1
|
Ibrahim R, Bahilo Martinez M, Dobson AJ. Rapamycin's lifespan effect is modulated by mito-nuclear epistasis in Drosophila. Aging Cell 2024; 23:e14328. [PMID: 39225061 PMCID: PMC11634709 DOI: 10.1111/acel.14328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 08/02/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
The macrolide drug rapamycin is a benchmark anti-ageing drug, which robustly extends lifespan of diverse organisms. For any health intervention, it is paramount to establish whether benefits are distributed equitably among individuals and populations, and ideally to match intervention to recipients' needs. However, how responses to rapamycin vary is surprisingly understudied. Here we investigate how among-population variation in both mitochondrial and nuclear genetics shapes rapamycin's effects on lifespan. We show that epistatic "mito-nuclear" interactions, between mitochondria and nuclei, modulate the response to rapamycin treatment. Differences manifest as differential demographic effects of rapamycin, with altered age-specific mortality rate. However, a fitness cost of rapamycin early in life does not show a correlated response, suggesting that mito-nuclear epistasis can decouple costs and benefits of treatment. These findings suggest that a deeper understanding of how variation in mitochondrial and nuclear genomes shapes physiology may facilitate tailoring of anti-ageing therapy to individual need.
Collapse
Affiliation(s)
- Rita Ibrahim
- School of Molecular BiosciencesUniversity of GlasgowGlasgowUK
| | | | - Adam J. Dobson
- School of Molecular BiosciencesUniversity of GlasgowGlasgowUK
| |
Collapse
|
2
|
Lujan C, Tyler EJ, Ecker S, Webster AP, Stead ER, Martinez-Miguel VE, Milligan D, Garbe JC, Stampfer MR, Beck S, Lowe R, Bishop CL, Bjedov I. An expedited screening platform for the discovery of anti-ageing compounds in vitro and in vivo. Genome Med 2024; 16:85. [PMID: 38956711 PMCID: PMC11218148 DOI: 10.1186/s13073-024-01349-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 05/21/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Restraining or slowing ageing hallmarks at the cellular level have been proposed as a route to increased organismal lifespan and healthspan. Consequently, there is great interest in anti-ageing drug discovery. However, this currently requires laborious and lengthy longevity analysis. Here, we present a novel screening readout for the expedited discovery of compounds that restrain ageing of cell populations in vitro and enable extension of in vivo lifespan. METHODS Using Illumina methylation arrays, we monitored DNA methylation changes accompanying long-term passaging of adult primary human cells in culture. This enabled us to develop, test, and validate the CellPopAge Clock, an epigenetic clock with underlying algorithm, unique among existing epigenetic clocks for its design to detect anti-ageing compounds in vitro. Additionally, we measured markers of senescence and performed longevity experiments in vivo in Drosophila, to further validate our approach to discover novel anti-ageing compounds. Finally, we bench mark our epigenetic clock with other available epigenetic clocks to consolidate its usefulness and specialisation for primary cells in culture. RESULTS We developed a novel epigenetic clock, the CellPopAge Clock, to accurately monitor the age of a population of adult human primary cells. We find that the CellPopAge Clock can detect decelerated passage-based ageing of human primary cells treated with rapamycin or trametinib, well-established longevity drugs. We then utilise the CellPopAge Clock as a screening tool for the identification of compounds which decelerate ageing of cell populations, uncovering novel anti-ageing drugs, torin2 and dactolisib (BEZ-235). We demonstrate that delayed epigenetic ageing in human primary cells treated with anti-ageing compounds is accompanied by a reduction in senescence and ageing biomarkers. Finally, we extend our screening platform in vivo by taking advantage of a specially formulated holidic medium for increased drug bioavailability in Drosophila. We show that the novel anti-ageing drugs, torin2 and dactolisib (BEZ-235), increase longevity in vivo. CONCLUSIONS Our method expands the scope of CpG methylation profiling to accurately and rapidly detecting anti-ageing potential of drugs using human cells in vitro, and in vivo, providing a novel accelerated discovery platform to test sought after anti-ageing compounds and geroprotectors.
Collapse
Affiliation(s)
- Celia Lujan
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street London, London, WC1E 6DD, UK
| | - Eleanor Jane Tyler
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK
| | - Simone Ecker
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street London, London, WC1E 6DD, UK
| | - Amy Philomena Webster
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street London, London, WC1E 6DD, UK
- University of Exeter Medical School, Exeter, UK
| | - Eleanor Rachel Stead
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street London, London, WC1E 6DD, UK
| | - Victoria Eugenia Martinez-Miguel
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street London, London, WC1E 6DD, UK
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Deborah Milligan
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK
| | - James Charles Garbe
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Martha Ruskin Stampfer
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Stephan Beck
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street London, London, WC1E 6DD, UK.
| | - Robert Lowe
- Centre for Genomics and Child Health, Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK.
| | - Cleo Lucinda Bishop
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK.
| | - Ivana Bjedov
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street London, London, WC1E 6DD, UK.
| |
Collapse
|
3
|
Wang J, Zhou Y, Yu Y, Wang Y, Xue D, Zhou Y, Li X. A ginseng-derived rhamnogalacturonan I (RG-I) pectin promotes longevity via TOR signalling in Caenorhabditis elegans. Carbohydr Polym 2023; 312:120818. [PMID: 37059546 DOI: 10.1016/j.carbpol.2023.120818] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/07/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
Panax ginseng C. A. Meyer (ginseng), a traditional Chinese herb, is usually used to improve health and increase anti-aging activity for human. Polysaccharides are bioactive components of ginseng. Herein, using Caenorhabditis elegans as a model, we discovered a ginseng-derived rhamnogalacturonan I (RG-I) pectin WGPA-1-RG promoted longevity via TOR signalling pathway with transcription factors FOXO/DAF-16 and Nrf2/SKN-1 accumulated in the nucleus, where they activated target genes. And the WGPA-1-RG-mediated lifespan extension was dependent on endocytosis, rather than a bacterial metabolic process. Glycosidic linkage analyses combined with arabinose- and galactose-releasing enzyme hydrolyses identified the RG-I backbone of WGPA-1-RG was primarily substituted with α-1,5-linked arabinan, β-1,4-linked galactan and arabinogalactan II (AG-II) side chains. Feeding worms with the WGPA-1-RG-derived fractions which lost distinct structural elements by enzymatic digestions, we found the arabinan side chains prominently contributed to the longevity-promoting activity of WGPA-1-RG. These findings provide a novel ginseng-derived nutrient that potentially increases human longevity.
Collapse
|
4
|
Wu L, Xie X, Liang T, Ma J, Yang L, Yang J, Li L, Xi Y, Li H, Zhang J, Chen X, Ding Y, Wu Q. Integrated Multi-Omics for Novel Aging Biomarkers and Antiaging Targets. Biomolecules 2021; 12:39. [PMID: 35053186 PMCID: PMC8773837 DOI: 10.3390/biom12010039] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/17/2021] [Accepted: 12/19/2021] [Indexed: 12/12/2022] Open
Abstract
Aging is closely related to the occurrence of human diseases; however, its exact biological mechanism is unclear. Advancements in high-throughput technology provide new opportunities for omics research to understand the pathological process of various complex human diseases. However, single-omics technologies only provide limited insights into the biological mechanisms of diseases. DNA, RNA, protein, metabolites, and microorganisms usually play complementary roles and perform certain biological functions together. In this review, we summarize multi-omics methods based on the most relevant biomarkers in single-omics to better understand molecular functions and disease causes. The integration of multi-omics technologies can systematically reveal the interactions among aging molecules from a multidimensional perspective. Our review provides new insights regarding the discovery of aging biomarkers, mechanism of aging, and identification of novel antiaging targets. Overall, data from genomics, transcriptomics, proteomics, metabolomics, integromics, microbiomics, and systems biology contribute to the identification of new candidate biomarkers for aging and novel targets for antiaging interventions.
Collapse
Affiliation(s)
- Lei Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (L.W.); (X.X.); (T.L.); (L.Y.); (J.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (J.M.); (X.C.)
| | - Xinqiang Xie
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (L.W.); (X.X.); (T.L.); (L.Y.); (J.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
| | - Tingting Liang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (L.W.); (X.X.); (T.L.); (L.Y.); (J.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (J.M.); (X.C.)
| | - Jun Ma
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (J.M.); (X.C.)
| | - Lingshuang Yang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (L.W.); (X.X.); (T.L.); (L.Y.); (J.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
| | - Juan Yang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (L.W.); (X.X.); (T.L.); (L.Y.); (J.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (J.M.); (X.C.)
| | - Longyan Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (L.W.); (X.X.); (T.L.); (L.Y.); (J.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
| | - Yu Xi
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (L.W.); (X.X.); (T.L.); (L.Y.); (J.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
| | - Haixin Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (L.W.); (X.X.); (T.L.); (L.Y.); (J.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
| | - Jumei Zhang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (L.W.); (X.X.); (T.L.); (L.Y.); (J.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
| | - Xuefeng Chen
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (J.M.); (X.C.)
| | - Yu Ding
- Department of Food Science and Technology, Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, China
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (L.W.); (X.X.); (T.L.); (L.Y.); (J.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
| |
Collapse
|
5
|
Cellular senescence-an aging hallmark in chronic obstructive pulmonary disease pathogenesis. Respir Investig 2021; 60:33-44. [PMID: 34649812 DOI: 10.1016/j.resinv.2021.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/12/2021] [Accepted: 09/09/2021] [Indexed: 12/13/2022]
Abstract
Chronic obstructive pulmonary disease (COPD),1 a representative aging-related pulmonary disorder, is mainly caused by cigarette smoke (CS) exposure. Age is one of the most important risk factors for COPD development, and increased cellular senescence in tissues and organs is a component of aging. CS exposure can induce cellular senescence, as characterized by irreversible growth arrest and aberrant cytokine secretion of the senescence-associated secretory phenotype; thus, accumulation of senescent cells is widely implicated in COPD pathogenesis. CS-induced oxidative modifications to cellular components may be causally linked to accelerated cellular senescence, especially during accumulation of damaged macromolecules. Autophagy is a conserved mechanism whereby cytoplasmic components are sent for lysosomal degradation to maintain proteostasis. Autophagy diminishes with age, and loss of proteostasis is one of the hallmarks of aging. We have reported the involvement of insufficient autophagy in regulating CS-induced cellular senescence with respect to COPD pathogenesis. However, the role of autophagy in COPD pathogenesis can vary based on levels of cell stress and type of selective autophagy because excessive activation of autophagy can be responsible for inducing regulated cell death. Senotherapies targeting cellular senescence may be effective COPD treatments. Autophagy activation could be a promising sonotherapeutic approach, but the optimal modality of autophagy activation should be examined in future studies.
Collapse
|
6
|
The Regulating Effect of Autophagy-Related MiRNAs in Kidney, Bladder, and Prostate Cancer. JOURNAL OF ONCOLOGY 2021; 2021:5510318. [PMID: 33976697 PMCID: PMC8084683 DOI: 10.1155/2021/5510318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/26/2021] [Accepted: 03/31/2021] [Indexed: 12/17/2022]
Abstract
Autophagy is a treatment target for many disorders, including cancer, and its specific role is becoming increasingly well known. In tumors, researchers pay attention to microribonucleic acids (miRNAs) with regulatory effects to develop more effective therapeutic drugs for autophagy and find new therapeutic targets. Various studies have shown that autophagy-related miRNAs play an irreplaceable role in different tumors, such as miR-495, miR-30, and miR-101. These miRNAs are associated with autophagy resistance in gastric cancer, non-small cell lung cancer, and cervical cancer. In recent years, autophagy-related miRNAs have also been reported to play a role in autophagy in urinary system tumors. This article reviews the regulatory effects of autophagy-related miRNAs in kidney, bladder, and prostate cancer and provides new ideas for targeted therapy of the three major tumors of the urinary system.
Collapse
|
7
|
Rapamycin Treatment of Tendon Stem/Progenitor Cells Reduces Cellular Senescence by Upregulating Autophagy. Stem Cells Int 2021; 2021:6638249. [PMID: 33603790 PMCID: PMC7870298 DOI: 10.1155/2021/6638249] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/30/2020] [Accepted: 01/08/2021] [Indexed: 12/21/2022] Open
Abstract
The elderly population is prone to tendinopathy due to aging-related tendon changes such as cellular senescence and a decreased ability to modulate inflammation. Aging can render tendon stem/progenitor cells (TSCs) into premature senescence. We investigated the effects of rapamycin, a specific mTOR inhibitor, on the senescence of TSCs. We first showed that after treatment with bleomycin in vitro, rat patellar TSCs (PTSCs) underwent senescence, characterized by morphological alterations, induction of senescence-associated β-galactosidase (SA-β-gal) activity, and an increase in p53, p21, and p62 protein expression. Senescence of PTSCs was also characterized by the elevated expression of MMP-13 and TNF-α genes, both of which are molecular hallmarks of chronic tendinopathy. We then showed that rapamycin treatment was able to reverse the above senescent phenotypes and increase autophagy in the senescent PTSCs. The activation of autophagy and senescence rescue was, at least partly, due to the translocation of HMGB1 from the nucleus to the cytosol that functions as an autophagy promoter. By reducing TSC senescence, rapamycin may be used as a therapeutic to inhibit tendinopathy development in the aging population by promoting autophagy.
Collapse
|
8
|
Zhang W, Feng C, Jiang H. Novel target for treating Alzheimer's Diseases: Crosstalk between the Nrf2 pathway and autophagy. Ageing Res Rev 2021; 65:101207. [PMID: 33144123 DOI: 10.1016/j.arr.2020.101207] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 10/02/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023]
Abstract
In mammals, the Keap1-Nrf2-ARE pathway (henceforth, "the Nrf2 pathway") and autophagy are major intracellular defence systems that combat oxidative damage and maintain homeostasis. p62/SQSTM1, a ubiquitin-binding autophagy receptor protein, links the Nrf2 pathway and autophagy. Phosphorylation of p62 dramatically enhances its affinity for Keap1, which induces Keap1 to release Nrf2, and the p62-Keap1 heterodimer recruits LC3 and mediates the permanent degradation of Keap1 in the selective autophagy pathway. Eventually, Nrf2 accumulates in the cytoplasm and then translocates into the nucleus to activate the transcription of downstream genes that encode antioxidant enzymes, which protect cells from oxidative damage. Since Nrf2 also upregulates the expression of the p62 gene, a p62-Keap1-Nrf2 positive feedback loop is created that further enhances the protective effect on cells. Studies have shown that the p62-activated noncanonical Nrf2 pathway is an important marker of neurodegenerative diseases. The p62-Keap1-Nrf2 positive feedback loop and the Nrf2 pathway are involved in eliminating the ROS and protein aggregates induced by AD. Therefore, maintaining the homeostasis of the p62-Keap1-Nrf2 positive feedback loop, which is a bridge between the Nrf2 pathway and autophagy, may be a potential target for the treatment of AD.
Collapse
Affiliation(s)
- Weiwei Zhang
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, People's Republic of China
| | - Cong Feng
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, People's Republic of China
| | - Hong Jiang
- Department of Health Laboratory Technology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, People's Republic of China.
| |
Collapse
|
9
|
Moretti CH, Schiffer TA, Montenegro MF, Larsen FJ, Tsarouhas V, Carlström M, Samakovlis C, Weitzberg E, Lundberg JO. Dietary nitrite extends lifespan and prevents age-related locomotor decline in the fruit fly. Free Radic Biol Med 2020; 160:860-870. [PMID: 32980539 DOI: 10.1016/j.freeradbiomed.2020.09.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/24/2020] [Accepted: 09/14/2020] [Indexed: 02/08/2023]
Abstract
Aging is associated with decreased nitric oxide (NO) bioavailability and signalling. Boosting of a dietary nitrate-nitrite-NO pathway e.g. by ingestion of leafy green vegetables, improves cardiometabolic function, mitochondrial efficiency and reduces oxidative stress in humans and rodents, making dietary nitrate and nitrite an appealing intervention to address age-related disorders. On the other hand, these anions have long been implicated in detrimental health effects of our diet, particularly in formation of carcinogenic nitrosamines. The aim of this study was to assess whether inorganic nitrite affects lifespan in Drosophila melanogaster and investigate possible mechanisms underlying any such effect. In a survival assay, female flies fed a nitrite supplemented diet showed lifespan extension by 9 and 15% with 0.1 and 1 μM nitrite respectively, with no impact of nitrite on reproductive output. Interestingly, nitrite could also protect female flies from age-dependent locomotor decline, indicating a protective effect on healthspan. NO generation from nitrite involved Drosophila commensal bacteria and was indicated by a fluorescent probe as well as direct measurements of NO gas formation with chemiluminescence. Nutrient sensing pathways such as TOR and sirtuins, have been strongly implicated in lifespan extension. In aged flies, nitrite supplementation significantly downregulated dTOR and upregulated dSir2 gene expression. Total triglycerides and glucose were decreased, a described downstream effect of both TOR and sirtuin pathways. In conclusion, we demonstrate that very low doses of dietary nitrite extend lifespan and favour healthspan in female flies. We propose modulation of nutrient sensing pathways as driving mechanisms for such effects.
Collapse
Affiliation(s)
- Chiara H Moretti
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 171 77, Sweden.
| | - Tomas A Schiffer
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Marcelo F Montenegro
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Filip J Larsen
- The Swedish School of Sport and Health Sciences, Stockholm, 114 86, Sweden
| | - Vasilios Tsarouhas
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm, 106 91, Sweden
| | - Mattias Carlström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Christos Samakovlis
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm, 106 91, Sweden
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 171 77, Sweden.
| |
Collapse
|
10
|
Bjedov I, Cochemé HM, Foley A, Wieser D, Woodling NS, Castillo-Quan JI, Norvaisas P, Lujan C, Regan JC, Toivonen JM, Murphy MP, Thornton J, Kinghorn KJ, Neufeld TP, Cabreiro F, Partridge L. Fine-tuning autophagy maximises lifespan and is associated with changes in mitochondrial gene expression in Drosophila. PLoS Genet 2020; 16:e1009083. [PMID: 33253201 PMCID: PMC7738165 DOI: 10.1371/journal.pgen.1009083] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 12/15/2020] [Accepted: 08/26/2020] [Indexed: 01/26/2023] Open
Abstract
Increased cellular degradation by autophagy is a feature of many interventions that delay ageing. We report here that increased autophagy is necessary for reduced insulin-like signalling (IIS) to extend lifespan in Drosophila and is sufficient on its own to increase lifespan. We first established that the well-characterised lifespan extension associated with deletion of the insulin receptor substrate chico was completely abrogated by downregulation of the essential autophagy gene Atg5. We next directly induced autophagy by over-expressing the major autophagy kinase Atg1 and found that a mild increase in autophagy extended lifespan. Interestingly, strong Atg1 up-regulation was detrimental to lifespan. Transcriptomic and metabolomic approaches identified specific signatures mediated by varying levels of autophagy in flies. Transcriptional upregulation of mitochondrial-related genes was the signature most specifically associated with mild Atg1 upregulation and extended lifespan, whereas short-lived flies, possessing strong Atg1 overexpression, showed reduced mitochondrial metabolism and up-regulated immune system pathways. Increased proteasomal activity and reduced triacylglycerol levels were features shared by both moderate and high Atg1 overexpression conditions. These contrasting effects of autophagy on ageing and differential metabolic profiles highlight the importance of fine-tuning autophagy levels to achieve optimal healthspan and disease prevention.
Collapse
Affiliation(s)
- Ivana Bjedov
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- UCL Cancer Institute, Paul O'Gorman Building, London United Kingdom
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Helena M. Cochemé
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- MRC London Institute of Medical Sciences, Imperial College London, Du Cane Road, London, United Kingdom
| | - Andrea Foley
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- MRC London Institute of Medical Sciences, Imperial College London, Du Cane Road, London, United Kingdom
| | - Daniela Wieser
- EMBL European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Nathaniel S. Woodling
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| | - Jorge Iván Castillo-Quan
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Section on Islet Cell and Regenerative Biology, Research Division, Joslin Diabetes Center, Boston MA, United States of America
- Department of Genetics and Harvard Stem Cell Institute, Harvard Medical School, Boston MA, United States of America
| | - Povilas Norvaisas
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Celia Lujan
- UCL Cancer Institute, Paul O'Gorman Building, London United Kingdom
| | - Jennifer C. Regan
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Janne M. Toivonen
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- LAGENBIO, Facultad de Veterinaria-IIS, IA2-CITA, CIBERNED, Universidad de Zaragoza, Zaragoza, Spain
| | - Michael P. Murphy
- MRC Mitochondrial Biology Unit, the Keith Peters Building, University of Cambridge, Cambridge, United Kingdom
| | - Janet Thornton
- EMBL European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Kerri J. Kinghorn
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| | - Thomas P. Neufeld
- Department of Genetics, Cell Biology and Development, University of Minnesota, 321 Church St. SE, Minneapolis, MN, United States of America
| | - Filipe Cabreiro
- MRC London Institute of Medical Sciences, Imperial College London, Du Cane Road, London, United Kingdom
- Institute of Structural and Molecular Biology, University College London, London, United Kingdom
| | - Linda Partridge
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| |
Collapse
|
11
|
Xu T, Sun D, Chen Y, Ouyang L. Targeting mTOR for fighting diseases: A revisited review of mTOR inhibitors. Eur J Med Chem 2020; 199:112391. [DOI: 10.1016/j.ejmech.2020.112391] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/24/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023]
|
12
|
Moatt JP, Savola E, Regan JC, Nussey DH, Walling CA. Lifespan Extension Via Dietary Restriction: Time to Reconsider the Evolutionary Mechanisms? Bioessays 2020; 42:e1900241. [DOI: 10.1002/bies.201900241] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 04/24/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Joshua P. Moatt
- Institute of Evolutionary BiologySchool of Biological ScienceUniversity of Edinburgh Edinburgh EH9 3FL UK
| | - Eevi Savola
- Institute of Evolutionary BiologySchool of Biological ScienceUniversity of Edinburgh Edinburgh EH9 3FL UK
| | - Jennifer C. Regan
- Institute for Immunology and InfectionSchool of Biological ScienceUniversity of Edinburgh Edinburgh EH9 3FL UK
| | - Daniel H. Nussey
- Institute of Evolutionary BiologySchool of Biological ScienceUniversity of Edinburgh Edinburgh EH9 3FL UK
| | - Craig A. Walling
- Institute of Evolutionary BiologySchool of Biological ScienceUniversity of Edinburgh Edinburgh EH9 3FL UK
| |
Collapse
|
13
|
Abstract
Cellular parabiosis is tissue-based phenotypic suppression of cellular dysfunction by intercellular molecular traffic keeping initiated age-related diseases and conditions in long latency. Interruption of cellular parabiosis (e.g. by chronic inflammation) promotes the onset of initiated pathologies. The stability of initiated latent cancers and other age-related diseases (ARD) hints to phenotypically silent genome alterations. I propose that latency in the onset of ageing and ARD is largely due to phenotypic suppression of cellular dysfunctions via molecular traffic among neighbouring cells. Intercellular trafficking ranges from the transfer of ions and metabolites (via gap junctions) to entire organelles (via tunnelling nanotubes). Any mechanism of cell-to-cell communication resulting in functional cross-complementation among the cells is called cellular parabiosis. Such ‘cellular solidarity’ creates tissue homeostasis by buffering defects and averaging cellular functions within the tissues. Chronic inflammation is known to (i) interrupt cellular parabiosis by the activity of extracellular proteases, (ii) activate dormant pathologies and (iii) shorten disease latency, as in tumour promotion and inflammaging. Variation in cellular parabiosis and protein oxidation can account for interspecies correlations between body mass, ARD latency and longevity. Now, prevention of ARD onset by phenotypic suppression, and healing by phenotypic reversion, become conceivable.
Collapse
Affiliation(s)
- Miroslav Radman
- 1 Mediterranean Institute for Life Sciences (MedILS) , 21000 Split , Croatia.,2 Naos Institute for Life Sciences , 13290 Aix-en-Provence , France.,3 Inserm u-1001, University R. Descartes Medical School , Cochin Site, 75014 Paris , France
| |
Collapse
|
14
|
Rodríguez-López M, Gonzalez S, Hillson O, Tunnacliffe E, Codlin S, Tallada VA, Bähler J, Rallis C. The GATA Transcription Factor Gaf1 Represses tRNAs, Inhibits Growth, and Extends Chronological Lifespan Downstream of Fission Yeast TORC1. Cell Rep 2020; 30:3240-3249.e4. [PMID: 32160533 PMCID: PMC7068653 DOI: 10.1016/j.celrep.2020.02.058] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 12/17/2019] [Accepted: 02/13/2020] [Indexed: 12/13/2022] Open
Abstract
Target of Rapamycin Complex 1 (TORC1) signaling promotes growth and aging. Inhibition of TORC1 leads to reduced protein translation, which promotes longevity. TORC1-dependent post-transcriptional regulation of protein translation has been well studied, while analogous transcriptional regulation is less understood. Here we screen fission yeast mutants for resistance to Torin1, which inhibits TORC1 and cell growth. Cells lacking the GATA factor Gaf1 (gaf1Δ) grow normally even in high doses of Torin1. The gaf1Δ mutation shortens the chronological lifespan of non-dividing cells and diminishes Torin1-mediated longevity. Expression profiling and genome-wide binding experiments show that upon TORC1 inhibition, Gaf1 directly upregulates genes for small-molecule metabolic pathways and indirectly represses genes for protein translation. Surprisingly, Gaf1 binds to and downregulates the tRNA genes, so it also functions as a transcription factor for RNA polymerase III. Thus, Gaf1 controls the transcription of both protein-coding and tRNA genes to inhibit translation and growth downstream of TORC1.
Collapse
Affiliation(s)
- María Rodríguez-López
- Institute of Healthy Ageing and Department of Genetics, Evolution & Environment, University College London, London WC1E 6BT, UK
| | - Suam Gonzalez
- School of Health, Sport and Bioscience, University of East London, Stratford Campus, London E14 4LZ, UK
| | - Olivia Hillson
- School of Health, Sport and Bioscience, University of East London, Stratford Campus, London E14 4LZ, UK
| | - Edward Tunnacliffe
- Institute of Healthy Ageing and Department of Genetics, Evolution & Environment, University College London, London WC1E 6BT, UK
| | - Sandra Codlin
- Institute of Healthy Ageing and Department of Genetics, Evolution & Environment, University College London, London WC1E 6BT, UK
| | - Victor A Tallada
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide/CSIC, 41013 Sevilla, Spain
| | - Jürg Bähler
- Institute of Healthy Ageing and Department of Genetics, Evolution & Environment, University College London, London WC1E 6BT, UK.
| | - Charalampos Rallis
- Institute of Healthy Ageing and Department of Genetics, Evolution & Environment, University College London, London WC1E 6BT, UK; School of Health, Sport and Bioscience, University of East London, Stratford Campus, London E14 4LZ, UK; School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK.
| |
Collapse
|
15
|
Novel compounds for the modulation of mTOR and autophagy to treat neurodegenerative diseases. Cell Signal 2020; 65:109442. [DOI: 10.1016/j.cellsig.2019.109442] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 12/16/2022]
|
16
|
Stead ER, Castillo-Quan JI, Miguel VEM, Lujan C, Ketteler R, Kinghorn KJ, Bjedov I. Agephagy - Adapting Autophagy for Health During Aging. Front Cell Dev Biol 2019; 7:308. [PMID: 31850344 PMCID: PMC6892982 DOI: 10.3389/fcell.2019.00308] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 11/12/2019] [Indexed: 12/11/2022] Open
Abstract
Autophagy is a major cellular recycling process that delivers cellular material and entire organelles to lysosomes for degradation, in a selective or non-selective manner. This process is essential for the maintenance of cellular energy levels, components, and metabolites, as well as the elimination of cellular molecular damage, thereby playing an important role in numerous cellular activities. An important function of autophagy is to enable survival under starvation conditions and other stresses. The majority of factors implicated in aging are modifiable through the process of autophagy, including the accumulation of oxidative damage and loss of proteostasis, genomic instability and epigenetic alteration. These primary causes of damage could lead to mitochondrial dysfunction, deregulation of nutrient sensing pathways and cellular senescence, finally causing a variety of aging phenotypes. Remarkably, advances in the biology of aging have revealed that aging is a malleable process: a mild decrease in signaling through nutrient-sensing pathways can improve health and extend lifespan in all model organisms tested. Consequently, autophagy is implicated in both aging and age-related disease. Enhancement of the autophagy process is a common characteristic of all principal, evolutionary conserved anti-aging interventions, including dietary restriction, as well as inhibition of target of rapamycin (TOR) and insulin/IGF-1 signaling (IIS). As an emerging and critical process in aging, this review will highlight how autophagy can be modulated for health improvement.
Collapse
Affiliation(s)
- Eleanor R Stead
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Jorge I Castillo-Quan
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA, United States.,Department of Genetics, Harvard Medical School, Boston, MA, United States
| | | | - Celia Lujan
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Robin Ketteler
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Kerri J Kinghorn
- Institute of Healthy Ageing, University College London, London, United Kingdom.,Department of Genetics, Evolution and Environment, University College London, London, United Kingdom.,Institute of Neurology, University College London, London, United Kingdom
| | - Ivana Bjedov
- UCL Cancer Institute, University College London, London, United Kingdom
| |
Collapse
|
17
|
Fan F, Sam R, Ryan E, Alvarado K, Villa-Cuesta E. Rapamycin as a potential treatment for succinate dehydrogenase deficiency. Heliyon 2019; 5:e01217. [PMID: 30805566 PMCID: PMC6374580 DOI: 10.1016/j.heliyon.2019.e01217] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 01/04/2019] [Accepted: 02/04/2019] [Indexed: 11/21/2022] Open
Abstract
Drosophila melanogaster is a powerful model to study mitochondrial respiratory chain defects, particularly succinate dehydrogenase (SDH) deficiency. Mutations in sdh genes cause degenerative disorders and often lead to death. Therapies for such pathologies are based on a combination of vitamins and dietary supplements, and are rarely effective. In Drosophila, mutations in several of the genes encoding SDH resemble the pathology of SDH deficiency in humans, enabling the Drosophila model to be used in finding treatments for this condition. Here we show that exposure to the drug rapamycin improves the survival of sdh mutant strains, the activity of SDH and the impaired climbing associated with sdh mutations. However, the production of reactive oxygen species, the oxygen consumption of isolated mitochondria and the resistance to hyperoxia were minimally affected. Our results contribute to the current research seeking a treatment for mitochondrial disease.
Collapse
Affiliation(s)
- Frances Fan
- Biology Department, Adelphi University, Garden City, NY, USA
- Honors College, Adelphi University, Garden City, NY, USA
| | - Rheba Sam
- Biology Department, Adelphi University, Garden City, NY, USA
- Honors College, Adelphi University, Garden City, NY, USA
| | - Emma Ryan
- Biology Department, Adelphi University, Garden City, NY, USA
- Honors College, Adelphi University, Garden City, NY, USA
| | | | - Eugenia Villa-Cuesta
- Biology Department, Adelphi University, Garden City, NY, USA
- NYU Winthrop Research Institute, Mineola, NY, USA
| |
Collapse
|
18
|
The lysosome as a cellular centre for signalling, metabolism and quality control. Nat Cell Biol 2019; 21:133-142. [DOI: 10.1038/s41556-018-0244-7] [Citation(s) in RCA: 409] [Impact Index Per Article: 68.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/31/2018] [Indexed: 02/06/2023]
|
19
|
Abstract
Chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis are regarded as a diseases of accelerated lung ageing and show all of the hallmarks of ageing, including telomere shortening, cellular senescence, activation of PI3 kinase-mTOR signaling, impaired autophagy, mitochondrial dysfunction, stem cell exhaustion, epigenetic changes, abnormal microRNA profiles, immunosenescence and a low grade chronic inflammation due to senescence-associated secretory phenotype (SASP). Many of these ageing mechanisms are driven by exogenous and endogenous oxidative stress. There is also a reduction in anti-ageing molecules, such as sirtuins and Klotho, which further accelerate the ageing process. Understanding these molecular mechanisms has identified several novel therapeutic targets and several drugs and dietary interventions are now in development to treat chronic lung disease.
Collapse
Affiliation(s)
- Peter J Barnes
- Airway Disease Section, National Heart and Lung Institute, Imperial College, London, UK.
| |
Collapse
|
20
|
Wang Y, Li Y, He C, Gou B, Song M. Mitochondrial regulation of cardiac aging. Biochim Biophys Acta Mol Basis Dis 2018; 1865:1853-1864. [PMID: 30593894 DOI: 10.1016/j.bbadis.2018.12.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 12/13/2022]
Abstract
Aging is associated with progressive decline in cardiac structure and function. Accumulating evidence in model organisms and humans links cardiac aging to mitochondrial regulation, encompassing a complex interplay of mitochondrial morphology, mitochondrial ROS, mitochondrial DNA mutations, mitochondrial unfolded protein response, nicotinamide adenine dinucleotide levels and sirtuins, as well as mitophagy. This review summarizes the recent discoveries on the mitochondrial regulation of cardiac aging and the possible molecular mechanisms underlying the anti-aging effects, as well as the potential interventions that alleviate aging-related cardiac diseases and attenuate cardiac aging via the regulation of mitochondria.
Collapse
Affiliation(s)
- Yuhan Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Forestry University, Beijing 100083, China
| | - Yujing Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Science and Technology of China, Anhui 230026, China
| | - Chuting He
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bo Gou
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
21
|
Metaxakis A, Ploumi C, Tavernarakis N. Autophagy in Age-Associated Neurodegeneration. Cells 2018; 7:cells7050037. [PMID: 29734735 PMCID: PMC5981261 DOI: 10.3390/cells7050037] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 04/23/2018] [Accepted: 05/03/2018] [Indexed: 12/12/2022] Open
Abstract
The elimination of abnormal and dysfunctional cellular constituents is an essential prerequisite for nerve cells to maintain their homeostasis and proper function. This is mainly achieved through autophagy, a process that eliminates abnormal and dysfunctional cellular components, including misfolded proteins and damaged organelles. Several studies suggest that age-related decline of autophagy impedes neuronal homeostasis and, subsequently, leads to the progression of neurodegenerative disorders due to the accumulation of toxic protein aggregates in neurons. Here, we discuss the involvement of autophagy perturbation in neurodegeneration and present evidence indicating that upregulation of autophagy holds potential for the development of therapeutic interventions towards confronting neurodegenerative diseases in humans.
Collapse
Affiliation(s)
- Athanasios Metaxakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion 70013, Crete, Greece.
| | - Christina Ploumi
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion 70013, Crete, Greece.
- Department of Basic Sciences, Faculty of Medicine, University of Crete, Heraklion 70013, Crete, Greece.
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion 70013, Crete, Greece.
- Department of Basic Sciences, Faculty of Medicine, University of Crete, Heraklion 70013, Crete, Greece.
| |
Collapse
|
22
|
Selman C, Sinclair A, Pedroni SMA, Irvine EE, Michie AM, Withers DJ. Evidence that hematopoietic stem cell function is preserved during aging in long-lived S6K1 mutant mice. Oncotarget 2017; 7:29937-43. [PMID: 27083004 PMCID: PMC5058654 DOI: 10.18632/oncotarget.8729] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 04/05/2016] [Indexed: 11/25/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) signalling pathway plays a highly conserved role in aging; mice lacking ribosomal protein S6 kinase 1 (S6K1−/−) have extended lifespan and healthspan relative to wild type (WT) controls. Exactly how reduced mTOR signalling induces such effects is unclear, although preservation of stem cell function may be important. We show, using gene expression analyses, that there was a reduction in expression of cell cycle genes in young (12 week) and aged (80 week) S6K1−/− BM-derived c-Kit+ cells when compared to age-matched WT mice, suggesting that these cells are more quiescent in S6K1−/− mice. In addition, we investigated hematopoietic stem cell (HSC) frequency and function in young and aged S6K1−/− and WT mice. Young, but not aged, S6K1−/− mice had more LSK (lineage−, c-Kit+, Sca-1+) cells (% of bone marrow (BM)), including the most primitive long-term repopulating HSCs (LT-HSC) relative to WT controls. Donor-derived engraftment of LT-HSCs in recipient mice was unaffected by genotype in young mice, but was enhanced in transplants using LT-HSCs derived from aged S6K1−/− mice. Our results are the first to provide evidence that age-associated HSC functional decline is ameliorated in a long-lived mTOR mutant mouse.
Collapse
Affiliation(s)
- Colin Selman
- Glasgow Ageing Research Network (GARNER), Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Amy Sinclair
- Glasgow Ageing Research Network (GARNER), Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Silvia M A Pedroni
- Metabolic Signaling Group, Medical Research Council Clinical Sciences Centre, Imperial College, London, UK
| | - Elaine E Irvine
- Metabolic Signaling Group, Medical Research Council Clinical Sciences Centre, Imperial College, London, UK
| | - Alison M Michie
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Dominic J Withers
- Metabolic Signaling Group, Medical Research Council Clinical Sciences Centre, Imperial College, London, UK
| |
Collapse
|
23
|
Abstract
Cell size is amenable by genetic and environmental factors. The highly conserved nutrient-responsive Target of Rapamycin (TOR) signaling pathway regulates cellular metabolic status and growth in response to numerous inputs. Timing and duration of TOR pathway activity is pivotal for both cell mass built up as well as cell cycle progression and is controlled and fine-tuned by the abundance and quality of nutrients, hormonal signals, growth factors, stress, and oxygen. TOR kinases function within two functionally and structurally discrete multiprotein complexes, TORC1 and TORC2, that are implicated in temporal and spatial control of cell size and growth respectively; however, recent data indicate that such functional distinctions are much more complex. Here, we briefly review roles of the two complexes in cellular growth and cytoarchitecture in various experimental model systems.
Collapse
Affiliation(s)
- Suam Gonzalez
- School of Health, Sport and Bioscience, University of East LondonLondon, United Kingdom
| | - Charalampos Rallis
- School of Health, Sport and Bioscience, University of East LondonLondon, United Kingdom
| |
Collapse
|
24
|
[Anti-ageing therapies in Alzheimer's disease]. Rev Esp Geriatr Gerontol 2017; 53:45-53. [PMID: 28549745 DOI: 10.1016/j.regg.2017.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 02/20/2017] [Accepted: 02/21/2017] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease is the most common cause of dementia in the elderly population. Currently, there are no effective treatments to prevent or delay the natural course of the disease. Numerous studies have provided information about the molecular processes underlying biological ageing and, perhaps more importantly, potential interventions to slow ageing and promote healthy longevity in laboratory model systems. The main issue addressed in this review is whether an intervention that has anti-ageing properties can alter the appearance and/or progression of Alzheimer's disease, a disease in which age is the biggest risk factor. Different anti-ageing interventions have been shown to prevent (and in some cases possibly restore) several parameters recognised as central symptoms to the development of Alzheimer's disease. In addition, they are taking the first steps towards translating these laboratory discoveries into clinical applications.
Collapse
|
25
|
Williams AC, Hill LJ. Meat and Nicotinamide: A Causal Role in Human Evolution, History, and Demographics. Int J Tryptophan Res 2017; 10:1178646917704661. [PMID: 28579800 PMCID: PMC5417583 DOI: 10.1177/1178646917704661] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/15/2017] [Indexed: 01/15/2023] Open
Abstract
Hunting for meat was a critical step in all animal and human evolution. A key brain-trophic element in meat is vitamin B3 / nicotinamide. The supply of meat and nicotinamide steadily increased from the Cambrian origin of animal predators ratcheting ever larger brains. This culminated in the 3-million-year evolution of Homo sapiens and our overall demographic success. We view human evolution, recent history, and agricultural and demographic transitions in the light of meat and nicotinamide intake. A biochemical and immunological switch is highlighted that affects fertility in the 'de novo' tryptophan-to-kynurenine-nicotinamide 'immune tolerance' pathway. Longevity relates to nicotinamide adenine dinucleotide consumer pathways. High meat intake correlates with moderate fertility, high intelligence, good health, and longevity with consequent population stability, whereas low meat/high cereal intake (short of starvation) correlates with high fertility, disease, and population booms and busts. Too high a meat intake and fertility falls below replacement levels. Reducing variances in meat consumption might help stabilise population growth and improve human capital.
Collapse
Affiliation(s)
- Adrian C Williams
- Department of Neurology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Lisa J Hill
- Neuroscience and Ophthalmology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| |
Collapse
|
26
|
Abstract
Chronic obstructive pulmonary disease (COPD) is regarded as a disease of accelerated lung aging. This affliction shows all of the hallmarks of aging, including telomere shortening, cellular senescence, activation of PI3 kinase-mTOR signaling, impaired autophagy, mitochondrial dysfunction, stem cell exhaustion, epigenetic changes, abnormal microRNA profiles, immunosenescence, and a low-grade chronic inflammation (inflammaging). Many of these pathways are driven by chronic exogenous and endogenous oxidative stress. There is also a reduction in antiaging molecules, such as sirtuins and Klotho, which further accelerate the aging process. COPD is associated with several comorbidities (multimorbidity), such as cardiovascular and metabolic diseases, that share the same pathways of accelerated aging. Understanding these mechanisms has helped identify several novel therapeutic targets, and several drugs and dietary interventions are now in development to treat multimorbidity.
Collapse
Affiliation(s)
- Peter J. Barnes
- National Heart and Lung Institute, Imperial College, London SW3 6LY, United Kingdom
| |
Collapse
|
27
|
Justice J, Miller JD, Newman JC, Hashmi SK, Halter J, Austad SN, Barzilai N, Kirkland JL. Frameworks for Proof-of-Concept Clinical Trials of Interventions That Target Fundamental Aging Processes. J Gerontol A Biol Sci Med Sci 2016; 71:1415-1423. [PMID: 27535966 PMCID: PMC5055651 DOI: 10.1093/gerona/glw126] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/16/2016] [Indexed: 01/09/2023] Open
Abstract
Therapies targeted at fundamental processes of aging may hold great promise for enhancing the health of a wide population by delaying or preventing a range of age-related diseases and conditions—a concept dubbed the “geroscience hypothesis.” Early, proof-of-concept clinical trials will be a key step in the translation of therapies emerging from model organism and preclinical studies into clinical practice. This article summarizes the outcomes of an international meeting partly funded through the NIH R24 Geroscience Network, whose purpose was to generate concepts and frameworks for early, proof-of-concept clinical trials for therapeutic interventions that target fundamental processes of aging. The goals of proof-of-concept trials include generating preliminary signals of efficacy in an aging-related disease or outcome that will reduce the risk of conducting larger trials, contributing data and biological samples to support larger-scale research by strategic networks, and furthering a dialogue with regulatory agencies on appropriate registration indications. We describe three frameworks for proof-of-concept trials that target age-related chronic diseases, geriatric syndromes, or resilience to stressors. We propose strategic infrastructure and shared resources that could accelerate development of therapies that target fundamental aging processes.
Collapse
Affiliation(s)
- Jamie Justice
- Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Jordan D Miller
- Department of Surgery.,Department of Physiology and Biomedical Engineering and.,The Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - John C Newman
- Division of Geriatrics, University of California San Francisco
| | - Shahrukh K Hashmi
- Department of Hematology and Transplant Center, Mayo Clinic, Rochester, Minnesota
| | - Jeffrey Halter
- Geriatrics Center and Institute of Gerontology, University of Michigan, Ann Arbor
| | - Steve N Austad
- Department of Biology, University of Alabama at Birmingham
| | - Nir Barzilai
- Department of Medicine, Division of Endocrinology and.,Institute for Aging Research, Albert Einstein College of Medicine, New York
| | - James L Kirkland
- Department of Physiology and Biomedical Engineering and .,The Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
28
|
All-trans retinoic acid and rapamycin normalize Hutchinson Gilford progeria fibroblast phenotype. Oncotarget 2016; 6:29914-28. [PMID: 26359359 PMCID: PMC4745772 DOI: 10.18632/oncotarget.4939] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 07/31/2015] [Indexed: 11/25/2022] Open
Abstract
Hutchinson Gilford progeria syndrome is a fatal disorder characterized by accelerated aging, bone resorption and atherosclerosis, caused by a LMNA mutation which produces progerin, a mutant lamin A precursor. Progeria cells display progerin and prelamin A nuclear accumulation, altered histone methylation pattern, heterochromatin loss, increased DNA damage and cell cycle alterations. Since the LMNA promoter contains a retinoic acid responsive element, we investigated if all-trans retinoic acid administration could lower progerin levels in cultured fibroblasts. We also evaluated the effect of associating rapamycin, which induces autophagic degradation of progerin and prelamin A. We demonstrate that all-trans retinoic acid acts synergistically with low-dosage rapamycin reducing progerin and prelamin A, via transcriptional downregulation associated with protein degradation, and increasing the lamin A to progerin ratio. These effects rescue cell dynamics and cellular proliferation through recovery of DNA damage response factor PARP1 and chromatin-associated nuclear envelope proteins LAP2α and BAF. The combined all-trans retinoic acid-rapamycin treatment is dramatically efficient, highly reproducible, represents a promising new approach in Hutchinson-Gilford Progeria therapy and deserves investigation in ageing-associated disorders.
Collapse
|
29
|
Scialò F, Sriram A, Naudí A, Ayala V, Jové M, Pamplona R, Sanz A. Target of rapamycin activation predicts lifespan in fruit flies. Cell Cycle 2016; 14:2949-58. [PMID: 26259964 PMCID: PMC4630862 DOI: 10.1080/15384101.2015.1071745] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Aging and age-related diseases are one of the most important health issues that the world will confront during the 21st century. Only by understanding the proximal causes will we be able to find treatments to reduce or delay the onset of degenerative diseases associated with aging. Currently, the prevalent paradigm in the field is the accumulation of damage. However, a new theory that proposes an alternative explanation is gaining momentum. The hyperfunction theory proposes that aging is not a consequence of a wear and tear process, but a result of the continuation of developmental programs during adulthood. Here we use Drosophila melanogaster, where evidence supporting both paradigms has been reported, to identify which parameters that have been previously related with lifespan best predict the rate of aging in wild type flies cultured at different temperatures. We find that mitochondrial function and mitochondrial reactive oxygen species (mtROS) generation correlates with metabolic rate, but not with the rate of aging. Importantly, we find that activation of nutrient sensing pathways (i.e. insulin-PI3K/Target of rapamycin (Tor) pathway) correlates with lifespan, but not with metabolic rate. Our results, dissociate metabolic rate and lifespan in wild type flies and instead link nutrient sensing signaling with longevity as predicted by the hyperfunction theory.
Collapse
Affiliation(s)
- Filippo Scialò
- a Institute for Cell and Molecular Biosciences; Newcastle University Institute for Ageing; Newcastle University ; Newcastle-Upon-Tyne , UK
| | - Ashwin Sriram
- a Institute for Cell and Molecular Biosciences; Newcastle University Institute for Ageing; Newcastle University ; Newcastle-Upon-Tyne , UK
| | - Alba Naudí
- b Department of Experimental Medicine ; University of Lleida-IRB ; Lleida , Spain
| | - Victoria Ayala
- b Department of Experimental Medicine ; University of Lleida-IRB ; Lleida , Spain
| | - Mariona Jové
- b Department of Experimental Medicine ; University of Lleida-IRB ; Lleida , Spain
| | - Reinald Pamplona
- b Department of Experimental Medicine ; University of Lleida-IRB ; Lleida , Spain
| | - Alberto Sanz
- a Institute for Cell and Molecular Biosciences; Newcastle University Institute for Ageing; Newcastle University ; Newcastle-Upon-Tyne , UK
| |
Collapse
|
30
|
Leontieva OV, Demidenko ZN, Blagosklonny MV. Dual mTORC1/C2 inhibitors suppress cellular geroconversion (a senescence program). Oncotarget 2016; 6:23238-48. [PMID: 26177051 PMCID: PMC4695114 DOI: 10.18632/oncotarget.4836] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 07/24/2015] [Indexed: 11/30/2022] Open
Abstract
In proliferating cells, mTOR is active and promotes cell growth. When the cell cycle is arrested, then mTOR converts reversible arrest to senescence (geroconversion). Rapamycin and other rapalogs suppress geroconversion, maintaining quiescence instead. Here we showed that ATP-competitive kinase inhibitors (Torin1 and PP242), which inhibit both mTORC1 and TORC2, also suppressed geroconversion. Despite inhibition of proliferation (in proliferating cells), mTOR inhibitors preserved re-proliferative potential (RP) in arrested cells. In p21-arrested cells, Torin 1 and PP242 detectably suppressed geroconversion at concentrations as low as 1-3 nM and 10-30 nM, reaching maximal gerosuppression at 30 nM and 300 nM, respectively. Near-maximal gerosuppression coincided with inhibition of p-S6K(T389) and p-S6(S235/236). Dual mTOR inhibitors prevented senescent morphology and hypertrophy. Our study warrants investigation into whether low doses of dual mTOR inhibitors will prolong animal life span and delay age-related diseases. A new class of potential anti-aging drugs can be envisioned.
Collapse
Affiliation(s)
- Olga V Leontieva
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Zoya N Demidenko
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | |
Collapse
|
31
|
Rejuvenating immunity: "anti-aging drug today" eight years later. Oncotarget 2016; 6:19405-12. [PMID: 25844603 PMCID: PMC4637294 DOI: 10.18632/oncotarget.3740] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 03/28/2015] [Indexed: 01/02/2023] Open
Abstract
The 2014 year ended with celebration: Everolimus, a rapamycin analog, was shown to improve immunity in old humans, heralding ‘a turning point’ in research and new era in human quest for immortality. Yet, this turning point was predicted a decade ago. But what will cause human death, when aging will be abolished?
Collapse
|
32
|
Redmann M, Darley-Usmar V, Zhang J. The Role of Autophagy, Mitophagy and Lysosomal Functions in Modulating Bioenergetics and Survival in the Context of Redox and Proteotoxic Damage: Implications for Neurodegenerative Diseases. Aging Dis 2016; 7:150-62. [PMID: 27114848 PMCID: PMC4809607 DOI: 10.14336/ad.2015.0820] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 08/20/2015] [Indexed: 12/21/2022] Open
Abstract
Redox and proteotoxic stress contributes to age-dependent accumulation of dysfunctional mitochondria and protein aggregates, and is associated with neurodegeneration. The free radical theory of aging inspired many studies using reactive species scavengers such as alpha-tocopherol, ascorbate and coenzyme Q to suppress the initiation of oxidative stress. However, clinical trials have had limited success in the treatment of neurodegenerative diseases. We ascribe this to the emerging literature which suggests that the oxidative stress hypothesis does not encompass the role of reactive species in cell signaling and therefore the interception with reactive species with antioxidant supplementation may result in disruption of redox signaling. In addition, the accumulation of redox modified proteins or organelles cannot be reversed by oxidant intercepting antioxidants and must then be removed by alternative mechanisms. We have proposed that autophagy serves this essential function in removing damaged or dysfunctional proteins and organelles thus preserving neuronal function and survival. In this review, we will highlight observations regarding the impact of autophagy regulation on cellular bioenergetics and survival in response to reactive species or reactive species generating compounds, and in response to proteotoxic stress.
Collapse
Affiliation(s)
- Matthew Redmann
- Center for Free Radical Biology,; Department of Pathology, University of Alabama at Birmingham
| | - Victor Darley-Usmar
- Center for Free Radical Biology,; Department of Pathology, University of Alabama at Birmingham
| | - Jianhua Zhang
- Center for Free Radical Biology,; Department of Pathology, University of Alabama at Birmingham,; Department of Veterans Affairs, Birmingham VA Medical Center, Birmingham, Alabama 35294, USA
| |
Collapse
|
33
|
López-Domínguez JA, Cánovas Á, Medrano JF, Islas-Trejo A, Kim K, Taylor SL, Villalba JM, López-Lluch G, Navas P, Ramsey JJ. Omega-3 fatty acids partially revert the metabolic gene expression profile induced by long-term calorie restriction. Exp Gerontol 2016; 77:29-37. [PMID: 26875793 DOI: 10.1016/j.exger.2016.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/03/2016] [Accepted: 02/08/2016] [Indexed: 11/18/2022]
Abstract
Calorie restriction (CR) consistently extends longevity and delays age-related diseases across several animal models. We have previously shown that different dietary fat sources can modulate life span and mitochondrial ultrastructure, function and membrane fatty acid composition in mice maintained on a 40% CR. In particular, animals consuming lard as the main fat source (CR-Lard) lived longer than CR mice consuming diets with soybean oil (CR-Soy) or fish oil (CR-Fish) as the predominant lipid source. In the present work, a transcriptomic analysis in the liver and skeletal muscle was performed in order to elucidate possible mechanisms underlying the changes in energy metabolism and longevity induced by dietary fat in CR mice. After 8 months of CR, transcription downstream of several mediators of inflammation was inhibited in liver. In contrast, proinflammatory signaling was increased in the CR-Fish versus other CR groups. Dietary fish oil induced a gene expression pattern consistent with increased transcriptional regulation by several cytokines (TNF, GM-CSF, TGF-β) and sex hormones when compared to the other CR groups. The CR-Fish also had lower expression of genes involved in fatty acid biosynthesis and increased expression of mitochondrial and peroxisomal fatty acid β-oxidation genes than the other CR diet groups. Our data suggest that a diet high in n-3 PUFA, partially reverts CR-related changes in gene expression of key processes, such as inflammation and steroid hormone signaling, and this may mitigate life span extension with CR in mice consuming diets high in fish oil.
Collapse
Affiliation(s)
| | - Ángela Cánovas
- Department of Animal Science, University of California, Davis, USA
| | - Juan F Medrano
- Department of Animal Science, University of California, Davis, USA
| | - Alma Islas-Trejo
- Department of Animal Science, University of California, Davis, USA
| | - Kyoungmi Kim
- Department of Public Health, School of Medicine, University of California, Davis, USA
| | - Sandra L Taylor
- Department of Public Health, School of Medicine, University of California, Davis, USA
| | - José Manuel Villalba
- Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Campus de Excelencia Internacional Agroalimentario, ceiA3, Spain
| | - Guillermo López-Lluch
- Centro Andaluz de Biología del Desarrollo (CABD), Universidad Pablo de Olavide-CSIC, CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Plácido Navas
- Centro Andaluz de Biología del Desarrollo (CABD), Universidad Pablo de Olavide-CSIC, CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Jon J Ramsey
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, USA
| |
Collapse
|
34
|
The Progress of Mitophagy and Related Pathogenic Mechanisms of the Neurodegenerative Diseases and Tumor. NEUROSCIENCE JOURNAL 2015; 2015:543758. [PMID: 26779531 PMCID: PMC4686711 DOI: 10.1155/2015/543758] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 10/14/2015] [Accepted: 10/25/2015] [Indexed: 12/29/2022]
Abstract
Mitochondrion, an organelle with two layers of membrane, is extremely vital to eukaryotic cell. Its major functions are energy center and apoptosis censor inside cell. The intactness of mitochondrial membrane is important to maintain its structure and function. Mitophagy is one kind of autophagy. In recent years, studies of mitochondria have shown that mitophagy is regulated by various factors and is an important regulation mechanism for organisms to maintain their normal state. In addition, abnormal mitophagy is closely related to several neurodegenerative diseases and tumor. However, the related signal pathway and its regulation mechanism still remain unclear. As a result, summarizing the progress of mitophagy and its related pathogenic mechanism not only helps to reveal the complicated molecular mechanism, but also helps to find a new target to treat the related diseases.
Collapse
|
35
|
Eiden AM, Zhang S, Gary JM, Simmons JK, Mock BA. Molecular Pathways: Increased Susceptibility to Infection Is a Complication of mTOR Inhibitor Use in Cancer Therapy. Clin Cancer Res 2015; 22:277-83. [PMID: 26607598 DOI: 10.1158/1078-0432.ccr-14-3239] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 11/04/2015] [Indexed: 12/20/2022]
Abstract
As one of the earliest examples of "chemical biology," the M: echanistic T: arget of R: apamycin (mTOR) protein and its chemical inhibitors have been extensively studied across a spectrum of physiologic and pathologic processes at the molecular, organismal, and patient population levels. There are several FDA-approved mTOR inhibitors (sirolimus, everolimus, and temsirolimus) with indications for cancer treatment and for prevention of solid organ rejection. Dozens of mTOR inhibitors are currently being evaluated in hundreds of ongoing clinical trials across a spectrum of diseases, including numerous cancer indications, autoimmune diseases, and a number of congenital disorders. As many of the approved and investigational indications for mTOR inhibitors require long-term treatment, the magnitude and incidence of particular side effects differ from those observed in shorter-term treatments. Here, we focus on the increased risk of infections in patients being treated with mTOR inhibitors. While increased infection rates might be expected from a class of drugs approved as posttransplant immunosuppressants, we review reports from clinical, mechanistic, and genetically engineered mouse model studies detailing a much more nuanced view of mTOR inhibitor drug action and target biology.
Collapse
Affiliation(s)
- Adrian M Eiden
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Shuling Zhang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Joy M Gary
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - John K Simmons
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Beverly A Mock
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
36
|
Castillo-Quan JI, Kinghorn KJ, Bjedov I. Genetics and pharmacology of longevity: the road to therapeutics for healthy aging. ADVANCES IN GENETICS 2015; 90:1-101. [PMID: 26296933 DOI: 10.1016/bs.adgen.2015.06.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aging can be defined as the progressive decline in tissue and organismal function and the ability to respond to stress that occurs in association with homeostatic failure and the accumulation of molecular damage. Aging is the biggest risk factor for human disease and results in a wide range of aging pathologies. Although we do not completely understand the underlying molecular basis that drives the aging process, we have gained exceptional insights into the plasticity of life span and healthspan from the use of model organisms such as the worm Caenorhabditis elegans and the fruit fly Drosophila melanogaster. Single-gene mutations in key cellular pathways that regulate environmental sensing, and the response to stress, have been identified that prolong life span across evolution from yeast to mammals. These genetic manipulations also correlate with a delay in the onset of tissue and organismal dysfunction. While the molecular genetics of aging will remain a prosperous and attractive area of research in biogerontology, we are moving towards an era defined by the search for therapeutic drugs that promote healthy aging. Translational biogerontology will require incorporation of both therapeutic and pharmacological concepts. The use of model organisms will remain central to the quest for drug discovery, but as we uncover molecular processes regulated by repurposed drugs and polypharmacy, studies of pharmacodynamics and pharmacokinetics, drug-drug interactions, drug toxicity, and therapeutic index will slowly become more prevalent in aging research. As we move from genetics to pharmacology and therapeutics, studies will not only require demonstration of life span extension and an underlying molecular mechanism, but also the translational relevance for human health and disease prevention.
Collapse
Affiliation(s)
- Jorge Iván Castillo-Quan
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK; Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Kerri J Kinghorn
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK; Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Ivana Bjedov
- Cancer Institute, University College London, London, UK
| |
Collapse
|
37
|
Diet-induced changes in brain structure and behavior in old gerbils. Nutr Diabetes 2015; 5:e163. [PMID: 26075640 PMCID: PMC4491854 DOI: 10.1038/nutd.2015.13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 04/05/2015] [Accepted: 04/24/2015] [Indexed: 11/25/2022] Open
Abstract
Background/Objectives: Aging is associated with many physiological alterations such as changes in metabolism, food intake and brain dysfunction. Possible ways to correct age-related brain dysfunction using dietary treatments still remains undeveloped. The aim of our research was to investigate whether long-term dietary treatment with 2-oxoglutarate (2-OX), which is involved in many regulatory pathways, together with pancreatic-like enzymes of microbial origin (PLEM), which ensure appropriate digestion and absorption of nutrients, affects age-related changes in the brain morphology and cognitive function in old Mongolian gerbils. Materials/methods: Experiment was comprised of two separate studies. Samples of the hippocampus were obtained from male Mongolian gerbils of different ages (n=63 in the first study, n=74 in the second study). Immunohistochemistry was used for visualization of the nestin/NeuN-positive neuronal progenitors. Changes in amount of neural cell adhesion molecules (NCAMs) were estimated using enzyme-linked immunosorbent assay. For assessment of cognitive and sensorimotor functions, the T-maze spontaneous alternation test and the adhesive removal test (ART) were used. The ultrastructure of the CA1 hippocampal area was visualized using transmission electron microscopy. Results: Long-term treatment with 2-OX+PLEM led to a significantly increased amount of nestin/NeuN-positive cells in the CA1 hippocampal area and positive changes in learning and sensorimotor functions. As for synaptic transmission, changes in the spatial distribution of synaptic vesicles, as well as the redistribution of NCAM forms, were observed in the hippocampal synapses of the old gerbils. Conclusions: Taken together, our data show that dietary supplementation with 2-OX+PLEM not only enhances the proliferation and differentiation of neuronal progenitors, but also improves age-related deficits in the morphological and functional state of the brain of old gerbils. Thus, suggesting that a 2-OX+PLEM-enriched diet could also improve brain functions that have deteriorated with age.
Collapse
|
38
|
Millson SH, Piper PW. Insights from yeast into whether the inhibition of heat shock transcription factor (Hsf1) by rapamycin can prevent the Hsf1 activation that results from treatment with an Hsp90 inhibitor. Oncotarget 2015; 5:5054-64. [PMID: 24970820 PMCID: PMC4148121 DOI: 10.18632/oncotarget.2077] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
In human cells TORC1 mTOR (target of rapamycin) protein kinase complex renders heat shock transcription factor 1 (Hsf1) competent for stress activation. In such cells, as well as in yeast, the selective TORC1 inhibitor rapamycin blocks this activation in contrast to Hsp90 inhibitors which potently activate Hsf1. Potentially therefore rapamycin could prevent the Hsf1 activation that frequently compromises the efficiency of Hsp90 inhibitor cancer drugs. Little synergy was found between the effects of rapamycin and the Hsp90 inhibitor radicicol on yeast growth. However certain rapamycin resistance mutations sensitised yeast to Hsp90 inhibitor treatment and an Hsp90 mutation that overactivates Hsf1 sensitised cells to rapamycin. Rapamycin inhibition of the yeast Hsf1 was abolished by this Hsp90 mutation, as well as with the loss of Ppt1, the Hsp90-interacting protein phosphatase that is the ortholog of mammalian PP5. Unexpectedly Hsf1 activation was found to have a requirement for the rapamycin binding immunophilin FKBP12 even in the absence of rapamycin, while TORC1 “bypass” strains revealed that the rapamycin inhibition of yeast Hsf1 is not exerted through two of the major downstream targets of TORC1, the protein phosphatase regulator Tap42 and the protein kinase Sch9 – the latter the ortholog of human S6 protein kinase 1. Significance: A problem with most of the Hsp90 inhibitor drugs now in cancer clinic trials is that they potently activate Hsf1. This leads to an induction of heat shock proteins, many of which have a “pro-survival” role in that they help to protect cells from apopotosis. As the activation of Hsf1 requires TORC1, inhibitors of mTOR kinase could potentially block this activation of Hsf1 and be of value when used in combination drug therapies with Hsp90 inhibitors. However many of the mechanistic details of the TORC1 regulation of Hsf1, as well as the interplay between cellular resistances to rapamycin and to Hsp90 inhibitors, still remain to be resolved.
Collapse
Affiliation(s)
- Stefan H Millson
- Dept. of Molecular Biology and Biotechnology, University of Sheffield, Western Bank, Sheffield, United Kingdom
| | - Peter W Piper
- Dept. of Molecular Biology and Biotechnology, University of Sheffield, Western Bank, Sheffield, United Kingdom
| |
Collapse
|
39
|
Huang X, Leggas M, Dickson RC. Drug synergy drives conserved pathways to increase fission yeast lifespan. PLoS One 2015; 10:e0121877. [PMID: 25786258 PMCID: PMC4364780 DOI: 10.1371/journal.pone.0121877] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 02/11/2015] [Indexed: 01/02/2023] Open
Abstract
Aging occurs over time with gradual and progressive loss of physiological function. Strategies to reduce the rate of functional loss and mitigate the subsequent onset of deadly age-related diseases are being sought. We demonstrated previously that a combination of rapamycin and myriocin reduces age-related functional loss in the Baker’s yeast Saccharomyces cerevisiae and produces a synergistic increase in lifespan. Here we show that the same drug combination also produces a synergistic increase in the lifespan of the fission yeast Schizosaccharomyces pombe and does so by controlling signal transduction pathways conserved across a wide evolutionary time span ranging from yeasts to mammals. Pathways include the target of rapamycin complex 1 (TORC1) protein kinase, the protein kinase A (PKA) and a stress response pathway, which in fission yeasts contains the Sty1 protein kinase, an ortholog of the mammalian p38 MAP kinase, a type of Stress Activated Protein Kinase (SAPK). These results along with previous studies in S. cerevisiae support the premise that the combination of rapamycin and myriocin enhances lifespan by regulating signaling pathways that couple nutrient and environmental conditions to cellular processes that fine-tune growth and stress protection in ways that foster long term survival. The molecular mechanisms for fine-tuning are probably species-specific, but since they are driven by conserved nutrient and stress sensing pathways, the drug combination may enhance survival in other organisms.
Collapse
Affiliation(s)
- Xinhe Huang
- Department of Molecular and Cellular Biochemistry and the Lucille Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- * E-mail: (RCD); (XH)
| | - Markos Leggas
- Department of Pharmaceutical Sciences and the Lucille Markey Cancer Center, College of Pharmacy, University of Kentucky, Lexington, Kentucky, United States of America
| | - Robert C. Dickson
- Department of Molecular and Cellular Biochemistry and the Lucille Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- * E-mail: (RCD); (XH)
| |
Collapse
|
40
|
Carvalhal Marques F, Volovik Y, Cohen E. The Roles of Cellular and Organismal Aging in the Development of Late-Onset Maladies. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2015; 10:1-23. [DOI: 10.1146/annurev-pathol-012414-040508] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Filipa Carvalhal Marques
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada, Hebrew University School of Medicine, 91120 Jerusalem, Israel;
- Centre of Ophthalmology and Vision Sciences, Institute for Biomedical Imaging and Life Sciences, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Yuli Volovik
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada, Hebrew University School of Medicine, 91120 Jerusalem, Israel;
| | - Ehud Cohen
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada, Hebrew University School of Medicine, 91120 Jerusalem, Israel;
| |
Collapse
|
41
|
Abstract
In ageing populations many patients have multiple diseases characterised by acceleration of the normal ageing process. Better understanding of the signalling pathways and cellular events involved in ageing shows that these are characteristic of many chronic degenerative diseases, such as chronic obstructive pulmonary disease (COPD), chronic cardiovascular and metabolic diseases, and neurodegeneration. Common mechanisms have now been identified in these diseases, which show evidence of cellular senescence with telomere shortening, activation of PI3K–AKT–mTOR signalling, impaired autophagy, mitochondrial dysfunction, stem cell exhaustion, epigenetic changes, abnormal microRNA profiles, immunosenescence and low grade chronic inflammation (“inflammaging”). Many of these pathways are driven by chronic oxidative stress. There is also a reduction in anti-ageing molecules, such as sirtuins and Klotho, which further accelerates the ageing process. Understanding these molecular mechanisms has identified several novel therapeutic targets and several drugs have already been developed that may slow the ageing process, as well as lifestyle interventions, such as diet and physical activity. This indicates that in the future new treatment approaches may target the common pathways involved in multimorbidity and this area of research should be given high priority. Thus, COPD should be considered as a component of multimorbidity and common disease pathways, particularly accelerated ageing, should be targeted.
Collapse
|
42
|
Teaching the basics of autophagy and mitophagy to redox biologists--mechanisms and experimental approaches. Redox Biol 2015; 4:242-59. [PMID: 25618581 PMCID: PMC4803799 DOI: 10.1016/j.redox.2015.01.003] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 12/24/2014] [Accepted: 01/01/2015] [Indexed: 02/08/2023] Open
Abstract
Autophagy is a lysosomal mediated degradation activity providing an essential mechanism for recycling cellular constituents, and clearance of excess or damaged lipids, proteins and organelles. Autophagy involves more than 30 proteins and is regulated by nutrient availability, and various stress sensing signaling pathways. This article provides an overview of the mechanisms and regulation of autophagy, its role in health and diseases, and methods for its measurement. Hopefully this teaching review together with the graphic illustrations will be helpful for instructors teaching graduate students who are interested in grasping the concepts and major research areas and introducing recent developments in the field. mTOR, Beclin–VPS34, LC3 homologs, and adaptor proteins in autophagy. Autophagosomal membranes may derive from multiple sources. Autophagosomal–lysosomal fusion contributes to the control of autophagic flux. Assess autophagy by autophagosomal and protein turnover, and morphological alterations. Autophagy adysfunction in cancer, aging, neurodegeneration and infection.
Collapse
|
43
|
Gillespie ZE, MacKay K, Sander M, Trost B, Dawicki W, Wickramarathna A, Gordon J, Eramian M, Kill IR, Bridger JM, Kusalik A, Mitchell JA, Eskiw CH. Rapamycin reduces fibroblast proliferation without causing quiescence and induces STAT5A/B-mediated cytokine production. Nucleus 2015; 6:490-506. [PMID: 26652669 PMCID: PMC4915505 DOI: 10.1080/19491034.2015.1128610] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/24/2015] [Accepted: 11/30/2015] [Indexed: 12/25/2022] Open
Abstract
Rapamycin is a well-known inhibitor of the Target of Rapamycin (TOR) signaling cascade; however, the impact of this drug on global genome function and organization in normal primary cells is poorly understood. To explore this impact, we treated primary human foreskin fibroblasts with rapamycin and observed a decrease in cell proliferation without causing cell death. Upon rapamycin treatment chromosomes 18 and 10 were repositioned to a location similar to that of fibroblasts induced into quiescence by serum reduction. Although similar changes in positioning occurred, comparative transcriptome analyses demonstrated significant divergence in gene expression patterns between rapamycin-treated and quiescence-induced fibroblasts. Rapamycin treatment induced the upregulation of cytokine genes, including those from the Interleukin (IL)-6 signaling network, such as IL-8 and the Leukemia Inhibitory Factor (LIF), while quiescent fibroblasts demonstrated up-regulation of genes involved in the complement and coagulation cascade. In addition, genes significantly up-regulated by rapamycin treatment demonstrated increased promoter occupancy of the transcription factor Signal Transducer and Activator of Transcription 5A/B (STAT5A/B). In summary, we demonstrated that the treatment of fibroblasts with rapamycin decreased proliferation, caused chromosome territory repositioning and induced STAT5A/B-mediated changes in gene expression enriched for cytokines.
Collapse
Affiliation(s)
- Zoe E Gillespie
- Department of Food and Bioproduct Sciences; University of Saskatchewan; Saskatoon, Canada
- Institute of Environment, Health and Societies; Brunel University; London, Uxbridge, United Kingdom
| | - Kimberly MacKay
- Department of Computer Science; University of Saskatchewan; Saskatoon, Canada
| | - Michelle Sander
- Department of Food and Bioproduct Sciences; University of Saskatchewan; Saskatoon, Canada
| | - Brett Trost
- Department of Computer Science; University of Saskatchewan; Saskatoon, Canada
| | - Wojciech Dawicki
- Department of Medicine; Division of Respirology, Critical Care and Sleep Medicine; Royal University Hospital; Saskatoon, Canada
| | - Aruna Wickramarathna
- Department of Food and Bioproduct Sciences; University of Saskatchewan; Saskatoon, Canada
| | - John Gordon
- Department of Medicine; Division of Respirology, Critical Care and Sleep Medicine; Royal University Hospital; Saskatoon, Canada
| | - Mark Eramian
- Department of Computer Science; University of Saskatchewan; Saskatoon, Canada
| | - Ian R Kill
- Institute of Environment, Health and Societies; Brunel University; London, Uxbridge, United Kingdom
| | - Joanna M Bridger
- Institute of Environment, Health and Societies; Brunel University; London, Uxbridge, United Kingdom
| | - Anthony Kusalik
- Department of Computer Science; University of Saskatchewan; Saskatoon, Canada
| | - Jennifer A Mitchell
- Department of Cell and Systems Biology; University of Toronto; Toronto, Canada
- Centre for the Analysis of Genome Evolution and Function; University of Toronto, Toronto, ON, Canada
| | - Christopher H Eskiw
- Department of Food and Bioproduct Sciences; University of Saskatchewan; Saskatoon, Canada
- Institute of Environment, Health and Societies; Brunel University; London, Uxbridge, United Kingdom
| |
Collapse
|
44
|
Ito K, Mercado N. STOP accelerating lung aging for the treatment of COPD. Exp Gerontol 2014; 59:21-7. [DOI: 10.1016/j.exger.2014.03.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 03/15/2014] [Accepted: 03/18/2014] [Indexed: 11/28/2022]
|
45
|
Abstract
Ageing is associated with a progressive degeneration of the tissues, which has a negative impact on the structure and function of vital organs and is among the most important known risk factors for most chronic diseases. Since the proportion of the world's population aged >60 years will double in the next four decades, this will be accompanied by an increased incidence of chronic age-related diseases that will place a huge burden on healthcare resources. There is increasing evidence that many chronic inflammatory diseases represent an acceleration of the ageing process. Chronic pulmonary diseases represents an important component of the increasingly prevalent multiple chronic debilitating diseases, which are a major cause of morbidity and mortality, particularly in the elderly. The lungs age and it has been suggested that chronic obstructive pulmonary disease (COPD) is a condition of accelerated lung ageing and that ageing may provide a mechanistic link between COPD and many of its extrapulmonary effects and comorbidities. In this article we will describe the physiological changes and mechanisms of ageing, with particular focus on the pulmonary effects of ageing and how these may be relevant to the development of COPD and its major extrapulmonary manifestations.
Collapse
Affiliation(s)
- William MacNee
- ELEGI Colt Research Laboratories, MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Roberto A Rabinovich
- ELEGI Colt Research Laboratories, MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Gourab Choudhury
- ELEGI Colt Research Laboratories, MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
46
|
Klermund J, Bender K, Luke B. High nutrient levels and TORC1 activity reduce cell viability following prolonged telomere dysfunction and cell cycle arrest. Cell Rep 2014; 9:324-335. [PMID: 25263563 DOI: 10.1016/j.celrep.2014.08.053] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Revised: 08/08/2014] [Accepted: 08/22/2014] [Indexed: 12/13/2022] Open
Abstract
Cells challenged with DNA damage activate checkpoints to arrest the cell cycle and allow time for repair. Successful repair coupled to subsequent checkpoint inactivation is referred to as recovery. When DNA damage cannot be repaired, a choice between permanent arrest and cycling in the presence of damage (checkpoint adaptation) must be made. While permanent arrest jeopardizes future lineages, continued proliferation is associated with the risk of genome instability. We demonstrate that nutritional signaling through target of rapamycin complex 1 (TORC1) influences the outcome of this decision. Rapamycin-mediated TORC1 inhibition prevents checkpoint adaptation via both Cdc5 inactivation and autophagy induction. Preventing adaptation results in increased cell viability and hence proliferative potential. In accordance, the ability of rapamycin to increase longevity is dependent upon the DNA damage checkpoint. The crosstalk between TORC1 and the DNA damage checkpoint may have important implications in terms of therapeutic alternatives for diseases associated with genome instability.
Collapse
Affiliation(s)
- Julia Klermund
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Katharina Bender
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Brian Luke
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany.
| |
Collapse
|
47
|
Allison DB, Antoine LH, Ballinger SW, Bamman MM, Biga P, Darley-Usmar VM, Fisher G, Gohlke JM, Halade GV, Hartman JL, Hunter GR, Messina JL, Nagy TR, Plaisance EP, Powell ML, Roth KA, Sandel MW, Schwartz TS, Smith DL, Sweatt JD, Tollefsbol TO, Watts SA, Yang Y, Zhang J, Austad SN. Aging and energetics' 'Top 40' future research opportunities 2010-2013. F1000Res 2014; 3:219. [PMID: 25324965 PMCID: PMC4197746 DOI: 10.12688/f1000research.5212.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/08/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND As part of a coordinated effort to expand our research activity at the interface of Aging and Energetics a team of investigators at The University of Alabama at Birmingham systematically assayed and catalogued the top research priorities identified in leading publications in that domain, believing the result would be useful to the scientific community at large. OBJECTIVE To identify research priorities and opportunities in the domain of aging and energetics as advocated in the 40 most cited papers related to aging and energetics in the last 4 years. DESIGN The investigators conducted a search for papers on aging and energetics in Scopus, ranked the resulting papers by number of times they were cited, and selected the ten most-cited papers in each of the four years that include 2010 to 2013, inclusive. RESULTS Ten research categories were identified from the 40 papers. These included: (1) Calorie restriction (CR) longevity response, (2) role of mTOR (mechanistic target of Rapamycin) and related factors in lifespan extension, (3) nutrient effects beyond energy (especially resveratrol, omega-3 fatty acids, and selected amino acids), 4) autophagy and increased longevity and health, (5) aging-associated predictors of chronic disease, (6) use and effects of mesenchymal stem cells (MSCs), (7) telomeres relative to aging and energetics, (8) accretion and effects of body fat, (9) the aging heart, and (10) mitochondria, reactive oxygen species, and cellular energetics. CONCLUSION The field is rich with exciting opportunities to build upon our existing knowledge about the relations among aspects of aging and aspects of energetics and to better understand the mechanisms which connect them.
Collapse
Affiliation(s)
- David B. Allison
- Office of Energetics, University of Alabama at Birmingham, Birmingham, USA
- School of Public Health, University of Alabama at Birmingham, Birmingham, USA
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Lisa H. Antoine
- Office of Energetics, University of Alabama at Birmingham, Birmingham, USA
- School of Engineering, University of Alabama at Birmingham, Birmingham, USA
- Department of Environmental Health Sciences, University of Alabama at Birmingham, Birmingham, USA
| | - Scott W. Ballinger
- Department of Pathology, University of Alabama at Birmingham, Birmingham, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Marcas M. Bamman
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Cell, Developmental, & Integrative Biology, University of Alabama at Birmingham, Birmingham, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, USA
- Birmingham VA Medical Center, Birmingham, USA
| | - Peggy Biga
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Victor M. Darley-Usmar
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Pathology, University of Alabama at Birmingham, Birmingham, USA
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Gordon Fisher
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Human Studies, University of Alabama at Birmingham, Birmingham, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Julia M. Gohlke
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Environmental Health Sciences, University of Alabama at Birmingham, Birmingham, USA
| | - Ganesh V. Halade
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Medicine – Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, USA
| | - John L. Hartman
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Genetics, University of Alabama at Birmingham, Birmingham, USA
| | - Gary R. Hunter
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Human Studies, University of Alabama at Birmingham, Birmingham, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Joseph L. Messina
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Pathology, University of Alabama at Birmingham, Birmingham, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, USA
- Birmingham VA Medical Center, Birmingham, USA
| | - Tim R. Nagy
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, USA
- Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Eric P. Plaisance
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Human Studies, University of Alabama at Birmingham, Birmingham, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Mickie L. Powell
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Kevin A. Roth
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Michael W. Sandel
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, USA
| | - Tonia S. Schwartz
- School of Public Health, University of Alabama at Birmingham, Birmingham, USA
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
| | - Daniel L. Smith
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, USA
| | - J. David Sweatt
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Trygve O. Tollefsbol
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Stephen A. Watts
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Yongbin Yang
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, USA
| | - Jianhua Zhang
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Steven N. Austad
- Nutrition and Obesity Research Center, University of Alabama at Birmingham, Birmingham, USA
- Department of Biology, University of Alabama at Birmingham, Birmingham, USA
- Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, USA
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, USA
| |
Collapse
|
48
|
de Cabo R, Carmona-Gutierrez D, Bernier M, Hall MN, Madeo F. The search for antiaging interventions: from elixirs to fasting regimens. Cell 2014; 157:1515-26. [PMID: 24949965 DOI: 10.1016/j.cell.2014.05.031] [Citation(s) in RCA: 251] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Indexed: 10/25/2022]
Abstract
The phenomenon of aging is an intrinsic feature of life. Accordingly, the possibility to manipulate it has fascinated humans likely since time immemorial. Recent evidence is shaping a picture where low caloric regimes and exercise may improve healthy senescence, and several pharmacological strategies have been suggested to counteract aging. Surprisingly, the most effective interventions proposed to date converge on only a few cellular processes, in particular nutrient signaling, mitochondrial efficiency, proteostasis, and autophagy. Here, we critically examine drugs and behaviors to which life- or healthspan-extending properties have been ascribed and discuss the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA.
| | | | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Michael N Hall
- Biozentrum, University of Basel, Basel 4056, Switzerland
| | - Frank Madeo
- Institute of Molecular Biosciences, University of Graz, Graz 8010, Austria.
| |
Collapse
|
49
|
Extension of Saccharomyces paradoxus chronological lifespan by retrotransposons in certain media conditions is associated with changes in reactive oxygen species. Genetics 2014; 198:531-45. [PMID: 25106655 DOI: 10.1534/genetics.114.168799] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Retrotransposons are mobile DNA elements present throughout eukaryotic genomes that can cause mutations and genome rearrangements when they replicate through reverse transcription. Increased expression and/or mobility of retrotransposons has been correlated with aging in yeast, Caenorhabditis elegans, Drosophila melanogaster, and mammals. The many copies of retrotransposons in humans and various model organisms complicate further pursuit of this relationship. The Saccharomyces cerevisiae Ty1 retrotransposon was introduced into a strain of S. paradoxus that completely lacks retrotransposons to compare chronological lifespans (CLSs) of yeast strains with zero, low, or high Ty1 copy number. Yeast chronological lifespan reflects the progressive loss of cell viability in a nondividing state. Chronological lifespans for the strains were not different in rich medium, but were extended in high Ty1 copy-number strains in synthetic medium and in rich medium containing a low dose of hydroxyurea (HU), an agent that depletes deoxynucleoside triphosphates. Lifespan extension was not strongly correlated with Ty1 mobility or mutation rates for a representative gene. Buffering deoxynucleoside triphosphate levels with threonine supplementation did not substantially affect this lifespan extension, and no substantial differences in cell cycle arrest in the nondividing cells were observed. Lifespan extension was correlated with reduced reactive oxygen species during early stationary phase in high Ty1 copy strains, and antioxidant treatment allowed the zero Ty1 copy strain to live as long as high Ty1 copy-number strains in rich medium with hydroxyurea. This exceptional yeast system has identified an unexpected longevity-promoting role for retrotransposons that may yield novel insights into mechanisms regulating lifespan.
Collapse
|
50
|
Leontieva OV, Paszkiewicz GM, Blagosklonny MV. Weekly administration of rapamycin improves survival and biomarkers in obese male mice on high-fat diet. Aging Cell 2014; 13:616-22. [PMID: 24655348 PMCID: PMC4326934 DOI: 10.1111/acel.12211] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2014] [Indexed: 01/03/2023] Open
Abstract
Recent discoveries have revealed the key role of mTOR (target of rapamycin) in aging. Furthermore, rapamycin extends lifespan in mice, especially in female mice. Here, we treated obese male mice on high-fat diet with rapamycin given intermittently: either weekly (once a week) or alternating bi-weekly (three injections every other week). While only marginally reducing obesity, intermittent administration of rapamycin significantly extended lifespan. Significance was achieved for weekly treated group and for the three rapamycin-received groups combined. In weekly treatment group, 100% mice were alive by the age of 2 years, whereas 60% of mice died in untreated group by this age. The effect of weekly treatment on survival was highly significant and cannot be fully explained by partial reduction in obesity. Alternating bi-weekly treatments seem to be less effective than weekly treatment, although effects of additional factors (see Discussion) may not be excluded. After one year of treatment, all survived mice were sacrificed 8 days after the last administration of rapamycin to avoid its direct interference with parameters examined. Fasting levels of cardiac and hepatic p-S6, a marker of mTORC1 activity, were lower in weekly treatment group compared with control mice. In contrast, levels of p-Akt (S473), glucose, triglycerides and insulin were unchanged, whereas leptin and IGF-1 tended to be lower. Thus, weekly treatment with rapamycin may slow down aging in obese male mice on high-fat diet.
Collapse
Affiliation(s)
- Olga V. Leontieva
- Cell Stress Biology Roswell Park Cancer Institute Buffalo NY 14263USA
| | | | | |
Collapse
|