1
|
Yeewa R, Pohsa S, Yamsri T, Wongkummool W, Jantaree P, Potikanond S, Nimlamool W, Shotelersuk V, Lo Piccolo L, Jantrapirom S. The histone acylation reader ENL/AF9 regulates aging in Drosophila melanogaster. Neurobiol Aging 2024; 144:153-162. [PMID: 39405796 DOI: 10.1016/j.neurobiolaging.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 10/05/2024] [Accepted: 10/05/2024] [Indexed: 10/21/2024]
Abstract
Histone acylation plays a pivotal role in modulating gene expression, ensuring proper neurogenesis and responsiveness to various signals. Recently, the evolutionary conserved YAF9, ENL, AF9, TAF41, SAS5 (YEATS) domain found in four human paralogs, has emerged as a new class of histone acylation reader with a preference for the bulkier crotonyl group lysine over acetylation. Despite advancements, the role of either histone crotonylation or its readers in neurons remains unclear. In this study, we employed Drosophila melanogaster to investigate the role of ENL/AF9 (dENL/AF9) in the nervous system. Pan-neuronal dENL/AF9 knockdown not only extended the lifespan of flies but also enhanced their overall fitness during aging, including improved sleep quality and locomotion. Moreover, a decreased activity of dENL/AF9 in neurons led to an up-regulation of catalase gene expression which combined with reduced levels of malondialdehyde (MDA) and an enhanced tolerance to oxidative stress in aging flies. This study unveiled a novel function of histone crotonylation readers in aging with potential implications for understanding age-related conditions in humans.
Collapse
Affiliation(s)
- Ranchana Yeewa
- Centre of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sureena Pohsa
- Centre of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Titaree Yamsri
- Centre of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Wasinee Wongkummool
- Centre of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Phatcharida Jantaree
- Centre of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Saranyapin Potikanond
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Wutigri Nimlamool
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Vorasuk Shotelersuk
- Centre of Excellence for Medical Genomics, Medical Genomics Cluster, Department of Paediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Excellence Centre for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, the Thai Red Cross Society, Bangkok, Thailand
| | - Luca Lo Piccolo
- Centre of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| | - Salinee Jantrapirom
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Drosophila Centre for Human Diseases and Drug Discovery (DHD), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
2
|
Lind MI, Mautz BS, Carlsson H, Hinas A, Gudmunds E, Maklakov AA. Sex-specific growth and lifespan effects of germline removal in the dioecious nematode Caenorhabditis remanei. Aging Cell 2024; 23:e14290. [PMID: 39082232 DOI: 10.1111/acel.14290] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 11/15/2024] Open
Abstract
Germline regulates the expression of life-history traits and mediates the trade-off between reproduction and somatic maintenance. However, germline maintenance in itself can be costly, and the costs can vary between the sexes depending on the number of gametes produced across the lifetime. We tested this directly by germline ablation using glp-1 RNA interference (RNAi) in a dioecious nematode Caenorhabditis remanei. Germline removal strongly increased heat-shock resistance in both sexes, thus confirming the role of the germline in regulating somatic maintenance. However, germline removal resulted in increased lifespan only in males. High costs of mating strongly reduced lifespan in both sexes and obliterated the survival benefit of germline-less males even though neither sex produced any offspring. Furthermore, germline removal reduced male growth before maturation but not in adulthood, while female growth rate was reduced both before and especially after maturation. Thus, germline removal improves male lifespan without major growth costs, while germline-less females grow slower and do not live longer than reproductively functional counterparts in the absence of environmental stress. Overall, these results suggest that germline maintenance is costlier for males than for females in C. remanei.
Collapse
Affiliation(s)
- Martin I Lind
- Animal Ecology, Department of Ecology and Genetics, Uppsala University, Uppsala, Sweden
- Department of Environmental and Biosciences, Halmstad University, Halmstad, Sweden
| | - Brian S Mautz
- Animal Ecology, Department of Ecology and Genetics, Uppsala University, Uppsala, Sweden
- Population Analytics & Insights, Data Sciences Analytics & Insights, Innovative Medicine Research & Development, Johnson & Johnson, Spring House, Pennsylvania, USA
| | - Hanne Carlsson
- Animal Ecology, Department of Ecology and Genetics, Uppsala University, Uppsala, Sweden
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Andrea Hinas
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Erik Gudmunds
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
- Evolutionary Biology, Department of Ecology and Genetics, Uppsala University, Uppsala, Sweden
| | - Alexei A Maklakov
- Animal Ecology, Department of Ecology and Genetics, Uppsala University, Uppsala, Sweden
- School of Biological Sciences, University of East Anglia, Norwich, UK
| |
Collapse
|
3
|
Liu M, Chen J, Cui G, Dai Y, Song M, Zhou C, Hu Q, Chen Q, Wang H, Chen W, Han JJ, Peng G, Jing N, Shen Y. Germline loss in C. elegans enhances longevity by disrupting adhesion between niche and stem cells. EMBO J 2024; 43:4000-4019. [PMID: 39060516 PMCID: PMC11405865 DOI: 10.1038/s44318-024-00185-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 07/03/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Ageing and fertility are intertwined. Germline loss extends the lifespan in various organisms, termed gonadal longevity. However, the original longevity signal from the somatic gonad remains poorly understood. Here, we focused on the interaction between germline stem cells (GSCs) and their niche, the distal tip cells (DTCs), to explore the barely known longevity signal from the somatic gonad in C. elegans. We found that removing germline disrupts the cell adhesions between GSC and DTC, causing a significant transcriptomic change in DTC through hmp-2/β-catenin and two GATA transcription factors, elt-3 and pqm-1 in this niche cell. Inhibiting elt-3 and pqm-1 in DTC suppresses gonadal longevity. Moreover, we further identified the TGF-β ligand, tig-2, as the cytokine from DTC upon the loss of germline, which evokes the downstream gonadal longevity signalling throughout the body. Our findings thus reveal the source of the longevity signalling in response to germline removal, highlighting the stem cell niche as a critical signalling hub in ageing.
Collapse
Affiliation(s)
- Meng Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Jiehui Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Guizhong Cui
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
- Guangzhou Laboratory, 510005, Guangzhou, China
| | - Yumin Dai
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Mengjiao Song
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Chunyu Zhou
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, 102213, Beijing, China
| | - Qingyuan Hu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Qingxia Chen
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Institute of Early Life Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 200092, Shanghai, China
| | - Hongwei Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Wanli Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Jingdong Jackie Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, 102213, Beijing, China
| | - Guangdun Peng
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Guangzhou Laboratory, 510005, Guangzhou, China
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Naihe Jing
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Guangzhou Laboratory, 510005, Guangzhou, China
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Yidong Shen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 200031, Shanghai, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
| |
Collapse
|
4
|
Eder M, Martin OMF, Oswal N, Sedlackova L, Moutinho C, Del Carmen-Fabregat A, Menendez Bravo S, Sebé-Pedrós A, Heyn H, Stroustrup N. Systematic mapping of organism-scale gene-regulatory networks in aging using population asynchrony. Cell 2024; 187:3919-3935.e19. [PMID: 38908368 DOI: 10.1016/j.cell.2024.05.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 04/02/2024] [Accepted: 05/27/2024] [Indexed: 06/24/2024]
Abstract
In aging, physiologic networks decline in function at rates that differ between individuals, producing a wide distribution of lifespan. Though 70% of human lifespan variance remains unexplained by heritable factors, little is known about the intrinsic sources of physiologic heterogeneity in aging. To understand how complex physiologic networks generate lifespan variation, new methods are needed. Here, we present Asynch-seq, an approach that uses gene-expression heterogeneity within isogenic populations to study the processes generating lifespan variation. By collecting thousands of single-individual transcriptomes, we capture the Caenorhabditis elegans "pan-transcriptome"-a highly resolved atlas of non-genetic variation. We use our atlas to guide a large-scale perturbation screen that identifies the decoupling of total mRNA content between germline and soma as the largest source of physiologic heterogeneity in aging, driven by pleiotropic genes whose knockdown dramatically reduces lifespan variance. Our work demonstrates how systematic mapping of physiologic heterogeneity can be applied to reduce inter-individual disparities in aging.
Collapse
Affiliation(s)
- Matthias Eder
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
| | - Olivier M F Martin
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Natasha Oswal
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Lucia Sedlackova
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
| | - Cátia Moutinho
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona, Spain
| | - Andrea Del Carmen-Fabregat
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Simon Menendez Bravo
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain
| | - Arnau Sebé-Pedrós
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; ICREA, Pg. Lluis Companys 23, Barcelona 08010, Spain
| | - Holger Heyn
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona, Spain; ICREA, Pg. Lluis Companys 23, Barcelona 08010, Spain
| | - Nicholas Stroustrup
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain.
| |
Collapse
|
5
|
Easwaran S, Montell DJ. A genome-wide association study implicates the olfactory system in Drosophila melanogaster diapause-associated lifespan extension and fecundity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.10.584341. [PMID: 39005458 PMCID: PMC11244867 DOI: 10.1101/2024.03.10.584341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The effects of environmental stress on animal life are gaining importance with climate change. Diapause is a dormancy program that occurs in response to an adverse environment, followed by resumption of development and reproduction upon the return of favorable conditions. Diapause is a complex trait, so we leveraged the Drosophila genetic reference panel (DGRP) lines and conducted a Genome-Wide Association Study (GWAS) to characterize the genetic basis of diapause. We assessed post-diapause and non-diapause fecundity across 193 DGRP lines. GWAS revealed 546 genetic variants, encompassing single nucleotide polymorphisms, insertions and deletions associated with post-diapause fecundity. We identified 291 candidate diapause-associated genes, 40 of which had previously been associated with diapause. 89 of the candidates were associated with more than one SNP. Gene network analysis indicated that the diapause-associated genes were primarily linked to neuronal and reproductive system development. Similarly, comparison with results from other fly GWAS revealed the greatest overlap with olfactory-behavior-associated and fecundity-and-lifespan-associated genes. An RNAi screen of the top candidates identified two neuronal genes, Dip-γ and Scribbler, to be required during recovery for post-diapause fecundity. We complemented the genetic analysis with a test of which neurons are required for successful diapause. We found that although amputation of the antenna had little to no effect on non-diapause lifespan, it reduced diapause lifespan and postdiapause fecundity. We further show that olfactory receptor neurons and temperature-sensing neurons are required for successful recovery from diapause. Our results provide insights into the molecular, cellular, and genetic basis of adult reproductive diapause in Drosophila .
Collapse
|
6
|
Sala AJ, Grant RA, Imran G, Morton C, Brielmann RM, Gorgoń S, Watts J, Bott LC, Morimoto RI. Nuclear receptor signaling via NHR-49/MDT-15 regulates stress resilience and proteostasis in response to reproductive and metabolic cues. Genes Dev 2024; 38:380-392. [PMID: 38816072 PMCID: PMC11216168 DOI: 10.1101/gad.351829.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 05/16/2024] [Indexed: 06/01/2024]
Abstract
The ability to sense and respond to proteotoxic insults declines with age, leaving cells vulnerable to chronic and acute stressors. Reproductive cues modulate this decline in cellular proteostasis to influence organismal stress resilience in Caenorhabditis elegans We previously uncovered a pathway that links the integrity of developing embryos to somatic health in reproductive adults. Here, we show that the nuclear receptor NHR-49, an ortholog of mammalian peroxisome proliferator-activated receptor α (PPARα), regulates stress resilience and proteostasis downstream from embryo integrity and other pathways that influence lipid homeostasis and upstream of HSF-1. Disruption of the vitelline layer of the embryo envelope, which activates a proteostasis-enhancing intertissue pathway in somatic cells, triggers changes in lipid catabolism gene expression that are accompanied by an increase in fat stores. NHR-49, together with its coactivator, MDT-15, contributes to this remodeling of lipid metabolism and is also important for the elevated stress resilience mediated by inhibition of the embryonic vitelline layer. Our findings indicate that NHR-49 also contributes to stress resilience in other pathways known to change lipid homeostasis, including reduced insulin-like signaling and fasting, and that increased NHR-49 activity is sufficient to improve proteostasis and stress resilience in an HSF-1-dependent manner. Together, our results establish NHR-49 as a key regulator that links lipid homeostasis and cellular resilience to proteotoxic stress.
Collapse
Affiliation(s)
- Ambre J Sala
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208, USA;
- Institute for Integrative Biology of the Cell (I2BC), Commissariat à l'Énergie Atomique et Aux Énergies Alternatives (CEA), Centre National de la Recherche Scientifique (CNRS), Université Paris-Saclay, Gif-sur-Yvette 91190, France
| | - Rogan A Grant
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Ghania Imran
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208, USA
| | - Claire Morton
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208, USA
| | - Renee M Brielmann
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208, USA
| | - Szymon Gorgoń
- Institute for Integrative Biology of the Cell (I2BC), Commissariat à l'Énergie Atomique et Aux Énergies Alternatives (CEA), Centre National de la Recherche Scientifique (CNRS), Université Paris-Saclay, Gif-sur-Yvette 91190, France
| | - Jennifer Watts
- School of Molecular Biosciences, Washington State University, Pullman, Washington 99164, USA
| | - Laura C Bott
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208, USA
| | - Richard I Morimoto
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208, USA;
| |
Collapse
|
7
|
Abe K, Ino H, Niwa T, Semmy D, Takaochi A, Nishimura T, Mogi C, Uenaka M, Ishii M, Tanaka K, Ohkawa Y, Ishitani T. Sex-dependent regulation of vertebrate somatic growth and aging by germ cells. SCIENCE ADVANCES 2024; 10:eadi1621. [PMID: 38865462 PMCID: PMC11168456 DOI: 10.1126/sciadv.adi1621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 04/23/2024] [Indexed: 06/14/2024]
Abstract
The function of germ cells in somatic growth and aging has been demonstrated in invertebrate models but remains unclear in vertebrates. We demonstrated sex-dependent somatic regulation by germ cells in the short-lived vertebrate model Nothobranchius furzeri. In females, germ cell removal shortened life span, decreased estrogen, and increased insulin-like growth factor 1 (IGF-1) signaling. In contrast, germ cell removal in males improved their health with increased vitamin D signaling. Body size increased in both sexes but was caused by different signaling pathways, i.e., IGF-1 and vitamin D in females and males, respectively. Thus, vertebrate germ cells regulate somatic growth and aging through different pathways of the endocrine system, depending on the sex, which may underlie the sexual difference in reproductive strategies.
Collapse
Affiliation(s)
- Kota Abe
- Department of Homeostatic Regulation, Division of Cellular and Molecular Biology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Hikaru Ino
- Department of Homeostatic Regulation, Division of Cellular and Molecular Biology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Tomomi Niwa
- Department of Homeostatic Regulation, Division of Cellular and Molecular Biology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Daniel Semmy
- Department of Homeostatic Regulation, Division of Cellular and Molecular Biology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Ayami Takaochi
- Department of Homeostatic Regulation, Division of Cellular and Molecular Biology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Takashi Nishimura
- Metabolic Regulation and Genetics, Department of Molecular and Cellular Biology, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Chihiro Mogi
- Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Maki Uenaka
- Department of Immunology and Cell Biology, Graduate School of Medicine / Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
- Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine / Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
- Immunology Frontier Research Center, Osaka University, Suita, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan
| | - Kaori Tanaka
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Tohru Ishitani
- Department of Homeostatic Regulation, Division of Cellular and Molecular Biology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan
| |
Collapse
|
8
|
Moses E, Atlan T, Sun X, Franěk R, Siddiqui A, Marinov GK, Shifman S, Zucker DM, Oron-Gottesman A, Greenleaf WJ, Cohen E, Ram O, Harel I. The killifish germline regulates longevity and somatic repair in a sex-specific manner. NATURE AGING 2024; 4:791-813. [PMID: 38750187 DOI: 10.1038/s43587-024-00632-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 04/10/2024] [Indexed: 05/22/2024]
Abstract
Classical evolutionary theories propose tradeoffs among reproduction, damage repair and lifespan. However, the specific role of the germline in shaping vertebrate aging remains largely unknown. In this study, we used the turquoise killifish (Nothobranchius furzeri) to genetically arrest germline development at discrete stages and examine how different modes of infertility impact life history. We first constructed a comprehensive single-cell gonadal atlas, providing cell-type-specific markers for downstream phenotypic analysis. We show here that germline depletion-but not arresting germline differentiation-enhances damage repair in female killifish. Conversely, germline-depleted males instead showed an extension in lifespan and rejuvenated metabolic functions. Through further transcriptomic analysis, we highlight enrichment of pro-longevity pathways and genes in germline-depleted male killifish and demonstrate functional conservation of how these factors may regulate longevity in germline-depleted Caenorhabditis elegans. Our results, therefore, demonstrate that different germline manipulation paradigms can yield pronounced sexually dimorphic phenotypes, implying alternative responses to classical evolutionary tradeoffs.
Collapse
Affiliation(s)
- Eitan Moses
- Department of Genetics, Silberman Institute, Hebrew University of Jerusalem, Givat Ram, Jerusalem, Israel
| | - Tehila Atlan
- Department of Genetics, Silberman Institute, Hebrew University of Jerusalem, Givat Ram, Jerusalem, Israel
| | - Xue Sun
- Department of Biochemistry, Silberman Institute, Hebrew University of Jerusalem, Givat Ram, Jerusalem, Israel
| | - Roman Franěk
- Department of Genetics, Silberman Institute, Hebrew University of Jerusalem, Givat Ram, Jerusalem, Israel
- South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, University of South Bohemia in Ceske Budejovice, Vodnany, Czech Republic
| | - Atif Siddiqui
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Hebrew University School of Medicine, Jerusalem, Israel
| | | | - Sagiv Shifman
- Department of Genetics, Silberman Institute, Hebrew University of Jerusalem, Givat Ram, Jerusalem, Israel
| | - David M Zucker
- Department of Statistics and Data Science, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Adi Oron-Gottesman
- Department of Genetics, Silberman Institute, Hebrew University of Jerusalem, Givat Ram, Jerusalem, Israel
| | - William J Greenleaf
- Department of Genetics, Stanford University, Stanford, CA, USA
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
- Department of Applied Physics, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Ehud Cohen
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Hebrew University School of Medicine, Jerusalem, Israel
| | - Oren Ram
- Department of Biochemistry, Silberman Institute, Hebrew University of Jerusalem, Givat Ram, Jerusalem, Israel
| | - Itamar Harel
- Department of Genetics, Silberman Institute, Hebrew University of Jerusalem, Givat Ram, Jerusalem, Israel.
| |
Collapse
|
9
|
Moses E, Atlan T, Sun X, Franek R, Siddiqui A, Marinov GK, Shifman S, Zucker DM, Oron-Gottesman A, Greenleaf WJ, Cohen E, Ram O, Harel I. The killifish germline regulates longevity and somatic repair in a sex-specific manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.18.572041. [PMID: 38187630 PMCID: PMC10769255 DOI: 10.1101/2023.12.18.572041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Classical evolutionary theories propose tradeoffs between reproduction, damage repair, and lifespan. However, the specific role of the germline in shaping vertebrate aging remains largely unknown. Here, we use the turquoise killifish ( N. furzeri ) to genetically arrest germline development at discrete stages, and examine how different modes of infertility impact life-history. We first construct a comprehensive single-cell gonadal atlas, providing cell-type-specific markers for downstream phenotypic analysis. Next, we show that germline depletion - but not arresting germline differentiation - enhances damage repair in female killifish. Conversely, germline-depleted males instead showed an extension in lifespan and rejuvenated metabolic functions. Through further transcriptomic analysis, we highlight enrichment of pro-longevity pathways and genes in germline-depleted male killifish and demonstrate functional conservation of how these factors may regulate longevity in germline-depleted C. elegans . Our results therefore demonstrate that different germline manipulation paradigms can yield pronounced sexually dimorphic phenotypes, implying alternative responses to classical evolutionary tradeoffs.
Collapse
|
10
|
Pires da Silva A, Kelleher R, Reynoldson L. Decoding lifespan secrets: the role of the gonad in Caenorhabditis elegans aging. FRONTIERS IN AGING 2024; 5:1380016. [PMID: 38605866 PMCID: PMC11008531 DOI: 10.3389/fragi.2024.1380016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 03/18/2024] [Indexed: 04/13/2024]
Abstract
The gonad has become a central organ for understanding aging in C. elegans, as removing the proliferating stem cells in the germline results in significant lifespan extension. Similarly, when starvation in late larval stages leads to the quiescence of germline stem cells the adult nematode enters reproductive diapause, associated with an extended lifespan. This review summarizes recent advancements in identifying the mechanisms behind gonad-mediated lifespan extension, including comparisons with other nematodes and the role of lipid signaling and transcriptional changes. Given that the gonad also mediates lifespan regulation in other invertebrates and vertebrates, elucidating the underlying mechanisms may help to gain new insights into the mechanisms and evolution of aging.
Collapse
|
11
|
Landis GN, Bell HS, Peng O, Bognar B, Tong A, Manea TD, Bao H, Han X, Tower J. Dhr96[1] mutation and maternal tudor[1] mutation increase life span and reduce the beneficial effects of mifepristone in mated female Drosophila. PLoS One 2023; 18:e0292820. [PMID: 38127988 PMCID: PMC10735022 DOI: 10.1371/journal.pone.0292820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/28/2023] [Indexed: 12/23/2023] Open
Abstract
Mating and receipt of male Sex Peptide hormone cause increased egg laying, increased midgut size and decreased life span in female Drosophila. Feeding mated females with the synthetic steroid mifepristone decreases egg production, reduces midgut size, and increases life span. Here, several gene mutations were assayed to investigate possible mechanisms for mifepristone action. Drosophila Dhr96 is a hormone receptor, and a key positive regulator of midgut lipid uptake and metabolism. Dhr96[1] null mutation increased female life span, and reduced the effects of mifepristone on life span, suggesting that Dhr96[1] mutation and mifepristone may act in part through the same mechanism. Consistent with this idea, lipidomics analysis revealed that mating increases whole-body levels of triglycerides and fatty-acids in triglycerides, and these changes are reversed by mifepristone. Maternal tudor[1] mutation results in females that lack the germ-line and produce no eggs. Maternal tudor[1] mutation increased mated female life span, and reduced but did not eliminate the effects of mating and mifepristone on life span. This indicates that decreased egg production may be related to the life span benefits of mifepristone, but is not essential. Mifepristone increases life span in w[1118] mutant mated females, but did not increase life span in w[1118] mutant virgin females. Mifepristone decreased egg production in w[1118] mutant virgin females, indicating that decreased egg production is not sufficient for mifepristone to increase life span. Mifepristone increases life span in virgin females of some, but not all, white[+] and mini-white[+] strains. Backcrossing of mini-white[+] transgenes into the w[1118] background was not sufficient to confer a life span response to mifepristone in virgin females. Taken together, the data support the hypothesis that mechanisms for mifepristone life span increase involve reduced lipid uptake and/or metabolism, and suggest that mifepristone may increase life span in mated females and virgin females through partly different mechanisms.
Collapse
Affiliation(s)
- Gary N. Landis
- Department of Biological Sciences, University of Southern California, Los Angeles, California, United States of America
| | - Hans S. Bell
- Department of Biological Sciences, University of Southern California, Los Angeles, California, United States of America
| | - Oscar Peng
- Department of Biological Sciences, University of Southern California, Los Angeles, California, United States of America
| | - Brett Bognar
- Department of Biological Sciences, University of Southern California, Los Angeles, California, United States of America
| | - Andy Tong
- Department of Biological Sciences, University of Southern California, Los Angeles, California, United States of America
| | - Tomás D. Manea
- Department of Biological Sciences, University of Southern California, Los Angeles, California, United States of America
| | - Hanmei Bao
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - John Tower
- Department of Biological Sciences, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
12
|
Hernández G, Vazquez-Pianzola P. eIF4E as a molecular wildcard in metazoans RNA metabolism. Biol Rev Camb Philos Soc 2023; 98:2284-2306. [PMID: 37553111 DOI: 10.1111/brv.13005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/01/2023] [Accepted: 07/25/2023] [Indexed: 08/10/2023]
Abstract
The evolutionary origin of eukaryotes spurred the transition from prokaryotic-like translation to a more sophisticated, eukaryotic translation. During this process, successive gene duplication of a single, primordial eIF4E gene encoding the mRNA cap-binding protein eukaryotic translation initiation factor 4E (eIF4E) gave rise to a plethora of paralog genes across eukaryotes that underwent further functional diversification in RNA metabolism. The ability to take different roles is due to eIF4E promiscuity in binding many partner proteins, rendering eIF4E a highly versatile and multifunctional player that functions as a molecular wildcard. Thus, in metazoans, eIF4E paralogs are involved in various processes, including messenger RNA (mRNA) processing, export, translation, storage, and decay. Moreover, some paralogs display differential expression in tissues and developmental stages and show variable biochemical properties. In this review, we discuss recent advances shedding light on the functional diversification of eIF4E in metazoans. We emphasise humans and two phylogenetically distant species which have become paradigms for studies on development, namely the fruit fly Drosophila melanogaster and the roundworm Caenorhabditis elegans.
Collapse
Affiliation(s)
- Greco Hernández
- mRNA and Cancer Laboratory, Unit of Biomedical Research on Cancer, National Institute of Cancer (Instituto Nacional de Cancerología, INCan), 22 San Fernando Ave., Tlalpan, Mexico City, 14080, Mexico
| | - Paula Vazquez-Pianzola
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, Berne, 3012, Switzerland
| |
Collapse
|
13
|
Liu T, Zhang L. Multigenerational effects of arsenate on development and reproduction in marine copepod Tigriopus japonicus. CHEMOSPHERE 2023; 342:140158. [PMID: 37709060 DOI: 10.1016/j.chemosphere.2023.140158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/21/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023]
Abstract
Arsenic (As) is a persistent toxic substance, however, its toxicity to marine zooplankton remains unclear. In this study, copepods were exposed to a series of dissolved arsenate (As(V)) for four generations (F0-F3) and subsequently depurated in clean seawater for two generations (F4-F5) to assess multigenerational toxicity of As(V). As(V) exposure prolonged copepod development. The development time were 1.9, 2.4, and 3.4 days longer than the control in F0 when exposed to 50, 100, and 500 μg/L As(V), respectively, and the toxicity increased with generations. Moreover, As(V) reduced the reproductive capacity of copepods, and this effect become more severe during generation succession. The 10-day fecundities were reduced from 80 to 85 eggs per female in the control to 42 eggs per female, the lowest level, in 500 μg/L As(V) exposure group in F3. Nevertheless, the fecundity was recovered to the control level in the offspring of the 50 and 100 μg/L As(V) exposed groups (F4), suggesting it was an acclimation effect of copepods during As(V) exposure. In addition, the survival rate, development time, and reproductive parameters were significantly correlated with the As accumulation in copepods. Overall, As(V) exposure caused As bioaccumulation which negatively affected copepods' survival, development, and reproductive traits, and this toxic effect was amplified with generations and concentrations. Therefore, the multigenerational toxicity of As should be considered in the environmental risk assessments.
Collapse
Affiliation(s)
- Tianrui Liu
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Li Zhang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301, China; Sanya Institute of Ocean Eco-Environmental Engineering, Sanya, 572025, China.
| |
Collapse
|
14
|
Kyriazis M, Swas L, Orlova T. The Impact of Hormesis, Neuronal Stress Response, and Reproduction, upon Clinical Aging: A Narrative Review. J Clin Med 2023; 12:5433. [PMID: 37629475 PMCID: PMC10455615 DOI: 10.3390/jcm12165433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/05/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
INTRODUCTION The primary objective of researchers in the biology of aging is to gain a comprehensive understanding of the aging process while developing practical solutions that can enhance the quality of life for older individuals. This involves a continuous effort to bridge the gap between fundamental biological research and its real-world applications. PURPOSE In this narrative review, we attempt to link research findings concerning the hormetic relationship between neurons and germ cells, and translate these findings into clinically relevant concepts. METHODS We conducted a literature search using PubMed, Embase, PLOS, Digital Commons Network, Google Scholar and Cochrane Library from 2000 to 2023, analyzing studies dealing with the relationship between hormetic, cognitive, and reproductive aspects of human aging. RESULTS The process of hormesis serves as a bridge between the biology of neuron-germ cell interactions on one hand, and the clinical relevance of these interactions on the other. Details concerning these processes are discussed here, emphasizing new research which strengthens the overall concept. CONCLUSIONS This review presents a scientifically and clinically relevant argument, claiming that maintaining a cognitively active lifestyle may decrease age-related degeneration, and improve overall health in aging. This is a totally novel approach which reflects current developments in several relevant aspects of our biology, technology, and society.
Collapse
|
15
|
Kim HS, Parker DJ, Hardiman MM, Munkácsy E, Jiang N, Rogers AN, Bai Y, Brent C, Mobley JA, Austad SN, Pickering AM. Early-adulthood spike in protein translation drives aging via juvenile hormone/germline signaling. Nat Commun 2023; 14:5021. [PMID: 37596266 PMCID: PMC10439225 DOI: 10.1038/s41467-023-40618-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 08/01/2023] [Indexed: 08/20/2023] Open
Abstract
Protein translation (PT) declines with age in invertebrates, rodents, and humans. It has been assumed that elevated PT at young ages is beneficial to health and PT ends up dropping as a passive byproduct of aging. In Drosophila, we show that a transient elevation in PT during early-adulthood exerts long-lasting negative impacts on aging trajectories and proteostasis in later-life. Blocking the early-life PT elevation robustly improves life-/health-span and prevents age-related protein aggregation, whereas transiently inducing an early-life PT surge in long-lived fly strains abolishes their longevity/proteostasis benefits. The early-life PT elevation triggers proteostatic dysfunction, silences stress responses, and drives age-related functional decline via juvenile hormone-lipid transfer protein axis and germline signaling. Our findings suggest that PT is adaptively suppressed after early-adulthood, alleviating later-life proteostatic burden, slowing down age-related functional decline, and improving lifespan. Our work provides a theoretical framework for understanding how lifetime PT dynamics shape future aging trajectories.
Collapse
Affiliation(s)
- Harper S Kim
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
- Medical Scientist Training Program, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Danitra J Parker
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, 77030, USA
| | - Madison M Hardiman
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Erin Munkácsy
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Nisi Jiang
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Aric N Rogers
- MDI Biological Laboratory, Bar Harbor, ME, 04672, USA
| | - Yidong Bai
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Colin Brent
- USDA-ARS Arid Land Agricultural Research Center, Maricopa, AZ, 85138, USA
| | - James A Mobley
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, 35249, USA
| | - Steven N Austad
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Nathan Shock Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Andrew M Pickering
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, 78229, USA.
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, 77030, USA.
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
16
|
Soo SK, Rudich ZD, Ko B, Moldakozhayev A, AlOkda A, Van Raamsdonk JM. Biological resilience and aging: Activation of stress response pathways contributes to lifespan extension. Ageing Res Rev 2023; 88:101941. [PMID: 37127095 DOI: 10.1016/j.arr.2023.101941] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/06/2023] [Accepted: 04/28/2023] [Indexed: 05/03/2023]
Abstract
While aging was traditionally viewed as a stochastic process of damage accumulation, it is now clear that aging is strongly influenced by genetics. The identification and characterization of long-lived genetic mutants in model organisms has provided insights into the genetic pathways and molecular mechanisms involved in extending longevity. Long-lived genetic mutants exhibit activation of multiple stress response pathways leading to enhanced resistance to exogenous stressors. As a result, lifespan exhibits a significant, positive correlation with resistance to stress. Disruption of stress response pathways inhibits lifespan extension in multiple long-lived mutants representing different pathways of lifespan extension and can also reduce the lifespan of wild-type animals. Combined, this suggests that activation of stress response pathways is a key mechanism by which long-lived mutants achieve their extended longevity and that many of these pathways are also required for normal lifespan. These results highlight an important role for stress response pathways in determining the lifespan of an organism.
Collapse
Affiliation(s)
- Sonja K Soo
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Zenith D Rudich
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Bokang Ko
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Alibek Moldakozhayev
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Abdelrahman AlOkda
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Jeremy M Van Raamsdonk
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
17
|
Wenzel M, Aquadro CF. Wolbachia genetically interacts with the bag of marbles germline stem cell gene in male D. melanogaster. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000845. [PMID: 37416893 PMCID: PMC10321140 DOI: 10.17912/micropub.biology.000845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/20/2023] [Accepted: 05/23/2023] [Indexed: 07/08/2023]
Abstract
The bacterial endosymbiont Wolbachia manipulates reproduction of its arthropod hosts to promote its own maternal vertical transmission. In female D. melanogaster , Wolbachia has been shown to genetically interact with three key reproductive genes ( bag of marbles ( bam ) , Sex-lethal, and mei-P26) , as it rescues the reduced female fertility or fecundity phenotype seen in partial loss-of-function mutants of these genes . Here, we show that Wolbachia also partially rescues male fertility in D. melanogaster carrying a new, largely sterile bam allele when in a bam null genetic background. This finding shows that the molecular mechanism of Wolbachia 's influence on its hosts' reproduction involves interaction with genes in males as well as females, at least in D. melanogaster .
Collapse
Affiliation(s)
- Miwa Wenzel
- Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States
| | - Charles F. Aquadro
- Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States
| |
Collapse
|
18
|
Sala AJ, Grant RA, Imran G, Morton C, Brielmann RM, Bott LC, Watts J, Morimoto RI. Nuclear receptor signaling via NHR-49/MDT-15 regulates stress resilience and proteostasis in response to reproductive and metabolic cues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.25.537803. [PMID: 37162952 PMCID: PMC10168274 DOI: 10.1101/2023.04.25.537803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The ability to sense and respond to proteotoxic insults declines with age, leaving cells vulnerable to chronic and acute stressors. Reproductive cues modulate this decline in cellular proteostasis to influence organismal stress resilience in C. elegans. We previously uncovered a pathway that links the integrity of developing embryos to somatic health in reproductive adults. Here, we show that the nuclear receptor NHR-49, a functional homolog of mammalian peroxisome proliferator-activated receptor alpha (PPARα), regulates stress resilience and proteostasis downstream of embryo integrity and other pathways that influence lipid homeostasis, and upstream of HSF-1. Disruption of the vitelline layer of the embryo envelope, which activates a proteostasis-enhancing inter-tissue pathway in somatic tissues, also triggers changes in lipid catabolism gene expression that are accompanied by an increase in fat stores. NHR-49 together with its co-activator MDT-15 contributes to this remodeling of lipid metabolism and is also important for the elevated stress resilience mediated by inhibition of the embryonic vitelline layer as well as by other pathways known to change lipid homeostasis, including reduced insulin-like signaling and fasting. Further, we show that increased NHR-49 activity is sufficient to suppress polyglutamine aggregation and improve stress resilience in an HSF-1-dependent manner. Together, our results establish NHR-49 as a key regulator that links lipid homeostasis and cellular resilience to proteotoxic stress.
Collapse
Affiliation(s)
- Ambre J. Sala
- Department of Molecular Biosciences, Northwestern University, Evanston IL, USA
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, Gifsur-Yvette, France
| | - Rogan A. Grant
- Department of Molecular Biosciences, Northwestern University, Evanston IL, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ghania Imran
- Department of Molecular Biosciences, Northwestern University, Evanston IL, USA
| | - Claire Morton
- Department of Molecular Biosciences, Northwestern University, Evanston IL, USA
| | - Renee M. Brielmann
- Department of Molecular Biosciences, Northwestern University, Evanston IL, USA
| | - Laura C. Bott
- Department of Molecular Biosciences, Northwestern University, Evanston IL, USA
| | - Jennifer Watts
- School of Molecular Biosciences, Washington State University, Pullman WA, USA
| | - Richard I. Morimoto
- Department of Molecular Biosciences, Northwestern University, Evanston IL, USA
| |
Collapse
|
19
|
King TL, Bryner BS, Underwood KB, Walters MR, Zimmerman SM, Johnson NK, Mason JB. Estradiol-independent restoration of T-cell function in post-reproductive females. Front Endocrinol (Lausanne) 2023; 14:1066356. [PMID: 36755910 PMCID: PMC9900006 DOI: 10.3389/fendo.2023.1066356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
Aging leads to a general decline in protective immunity. The most common age-associated effects are in seen T-cell mediated immune function. Adult mice whose immune systems show only moderate changes in T-cell subsets tend to live longer than age-matched siblings that display extensive T-cell subset aging. Importantly, at the time of reproductive decline, the increase in disease risks in women significantly outpace those of men. In female mice, there is a significant decline in central and peripheral naïve T-cell subsets at the time of reproductive failure. Available evidence indicates that this naïve T-cell decline is sensitive to ovarian function and can be reversed in post-reproductive females by transplantation of young ovaries. The restoration of naïve T-cell subsets due to ovarian transplantation was impressive compared with post-reproductive control mice, but represented only a partial recovery of what was lost from 6 months of age. Apparently, the influence of ovarian function on immune function may be an indirect effect, likely moderated by other physiological functions. Estradiol is significantly reduced in post-reproductive females, but was not increased in post-reproductive females that received new ovaries, suggesting an estradiol-independent, but ovarian-dependent influence on immune function. Further evidence for an estradiol-independent influence includes the restoration of immune function through the transplantation of young ovaries depleted of follicles and through the injection of isolated ovarian somatic cells into the senescent ovaries of old mice. While the restoration of naïve T-cell populations represents only a small part of the immune system, the ability to reverse this important functional parameter independent of estradiol may hold promise for the improvement of post-reproductive female immune health. Further studies of the non-reproductive influence of the ovary will be needed to elucidate the mechanisms of the relationship between the ovary and health.
Collapse
Affiliation(s)
- Tristin L. King
- Department of Animal, Dairy and Veterinary Sciences, Center for Integrated BioSystems, College of Veterinary Medicine, Utah State University, Logan, UT, United States
| | - B. Shaun Bryner
- Department of Animal, Dairy and Veterinary Sciences, Center for Integrated BioSystems, College of Veterinary Medicine, Utah State University, Logan, UT, United States
| | - Kaden B. Underwood
- Department of Animal, Dairy and Veterinary Sciences, Center for Integrated BioSystems, College of Veterinary Medicine, Utah State University, Logan, UT, United States
| | - McKenna R. Walters
- Department of Animal, Dairy and Veterinary Sciences, Center for Integrated BioSystems, College of Veterinary Medicine, Utah State University, Logan, UT, United States
| | - Shawn M. Zimmerman
- Utah Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Utah State University, Logan, UT, United States
| | - Nathan K. Johnson
- Department of Animal, Dairy and Veterinary Sciences, Center for Integrated BioSystems, College of Veterinary Medicine, Utah State University, Logan, UT, United States
| | - Jeffrey B. Mason
- Department of Animal, Dairy and Veterinary Sciences, Center for Integrated BioSystems, College of Veterinary Medicine, Utah State University, Logan, UT, United States
| |
Collapse
|
20
|
Shi C, Murphy CT. piRNAs regulate a Hedgehog germline-to-soma pro-aging signal. NATURE AGING 2023; 3:47-63. [PMID: 37118518 PMCID: PMC10154208 DOI: 10.1038/s43587-022-00329-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/03/2022] [Indexed: 04/30/2023]
Abstract
The reproductive system regulates somatic aging through competing anti- and pro-aging signals. Germline removal extends somatic lifespan through conserved pathways including insulin and mammalian target-of-rapamycin signaling, while germline hyperactivity shortens lifespan through unknown mechanisms. Here we show that mating-induced germline hyperactivity downregulates piRNAs, in turn desilencing their targets, including the Hedgehog-like ligand-encoding genes wrt-1 and wrt-10, ultimately causing somatic collapse and death. Germline-produced Hedgehog signals require PTR-6 and PTR-16 receptors for mating-induced shrinking and death. Our results reveal an unconventional role of the piRNA pathway in transcriptional regulation of Hedgehog signaling and a new role of Hedgehog signaling in the regulation of longevity and somatic maintenance: Hedgehog signaling is controlled by the tunable piRNA pathway to encode the previously unknown germline-to-soma pro-aging signal. Mating-induced piRNA downregulation in the germline and subsequent Hedgehog signaling to the soma enable the animal to tune somatic resource allocation to germline needs, optimizing reproductive timing and survival.
Collapse
Affiliation(s)
- Cheng Shi
- Department of Molecular Biology and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
- Department of Biological Sciences, University of New Orleans, New Orleans, LA, USA.
| | - Coleen T Murphy
- Department of Molecular Biology and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
21
|
Duxbury EML, Carlsson H, Sales K, Sultanova Z, Immler S, Chapman T, Maklakov AA. Multigenerational downregulation of insulin/IGF-1 signaling in adulthood improves lineage survival, reproduction, and fitness in Caenorhabditis elegans supporting the developmental theory of ageing. Evolution 2022; 76:2829-2845. [PMID: 36199198 PMCID: PMC10092551 DOI: 10.1111/evo.14640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/18/2022] [Accepted: 09/08/2022] [Indexed: 01/22/2023]
Abstract
Adulthood-only downregulation of insulin/IGF-1 signaling (IIS), an evolutionarily conserved pathway regulating resource allocation between somatic maintenance and reproduction, increases life span without fecundity cost in the nematode, Caenorhabditis elegans. However, long-term multigenerational effects of reduced IIS remain unexplored and are proposed to carry costs for offspring quality. To test this hypothesis, we ran a mutation accumulation (MA) experiment and downregulated IIS in half of the 400 MA lines by silencing daf-2 gene expression using RNA interference (RNAi) across 40 generations. Contrary to the prediction, adulthood-only daf-2 RNAi reduced extinction of MA lines both under UV-induced and spontaneous MA. Fitness of the surviving UV-induced MA lines was higher under daf-2 RNAi. Reduced IIS increased intergenerational F1 offspring fitness under UV stress but had no quantifiable transgenerational effects. Functional hrde-1 was required for the benefits of multigenerational daf-2 RNAi. Overall, we found net benefit to fitness from multigenerational reduction of IIS and the benefits became more apparent under stress. Because reduced daf-2 expression during development carries fitness costs, we suggest that our findings are best explained by the developmental theory of ageing, which maintains that the decline in the force of selection with age results in poorly regulated gene expression in adulthood.
Collapse
Affiliation(s)
- Elizabeth M L Duxbury
- School of Biological Sciences, University of East Anglia, Norwich, NR4 7TJ, United Kingdom
| | - Hanne Carlsson
- School of Biological Sciences, University of East Anglia, Norwich, NR4 7TJ, United Kingdom
| | - Kris Sales
- School of Biological Sciences, University of East Anglia, Norwich, NR4 7TJ, United Kingdom
| | - Zahida Sultanova
- School of Biological Sciences, University of East Anglia, Norwich, NR4 7TJ, United Kingdom
| | - Simone Immler
- School of Biological Sciences, University of East Anglia, Norwich, NR4 7TJ, United Kingdom
| | - Tracey Chapman
- School of Biological Sciences, University of East Anglia, Norwich, NR4 7TJ, United Kingdom
| | - Alexei A Maklakov
- School of Biological Sciences, University of East Anglia, Norwich, NR4 7TJ, United Kingdom
| |
Collapse
|
22
|
De-Souza EA, Cummins N, Taylor RC. IRE-1 endoribonuclease activity declines early in C. elegans adulthood and is not rescued by reduced reproduction. FRONTIERS IN AGING 2022; 3:1044556. [PMID: 36389122 PMCID: PMC9649906 DOI: 10.3389/fragi.2022.1044556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
Abstract
The proteome of a cell helps to define its functional specialization. Most proteins must be translated and properly folded to ensure their biological function, but with aging, animals lose their ability to maintain a correctly folded proteome. This leads to the accumulation of protein aggregates, decreased stress resistance, and the onset of age-related disorders. The unfolded protein response of the endoplasmic reticulum (UPRER) is a central protein quality control mechanism, the function of which is known to decline with age. Here, we show that age-related UPRER decline in Caenorhabditis elegans occurs at the onset of the reproductive period and is caused by a failure in IRE-1 endoribonuclease activities, affecting both the splicing of xbp-1 mRNA and regulated Ire1 dependent decay (RIDD) activity. Animals with a defect in germline development, previously shown to rescue the transcriptional activity of other stress responses during aging, do not show restored UPRER activation with age. This underlines the mechanistic difference between age-associated loss of UPRER activation and that of other stress responses in this system, and uncouples reproductive status from the activity of somatic maintenance pathways. These observations may aid in the development of strategies that aim to overcome the proteostasis decline observed with aging.
Collapse
Affiliation(s)
| | | | - Rebecca C. Taylor
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| |
Collapse
|
23
|
Brooks D, Bawa S, Bontrager A, Stetsiv M, Guo Y, Geisbrecht ER. Independent pathways control muscle tissue size and sarcomere remodeling. Dev Biol 2022; 490:1-12. [PMID: 35760368 PMCID: PMC9648737 DOI: 10.1016/j.ydbio.2022.06.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 06/03/2022] [Accepted: 06/21/2022] [Indexed: 01/09/2023]
Abstract
Cell growth and proliferation must be balanced during development to attain a final adult size with the appropriate proportions of internal organs to maximize fitness and reproduction. While multiple signaling pathways coordinate Drosophila development, it is unclear how multi-organ communication within and between tissues converge to regulate systemic growth. One such growth pathway, mediated by insulin-like peptides that bind to and activate the insulin receptor in multiple target tissues, is a primary mediator of organismal size. Here we uncover a signaling role for the NUAK serine/threonine kinase in muscle tissue that impinges upon insulin pathway activity to limit overall body size, including a reduction in the growth of individual organs. In skeletal muscle tissue, manipulation of NUAK or insulin pathway components influences sarcomere number concomitant with modulation of thin and thick filament lengths, possibly by modulating the localization of Lasp, a nebulin repeat protein known to set thin filament length. This mode of sarcomere remodeling does not occur in other mutants that also exhibit smaller muscles, suggesting that a sensing mechanism exists in muscle tissue to regulate sarcomere growth that is independent of tissue size control.
Collapse
Affiliation(s)
- David Brooks
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA
| | - Simranjot Bawa
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA
| | - Alexandria Bontrager
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA
| | - Marta Stetsiv
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA
| | - Yungui Guo
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA
| | - Erika R Geisbrecht
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA.
| |
Collapse
|
24
|
Ferreira JV, da Rosa Soares A, Pereira P. Cell Non-autonomous Proteostasis Regulation in Aging and Disease. Front Neurosci 2022; 16:878296. [PMID: 35757551 PMCID: PMC9220288 DOI: 10.3389/fnins.2022.878296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Aging is a risk factor for a number of diseases, being the more notorious ones perhaps neurodegenerative diseases such as Alzheimer's and Parkinson's. These and other age-related pathologies are often associated with accumulation of proteotoxic material inside cells, as well as with the accumulation of protein deposits extracellularly. It is widely accepted that this accumulation of toxic proteins trails a progressive decline in the mechanisms that regulate protein homeostasis, or proteostasis, during aging. However, despite significant efforts, the progress in terms of novel or improved therapies targeting accumulation of proteotoxic material has been rather limited. For example, clinical trials for new drugs aimed at treating Alzheimer's disease, by preventing accumulation of toxic proteins, have notoriously failed. On the other hand, it is becoming increasingly apparent that regulation of proteostasis is not a cell autonomous process. In fact, cells rely on complex transcellular networks to maintain tissue and organ homeostasis involving endocrine and paracrine signaling pathways. In this review we will discuss the impact of cell non-autonomous proteostasis mechanisms and their impact in aging and disease. We will focus on how transcellular proteostasis networks can shed new light into stablished paradigms about the aging of organisms.
Collapse
Affiliation(s)
- Joao Vasco Ferreira
- Proteostasis and Intercellular Communication Lab, Chronic Diseases Research Centre (CEDOC), NOVA Medical School, Faculdade de Ciencias Medicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Ana da Rosa Soares
- Proteostasis and Intercellular Communication Lab, Chronic Diseases Research Centre (CEDOC), NOVA Medical School, Faculdade de Ciencias Medicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Paulo Pereira
- Proteostasis and Intercellular Communication Lab, Chronic Diseases Research Centre (CEDOC), NOVA Medical School, Faculdade de Ciencias Medicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
25
|
Shaposhnikov MV, Guvatova ZG, Zemskaya NV, Koval LA, Schegoleva EV, Gorbunova AA, Golubev DA, Pakshina NR, Ulyasheva NS, Solovev IA, Bobrovskikh MA, Gruntenko NE, Menshanov PN, Krasnov GS, Kudryavseva AV, Moskalev AA. Molecular mechanisms of exceptional lifespan increase of Drosophila melanogaster with different genotypes after combinations of pro-longevity interventions. Commun Biol 2022; 5:566. [PMID: 35681084 PMCID: PMC9184560 DOI: 10.1038/s42003-022-03524-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 05/24/2022] [Indexed: 12/13/2022] Open
Abstract
Aging is one of the global challenges of our time. The search for new anti-aging interventions is also an issue of great actuality. We report on the success of Drosophila melanogaster lifespan extension under the combined influence of dietary restriction, co-administration of berberine, fucoxanthin, and rapamycin, photodeprivation, and low-temperature conditions up to 185 days in w1118 strain and up to 213 days in long-lived E(z)/w mutants. The trade-off was found between longevity and locomotion. The transcriptome analysis showed an impact of epigenetic alterations, lipid metabolism, cellular respiration, nutrient sensing, immune response, and autophagy in the registered effect. The lifespan of fruit flies can be extended up to 213 days under specialized conditions.
Collapse
|
26
|
Dominguez LJ, Barbagallo M. Antiageing strategies. PATHY'S PRINCIPLES AND PRACTICE OF GERIATRIC MEDICINE 2022:1442-1458. [DOI: 10.1002/9781119484288.ch115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
27
|
Abstract
The proteostasis network (PN) regulates protein synthesis, folding, and degradation and is critical for the health and function of all cells. The PN has been extensively studied in the context of aging and age-related diseases, and loss of proteostasis is regarded as a major contributor to many age-associated disorders. In contrast to somatic tissues, an important feature of germ cells is their ability to maintain a healthy proteome across generations. Accumulating evidence has now revealed multiple layers of PN regulation that support germ cell function, determine reproductive capacity during aging, and prioritize reproduction at the expense of somatic health. Here, we review recent insights into these different modes of regulation and their implications for reproductive and somatic aging.
Collapse
|
28
|
Suriyalaksh M, Raimondi C, Mains A, Segonds-Pichon A, Mukhtar S, Murdoch S, Aldunate R, Krueger F, Guimerà R, Andrews S, Sales-Pardo M, Casanueva O. Gene regulatory network inference in long-lived C. elegans reveals modular properties that are predictive of novel aging genes. iScience 2022; 25:103663. [PMID: 35036864 PMCID: PMC8753122 DOI: 10.1016/j.isci.2021.103663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 09/09/2021] [Accepted: 12/15/2021] [Indexed: 11/24/2022] Open
Abstract
We design a “wisdom-of-the-crowds” GRN inference pipeline and couple it to complex network analysis to understand the organizational principles governing gene regulation in long-lived glp-1/Notch Caenorhabditis elegans. The GRN has three layers (input, core, and output) and is topologically equivalent to bow-tie/hourglass structures prevalent among metabolic networks. To assess the functional importance of structural layers, we screened 80% of regulators and discovered 50 new aging genes, 86% with human orthologues. Genes essential for longevity—including ones involved in insulin-like signaling (ILS)—are at the core, indicating that GRN's structure is predictive of functionality. We used in vivo reporters and a novel functional network covering 5,497 genetic interactions to make mechanistic predictions. We used genetic epistasis to test some of these predictions, uncovering a novel transcriptional regulator, sup-37, that works alongside DAF-16/FOXO. We present a framework with predictive power that can accelerate discovery in C. elegans and potentially humans. Gene-regulatory inference provides global network of long-lived animals The large-scale topology of the network has an hourglass structure Membership to the core of the hourglass is a good predictor of functionality Discovered 50 novel aging genes, including sup-37, a DAF-16 dependent gene
Collapse
Affiliation(s)
| | | | - Abraham Mains
- Babraham Institute, Babraham, Cambridge CB22 3AT, UK
| | | | | | | | - Rebeca Aldunate
- Escuela de Biotecnología, Facultad de Ciencias, Universidad Santo Tomas, Santiago, Chile
| | - Felix Krueger
- Babraham Institute, Babraham, Cambridge CB22 3AT, UK
| | - Roger Guimerà
- ICREA, Barcelona 08010, Catalonia, Spain.,Department of Chemical Engineering, Universitat Rovira i Virgili, Tarragona 43007, Catalonia, Spain
| | - Simon Andrews
- Babraham Institute, Babraham, Cambridge CB22 3AT, UK
| | - Marta Sales-Pardo
- Department of Chemical Engineering, Universitat Rovira i Virgili, Tarragona 43007, Catalonia, Spain
| | | |
Collapse
|
29
|
Promislow DEL, Flatt T, Bonduriansky R. The Biology of Aging in Insects: From Drosophila to Other Insects and Back. ANNUAL REVIEW OF ENTOMOLOGY 2022; 67:83-103. [PMID: 34590891 PMCID: PMC8940561 DOI: 10.1146/annurev-ento-061621-064341] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
An enormous amount of work has been done on aging in Drosophila melanogaster, a classical genetic and molecular model system, but also in numerous other insects. However, these two extensive bodies of work remain poorly integrated to date. Studies in Drosophila often explore genetic, developmental, physiological, and nutrition-related aspects of aging in the lab, while studies in other insects often explore ecological, social, and somatic aspects of aging in both lab and natural populations. Alongside exciting genomic and molecular research advances in aging in Drosophila, many new studies have also been published on aging in various other insects, including studies on aging in natural populations of diverse species. However, no broad synthesis of these largely separate bodies of work has been attempted. In this review, we endeavor to synthesize these two semi-independent literatures to facilitate collaboration and foster the exchange of ideas and research tools. While lab studies of Drosophila have illuminated many fundamental aspects of senescence, the stunning diversity of aging patterns among insects, especially in the context of their rich ecology, remains vastlyunderstudied. Coupled with field studies and novel, more easily applicable molecular methods, this represents a major opportunity for deepening our understanding of the biology of aging in insects and beyond.
Collapse
Affiliation(s)
- Daniel E L Promislow
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington 98195, USA;
- Department of Biology, University of Washington, Seattle, Washington 98195, USA
| | - Thomas Flatt
- Department of Biology, University of Fribourg, CH-1700 Fribourg, Switzerland;
| | - Russell Bonduriansky
- Evolution and Ecology Research Centre, School of Biological, Earth and Environmental Sciences, University of New South Wales Sydney, New South Wales 2052, Australia;
| |
Collapse
|
30
|
Mack HID, Kremer J, Albertini E, Mack EKM, Jansen-Dürr P. Regulation of fatty acid desaturase- and immunity gene-expression by mbk-1/DYRK1A in Caenorhabditis elegans. BMC Genomics 2022; 23:25. [PMID: 34983389 PMCID: PMC8729107 DOI: 10.1186/s12864-021-08176-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/08/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In the nematode Caenorhabditis elegans, longevity in response to germline ablation, but not in response to reduced insulin/IGF1-like signaling, is strongly dependent on the conserved protein kinase minibrain-related kinase 1 (MBK-1). In humans, the MBK-1 ortholog DYRK1A is associated with a variety of disorders, most prominently with neurological defects observed in Down syndrome. To better understand mbk-1's physiological roles and their dependence on genetic background, we analyzed the influence of mbk-1 loss on the transcriptomes of wildtype and long-lived, germline-deficient or insulin-receptor defective, C. elegans strains by RNA-sequencing. RESULTS mbk-1 loss elicited global changes in transcription that were less pronounced in insulin-receptor mutant than in germline-deficient or wildtype C. elegans. Irrespective of genetic background, mbk-1 regulated genes were enriched for functions in biological processes related to organic acid metabolism and pathogen defense. qPCR-studies confirmed mbk-1 dependent induction of all three C. elegans Δ9-fatty acid desaturases, fat-5, fat-6 and fat-7, in wildtype, germline-deficient and insulin-receptor mutant strains. Conversely, mbk-1 dependent expression patterns of selected pathogen resistance genes, including asp-12, dod-24 and drd-50, differed across the genetic backgrounds examined. Finally, cth-1 and cysl-2, two genes which connect pathogen resistance to the metabolism of the gaseous messenger and lifespan regulator hydrogen sulfide (H2S), were commonly suppressed by mbk-1 loss only in wildtype and germline-deficient, but not in insulin-receptor mutant C. elegans. CONCLUSION Our work reveals previously unknown roles of C. elegans mbk-1 in the regulation of fatty acid desaturase- and H2S metabolic-genes. These roles are only partially dependent on genetic background. Considering the particular importance of fatty acid desaturation and H2S for longevity of germline-deficient C. elegans, we propose that these processes at least in part account for the previous observation that mbk-1 preferentially regulates lifespan in these worms.
Collapse
Affiliation(s)
- Hildegard I D Mack
- Institute for Biomedical Aging Research, University of Innsbruck, Rennweg 10, 6020, Innsbruck, Austria.
| | - Jennifer Kremer
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg, and University Hospital Giessen and Marburg, Baldingerstrasse, 35032, Marburg, Germany
| | - Eva Albertini
- Institute for Biomedical Aging Research, University of Innsbruck, Rennweg 10, 6020, Innsbruck, Austria
| | - Elisabeth K M Mack
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg, and University Hospital Giessen and Marburg, Baldingerstrasse, 35032, Marburg, Germany
| | - Pidder Jansen-Dürr
- Institute for Biomedical Aging Research, University of Innsbruck, Rennweg 10, 6020, Innsbruck, Austria.
| |
Collapse
|
31
|
Li R, Tao M, Xu T, Pan S, Xu X, Wu T. Small berries as health-promoting ingredients: a review on anti-aging effects and mechanisms in Caenorhabditis elegans. Food Funct 2021; 13:478-500. [PMID: 34927654 DOI: 10.1039/d1fo02184b] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Aging is an inevitable, irreversible, and complex process of damage accumulation and functional decline, increasing the risk of various chronic diseases. However, for now no drug can delay aging process nor cure aging-related diseases. Nutritional intervention is considered as a key and effective strategy to promote healthy aging and improve life quality. Small berries, as one of the most common and popular fruits, have been demonstrated to improve cognitive function and possess neuroprotective activities. However, the anti-aging effects of small berries have not been systematically elucidated yet. This review mainly focuses on small berries' anti-aging activity studies involving small berry types, active components, the utilized model organism Caenorhabditis elegans (C. elegans), related signaling pathways, and molecular mechanisms. The purpose of this review is to propose effective strategies to evaluate the anti-aging effects of small berries and provide guidance for the development of anti-aging supplements from small berries.
Collapse
Affiliation(s)
- Rong Li
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China.
| | - Mingfang Tao
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China.
| | - Tingting Xu
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China.
| | - Siyi Pan
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China.
| | - Xiaoyun Xu
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China.
| | - Ting Wu
- Key Laboratory of Environment Correlative Dietology (Ministry of Education), College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China.
| |
Collapse
|
32
|
Rodrigues MA, Merckelbach A, Durmaz E, Kerdaffrec E, Flatt T. Transcriptomic evidence for a trade-off between germline proliferation and immunity in Drosophila. Evol Lett 2021; 5:644-656. [PMID: 34917403 PMCID: PMC8645197 DOI: 10.1002/evl3.261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/07/2021] [Accepted: 09/12/2021] [Indexed: 11/08/2022] Open
Abstract
Life-history theory posits that investment into reproduction might occur at the expense of investment into somatic maintenance, including immune function. If so, reduced or curtailed reproductive effort might be expected to increase immunity. In support of this notion, work in Caenorhabditis elegans has shown that worms lacking a germline exhibit improved immunity, but whether the antagonistic relation between germline proliferation and immunity also holds for other organisms is less well understood. Here, we report that transgenic ablation of germ cells in late development or early adulthood in Drosophila melanogaster causes elevated baseline expression and increased induction of Toll and Imd immune genes upon bacterial infection, as compared to fertile flies with an intact germline. We also identify immune genes whose expression after infection differs between fertile and germline-less flies in a manner that is conditional on their mating status. We conclude that germline activity strongly impedes the expression and inducibility of immune genes and that this physiological trade-off might be evolutionarily conserved.
Collapse
Affiliation(s)
| | | | - Esra Durmaz
- Department of BiologyUniversity of FribourgCH‐1700 FribourgSwitzerland
| | - Envel Kerdaffrec
- Department of BiologyUniversity of FribourgCH‐1700 FribourgSwitzerland
| | - Thomas Flatt
- Department of BiologyUniversity of FribourgCH‐1700 FribourgSwitzerland
| |
Collapse
|
33
|
Mason JB, Habermehl TL, Underwood KB, Schneider A, Brieño-Enriquez MA, Masternak MM, Parkinson KC. The interrelationship between female reproductive aging and survival. J Gerontol A Biol Sci Med Sci 2021; 77:75-83. [PMID: 34528058 DOI: 10.1093/gerona/glab252] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Indexed: 01/08/2023] Open
Abstract
The link between survival and reproductive function is demonstrated across many species and is under both long-term evolutionary pressures and short-term environmental pressures. Loss of reproductive function is common in mammals and is strongly correlated with increased rates of disease in both males and females. However, the reproduction-associated change in disease rates is more abrupt and more severe in women, who benefit from a significant health advantage over men until the age of menopause. Young women with early ovarian failure also suffer from increased disease risks, further supporting the role of ovarian function in female health. Contemporary experiments where the influence of young ovarian tissue has been restored in post-reproductive-aged females with surgical manipulation were found to increase survival significantly. In these experiments, young, intact ovaries were used to replace the aged ovaries of females that had already reached reproductive cessation. As has been seen previously in primitive species, when the young mammalian ovaries were depleted of germ cells prior to transplantation to the post-reproductive female, survival was increased even further than with germ cell-containing young ovaries. Thus, extending reproductive potential significantly increases survival and appears to be germ cell and ovarian hormone-independent. The current review will discuss historical and contemporary observations and theories that support the link between reproduction and survival and provide hope for future clinical applications to decrease menopause-associated increases in disease risks.
Collapse
Affiliation(s)
- Jeffrey B Mason
- Department of Animal, Dairy and Veterinary Sciences, Center for Integrated BioSystems, School of Veterinary Medicine, Utah State University, 4700 Old Main Hill, Logan, UT 84322, USA
| | - Tracy L Habermehl
- Department of Animal, Dairy and Veterinary Sciences, Center for Integrated BioSystems, School of Veterinary Medicine, Utah State University, 4700 Old Main Hill, Logan, UT 84322, USA
| | - Kaden B Underwood
- Department of Animal, Dairy and Veterinary Sciences, Center for Integrated BioSystems, School of Veterinary Medicine, Utah State University, 4700 Old Main Hill, Logan, UT 84322, USA
| | - Augusto Schneider
- Departmento de Nutrição, Universidade Federal de Pelotas, RS, Pelotas, Brazil
| | - Miguel A Brieño-Enriquez
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michal M Masternak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA.,Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland
| | - Kate C Parkinson
- Department of Animal, Dairy and Veterinary Sciences, Center for Integrated BioSystems, School of Veterinary Medicine, Utah State University, 4700 Old Main Hill, Logan, UT 84322, USA
| |
Collapse
|
34
|
Yamamoto R, Palmer M, Koski H, Curtis-Joseph N, Tatar M. Aging modulated by the Drosophila insulin receptor through distinct structure-defined mechanisms. Genetics 2021; 217:6064149. [PMID: 33724413 PMCID: PMC8045697 DOI: 10.1093/genetics/iyaa037] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022] Open
Abstract
Mutations of the Drosophila melanogaster insulin/IGF signaling system slow aging, while also affecting growth and reproduction. To understand this pleiotropy, we produced an allelic series of single codon substitutions in the Drosophila insulin receptor, InR. We generated InR substitutions using homologous recombination and related each to emerging models of receptor tyrosine kinase structure and function. Three mutations when combined as trans-heterozygotes extended lifespan while retarding growth and fecundity. These genotypes reduced insulin-stimulated Akt phosphorylation, suggesting they impede kinase catalytic domain function. Among these genotypes, longevity was negatively correlated with egg production, consistent with life-history trade-off theory. In contrast, one mutation (InR353) was located in the kinase insert domain, a poorly characterized element found in all receptor tyrosine kinases. Remarkably, wild-type heterozygotes with InR353 robustly extended lifespan without affecting growth or reproduction and retained capacity to fully phosphorylate Akt. The Drosophila insulin receptor kinase insert domain contains a previously unrecognized SH2 binding motif. We propose the kinase insert domain interacts with SH2-associated adapter proteins to affect aging through mechanisms that retain insulin sensitivity and are independent of reproduction.
Collapse
Affiliation(s)
- Rochele Yamamoto
- Department of Ecology and Evolutionary Biology, Brown University, Providence, RI, USA
| | - Michael Palmer
- Department of Ecology and Evolutionary Biology, Brown University, Providence, RI, USA
| | - Helen Koski
- Department of Ecology and Evolutionary Biology, Brown University, Providence, RI, USA
| | - Noelle Curtis-Joseph
- Department of Ecology and Evolutionary Biology, Brown University, Providence, RI, USA
| | - Marc Tatar
- Department of Ecology and Evolutionary Biology, Brown University, Providence, RI, USA
| |
Collapse
|
35
|
Howard AC, Mir D, Snow S, Horrocks J, Sayed H, Ma Z, Rogers AN. Anabolic Function Downstream of TOR Controls Trade-offs Between Longevity and Reproduction at the Level of Specific Tissues in C. elegans. FRONTIERS IN AGING 2021; 2:725068. [PMID: 35340273 PMCID: PMC8953723 DOI: 10.3389/fragi.2021.725068] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/27/2021] [Indexed: 11/20/2022]
Abstract
As the most energetically expensive cellular process, translation must be finely tuned to environmental conditions. Dietary restriction attenuates signaling through the nutrient sensing mTOR pathway, which reduces translation and redirects resources to preserve the soma. These responses are associated with increased lifespan but also anabolic impairment, phenotypes also observed when translation is genetically suppressed. Here, we restricted translation downstream of mTOR separately in major tissues in C. elegans to better understand their roles in systemic adaptation and whether consequences to anabolic impairment were separable from positive effects on lifespan. Lowering translation in neurons, hypodermis, or germline tissue led to increased lifespan under well-fed conditions and improved survival upon withdrawal of food, indicating that these are key tissues coordinating enhanced survival when protein synthesis is reduced. Surprisingly, lowering translation in body muscle during development shortened lifespan while accelerating and increasing reproduction, a reversal of phenotypic trade-offs associated with systemic translation suppression. Suppressing mTORC1 selectively in body muscle also increased reproduction while slowing motility during development. In nature, this may be indicative of reduced energy expenditure related to foraging, acting as a "GO!" signal for reproduction. Together, results indicate that low translation in different tissues helps direct distinct systemic adaptations and suggest that unknown endocrine signals mediate these responses. Furthermore, mTOR or translation inhibitory therapeutics that target specific tissues may achieve desired interventions to aging without loss of whole-body anabolism.
Collapse
Affiliation(s)
- Amber C. Howard
- Mount Desert Island Biological Laboratory, Davis Center for Regenerative Biology and Medicine, Bar Harbor, ME, United States
- Department of Natural Sciences, Middle Georgia State University, Cochran, GA, United States
| | - Dilawar Mir
- Mount Desert Island Biological Laboratory, Davis Center for Regenerative Biology and Medicine, Bar Harbor, ME, United States
| | - Santina Snow
- Mount Desert Island Biological Laboratory, Davis Center for Regenerative Biology and Medicine, Bar Harbor, ME, United States
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, United States
| | - Jordan Horrocks
- Mount Desert Island Biological Laboratory, Davis Center for Regenerative Biology and Medicine, Bar Harbor, ME, United States
| | - Hussein Sayed
- Mount Desert Island Biological Laboratory, Davis Center for Regenerative Biology and Medicine, Bar Harbor, ME, United States
| | - Zhengxin Ma
- Mount Desert Island Biological Laboratory, Davis Center for Regenerative Biology and Medicine, Bar Harbor, ME, United States
| | - Aric N. Rogers
- Mount Desert Island Biological Laboratory, Davis Center for Regenerative Biology and Medicine, Bar Harbor, ME, United States
| |
Collapse
|
36
|
Gems D, Kern CC, Nour J, Ezcurra M. Reproductive Suicide: Similar Mechanisms of Aging in C. elegans and Pacific Salmon. Front Cell Dev Biol 2021; 9:688788. [PMID: 34513830 PMCID: PMC8430333 DOI: 10.3389/fcell.2021.688788] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/21/2021] [Indexed: 12/17/2022] Open
Abstract
In some species of salmon, reproductive maturity triggers the development of massive pathology resulting from reproductive effort, leading to rapid post-reproductive death. Such reproductive death, which occurs in many semelparous organisms (with a single bout of reproduction), can be prevented by blocking reproductive maturation, and this can increase lifespan dramatically. Reproductive death is often viewed as distinct from senescence in iteroparous organisms (with multiple bouts of reproduction) such as humans. Here we review the evidence that reproductive death occurs in C. elegans and discuss what this means for its use as a model organism to study aging. Inhibiting insulin/IGF-1 signaling and germline removal suppresses reproductive death and greatly extends lifespan in C. elegans, but can also extend lifespan to a small extent in iteroparous organisms. We argue that mechanisms of senescence operative in reproductive death exist in a less catastrophic form in iteroparous organisms, particularly those that involve costly resource reallocation, and exhibit endocrine-regulated plasticity. Thus, mechanisms of senescence in semelparous organisms (including plants) and iteroparous ones form an etiological continuum. Therefore understanding mechanisms of reproductive death in C. elegans can teach us about some mechanisms of senescence that are operative in iteroparous organisms.
Collapse
Affiliation(s)
- David Gems
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| | - Carina C. Kern
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| | - Joseph Nour
- Institute of Healthy Ageing, Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| | - Marina Ezcurra
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| |
Collapse
|
37
|
Saturated very long chain fatty acid configures glycosphingolipid for lysosome homeostasis in long-lived C. elegans. Nat Commun 2021; 12:5073. [PMID: 34417467 PMCID: PMC8379269 DOI: 10.1038/s41467-021-25398-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 07/28/2021] [Indexed: 01/21/2023] Open
Abstract
The contents of numerous membrane lipids change upon ageing. However, it is unknown whether and how any of these changes are causally linked to lifespan regulation. Acyl chains contribute to the functional specificity of membrane lipids. In this study, working with C. elegans, we identified an acyl chain-specific sphingolipid, C22 glucosylceramide, as a longevity metabolite. Germline deficiency, a conserved lifespan-extending paradigm, induces somatic expression of the fatty acid elongase ELO-3, and behenic acid (22:0) generated by ELO-3 is incorporated into glucosylceramide for lifespan regulation. Mechanistically, C22 glucosylceramide is required for the membrane localization of clathrin, a protein that regulates membrane budding. The reduction in C22 glucosylceramide impairs the clathrin-dependent autophagic lysosome reformation, which subsequently leads to TOR activation and longevity suppression. These findings reveal a mechanistic link between membrane lipids and ageing and suggest a model of lifespan regulation by fatty acid-mediated membrane configuration. The membrane lipids change with ageing and function as regulatory molecules, but the underlying mechanisms are incompletely understood. Here, the authors identify C22 glucosylceramide as a regulator of the longevity transcription factor SKN-1, and show that C22 glucosylceramide regulates lifespan by controlling lysosome homeostasis and subsequent TOR activation.
Collapse
|
38
|
Malod K, Roets PD, Bosua H, Archer CR, Weldon CW. Selecting on age of female reproduction affects lifespan in both sexes and age-dependent reproductive effort in female (but not male) Ceratitis cosyra. Behav Ecol Sociobiol 2021. [DOI: 10.1007/s00265-021-03063-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
39
|
Naim N, Amrit FRG, Ratnappan R, DelBuono N, Loose JA, Ghazi A. Cell nonautonomous roles of NHR-49 in promoting longevity and innate immunity. Aging Cell 2021; 20:e13413. [PMID: 34156142 PMCID: PMC8282243 DOI: 10.1111/acel.13413] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 05/06/2021] [Accepted: 05/14/2021] [Indexed: 02/06/2023] Open
Abstract
Aging and immunity are inextricably linked and many genes that extend life span also enhance immunoresistance. However, it remains unclear whether longevity-enhancing factors modulate immunity and longevity by discrete or shared mechanisms. Here, we demonstrate that the Caenorhabditis elegans pro-longevity factor, NHR-49, also promotes resistance against Pseudomonas aeruginosa but modulates immunity and longevity distinctly. NHR-49 expression increases upon germline ablation, an intervention that extends life span, but was lowered by Pseudomonas infection. The immunosusceptibility induced by nhr-49 loss of function was rescued by neuronal NHR-49 alone, whereas the longevity diminution was rescued by expression in multiple somatic tissues. The well-established NHR-49 target genes, acs-2 and fmo-2, were also differentially regulated following germline elimination or Pseudomonas exposure. Interestingly, neither gene conferred immunity toward Gram-negative Pseudomonas, unlike their known functions against gram-positive pathogens. Instead, genes encoding antimicrobial factors and xenobiotic-response proteins upregulated by NHR-49 contributed to resistance against Pseudomonas. Thus, NHR-49 is differentially regulated by interventions that bring about long-term changes (life span extension) versus short-term stress (pathogen exposure) and in response it orchestrates discrete outputs, including pathogen-specific transcriptional programs.
Collapse
Affiliation(s)
- Nikki Naim
- Department of Pediatrics University of Pittsburgh School of Medicine Pittsburgh PA USA
| | - Francis R. G. Amrit
- Department of Pediatrics University of Pittsburgh School of Medicine Pittsburgh PA USA
| | - Ramesh Ratnappan
- Department of Pediatrics University of Pittsburgh School of Medicine Pittsburgh PA USA
| | - Nicholas DelBuono
- Department of Pediatrics University of Pittsburgh School of Medicine Pittsburgh PA USA
| | - Julia A. Loose
- Department of Pediatrics University of Pittsburgh School of Medicine Pittsburgh PA USA
| | - Arjumand Ghazi
- Department of Pediatrics University of Pittsburgh School of Medicine Pittsburgh PA USA
- Departments of Developmental Biology and Cell Biology and Physiology University of Pittsburgh School of Medicine Pittsburgh PA USA
| |
Collapse
|
40
|
Bergero R, Ellis P, Haerty W, Larcombe L, Macaulay I, Mehta T, Mogensen M, Murray D, Nash W, Neale MJ, O'Connor R, Ottolini C, Peel N, Ramsey L, Skinner B, Suh A, Summers M, Sun Y, Tidy A, Rahbari R, Rathje C, Immler S. Meiosis and beyond - understanding the mechanistic and evolutionary processes shaping the germline genome. Biol Rev Camb Philos Soc 2021; 96:822-841. [PMID: 33615674 PMCID: PMC8246768 DOI: 10.1111/brv.12680] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022]
Abstract
The separation of germ cell populations from the soma is part of the evolutionary transition to multicellularity. Only genetic information present in the germ cells will be inherited by future generations, and any molecular processes affecting the germline genome are therefore likely to be passed on. Despite its prevalence across taxonomic kingdoms, we are only starting to understand details of the underlying micro-evolutionary processes occurring at the germline genome level. These include segregation, recombination, mutation and selection and can occur at any stage during germline differentiation and mitotic germline proliferation to meiosis and post-meiotic gamete maturation. Selection acting on germ cells at any stage from the diploid germ cell to the haploid gametes may cause significant deviations from Mendelian inheritance and may be more widespread than previously assumed. The mechanisms that affect and potentially alter the genomic sequence and allele frequencies in the germline are pivotal to our understanding of heritability. With the rise of new sequencing technologies, we are now able to address some of these unanswered questions. In this review, we comment on the most recent developments in this field and identify current gaps in our knowledge.
Collapse
Affiliation(s)
- Roberta Bergero
- Institute of Evolutionary BiologyUniversity of EdinburghEdinburghEH9 3JTU.K.
| | - Peter Ellis
- School of BiosciencesUniversity of KentCanterburyCT2 7NJU.K.
| | | | - Lee Larcombe
- Applied Exomics LtdStevenage Bioscience CatalystStevenageSG1 2FXU.K.
| | - Iain Macaulay
- Earlham InstituteNorwich Research ParkNorwichNR4 7UZU.K.
| | - Tarang Mehta
- Earlham InstituteNorwich Research ParkNorwichNR4 7UZU.K.
| | - Mette Mogensen
- School of Biological SciencesUniversity of East AngliaNorwich Research ParkNorwichNR4 7TJU.K.
| | - David Murray
- School of Biological SciencesUniversity of East AngliaNorwich Research ParkNorwichNR4 7TJU.K.
| | - Will Nash
- Earlham InstituteNorwich Research ParkNorwichNR4 7UZU.K.
| | - Matthew J. Neale
- Genome Damage and Stability Centre, School of Life SciencesUniversity of SussexBrightonBN1 9RHU.K.
| | | | | | - Ned Peel
- Earlham InstituteNorwich Research ParkNorwichNR4 7UZU.K.
| | - Luke Ramsey
- The James Hutton InstituteInvergowrieDundeeDD2 5DAU.K.
| | - Ben Skinner
- School of Life SciencesUniversity of EssexColchesterCO4 3SQU.K.
| | - Alexander Suh
- School of Biological SciencesUniversity of East AngliaNorwich Research ParkNorwichNR4 7TJU.K.
- Department of Organismal BiologyUppsala UniversityNorbyvägen 18DUppsala752 36Sweden
| | - Michael Summers
- School of BiosciencesUniversity of KentCanterburyCT2 7NJU.K.
- The Bridge Centre1 St Thomas Street, London BridgeLondonSE1 9RYU.K.
| | - Yu Sun
- Norwich Medical SchoolUniversity of East AngliaNorwich Research Park, Colney LnNorwichNR4 7UGU.K.
| | - Alison Tidy
- School of BiosciencesUniversity of Nottingham, Plant Science, Sutton Bonington CampusSutton BoningtonLE12 5RDU.K.
| | | | - Claudia Rathje
- School of BiosciencesUniversity of KentCanterburyCT2 7NJU.K.
| | - Simone Immler
- School of Biological SciencesUniversity of East AngliaNorwich Research ParkNorwichNR4 7TJU.K.
| |
Collapse
|
41
|
Gaddy MA, Kuang S, Alfhili MA, Lee MH. The soma-germline communication: implications for somatic and reproductive aging. BMB Rep 2021. [PMID: 33407997 PMCID: PMC8167245 DOI: 10.5483/bmbrep.2021.54.5.198] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Aging is characterized by a functional decline in most physiological processes, including alterations in cellular metabolism and defense mechanisms. Increasing evidence suggests that caloric restriction extends longevity and retards age-related diseases at least in part by reducing metabolic rate and oxidative stress in a variety of species, including yeast, worms, flies, and mice. Moreover, recent studies in invertebrates – worms and flies, highlight the intricate interrelation between reproductive longevity and somatic aging (known as disposable soma theory of aging), which appears to be conserved in vertebrates. This review is specifically focused on how the reproductive system modulates somatic aging and vice versa in genetic model systems. Since many signaling pathways governing the aging process are evolutionarily conserved, similar mechanisms may be involved in controlling soma and reproductive aging in vertebrates.
Collapse
Affiliation(s)
- Matthew A. Gaddy
- Department of Internal Medicine, Division of Hematology/Oncology, Brody School of Medicine at East Carolina University, Greenville, NC 27834, United States
| | - Swana Kuang
- Department of Internal Medicine, Division of Hematology/Oncology, Brody School of Medicine at East Carolina University, Greenville, NC 27834, United States
| | - Mohammad A. Alfhili
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| | - Myon Hee Lee
- Department of Internal Medicine, Division of Hematology/Oncology, Brody School of Medicine at East Carolina University, Greenville, NC 27834, United States
| |
Collapse
|
42
|
Séguret A, Stolle E, Fleites-Ayil FA, Quezada-Euán JJG, Hartfelder K, Meusemann K, Harrison MC, Soro A, Paxton RJ. Transcriptomic Signatures of Ageing Vary in Solitary and Social Forms of an Orchid Bee. Genome Biol Evol 2021; 13:6259147. [PMID: 33914875 PMCID: PMC8214409 DOI: 10.1093/gbe/evab075] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2021] [Indexed: 12/14/2022] Open
Abstract
Eusocial insect queens are remarkable in their ability to maximize both fecundity and longevity, thus escaping the typical trade-off between these two traits. Several mechanisms have been proposed to underlie the remolding of the trade-off, such as reshaping of the juvenile hormone (JH) pathway, or caste-specific susceptibility to oxidative stress. However, it remains a challenge to disentangle the molecular mechanisms underlying the remolding of the trade-off in eusocial insects from caste-specific physiological attributes that have subsequently arisen. The socially polymorphic orchid bee Euglossa viridissima represents an excellent model to address the role of sociality per se in longevity as it allows direct comparisons of solitary and social individuals within a common genetic background. We investigated gene expression and JH levels in young and old bees from both solitary and social nests. We found 902 genes to be differentially expressed with age in solitary females, including genes involved in oxidative stress, versus only 100 genes in social dominant females, and 13 genes in subordinate females. A weighted gene coexpression network analysis further highlights pathways related to ageing in this species, including the target of rapamycin pathway. Eleven genes involved in translation, apoptosis, and DNA repair show concurrent age-related expression changes in solitary but not in social females, representing potential differences based on social status. JH titers did not vary with age or social status. Our results represent an important step in understanding the proximate mechanisms underlying the remodeling of the fecundity/longevity trade-off that accompanies the evolutionary transition from solitary life to eusociality.
Collapse
Affiliation(s)
- Alice Séguret
- Institute of Biology, Martin-Luther-University Halle-Wittenberg, Halle, Germany.,Institute for Evolution and Biodiversity, Westfälische-Wilhelms University, Münster, Germany
| | - Eckart Stolle
- Institute of Biology, Martin-Luther-University Halle-Wittenberg, Halle, Germany.,Leibniz Institute of Animal Biodiversity, Zoological Research Museum Alexander Koenig, Center of Molecular Biodiversity Research, Bonn, Germany
| | | | - José Javier G Quezada-Euán
- Department of Apiculture, Campus of Biological Sciences and Animal Husbandry, Autonomous University of Yucatán, Mérida, Mexico
| | - Klaus Hartfelder
- Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil
| | - Karen Meusemann
- Evolutionary Biology and Ecology, Albert-Ludwigs-University Freiburg, Freiburg (i. Brsg.), Germany
| | - Mark C Harrison
- Institute for Evolution and Biodiversity, Westfälische-Wilhelms University, Münster, Germany
| | - Antonella Soro
- Institute of Biology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Robert J Paxton
- Institute of Biology, Martin-Luther-University Halle-Wittenberg, Halle, Germany.,German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Leipzig, Germany
| |
Collapse
|
43
|
Mutlu AS, Duffy J, Wang MC. Lipid metabolism and lipid signals in aging and longevity. Dev Cell 2021; 56:1394-1407. [PMID: 33891896 DOI: 10.1016/j.devcel.2021.03.034] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/05/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023]
Abstract
Lipids play crucial roles in regulating aging and longevity. In the past few decades, a series of genetic pathways have been discovered to regulate lifespan in model organisms. Interestingly, many of these regulatory pathways are linked to lipid metabolism and lipid signaling. Lipid metabolic enzymes undergo significant changes during aging and are regulated by different longevity pathways. Lipids also actively modulate lifespan and health span as signaling molecules. In this review, we summarize recent insights into the roles of lipid metabolism and lipid signaling in aging and discuss lipid-related interventions in promoting longevity.
Collapse
Affiliation(s)
- Ayse Sena Mutlu
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jonathon Duffy
- Developmental Biology Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Meng C Wang
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Developmental Biology Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
44
|
Ngo KS, R-Almási B, Barta Z, Tökölyi J. Experimental manipulation of body size alters life history in hydra. Ecol Lett 2021; 24:728-738. [PMID: 33606896 DOI: 10.1111/ele.13698] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 12/18/2020] [Accepted: 01/07/2021] [Indexed: 11/26/2022]
Abstract
Body size has fundamental impacts on animal ecology and physiology but has been strongly influenced by recent climate change and human activities, such as size-selective harvesting. Understanding the ecological and life history consequences of body size has proved difficult due to the inseparability of direct effects of body size from processes connected to it (such as growth rate and individual condition). Here, we used the cnidarian Hydra oligactis to directly manipulate body size and understand its causal effects on reproduction and senescence. We found that experimentally reducing size delayed sexual development and lowered fecundity, while post-reproductive survival increased, implying that smaller individuals can physiologically detect their reduced size and adjust life history decisions to achieve higher survival. Our experiment suggests that ecological or human-induced changes in body size will have immediate effects on life history and population dynamics through a growth-independent link between body size, reproduction and senescence.
Collapse
Affiliation(s)
- Kha Sach Ngo
- MTA-DE Behavioral Ecology Research Group, Department of Evolutionary Zoology, Univ. of Debrecen, Debrecen, Hungary
| | - Berta R-Almási
- MTA-DE Behavioral Ecology Research Group, Department of Evolutionary Zoology, Univ. of Debrecen, Debrecen, Hungary
| | - Zoltán Barta
- MTA-DE Behavioral Ecology Research Group, Department of Evolutionary Zoology, Univ. of Debrecen, Debrecen, Hungary
| | - Jácint Tökölyi
- MTA-DE Behavioral Ecology Research Group, Department of Evolutionary Zoology, Univ. of Debrecen, Debrecen, Hungary
| |
Collapse
|
45
|
Tatar M. Aging Regulated Through a Stability Model of Insulin/Insulin Growth Factor Receptor Function. Front Endocrinol (Lausanne) 2021; 12:649880. [PMID: 33776941 PMCID: PMC7991905 DOI: 10.3389/fendo.2021.649880] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/08/2021] [Indexed: 01/04/2023] Open
Abstract
Mutations of the insulin-like receptor in Drosophila extend lifespan. New research suggests this receptor operates in two modes. The first extends lifespan while slowing reproduction and reducing growth. The second strongly extends lifespan without impairing growth or reproduction; it confers longevity assurance. The mutation that confers longevity assurance resides in the kinase insert domain, which contains a potential SH2 binding site for substrate proteins. We apply a recent model for the function of receptor tyrosine kinases to propose how insulin receptor structure can modulate aging. This concept hypothesizes that strong insulin-like ligands promote phosphorylation of high threshold substrate binding sites to robustly induce reproduction, which impairs survival as a consequence of trade-offs. Lower levels of receptor stimulation provide less kinase dimer stability, which reduces reproduction and extends lifespan by avoiding reproductive costs. Environmental conditions that favor diapause alter the expression of insulin ligands to further repress the stability of the interacting kinase domains, block phosphorylation of low threshold substrates and thus induce a unique molecular program that confers longevity assurance. Mutations of the insulin receptor that block low-phosphorylation site interactions, such as within the kinase insert domain, can extend lifespan while maintaining overall dimer stability. These flies are long-lived while maintaining reproduction and growth. The kinase insert domain of Drosophila provides a novel avenue from which to seek signaling of the insulin/insulin-like growth factor system of humans that modulate aging without impacting reproduction and growth, or incurring insulin resistance pathology.
Collapse
|
46
|
Bora S, Vardhan GSH, Deka N, Khataniar L, Gogoi D, Baruah A. Paraquat exposure over generation affects lifespan and reproduction through mitochondrial disruption in C. elegans. Toxicology 2020; 447:152632. [PMID: 33197508 DOI: 10.1016/j.tox.2020.152632] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/22/2022]
Abstract
Paraquat (methyl viologen), is a non-selective contact herbicide and well known mitochondrial toxicant. Mitochondria are the center of cellular metabolism, and involved in the development, lifespan, and reproduction of an organism. Mitochondria are dynamic organelles that are inherited maternally through the germline and carry multiple copies of their own genome (mtDNA). It is important to understand the effects of acute and chronic stress caused by mitochondrial toxicants over multiple generations at the cellular and organism levels. Using the model nematode C. elegans, we show that acute and chronic exposure to paraquat affects reproduction, longevity, gene expression, and mitochondrial physiology. Acute exposure to paraquat in N2 (wild type) causes induction of mitochondrial unfolded protein response (mtUPR), increased expression of mitochondrial superoxide dismutase, decreased mitochondrial membrane potential (Δψm), a dose-dependent progression from linear to fragmented mitochondria, and dose-dependent changes in longevity. Chronic exposure to a low dose of paraquat (0.035 mM) over multiple generations in N2 causes a progressive decline of fertility, leading to complete loss of fertile embryo production by the third generation. The mutation in CEP-1 [cep-1(gk138)], a key regulator of stress-induced apoptosis in the germline, causes increased sensitivity to chronic paraquat relative to N2 with no fertile embryo production beyond the second generation. Whereas, mitochondrial electron transport chain (complex III) mutant [isp-1(qm150)], which display constitutive activation of mtUPR showed increased tolerance and produced fertile embryo out to the fourth generation. The N2, cep-1(gk138), and isp-1(qm150) strain's lifespan over multiple generations exposed to chronic paraquat were measured. Fertility and lifespan data together indicate a trade-off between reproduction and somatic maintenance during chronic paraquat exposure. We have proposed that mitochondrial signaling, dynamics, and CEP-1 mediated germline apoptosis is involved in this trade-off.
Collapse
Affiliation(s)
- Snigdha Bora
- Department of Agricultural Biotechnology, Assam Agricultural University, Jorhat-13, India
| | | | - Nikhita Deka
- DBT-NECAB, Assam Agricultural University, Jorhat-13, India
| | - Lipika Khataniar
- Department of Agricultural Biotechnology, Assam Agricultural University, Jorhat-13, India
| | - Debajani Gogoi
- DBT-NECAB, Assam Agricultural University, Jorhat-13, India
| | - Aiswarya Baruah
- Department of Agricultural Biotechnology, Assam Agricultural University, Jorhat-13, India; DBT-NECAB, Assam Agricultural University, Jorhat-13, India.
| |
Collapse
|
47
|
Abstract
Sexual interactions negatively impact health and longevity in many species across the animal kingdom. C. elegans has been established as a good model to study how mating and intense sexual interactions influence longevity of the individuals. In this chapter, we review the most recent discoveries in this field. We first describe the phenotypes caused by intense mating, including shrinking, fat loss, and glycogen loss. We then describe three major mechanisms underlying mating-induced killing: germline activation, seminal fluid transfer, and male pheromone-mediated toxicity. Next, we summarize the current knowledge of genetic pathways involved in regulating mating-induced death, including DAF-9/DAF-12 steroid signaling, Insulin/IGF-1 signaling (IIS), and TOR signaling. Finally, we discuss the possible fitness benefits of mating-induced death. Throughout this review, we compare and contrast mating-induced death between the sexes and among different species in an effort to discuss this phenomenon and underlying mechanisms from the evolutionary perspective. Further investigation using mated C. elegans will improve our understanding of sexual antagonism, as well as the coordination between reproduction and somatic longevity in response to various external signals. Due to the evolutionary conservation in many aspects of mating-induced death, what we learn from a short-lived mated worm could provide new strategies to improve our own fitness and longevity.
Collapse
Affiliation(s)
- Cheng Shi
- Lewis-Sigler Institute for Integrative Genomics and Department of Molecular Biology, Princeton University, Princeton, NJ, United States
| | - Coleen T Murphy
- Lewis-Sigler Institute for Integrative Genomics and Department of Molecular Biology, Princeton University, Princeton, NJ, United States.
| |
Collapse
|
48
|
Li HH, Cai Y, Li JC, Su MP, Liu WL, Cheng L, Chou SJ, Yu GY, Wang HD, Chen CH. C-Type Lectins Link Immunological and Reproductive Processes in Aedes aegypti. iScience 2020; 23:101486. [PMID: 32891883 PMCID: PMC7481239 DOI: 10.1016/j.isci.2020.101486] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/14/2020] [Accepted: 08/18/2020] [Indexed: 12/24/2022] Open
Abstract
Physiological trade-offs between mosquito immune response and reproductive capability can arise due to insufficient resource availability. C-type lectin family members may be involved in these processes. We established a GCTL-3-/- mutant Aedes aegypti using CRISPR/Cas9 to investigate the role of GCTL-3 in balancing the costs associated with immune responses to arboviral infection and reproduction. GCTL-3-/- mutants showed significantly reduced DENV-2 infection rate and gut commensal microbiota populations, as well as upregulated JAK/STAT, IMD, Toll, and AMPs immunological pathways. Mutants also had significantly shorter lifespans than controls and laid fewer eggs due to defective germ line development. dsRNA knock-down of Attacin and Gambicin, two targets of the AMPs pathway, partially rescued this reduction in reproductive capabilities. Upregulation of immune response following GCTL-3 knock-out therefore comes at a cost to reproductive fitness. Knock-out of other lectins may further improve our knowledge of the molecular and genetic mechanisms underlying reproduction-immunity trade-offs in mosquitoes.
Collapse
Affiliation(s)
- Hsing-Han Li
- Institution of Biotechnology, National Tsing Hua University, Hsinchu, 300044, Taiwan; National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350401, Taiwan
| | - Yu Cai
- Temasek Life Sciences Laboratory, National University of Singapore, 117604, Singapore; Department of Biological Sciences, National University of Singapore, 117558, Singapore
| | - Jian-Chiuan Li
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350401, Taiwan
| | - Matthew P Su
- Department of Biological Science, Nagoya University, Nagoya 464-8602, Japan
| | - Wei-Liang Liu
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli 350401, Taiwan
| | - Lie Cheng
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350401, Taiwan
| | - Shu-Jen Chou
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei 115201, Taiwan
| | - Guann-Yi Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350401, Taiwan
| | - Horng-Dar Wang
- Institution of Biotechnology, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Chun-Hong Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350401, Taiwan; National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli 350401, Taiwan.
| |
Collapse
|
49
|
Lien W, Chen Y, Li Y, Wu J, Huang K, Lin J, Lin S, Hou C, Wang H, Wu C, Huang S, Chan C. Lifespan regulation in α/β posterior neurons of the fly mushroom bodies by Rab27. Aging Cell 2020; 19:e13179. [PMID: 32627932 PMCID: PMC7431830 DOI: 10.1111/acel.13179] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/25/2020] [Indexed: 12/13/2022] Open
Abstract
Brain function has been implicated to control the aging process and modulate lifespan. However, continuous efforts remain for the identification of the minimal sufficient brain region and the underlying mechanism for neuronal regulation of longevity. Here, we show that the Drosophila lifespan is modulated by rab27 functioning in a small subset of neurons of the mushroom bodies (MB), a brain structure that shares analogous functions with mammalian hippocampus and hypothalamus. Depleting rab27 in the α/βp neurons of the MB is sufficient to extend lifespan, enhance systemic stress responses, and alter energy homeostasis, all without trade‐offs in major life functions. Within the α/βp neurons, rab27KO causes the mislocalization of phosphorylated S6K thus attenuates TOR signaling, resulting in decreased protein synthesis and reduced neuronal activity. Consistently, expression of dominant‐negative S6K in the α/βp neurons increases lifespan. Furthermore, the expression of phospho‐mimetic S6 in α/βp neurons of rab27KO rescued local protein synthesis and reversed lifespan extension. These findings demonstrate that inhibiting TOR‐mediated protein synthesis in α/βp neurons is sufficient to promote longevity.
Collapse
Affiliation(s)
- Wen‐Yu Lien
- Graduate Institute of Physiology College of Medicine National Taiwan University Taipei Taiwan
| | - Yu‐Ting Chen
- Graduate Institute of Physiology College of Medicine National Taiwan University Taipei Taiwan
| | - Yi‐Jhan Li
- Graduate Institute of Physiology College of Medicine National Taiwan University Taipei Taiwan
| | - Jie‐Kai Wu
- Department of Biochemistry and Graduate Institute of Biomedical Sciences College of Medicine Chang Gung University Taoyuan Taiwan
| | - Kuan‐Lin Huang
- Graduate Institute of Physiology College of Medicine National Taiwan University Taipei Taiwan
| | - Jian‐Rong Lin
- Graduate Institute of Physiology College of Medicine National Taiwan University Taipei Taiwan
| | - Shih‐Ching Lin
- Graduate Institute of Physiology College of Medicine National Taiwan University Taipei Taiwan
| | - Chia‐Chun Hou
- Graduate Institute of Physiology College of Medicine National Taiwan University Taipei Taiwan
| | - Horng‐Dar Wang
- Institute of Biotechnology National Tsing Hua University Hsinchu Taiwan
| | - Chia‐Lin Wu
- Department of Biochemistry and Graduate Institute of Biomedical Sciences College of Medicine Chang Gung University Taoyuan Taiwan
- Department of Neurology Linkou Chang Gung Memorial Hospital Taoyuan Taiwan
| | - Shu‐Yi Huang
- Department of Medical Research National Taiwan University Hospital Taipei Taiwan
| | - Chih‐Chiang Chan
- Graduate Institute of Physiology College of Medicine National Taiwan University Taipei Taiwan
| |
Collapse
|
50
|
Sepil I, Hopkins BR, Dean R, Bath E, Friedman S, Swanson B, Ostridge HJ, Harper L, Buehner NA, Wolfner MF, Konietzny R, Thézénas ML, Sandham E, Charles PD, Fischer R, Steinhauer J, Kessler BM, Wigby S. Male reproductive aging arises via multifaceted mating-dependent sperm and seminal proteome declines, but is postponable in Drosophila. Proc Natl Acad Sci U S A 2020; 117:17094-17103. [PMID: 32611817 PMCID: PMC7382285 DOI: 10.1073/pnas.2009053117] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Declining ejaculate performance with male age is taxonomically widespread and has broad fitness consequences. Ejaculate success requires fully functional germline (sperm) and soma (seminal fluid) components. However, some aging theories predict that resources should be preferentially diverted to the germline at the expense of the soma, suggesting differential impacts of aging on sperm and seminal fluid and trade-offs between them or, more broadly, between reproduction and lifespan. While harmful effects of male age on sperm are well known, we do not know how much seminal fluid deteriorates in comparison. Moreover, given the predicted trade-offs, it remains unclear whether systemic lifespan-extending interventions could ameliorate the declining performance of the ejaculate as a whole. Here, we address these problems using Drosophila melanogaster. We demonstrate that seminal fluid deterioration contributes to male reproductive decline via mating-dependent mechanisms that include posttranslational modifications to seminal proteins and altered seminal proteome composition and transfer. Additionally, we find that sperm production declines chronologically with age, invariant to mating activity such that older multiply mated males become infertile principally via reduced sperm transfer and viability. Our data, therefore, support the idea that both germline and soma components of the ejaculate contribute to male reproductive aging but reveal a mismatch in their aging patterns. Our data do not generally support the idea that the germline is prioritized over soma, at least, within the ejaculate. Moreover, we find that lifespan-extending systemic down-regulation of insulin signaling results in improved late-life ejaculate performance, indicating simultaneous amelioration of both somatic and reproductive aging.
Collapse
Affiliation(s)
- Irem Sepil
- Department of Zoology, University of Oxford, OX1 3SZ Oxford, United Kingdom;
| | - Ben R Hopkins
- Department of Zoology, University of Oxford, OX1 3SZ Oxford, United Kingdom
- Department of Ecology and Evolution, University of California, Davis, CA 95616
| | - Rebecca Dean
- Department of Genetics, Evolution and Environment, University College London, WC1E 6BT London, United Kingdom
| | - Eleanor Bath
- Department of Zoology, University of Oxford, OX1 3SZ Oxford, United Kingdom
| | | | - Ben Swanson
- Department of Zoology, University of Oxford, OX1 3SZ Oxford, United Kingdom
| | - Harrison J Ostridge
- Department of Zoology, University of Oxford, OX1 3SZ Oxford, United Kingdom
- Department of Genetics, Evolution and Environment, University College London, WC1E 6BT London, United Kingdom
| | - Lucy Harper
- Department of Zoology, University of Oxford, OX1 3SZ Oxford, United Kingdom
- School of Biology, University of St Andrews, KY16 9ST St Andrews, United Kingdom
| | - Norene A Buehner
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853
| | - Mariana F Wolfner
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853
| | - Rebecca Konietzny
- Nuffield Department of Medicine, TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of Oxford, OX3 7FZ Oxford, United Kingdom
| | - Marie-Laëtitia Thézénas
- Nuffield Department of Medicine, TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of Oxford, OX3 7FZ Oxford, United Kingdom
| | - Elizabeth Sandham
- Department of Zoology, University of Oxford, OX1 3SZ Oxford, United Kingdom
| | - Philip D Charles
- Nuffield Department of Medicine, TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of Oxford, OX3 7FZ Oxford, United Kingdom
| | - Roman Fischer
- Nuffield Department of Medicine, TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of Oxford, OX3 7FZ Oxford, United Kingdom
| | | | - Benedikt M Kessler
- Nuffield Department of Medicine, TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of Oxford, OX3 7FZ Oxford, United Kingdom
| | - Stuart Wigby
- Department of Zoology, University of Oxford, OX1 3SZ Oxford, United Kingdom
- Faculty Biology, Applied Zoology, Technische Universität Dresden, 01069 Dresden, Germany
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, L69 7ZB Liverpool, United Kingdom
| |
Collapse
|