] [Indexed: 10/21/2023]
Abstract
Autophagy plays a crucial role in maintaining endothelial cell homeostasis through the turnover of intracellular components during stress conditions in a lysosomal-dependent manner. The regeneration strategy involves several aspects, including autophagy. Autophagy is a catabolic degenerative lysosomal-dependent degradation of intracellular components. Autophagy modifies cellular and subcellular endothelial cell functions, including mitochondria stress, lysosomal stress, and endoplasmic reticulum unfolded protein response. Activation of common signaling pathways of autophagy and regeneration and enhancement of intracellular endothelial cell metabolism serve as the bases for the induction of endothelial regeneration. Endothelial progenitor cells include induced pluripotent stem cells (iPSC), embryonic stem cells, and somatic cells, such as fibroblasts. Future strategies of endothelial cell regeneration involve the induction of autophagy to minimize the metabolic degeneration of the endothelial cells and optimize the regeneration outcomes.
Collapse
Affiliation(s)
- Basheer Abdullah Marzoog
- World-Class Research Center «Digital Biodesign and Personalized Healthcare», I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119991, Russia
| |
Collapse
|
10
|
Wang Q, Su W, Liu J, Zhao D. Advances in the investigation of the role of autophagy in the etiology of chronic obstructive pulmonary disease: A review. Medicine (Baltimore) 2023; 102:e36390. [PMID: 38013266 PMCID: PMC10681501 DOI: 10.1097/md.0000000000036390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/09/2023] [Indexed: 11/29/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common chronic respiratory illness. It arises from emphysema and chronic bronchitis and is characterized by progressive and irreversible airflow limitation and chronic inflammation of the lungs, which eventually progresses to pulmonary hypertension, chronic pulmonary heart disease and respiratory failure. Autophagy is a highly conserved cellular homeostasis maintenance mechanism that involves the transport of damaged organelles and proteins to lysosomes for destruction. Dysregulation of autophagy is one of the pathogenic mechanisms of many diseases and is strongly associated with the development of COPD, although the precise mechanisms are unknown. In this paper, we focus on macroautophagy, a type of autophagy that has been thoroughly studied, and describe the characteristics, processes, regulatory pathways, and functions of autophagy, and discuss its relationship with COPD from the perspectives of inflammation, emphysema, mucus hypersecretion, cilia structure and function, airway remodeling, vascular remodeling, and bacterial infections, with a view to searching for the therapeutic targets of COPD from the perspective of autophagy, which is hoped to be helpful for the clinical treatment.
Collapse
Affiliation(s)
- Qianxinhong Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Wenlong Su
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Junnan Liu
- The Third Clinical Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Dongkai Zhao
- The Third Clinical Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| |
Collapse
|
11
|
Nithianandam V, Bukhari H, Leventhal MJ, Battaglia RA, Dong X, Fraenkel E, Feany MB. Integrative analysis reveals a conserved role for the amyloid precursor protein in proteostasis during aging. Nat Commun 2023; 14:7034. [PMID: 37923712 PMCID: PMC10624868 DOI: 10.1038/s41467-023-42822-1] [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: 04/18/2023] [Accepted: 10/23/2023] [Indexed: 11/06/2023] Open
Abstract
Aβ peptides derived from the amyloid precursor protein (APP) have been strongly implicated in the pathogenesis of Alzheimer's disease. However, the normal function of APP and the importance of that role in neurodegenerative disease is less clear. We recover the Drosophila ortholog of APP, Appl, in an unbiased forward genetic screen for neurodegeneration mutants. We perform comprehensive single cell transcriptional and proteomic studies of Appl mutant flies to investigate Appl function in the aging brain. We find an unexpected role for Appl in control of multiple cellular pathways, including translation, mitochondrial function, nucleic acid and lipid metabolism, cellular signaling and proteostasis. We mechanistically define a role for Appl in regulating autophagy through TGFβ signaling and document the broader relevance of our findings using mouse genetic, human iPSC and in vivo tauopathy models. Our results demonstrate a conserved role for APP in controlling age-dependent proteostasis with plausible relevance to Alzheimer's disease.
Collapse
Affiliation(s)
- Vanitha Nithianandam
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Hassan Bukhari
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Matthew J Leventhal
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, MA, USA
| | - Rachel A Battaglia
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Xianjun Dong
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Genomics and Bioinformatics Hub, Brigham and Women's Hospital, Boston, MA, USA
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
12
|
Bierlein M, Charles J, Polisuk-Balfour T, Bretscher H, Rice M, Zvonar J, Pohl D, Winslow L, Wasie B, Deurloo S, Van Wert J, Williams B, Ankney G, Harmon Z, Dann E, Azuz A, Guzman-Vargas A, Kuhns E, Neufeld TP, O’Connor MB, Amissah F, Zhu CC. Autophagy impairment and lifespan reduction caused by Atg1 RNAi or Atg18 RNAi expression in adult fruit flies (Drosophila melanogaster). Genetics 2023; 225:iyad154. [PMID: 37594076 PMCID: PMC11491525 DOI: 10.1093/genetics/iyad154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023] Open
Abstract
Autophagy, an autophagosome and lysosome-based eukaryotic cellular degradation system, has previously been implicated in lifespan regulation in different animal models. In this report, we show that expression of the RNAi transgenes targeting the transcripts of the key autophagy genes Atg1 or Atg18 in adult fly muscle or glia does not affect the overall levels of autophagosomes in those tissues and does not change the lifespan of the tested flies but the lifespan reduction phenotype has become apparent when Atg1 RNAi or Atg18 RNAi is expressed ubiquitously in adult flies or after autophagy is eradicated through the knockdown of Atg1 or Atg18 in adult fly adipocytes. Lifespan reduction was also observed when Atg1 or Atg18 was knocked down in adult fly enteroblasts and midgut stem cells. Overexpression of wild-type Atg1 in adult fly muscle or adipocytes reduces the lifespan and causes accumulation of high levels of ubiquitinated protein aggregates in muscles. Our research data have highlighted the important functions of the key autophagy genes in adult fly adipocytes, enteroblasts, and midgut stem cells and their undetermined roles in adult fly muscle and glia for lifespan regulation.
Collapse
Affiliation(s)
- Mariah Bierlein
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Joseph Charles
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | | | - Heidi Bretscher
- Department of Genetics, Cell Biology, and Developmental Biology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Micaela Rice
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Jacklyn Zvonar
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Drake Pohl
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Lindsey Winslow
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Brennah Wasie
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Sara Deurloo
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Jordan Van Wert
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Britney Williams
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Gabrielle Ankney
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Zachary Harmon
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Erica Dann
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Anna Azuz
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Alex Guzman-Vargas
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Elizabeth Kuhns
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Thomas P Neufeld
- Department of Genetics, Cell Biology, and Developmental Biology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael B O’Connor
- Department of Genetics, Cell Biology, and Developmental Biology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Felix Amissah
- School of Pharmacy, Ferris State University, Big Rapids, MI 49307, USA
| | - Changqi C Zhu
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| |
Collapse
|
13
|
Liu S, Yao S, Yang H, Liu S, Wang Y. Autophagy: Regulator of cell death. Cell Death Dis 2023; 14:648. [PMID: 37794028 PMCID: PMC10551038 DOI: 10.1038/s41419-023-06154-8] [Citation(s) in RCA: 247] [Impact Index Per Article: 123.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 09/05/2023] [Accepted: 09/14/2023] [Indexed: 10/06/2023]
Abstract
Autophagy is the process by which cells degrade and recycle proteins and organelles to maintain intracellular homeostasis. Generally, autophagy plays a protective role in cells, but disruption of autophagy mechanisms or excessive autophagic flux usually leads to cell death. Despite recent progress in the study of the regulation and underlying molecular mechanisms of autophagy, numerous questions remain to be answered. How does autophagy regulate cell death? What are the fine-tuned regulatory mechanisms underlying autophagy-dependent cell death (ADCD) and autophagy-mediated cell death (AMCD)? In this article, we highlight the different roles of autophagy in cell death and discuss six of the main autophagy-related cell death modalities, with a focus on the metabolic changes caused by excessive endoplasmic reticulum-phagy (ER-phagy)-induced cell death and the role of mitophagy in autophagy-mediated ferroptosis. Finally, we discuss autophagy enhancement in the treatment of diseases and offer a new perspective based on the use of autophagy for different functional conversions (including the conversion of autophagy and that of different autophagy-mediated cell death modalities) for the clinical treatment of tumors.
Collapse
Affiliation(s)
- ShiZuo Liu
- School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - ShuaiJie Yao
- School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Huan Yang
- The Second School of Clinical Medicine, Xinjiang Medical University, Urumqi, China
| | - ShuaiJie Liu
- School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - YanJiao Wang
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
14
|
Sharma R, Diwan B. Lipids and the hallmarks of ageing: From pathology to interventions. Mech Ageing Dev 2023; 215:111858. [PMID: 37652278 DOI: 10.1016/j.mad.2023.111858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/21/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
Lipids are critical structural and functional architects of cellular homeostasis. Change in systemic lipid profile is a clinical indicator of underlying metabolic pathologies, and emerging evidence is now defining novel roles of lipids in modulating organismal ageing. Characteristic alterations in lipid metabolism correlate with age, and impaired systemic lipid profile can also accelerate the development of ageing phenotype. The present work provides a comprehensive review of the extent of lipids as regulators of the modern hallmarks of ageing viz., cellular senescence, chronic inflammation, gut dysbiosis, telomere attrition, genome instability, proteostasis and autophagy, epigenetic alterations, and stem cells dysfunctions. Current evidence on the modulation of each of these hallmarks has been discussed with emphasis on inherent age-dependent deficiencies in lipid metabolism as well as exogenous lipid changes. There appears to be sufficient evidence to consider impaired lipid metabolism as key driver of the ageing process although much of knowledge is yet fragmented. Considering dietary lipids, the type and quantity of lipids in the diet is a significant, but often overlooked determinant that governs the effects of lipids on ageing. Further research using integrative approaches amidst the known aging hallmarks is highly desirable for understanding the therapeutics of lipids associated with ageing.
Collapse
Affiliation(s)
- Rohit Sharma
- Nutrigerontology Laboratory, Faculty of Applied Sciences & Biotechnology, Shoolini University, Solan 173229, India.
| | - Bhawna Diwan
- Nutrigerontology Laboratory, Faculty of Applied Sciences & Biotechnology, Shoolini University, Solan 173229, India
| |
Collapse
|
15
|
Morón-Oset J, Fischer LK, Carcolé M, Giblin A, Zhang P, Isaacs AM, Grönke S, Partridge L. Toxicity of C9orf72-associated dipeptide repeat peptides is modified by commonly used protein tags. Life Sci Alliance 2023; 6:e202201739. [PMID: 37308278 PMCID: PMC10262077 DOI: 10.26508/lsa.202201739] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 06/01/2023] [Accepted: 06/03/2023] [Indexed: 06/14/2023] Open
Abstract
Hexanucleotide repeat expansions in the C9orf72 gene are the most prevalent genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia. Transcripts of the expansions are translated into toxic dipeptide repeat (DPR) proteins. Most preclinical studies in cell and animal models have used protein-tagged polyDPR constructs to investigate DPR toxicity but the effects of tags on DPR toxicity have not been systematically explored. Here, we used Drosophila to assess the influence of protein tags on DPR toxicity. Tagging of 36 but not 100 arginine-rich DPRs with mCherry increased toxicity, whereas adding mCherry or GFP to GA100 completely abolished toxicity. FLAG tagging also reduced GA100 toxicity but less than the longer fluorescent tags. Expression of untagged but not GFP- or mCherry-tagged GA100 caused DNA damage and increased p62 levels. Fluorescent tags also affected GA100 stability and degradation. In summary, protein tags affect DPR toxicity in a tag- and DPR-dependent manner, and GA toxicity might be underestimated in studies using tagged GA proteins. Thus, including untagged DPRs as controls is important when assessing DPR toxicity in preclinical models.
Collapse
Affiliation(s)
| | | | - Mireia Carcolé
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, UCL Queen Square Institute of Neurology, London, UK
| | - Ashling Giblin
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, UCL Queen Square Institute of Neurology, London, UK
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, UK
| | - Pingze Zhang
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Adrian M Isaacs
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, UCL Queen Square Institute of Neurology, London, UK
| | | | - Linda Partridge
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, UK
| |
Collapse
|
16
|
Wilson N, Kataura T, Korsgen ME, Sun C, Sarkar S, Korolchuk VI. The autophagy-NAD axis in longevity and disease. Trends Cell Biol 2023; 33:788-802. [PMID: 36878731 DOI: 10.1016/j.tcb.2023.02.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 03/07/2023]
Abstract
Autophagy is an intracellular degradation pathway that recycles subcellular components to maintain metabolic homeostasis. NAD is an essential metabolite that participates in energy metabolism and serves as a substrate for a series of NAD+-consuming enzymes (NADases), including PARPs and SIRTs. Declining levels of autophagic activity and NAD represent features of cellular ageing, and consequently enhancing either significantly extends health/lifespan in animals and normalises metabolic activity in cells. Mechanistically, it has been shown that NADases can directly regulate autophagy and mitochondrial quality control. Conversely, autophagy has been shown to preserve NAD levels by modulating cellular stress. In this review we highlight the mechanisms underlying this bidirectional relationship between NAD and autophagy, and the potential therapeutic targets it provides for combatting age-related disease and promoting longevity.
Collapse
Affiliation(s)
- Niall Wilson
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Tetsushi Kataura
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Miriam E Korsgen
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Congxin Sun
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Sovan Sarkar
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | - Viktor I Korolchuk
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK.
| |
Collapse
|
17
|
Morón-Oset J, Fischer LKS, Jauré N, Zhang P, Jahn AJ, Supèr T, Pahl A, Isaacs AM, Grönke S, Partridge L. Repeat length of C9orf72-associated glycine-alanine polypeptides affects their toxicity. Acta Neuropathol Commun 2023; 11:140. [PMID: 37644512 PMCID: PMC10463776 DOI: 10.1186/s40478-023-01634-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 08/05/2023] [Indexed: 08/31/2023] Open
Abstract
G4C2 hexanucleotide repeat expansions in a non-coding region of the C9orf72 gene are the most common cause of familial amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). G4C2 insertion length is variable, and patients can carry up to several thousand repeats. Dipeptide repeat proteins (DPRs) translated from G4C2 transcripts are thought to be a main driver of toxicity. Experiments in model organisms with relatively short DPRs have shown that arginine-rich DPRs are most toxic, while polyGlycine-Alanine (GA) DPRs cause only mild toxicity. However, GA is the most abundant DPR in patient brains, and experimental work in animals has generally relied on the use of low numbers of repeats, with DPRs often tagged for in vivo tracking. Whether repeat length or tagging affect the toxicity of GA has not been systematically assessed. Therefore, we generated Drosophila fly lines expressing GA100, GA200 or GA400 specifically in adult neurons. Consistent with previous studies, expression of GA100 and GA200 caused only mild toxicity. In contrast, neuronal expression of GA400 drastically reduced climbing ability and survival of flies, indicating that long GA DPRs can be highly toxic in vivo. This toxicity could be abolished by tagging GA400. Proteomics analysis of fly brains showed a repeat-length-dependent modulation of the brain proteome, with GA400 causing earlier and stronger changes than shorter GA proteins. PolyGA expression up-regulated proteins involved in ER to Golgi trafficking, and down-regulated proteins involved in insulin signalling. Experimental down-regulation of Tango1, a highly conserved regulator of ER-to Golgi transport, partially rescued GA400 toxicity, suggesting that misregulation of this process contributes to polyGA toxicity. Experimentally increasing insulin signaling also rescued GA toxicity. In summary, our data show that long polyGA proteins can be highly toxic in vivo, and that they may therefore contribute to ALS/FTD pathogenesis in patients.
Collapse
Affiliation(s)
- Javier Morón-Oset
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9B, 50931, Cologne, Germany
| | | | - Nathalie Jauré
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9B, 50931, Cologne, Germany
| | - Pingze Zhang
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9B, 50931, Cologne, Germany
| | - Annika Julia Jahn
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9B, 50931, Cologne, Germany
| | - Tessa Supèr
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9B, 50931, Cologne, Germany
| | - André Pahl
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9B, 50931, Cologne, Germany
| | - Adrian M Isaacs
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Sebastian Grönke
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9B, 50931, Cologne, Germany.
| | - Linda Partridge
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9B, 50931, Cologne, Germany.
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, Darwin Building, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
18
|
Sporbeck K, Haas ML, Pastor-Maldonado CJ, Schüssele DS, Hunter C, Takacs Z, Diogo de Oliveira AL, Franz-Wachtel M, Charsou C, Pfisterer SG, Gubas A, Haller PK, Knorr RL, Kaulich M, Macek B, Eskelinen EL, Simonsen A, Proikas-Cezanne T. The ABL-MYC axis controls WIPI1-enhanced autophagy in lifespan extension. Commun Biol 2023; 6:872. [PMID: 37620393 PMCID: PMC10449903 DOI: 10.1038/s42003-023-05236-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 08/10/2023] [Indexed: 08/26/2023] Open
Abstract
Human WIPI β-propellers function as PI3P effectors in autophagy, with WIPI4 and WIPI3 being able to link autophagy control by AMPK and TORC1 to the formation of autophagosomes. WIPI1, instead, assists WIPI2 in efficiently recruiting the ATG16L1 complex at the nascent autophagosome, which in turn promotes lipidation of LC3/GABARAP and autophagosome maturation. However, the specific role of WIPI1 and its regulation are unknown. Here, we discovered the ABL-ERK-MYC signalling axis controlling WIPI1. As a result of this signalling, MYC binds to the WIPI1 promoter and represses WIPI1 gene expression. When ABL-ERK-MYC signalling is counteracted, increased WIPI1 gene expression enhances the formation of autophagic membranes capable of migrating through tunnelling nanotubes to neighbouring cells with low autophagic activity. ABL-regulated WIPI1 function is relevant to lifespan control, as ABL deficiency in C. elegans increased gene expression of the WIPI1 orthologue ATG-18 and prolonged lifespan in a manner dependent on ATG-18. We propose that WIPI1 acts as an enhancer of autophagy that is physiologically relevant for regulating the level of autophagic activity over the lifespan.
Collapse
Affiliation(s)
- Katharina Sporbeck
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Maximilian L Haas
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Carmen J Pastor-Maldonado
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - David S Schüssele
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Catherine Hunter
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Zsuzsanna Takacs
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- Institute of Molecular Biotechnology, A-1030, Vienna, Austria
| | - Ana L Diogo de Oliveira
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Mirita Franz-Wachtel
- Proteome Center Tübingen, Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Chara Charsou
- Institute of Basic Medical Sciences, University of Oslo, 0372, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 0316, Oslo, Norway
| | - Simon G Pfisterer
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- Department of Anatomy, Faculty of Medicine, University of Helsinki, FI-00290, Helsinki, Finland
| | - Andrea Gubas
- Institute of Biochemistry II, Frankfurt Cancer Institute, Goethe University Medical School, D-60590, Frankfurt, Germany
| | - Patricia K Haller
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Roland L Knorr
- Humboldt University of Berlin, Institute of Biology, D-10115, Berlin, Germany
- Graduate School and Faculty of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
- International Research Frontiers Initiative, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, 226-8503, Japan
| | - Manuel Kaulich
- Institute of Biochemistry II, Frankfurt Cancer Institute, Goethe University Medical School, D-60590, Frankfurt, Germany
| | - Boris Macek
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- Proteome Center Tübingen, Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Eeva-Liisa Eskelinen
- Department of Biosciences, University of Helsinki, Fl-00790, Helsinki, Finland
- Institute of Biomedicine, University of Turku, FI-20520, Turku, Finland
| | - Anne Simonsen
- Institute of Basic Medical Sciences, University of Oslo, 0372, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 0316, Oslo, Norway
| | - Tassula Proikas-Cezanne
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany.
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany.
| |
Collapse
|
19
|
Demir E, Kacew S. Drosophila as a Robust Model System for Assessing Autophagy: A Review. TOXICS 2023; 11:682. [PMID: 37624187 PMCID: PMC10458868 DOI: 10.3390/toxics11080682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/07/2023] [Accepted: 08/07/2023] [Indexed: 08/26/2023]
Abstract
Autophagy is the process through which a body breaks down and recycles its own cellular components, primarily inside lysosomes. It is a cellular response to starvation and stress, which plays decisive roles in various biological processes such as senescence, apoptosis, carcinoma, and immune response. Autophagy, which was first discovered as a survival mechanism during starvation in yeast, is now known to serve a wide range of functions in more advanced organisms. It plays a vital role in how cells respond to stress, starvation, and infection. While research on yeast has led to the identification of many key components of the autophagy process, more research into autophagy in more complex systems is still warranted. This review article focuses on the use of the fruit fly Drosophila melanogaster as a robust testing model in further research on autophagy. Drosophila provides an ideal environment for exploring autophagy in a living organism during its development. Additionally, Drosophila is a well-suited compact tool for genetic analysis in that it serves as an intermediate between yeast and mammals because evolution conserved the molecular machinery required for autophagy in this species. Experimental tractability of host-pathogen interactions in Drosophila also affords great convenience in modeling human diseases on analogous structures and tissues.
Collapse
Affiliation(s)
- Esref Demir
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
- Medical Laboratory Techniques Program, Department of Medical Services and Techniques, Vocational School of Health Services, Antalya Bilim University, 07190 Antalya, Turkey
| | - Sam Kacew
- R. Samuel McLaughllin Center for Population Health Risk Assessment, Institute of Population Health, University of Ottawa, 1 Stewart (320), Ottawa, ON K1N 6N5, Canada;
| |
Collapse
|
20
|
Metaxakis A, Pavlidis M, Tavernarakis N. Neuronal atg1 Coordinates Autophagy Induction and Physiological Adaptations to Balance mTORC1 Signalling. Cells 2023; 12:2024. [PMID: 37626835 PMCID: PMC10453232 DOI: 10.3390/cells12162024] [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: 07/11/2023] [Revised: 08/04/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
The mTORC1 nutrient-sensing pathway integrates metabolic and endocrine signals into the brain to evoke physiological responses to food deprivation, such as autophagy. Nevertheless, the impact of neuronal mTORC1 activity on neuronal circuits and organismal metabolism remains obscure. Here, we show that mTORC1 inhibition acutely perturbs serotonergic neurotransmission via proteostatic alterations evoked by the autophagy inducer atg1. Neuronal ATG1 alters the intracellular localization of the serotonin transporter, which increases the extracellular serotonin and stimulates the 5HTR7 postsynaptic receptor. 5HTR7 enhances food-searching behaviour and ecdysone-induced catabolism in Drosophila. Along similar lines, the pharmacological inhibition of mTORC1 in zebrafish also stimulates food-searching behaviour via serotonergic activity. These effects occur in parallel with neuronal autophagy induction, irrespective of the autophagic activity and the protein synthesis reduction. In addition, ectopic neuronal atg1 expression enhances catabolism via insulin pathway downregulation, impedes peptidergic secretion, and activates non-cell autonomous cAMP/PKA. The above exert diverse systemic effects on organismal metabolism, development, melanisation, and longevity. We conclude that neuronal atg1 aligns neuronal autophagy induction with distinct physiological modulations, to orchestrate a coordinated physiological response against reduced mTORC1 activity.
Collapse
Affiliation(s)
- Athanasios Metaxakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
| | - Michail Pavlidis
- Department of Biology, University of Crete, 71409 Heraklion, Crete, Greece;
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
- Department of Basic Sciences, Faculty of Medicine, University of Crete, 71110 Heraklion, Crete, Greece
| |
Collapse
|
21
|
Yin Z, Zhang Z, Lei Y, Klionsky DJ. Bidirectional roles of the Ccr4-Not complex in regulating autophagy before and after nitrogen starvation. Autophagy 2023; 19:415-425. [PMID: 35167422 PMCID: PMC9851207 DOI: 10.1080/15548627.2022.2036476] [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: 10/01/2021] [Revised: 01/12/2022] [Accepted: 01/28/2022] [Indexed: 01/22/2023] Open
Abstract
Macroautophagy/autophagy is a highly conserved catabolic process by which cytoplasmic constituents are delivered to the vacuole/lysosome for degradation and recycling. To maintain cellular homeostasis and prevent pathologies, the induction and amplitude of autophagy activity are finely controlled through regulation of ATG gene expression. Here we report that the Ccr4-Not complex in Saccharomyces cerevisiae has bidirectional roles in regulating autophagy before and after nutrient deprivation. Under nutrient-rich conditions, Ccr4-Not directly targets the mRNAs of several ATG genes in the core autophagy machinery to promote their degradation through deadenylation, thus contributing to maintaining autophagy at the basal level. Upon starvation, Ccr4-Not releases its repression of these ATG genes and switches its role to promote the expression of a different subset of ATG genes, which is required for sufficient autophagy induction and activity. These results reveal that the Ccr4-Not complex is indispensable to maintain autophagy at the appropriate amplitude in both basal and stress conditions.Abbreviations: AID, auxin-inducible degron; Ape1, aminopeptidase I; Atg, autophagy related; Cvt, cytoplasm-to-vacuole targeting; DMSO, dimethyl sulfoxide; IAA, indole-3-acetic acid; PA, protein A; RIP, RNA immunoprecipitation.
Collapse
Affiliation(s)
- Zhangyuan Yin
- Life Sciences Institute, and the Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Zhihai Zhang
- Life Sciences Institute, and the Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Yuchen Lei
- Life Sciences Institute, and the Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Daniel J. Klionsky
- Life Sciences Institute, and the Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
22
|
Mishra S, Raval M, Kachhawaha AS, Tiwari BS, Tiwari AK. Aging: Epigenetic modifications. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 197:171-209. [PMID: 37019592 DOI: 10.1016/bs.pmbts.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
Abstract
Aging is one of the most complex and irreversible health conditions characterized by continuous decline in physical/mental activities that eventually poses an increased risk of several diseases and ultimately death. These conditions cannot be ignored by anyone but there are evidences that suggest that exercise, healthy diet and good routines may delay the Aging process significantly. Several studies have demonstrated that Epigenetics plays a key role in Aging and Aging-associated diseases through methylation of DNA, histone modification and non-coding RNA (ncRNA). Comprehension and relevant alterations in these epigenetic modifications can lead to new therapeutic avenues of age-delaying contrivances. These processes affect gene transcription, DNA replication and DNA repair, comprehending epigenetics as a key factor in understanding Aging and developing new avenues for delaying Aging, clinical advancements in ameliorating aging-related diseases and rejuvenating health. In the present article, we have described and advocated the epigenetic role in Aging and associated diseases.
Collapse
|
23
|
Zhang Y, Zhang J, Fu Z. Role of autophagy in lung diseases and ageing. Eur Respir Rev 2022; 31:31/166/220134. [PMID: 36543345 PMCID: PMC9879344 DOI: 10.1183/16000617.0134-2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 11/08/2022] [Indexed: 12/24/2022] Open
Abstract
The lungs face ongoing chemical, mechanical, biological, immunological and xenobiotic stresses over a lifetime. Advancing age progressively impairs lung function. Autophagy is a "housekeeping" survival strategy involved in numerous physiological and pathological processes in all eukaryotic cells. Autophagic activity decreases with age in several species, whereas its basic activity extends throughout the lifespan of most animals. Dysregulation of autophagy has been proven to be closely related to the pathogenesis of several ageing-related pulmonary diseases. This review summarises the role of autophagy in the pathogenesis of pulmonary diseases associated with or occurring in the context of ageing, including acute lung injury, chronic obstructive pulmonary disease, asthma and pulmonary fibrosis, and describes its potential as a therapeutic target.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jin Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhiling Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China,Corresponding author: Zhiling Fu ()
| |
Collapse
|
24
|
Tabibzadeh S. Role of autophagy in aging: The good, the bad, and the ugly. Aging Cell 2022; 22:e13753. [PMID: 36539927 PMCID: PMC9835585 DOI: 10.1111/acel.13753] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/10/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Autophagy (self-eating) is a conserved catabolic homeostatic process required for cellular metabolic demands by removal of the damaged molecules and organelles and for alleviation of stress initiated by pathology and infection. By such actions, autophagy is essential for the prevention of aging, disease, and cancer. Genetic defects of autophagy genes lead to a host of developmental, metabolic, and pathological aberrations. Similarly, the age-induced decline in autophagy leads to the loss of cellular homeostatic control. Paradoxically, such a valuable mechanism is hijacked by diseases, during tumor progression and by senescence, presumably due to high levels of metabolic demand. Here, we review both the role of autophagy in preventing cellular decline in aging by fulfillment of cellular bioenergetic demands and its contribution to the maintenance of the senescent state and SASP by acting on energy and nutritional sensors and diverse signaling pathways.
Collapse
Affiliation(s)
- Siamak Tabibzadeh
- Frontiers in Bioscience Research Institute in Aging and CancerIrvineCaliforniaUSA
| |
Collapse
|
25
|
Wang Q, Zhang J, Zhuang J, Shen F, Zhao M, Du J, Yu P, Zhong H, Feng F. Soft-Shelled Turtle Peptides Extend Lifespan and Healthspan in Drosophila. Nutrients 2022; 14:nu14245205. [PMID: 36558363 PMCID: PMC9781693 DOI: 10.3390/nu14245205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/02/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022] Open
Abstract
In traditional Chinese medicine, soft-shelled turtle protein and peptides serve as a nutraceutical for prolonging the lifespan. However, their effects on anti-aging have not been clarified scientifically in vivo. This study aimed to determine whether soft-shelled turtle peptides (STP) could promote the lifespan and healthspan in Drosophila melanogaster and the underlying molecular mechanisms. Herein, STP supplementation prolonged the mean lifespan by 20.23% and 9.04% in males and females, respectively, delaying the aging accompanied by climbing ability decline, enhanced gut barrier integrity, and improved anti-oxidation, starvation, and heat stress abilities, while it did not change the daily food intake. Mechanistically, STP enhanced autophagy and decreased oxidative stress by downregulating the target of rapamycin (TOR) signaling pathway. In addition, 95.18% of peptides from the identified sequences in STP could exert potential inhibitory effects on TOR through hydrogen bonds, van der Walls, hydrophobic interactions, and electrostatic interactions. The current study could provide a theoretical basis for the full exploitation of soft-shelled turtle aging prevention.
Collapse
Affiliation(s)
- Qianqian Wang
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Junhui Zhang
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Jiachen Zhuang
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Fei Shen
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Minjie Zhao
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Juan Du
- Zhejiang Nuoyan Biotechnology Co., Ltd., Huzhou 313000, China
| | - Peng Yu
- Yuyao Lengjiang Turtle Industry, Ningbo 315400, China
| | - Hao Zhong
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
- Correspondence: (H.Z.); (F.F.)
| | - Fengqin Feng
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
- Correspondence: (H.Z.); (F.F.)
| |
Collapse
|
26
|
González-Rodríguez P, Cheray M, Keane L, Engskog-Vlachos P, Joseph B. ULK3-dependent activation of GLI1 promotes DNMT3A expression upon autophagy induction. Autophagy 2022; 18:2769-2780. [PMID: 35226587 PMCID: PMC9673947 DOI: 10.1080/15548627.2022.2039993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Macroautophagy/autophagy is a tightly regulated catabolic process, which contributes at baseline level to cellular homeostasis, and upon its stimulation to the adaptive cellular response to intra- and extracellular stress stimuli. Decrease of autophagy activity is occurring upon aging and thought to contribute to age-related-diseases. Recently, we uncovered, upon autophagy induction, the role of de novo DNMT3A (DNA methyltransferase 3 alpha)-mediated DNA methylation on expression of the MAP1LC3 (microtubule associated protein 1 light chain 3) proteins, core components of the autophagy pathway, which resulted in reduced baseline autophagy activity. Here, we report that serine/threonine kinase ULK3 (unc-51 like kinase 3)-dependent activation of GLI1 (GLI family zinc finger 1) contributes to the transcriptional upregulation of DNMT3A gene expression upon autophagy induction, thereby bringing additional understanding of the long-term effect of autophagy induction and a possible mechanism for its decline upon aging, pathological conditions, or in response to treatment interventions.Abbreviations: CBZ: carbamazepine; ChIP: chromatin immunoprecipitation; Clon: clonidine; DNMT3A: DNA methyltransferase 3 alpha; GLI1: GLI family zinc finger 1; GLI2: GLI family zinc finger 2; MAP1LC3: microtubule associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; PLA: proximity ligation assay; RT-qPCR: quantitative reverse transcription PCR; shRNA: small hairpin RNA; siRNA: small interfering RNA; Treh: trehalose; ULK3: unc-51 like kinase 3.
Collapse
Affiliation(s)
| | - Mathilde Cheray
- Institute of Environmental MedicineToxicology Unit, Karolinska Institutet, StockholmSweden
| | - Lily Keane
- Institute of Environmental MedicineToxicology Unit, Karolinska Institutet, StockholmSweden
| | | | - Bertrand Joseph
- Institute of Environmental MedicineToxicology Unit, Karolinska Institutet, StockholmSweden,Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway,CONTACT Bertrand Joseph Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm17177, Sweden; Division of Biochemistry, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
27
|
Hofer SJ, Simon AK, Bergmann M, Eisenberg T, Kroemer G, Madeo F. Mechanisms of spermidine-induced autophagy and geroprotection. NATURE AGING 2022; 2:1112-1129. [PMID: 37118547 DOI: 10.1038/s43587-022-00322-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 10/28/2022] [Indexed: 04/30/2023]
Abstract
Aging involves the systemic deterioration of all known cell types in most eukaryotes. Several recently discovered compounds that extend the healthspan and lifespan of model organisms decelerate pathways that govern the aging process. Among these geroprotectors, spermidine, a natural polyamine ubiquitously found in organisms from all kingdoms, prolongs the lifespan of fungi, nematodes, insects and rodents. In mice, it also postpones the manifestation of various age-associated disorders such as cardiovascular disease and neurodegeneration. The specific features of spermidine, including its presence in common food items, make it an interesting candidate for translational aging research. Here, we review novel insights into the geroprotective mode of action of spermidine at the molecular level, as we discuss strategies for elucidating its clinical potential.
Collapse
Affiliation(s)
- Sebastian J Hofer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Anna Katharina Simon
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Max Delbrück Center, Berlin, Germany
| | - Martina Bergmann
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Tobias Eisenberg
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria.
- Field of Excellence BioHealth, University of Graz, Graz, Austria.
- BioTechMed Graz, Graz, Austria.
| |
Collapse
|
28
|
Singh A, Agrawal N. Progressive transcriptional changes in metabolic genes and altered fatbody homeostasis in Drosophila model of Huntington's disease. Metab Brain Dis 2022; 37:2783-2792. [PMID: 36121619 DOI: 10.1007/s11011-022-01078-2] [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: 06/10/2022] [Accepted: 08/30/2022] [Indexed: 10/14/2022]
Abstract
Huntington's disease (HD) is an autosomal-dominant neurodegenerative disorder marked by progressive neuronal atrophy, particularly in striatum and cerebral cortex. Although predominant manifestations of the disease include loss in the triad of motor, cognitive and behavioral capabilities, metabolic dysfunction in patients and HD models are being increasingly recognized. Patients display progressive body weight loss, which aggravates the disease and leads to cachexia in the terminal stages. Using the Drosophila model of HD, we have earlier reported that diseased flies exhibit an atypical pattern of lipid gain and loss with progression along with exhibiting extensive mitochondrial dysfunction, impaired calcium homeostasis and heightened apoptosis in the fatbody. Here, we first monitored the structural changes that abdominal fatbody undergoes with disease progression. Further, we checked the transcriptional changes of key metabolic genes in whole fly as well as genes regulating mitochondrial function, apoptosis, autophagy and calcium homeostasis in the abdominal fatbody. We found extensive alterations in whole-body and fatbody-specific transcriptional profile of the diseased flies, which was in consort with their stage-specific physiological state. Additionally, we also assessed lysosome-mediated autophagy in the fatbody of diseased flies in order to ascertain the mechanisms contributing to fatbody atrophy at the terminal stage. Interestingly, we found elevated autophagy in fatbody of flies throughout disease progression. This study provides new insights into the effect on peripheral metabolism due to degeneration of neurons in the neurodegenerative disease, thereby discerns novel mechanisms leading to cachexia in diseased flies and advocates for the need of managing metabolic dysfunctions in HD.
Collapse
Affiliation(s)
- Akanksha Singh
- Department of Zoology, University of Delhi, 110007, New Delhi, India
| | - Namita Agrawal
- Department of Zoology, University of Delhi, 110007, New Delhi, India.
| |
Collapse
|
29
|
Duan H, Li J, Yu L, Fan L. The road ahead of dietary restriction on anti-aging: focusing on personalized nutrition. Crit Rev Food Sci Nutr 2022; 64:891-908. [PMID: 35950606 DOI: 10.1080/10408398.2022.2110034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Dietary restriction (DR), including caloric restriction (CR), intermittent fasting (IF), and restriction of specific food compositions, can delay aging, and the main mechanisms include regulation of nutrient-sensing pathways and gut microbiota. However, the effects of DR regimens on longevity remain controversial, as some studies have demonstrated that IF, rather than CR or diet composition, influences longevity, while other studies have shown that the restricted-carbohydrate or -protein diets, rather than CR, determine health and longevity. Many factors, including DR-related factors (carbohydrate or protein composition, degree and duration of DR), and individual differences (health status, sex, genotype, and age of starting DR), would be used to explain the controversial anti-aging effects of DR, thus highlighting the necessity of precise DR intervention for anti-aging. Personalized DR intervention in humans is challenging because of the lack of accurate aging molecular biomarkers and vast individual variability. Using machine learning to build a predictive model based on the data set of clinical features, gut microbiome and metabolome, may be a good method to achieve precise DR intervention. Therefore, this review analyzed the anti-aging effects of various DR regimens, summarized their mechanisms and influencing factors, and proposed a future research direction for achieving personalized DR regimens for slowing aging.
Collapse
Affiliation(s)
- Hui Duan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Jinwei Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Research Laboratory for Probiotics at, Jiangnan University, Wuxi, China
| | - Liuping Fan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| |
Collapse
|
30
|
Duan H, Pan J, Guo M, Li J, Yu L, Fan L. Dietary strategies with anti-aging potential: dietary patterns and supplements. Food Res Int 2022; 158:111501. [DOI: 10.1016/j.foodres.2022.111501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/05/2022] [Accepted: 06/09/2022] [Indexed: 11/04/2022]
|
31
|
Regulation of Aging and Longevity by Ion Channels and Transporters. Cells 2022; 11:cells11071180. [PMID: 35406743 PMCID: PMC8997527 DOI: 10.3390/cells11071180] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/22/2022] [Accepted: 03/29/2022] [Indexed: 12/10/2022] Open
Abstract
Despite significant advances in our understanding of the mechanisms that underlie age-related physiological decline, our ability to translate these insights into actionable strategies to extend human healthspan has been limited. One of the major reasons for the existence of this barrier is that with a few important exceptions, many of the proteins that mediate aging have proven to be undruggable. The argument put forth here is that the amenability of ion channels and transporters to pharmacological manipulation could be leveraged to develop novel therapeutic strategies to combat aging. This review delves into the established roles for ion channels and transporters in the regulation of aging and longevity via their influence on membrane excitability, Ca2+ homeostasis, mitochondrial and endolysosomal function, and the transduction of sensory stimuli. The goal is to provide the reader with an understanding of emergent themes, and prompt further investigation into how the activities of ion channels and transporters sculpt the trajectories of cellular and organismal aging.
Collapse
|
32
|
Kakanj P, Bhide S, Moussian B, Leptin M. Autophagy-mediated plasma membrane removal promotes the formation of epithelial syncytia. EMBO J 2022; 41:e109992. [PMID: 35262206 PMCID: PMC9194749 DOI: 10.15252/embj.2021109992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 01/18/2023] Open
Abstract
Epithelial wound healing in Drosophila involves the formation of multinucleate cells surrounding the wound. We show that autophagy, a cellular degradation process often deployed in stress responses, is required for the formation of a multinucleated syncytium during wound healing, and that autophagosomes that appear near the wound edge acquire plasma membrane markers. In addition, uncontrolled autophagy in the unwounded epidermis leads to the degradation of endo‐membranes and the lateral plasma membrane, while apical and basal membranes and epithelial barrier function remain intact. Proper functioning of TORC1 is needed to prevent destruction of the larval epidermis by autophagy, in a process that depends on phagophore initiation and expansion but does not require autophagosomes fusion with lysosomes. Autophagy induction can also affect other sub‐cellular membranes, as shown by its suppression of experimentally induced laminopathy‐like nuclear defects. Our findings reveal a function for TORC1‐mediated regulation of autophagy in maintaining membrane integrity and homeostasis in the epidermis and during wound healing.
Collapse
Affiliation(s)
- Parisa Kakanj
- Institute for Genetics, University of Cologne, Cologne, Germany.,Director's Research Unit, European Molecular Biology Laboratory, Heidelberg, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Sourabh Bhide
- Director's Research Unit, European Molecular Biology Laboratory, Heidelberg, Germany.,Faculty of Biosciences, Collaboration for Joint PhD degree between EMBL and Heidelberg University, Heidelberg, Germany
| | | | - Maria Leptin
- Institute for Genetics, University of Cologne, Cologne, Germany.,Director's Research Unit, European Molecular Biology Laboratory, Heidelberg, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| |
Collapse
|
33
|
Yin Z, Klionsky DJ. Intermittent time-restricted feeding promotes longevity through circadian autophagy. Autophagy 2022; 18:471-472. [PMID: 35220894 PMCID: PMC9037462 DOI: 10.1080/15548627.2022.2039524] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Indexed: 11/02/2022] Open
Abstract
Macroautophagy/autophagy plays crucial roles in aging and the pathogenesis of age-related diseases. Studies in various animal models demonstrate the conserved requirement for autophagy-related genes in multiple anti-aging interventions. A recent study from the Shirasu-Hiza lab showed that a newly designed intermittent time-restricted feeding (iTRF) dietary regimen can robustly extend fly healthspan and lifespan through circadian rhythm-dependent activation of autophagy. The night-specific induction of autophagy is both necessary and sufficient for iTRF-mediated health benefits. The study provides the intriguing possibility that novel behavioral or pharmaceutical interventions that promote night-specific autophagy can be used to promote healthy aging.
Collapse
Affiliation(s)
- Zhangyuan Yin
- Life Sciences Institute, and the Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Daniel J. Klionsky
- Life Sciences Institute, and the Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
34
|
Abstract
Autophagy is an evolutionarily conserved, lysosome-dependent catabolic process whereby cytoplasmic components, including damaged organelles, protein aggregates and lipid droplets, are degraded and their components recycled. Autophagy has an essential role in maintaining cellular homeostasis in response to intracellular stress; however, the efficiency of autophagy declines with age and overnutrition can interfere with the autophagic process. Therefore, conditions such as sarcopenic obesity, insulin resistance and type 2 diabetes mellitus (T2DM) that are characterized by metabolic derangement and intracellular stresses (including oxidative stress, inflammation and endoplasmic reticulum stress) also involve the accumulation of damaged cellular components. These conditions are prevalent in ageing populations. For example, sarcopenia is an age-related loss of skeletal muscle mass and strength that is involved in the pathogenesis of both insulin resistance and T2DM, particularly in elderly people. Impairment of autophagy results in further aggravation of diabetes-related metabolic derangements in insulin target tissues, including the liver, skeletal muscle and adipose tissue, as well as in pancreatic β-cells. This Review summarizes the role of autophagy in the pathogenesis of metabolic diseases associated with or occurring in the context of ageing, including insulin resistance, T2DM and sarcopenic obesity, and describes its potential as a therapeutic target.
Collapse
Affiliation(s)
- Munehiro Kitada
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
- Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Daisuke Koya
- Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Ishikawa, Japan.
- Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa, Japan.
- Department of General Internal Medicine, Kusatsu General Hospital, Kusatsu, Shiga, Japan.
| |
Collapse
|
35
|
Fernandes SA, Demetriades C. The Multifaceted Role of Nutrient Sensing and mTORC1 Signaling in Physiology and Aging. FRONTIERS IN AGING 2021; 2:707372. [PMID: 35822019 PMCID: PMC9261424 DOI: 10.3389/fragi.2021.707372] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 08/12/2021] [Indexed: 01/10/2023]
Abstract
The mechanistic Target of Rapamycin (mTOR) is a growth-related kinase that, in the context of the mTOR complex 1 (mTORC1), touches upon most fundamental cellular processes. Consequently, its activity is a critical determinant for cellular and organismal physiology, while its dysregulation is commonly linked to human aging and age-related disease. Presumably the most important stimulus that regulates mTORC1 activity is nutrient sufficiency, whereby amino acids play a predominant role. In fact, mTORC1 functions as a molecular sensor for amino acids, linking the cellular demand to the nutritional supply. Notably, dietary restriction (DR), a nutritional regimen that has been shown to extend lifespan and improve healthspan in a broad spectrum of organisms, works via limiting nutrient uptake and changes in mTORC1 activity. Furthermore, pharmacological inhibition of mTORC1, using rapamycin or its analogs (rapalogs), can mimic the pro-longevity effects of DR. Conversely, nutritional amino acid overload has been tightly linked to aging and diseases, such as cancer, type 2 diabetes and obesity. Similar effects can also be recapitulated by mutations in upstream mTORC1 regulators, thus establishing a tight connection between mTORC1 signaling and aging. Although the role of growth factor signaling upstream of mTORC1 in aging has been investigated extensively, the involvement of signaling components participating in the nutrient sensing branch is less well understood. In this review, we provide a comprehensive overview of the molecular and cellular mechanisms that signal nutrient availability to mTORC1, and summarize the role that nutrients, nutrient sensors, and other components of the nutrient sensing machinery play in cellular and organismal aging.
Collapse
Affiliation(s)
- Stephanie A. Fernandes
- Max Planck Institute for Biology of Ageing (MPI-AGE), Cologne, Germany
- Cologne Graduate School for Ageing Research (CGA), Cologne, Germany
| | - Constantinos Demetriades
- Max Planck Institute for Biology of Ageing (MPI-AGE), Cologne, Germany
- Cologne Graduate School for Ageing Research (CGA), Cologne, Germany
- University of Cologne, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| |
Collapse
|
36
|
Aman Y, Schmauck-Medina T, Hansen M, Morimoto RI, Simon AK, Bjedov I, Palikaras K, Simonsen A, Johansen T, Tavernarakis N, Rubinsztein DC, Partridge L, Kroemer G, Labbadia J, Fang EF. Autophagy in healthy aging and disease. NATURE AGING 2021; 1:634-650. [PMID: 34901876 PMCID: PMC8659158 DOI: 10.1038/s43587-021-00098-4] [Citation(s) in RCA: 715] [Impact Index Per Article: 178.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 07/07/2021] [Indexed: 12/12/2022]
Abstract
Autophagy is a fundamental cellular process that eliminates molecules and subcellular elements, including nucleic acids, proteins, lipids and organelles, via lysosome-mediated degradation to promote homeostasis, differentiation, development and survival. While autophagy is intimately linked to health, the intricate relationship among autophagy, aging and disease remains unclear. This Review examines several emerging features of autophagy and postulates how they may be linked to aging as well as to the development and progression of disease. In addition, we discuss current preclinical evidence arguing for the use of autophagy modulators as suppressors of age-related pathologies such as neurodegenerative diseases. Finally, we highlight key questions and propose novel research avenues that will likely reveal new links between autophagy and the hallmarks of aging. Understanding the precise interplay between autophagy and the risk of age-related pathologies across organisms will eventually facilitate the development of clinical applications that promote long-term health.
Collapse
Affiliation(s)
- Yahyah Aman
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
- These authors contributed equally: Yahyah Aman, Tomas Schmauck-Medina
| | - Tomas Schmauck-Medina
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
- These authors contributed equally: Yahyah Aman, Tomas Schmauck-Medina
| | - Malene Hansen
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Richard I. Morimoto
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL, USA
| | | | - Ivana Bjedov
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
- UCL Cancer Institute, University College London, London, UK
| | - Konstantinos Palikaras
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Anne Simonsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, The University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Oslo, Norway
| | - Terje Johansen
- Molecular Cancer Research Group, Institute of Medical Biology, University of Tromsø–The Arctic University of Norway, Tromsø, Norway
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology–Hellas, Heraklion, Greece
- Department of Basic Sciences, School of Medicine, University of Crete, Heraklion, Greece
| | - David C. Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge, UK
| | - Linda Partridge
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
- Department of Biological Mechanisms of Ageing, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
- Karolinska Institute, Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
| | - John Labbadia
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Evandro F. Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
- The Norwegian Centre on Healthy Ageing (NO-Age), Oslo, Norway
| |
Collapse
|
37
|
Šonský I, Vodička P, Vodičková Kepková K, Hansíková H. Mitophagy in Huntington's disease. Neurochem Int 2021; 149:105147. [PMID: 34329735 DOI: 10.1016/j.neuint.2021.105147] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 01/26/2023]
Abstract
Huntington's disease (HD), as well as Parkinson's disease and Alzheimer's disease, belong to a group of neurodegenerative diseases characterized by common features, such as the progressive loss of neurons and the presence of pathogenic forms of misfolded protein aggregates. A quality control system such as autophagy is crucial for the clearance of protein aggregates and dysfunctional organelles and thus essential for the maintenance of neuronal homeostasis. The constant high energy demand of neuronal tissue links neurodegeneration to mitochondria. Inefficient removal of damaged mitochondria is thought to contribute to the pathogenesis of neurodegenerative diseases such as HD. In addition, direct involvement of the huntingtin protein in the autophagic machinery has been described. In this review, we focus on mitophagy, a selective form of autophagy responsible for mitochondrial turnover. We also discuss the relevance of pharmacological regulation of mitophagy in the future therapeutic approach to neurodegenerations, including HD.
Collapse
Affiliation(s)
- I Šonský
- Laboratory for Study of Mitochondrial Disorders, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - P Vodička
- Laboratory of Applied Proteome Analyses, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic
| | - K Vodičková Kepková
- Laboratory of Applied Proteome Analyses, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic
| | - H Hansíková
- Laboratory for Study of Mitochondrial Disorders, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| |
Collapse
|
38
|
Bolukbasi E, Woodling NS, Ivanov DK, Adcott J, Foley A, Rajasingam A, Gittings LM, Aleyakpo B, Niccoli T, Thornton JM, Partridge L. Cell type-specific modulation of healthspan by Forkhead family transcription factors in the nervous system. Proc Natl Acad Sci U S A 2021; 118:2011491118. [PMID: 33593901 PMCID: PMC7923679 DOI: 10.1073/pnas.2011491118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Reduced activity of insulin/insulin-like growth factor signaling (IIS) increases healthy lifespan among diverse animal species. Downstream of IIS, multiple evolutionarily conserved transcription factors (TFs) are required; however, distinct TFs are likely responsible for these effects in different tissues. Here we have asked which TFs can extend healthy lifespan within distinct cell types of the adult nervous system in Drosophila Starting from published single-cell transcriptomic data, we report that forkhead (FKH) is endogenously expressed in neurons, whereas forkhead-box-O (FOXO) is expressed in glial cells. Accordingly, we find that neuronal FKH and glial FOXO exert independent prolongevity effects. We have further explored the role of neuronal FKH in a model of Alzheimer's disease-associated neuronal dysfunction, where we find that increased neuronal FKH preserves behavioral function and reduces ubiquitinated protein aggregation. Finally, using transcriptomic profiling, we identify Atg17, a member of the Atg1 autophagy initiation family, as one FKH-dependent target whose neuronal overexpression is sufficient to extend healthy lifespan. Taken together, our results underscore the importance of cell type-specific mapping of TF activity to preserve healthy function with age.
Collapse
Affiliation(s)
- Ekin Bolukbasi
- Institute of Healthy Ageing, University College London, London WC1E 6BT, United Kingdom
- Department of Genetics, Evolution and Environment, University College London, London WC1E 6BT, United Kingdom
| | - Nathaniel S Woodling
- Institute of Healthy Ageing, University College London, London WC1E 6BT, United Kingdom
- Department of Genetics, Evolution and Environment, University College London, London WC1E 6BT, United Kingdom
| | - Dobril K Ivanov
- European Bioinformatics Institute, European Molecular Biology Laboratory, Cambridge CB10 1SD, United Kingdom
- UK Dementia Research Institute, Cardiff University, Cardiff CF24 4HQ, United Kingdom
| | - Jennifer Adcott
- Institute of Healthy Ageing, University College London, London WC1E 6BT, United Kingdom
- Department of Genetics, Evolution and Environment, University College London, London WC1E 6BT, United Kingdom
| | - Andrea Foley
- Institute of Healthy Ageing, University College London, London WC1E 6BT, United Kingdom
- Department of Genetics, Evolution and Environment, University College London, London WC1E 6BT, United Kingdom
| | - Arjunan Rajasingam
- Institute of Healthy Ageing, University College London, London WC1E 6BT, United Kingdom
- Department of Genetics, Evolution and Environment, University College London, London WC1E 6BT, United Kingdom
| | - Lauren M Gittings
- Institute of Healthy Ageing, University College London, London WC1E 6BT, United Kingdom
- Department of Genetics, Evolution and Environment, University College London, London WC1E 6BT, United Kingdom
| | - Benjamin Aleyakpo
- Institute of Healthy Ageing, University College London, London WC1E 6BT, United Kingdom
- Department of Genetics, Evolution and Environment, University College London, London WC1E 6BT, United Kingdom
| | - Teresa Niccoli
- Institute of Healthy Ageing, University College London, London WC1E 6BT, United Kingdom
- Department of Genetics, Evolution and Environment, University College London, London WC1E 6BT, United Kingdom
| | - Janet M Thornton
- European Bioinformatics Institute, European Molecular Biology Laboratory, Cambridge CB10 1SD, United Kingdom
| | - Linda Partridge
- Institute of Healthy Ageing, University College London, London WC1E 6BT, United Kingdom;
- Department of Genetics, Evolution and Environment, University College London, London WC1E 6BT, United Kingdom
- Department of Biological Mechanisms of Ageing, Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany
| |
Collapse
|