1
|
Zhou S, Novak KE, Kaletsky R, Weng Y, Ange JS, Stevenson ME, Toraason E, Zhang Y, Zhang W, Dong MQ, Murphy CT. Body-to-brain insulin and Notch signaling regulates memory through neuronal CREB activity. NATURE AGING 2025:10.1038/s43587-025-00873-7. [PMID: 40425807 DOI: 10.1038/s43587-025-00873-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 04/11/2025] [Indexed: 05/29/2025]
Abstract
While memory regulation is predominantly understood as autonomous to neurons, factors outside the brain can also affect neuronal function. In Caenorhabditis elegans, the insulin/IGF-1-like signaling (IIS) pathway regulates longevity, metabolism and memory: long-lived daf-2 insulin/IGF-1 receptor mutants more than double memory duration after a single training session, and it was assumed that memory regulation was strictly neuronal. However, here we show that degradation of DAF-2 in the hypodermis also greatly extends memory, via expression of the diffusible Notch ligand, OSM-11, which in turn activates Notch signaling in neurons. Single-nucleus RNA sequencing of neurons revealed increased expression of CREB and other memory genes. Furthermore, in aged animals, activation of the hypodermal IIS-Notch pathway as well as OSM-11 overexpression rescue both memory and learning via CREB activity. Thus, insulin signaling in the liver-like hypodermis non-autonomously regulates neuronal function, providing a systemic connection between metabolism and memory through IIS-Notch-CREB signaling from the body to the brain.
Collapse
Affiliation(s)
- Shiyi Zhou
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- LSI Genomics, Princeton University, Princeton, NJ, USA
| | - Katherine E Novak
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- LSI Genomics, Princeton University, Princeton, NJ, USA
| | - Rachel Kaletsky
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- LSI Genomics, Princeton University, Princeton, NJ, USA
| | - Yifei Weng
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- LSI Genomics, Princeton University, Princeton, NJ, USA
| | - Jonathan St Ange
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- LSI Genomics, Princeton University, Princeton, NJ, USA
| | - Morgan E Stevenson
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- LSI Genomics, Princeton University, Princeton, NJ, USA
| | - Erik Toraason
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- LSI Genomics, Princeton University, Princeton, NJ, USA
| | - Yanping Zhang
- National Institute of Biological Sciences, Beijing, China
- Beijing Key Laboratory of the Cell Biology of Animal Aging, Beijing, China
| | - Wenhong Zhang
- National Institute of Biological Sciences, Beijing, China
- Beijing Key Laboratory of the Cell Biology of Animal Aging, Beijing, China
| | - Meng-Qiu Dong
- National Institute of Biological Sciences, Beijing, China
- Beijing Key Laboratory of the Cell Biology of Animal Aging, Beijing, China
| | - Coleen T Murphy
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
- LSI Genomics, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
2
|
Zhu M, Wang J, Zhu L, Zhu M. Investigations of forgetting in Caenorhabditis elegans. Neurobiol Learn Mem 2025; 220:108061. [PMID: 40350072 DOI: 10.1016/j.nlm.2025.108061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 04/17/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025]
Abstract
The traditional view considered forgetting as a passive process where memory traces gradually fade due to the natural weakening of neural connections. However, studies on olfactory memory in Drosophila have revealed that forgetting is an active process controlled by specific neural circuits. Caenorhabditis elegans is a widely used model organism in neurobiological research due to its relatively simple nervous system. Despite its simplicity, C. elegans exhibits complex behaviors influenced by environmental factors and prior experiences. Similar to Drosophila, C. elegans can actively initiate neural circuits based on the type of memory that needs to be forgotten, which supports using C. elegans as a model for studying forgetting. These characteristics facilitate the identification of genes and pathways involved in forgetting in C. elegans. In this review, we discuss recent advances in understanding forgetting mechanisms in C. elegans through three well-characterized olfactory learning paradigms. The insights derived from C. elegans offer a valuable framework for understanding the molecular and cellular mechanisms underlying forgetting, with potentially broader implications for memory regulation in more complex organisms.
Collapse
Affiliation(s)
- Man Zhu
- College of Biological and Food Engineering, Qujing Normal University, Qujing 655011, China
| | - Jiayi Wang
- College of Biological and Food Engineering, Qujing Normal University, Qujing 655011, China
| | - Ling Zhu
- College of Biological and Food Engineering, Qujing Normal University, Qujing 655011, China
| | - Man Zhu
- College of Biological and Food Engineering, Qujing Normal University, Qujing 655011, China.
| |
Collapse
|
3
|
Wang PZ, Ge MH, Su P, Wu PP, Wang L, Zhu W, Li R, Liu H, Wu JJ, Xu Y, Zhao JL, Li SJ, Wang Y, Chen LM, Wu TH, Wu ZX. Sensory plasticity caused by up-down regulation encodes the information of short-term learning and memory. iScience 2025; 28:112215. [PMID: 40224011 PMCID: PMC11987006 DOI: 10.1016/j.isci.2025.112215] [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: 09/12/2024] [Revised: 11/26/2024] [Accepted: 03/10/2025] [Indexed: 04/15/2025] Open
Abstract
Learning and memory are essential for animals' well-being and survival. The underlying mechanisms are a major task of neuroscience studies. In this study, we identified a circuit consisting of ASER, RIC, RIS, and AIY, is required for short-term salt chemotaxis learning (SCL) in C. elegans. ASER NaCl-sensation possesses are remodeled by salt/food-deprivation pared conditioning. RIC integrates the sensory information of NaCl and food availability. It excites ASER and inhibits AIY by tyramine/TYRA-2 and octopamine/OCTR-1 signaling pathways, respectively. By the salt conditioning, RIC NaCl calcium response to NaCl is depressed, thus, the RIC excitation of ASER and inhibition of AIY are suppressed. ASER excites RIS by FLP-14/FRPR-10 signaling. RIS inhibits ASER via PDF-2/PDFR-1 signaling in negative feedback. ASER sensory plasticity caused by RIC plasticity and RIS negative feedback are required for both learning and memory recall. Thus, the sensation plasticity encodes the information of the short-term SCL that facilitates animal adaptation to dynamic environments.
Collapse
Affiliation(s)
- Ping-Zhou Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, Institute of Biophysics and Biochemistry, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Ming-Hai Ge
- Key Laboratory of Molecular Biophysics of Ministry of Education, Institute of Biophysics and Biochemistry, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Pan Su
- Key Laboratory of Molecular Biophysics of Ministry of Education, Institute of Biophysics and Biochemistry, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Piao-Ping Wu
- Key Laboratory of Molecular Biophysics of Ministry of Education, Institute of Biophysics and Biochemistry, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, Institute of Biophysics and Biochemistry, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Zhu
- Key Laboratory of Molecular Biophysics of Ministry of Education, Institute of Biophysics and Biochemistry, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Li
- Key Laboratory of Molecular Biophysics of Ministry of Education, Institute of Biophysics and Biochemistry, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, Institute of Biophysics and Biochemistry, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Jing-Jing Wu
- Key Laboratory of Molecular Biophysics of Ministry of Education, Institute of Biophysics and Biochemistry, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Xu
- Key Laboratory of Molecular Biophysics of Ministry of Education, Institute of Biophysics and Biochemistry, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Lu Zhao
- Key Laboratory of Molecular Biophysics of Ministry of Education, Institute of Biophysics and Biochemistry, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Si-Jia Li
- Key Laboratory of Molecular Biophysics of Ministry of Education, Institute of Biophysics and Biochemistry, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, Institute of Biophysics and Biochemistry, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Li-Ming Chen
- Key Laboratory of Molecular Biophysics of Ministry of Education, Institute of Biophysics and Biochemistry, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Tai-Hong Wu
- Hunan Research Center of the Basic Discipline for Cell Signaling, State Key Laboratory of Chemo and Biosensing, College of Biology, Hunan University, Changsha, China
| | - Zheng-Xing Wu
- Key Laboratory of Molecular Biophysics of Ministry of Education, Institute of Biophysics and Biochemistry, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Tomioka M. High-throughput assessment of the behavioral responses to toxic organic solvents in Caenorhabditis elegans. PLoS One 2025; 20:e0311460. [PMID: 40245001 PMCID: PMC12005522 DOI: 10.1371/journal.pone.0311460] [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: 09/18/2024] [Accepted: 03/10/2025] [Indexed: 04/19/2025] Open
Abstract
Novel chemical compounds are continuously being developed for use in various industries and daily life. Workers in these industries assess and avoid chemical hazards based on published information about chemical toxicities. However, avoiding the hazards associated with chemicals with unknown toxicity is difficult. Therefore, understanding the toxicities of chemicals in a high-throughput, multifaceted manner is essential. In this study, I developed a high-throughput method for assessing chemical toxicities through quantitative measurement of behavior in Caenorhabditis elegans. I determined the acute response to 30 organic solvents, including alcohols, cellosolves, ethers, ketones, and acetate esters, which are widely used in industries, with motility as an endpoint. Exposure to 0.5%-6% organic solvents caused a dramatic decrease in locomotion speed. The adverse effects of organic solvents on motility were proportional to the lipid solubility of the chemicals, similar to the positive relationship between the anesthetic effects of volatile organic chemicals and their lipid solubility in organisms, including humans. In addition to their effects on motility, organic solvents affect posture during locomotion in different ways depending on the chemical's functional group. Solvents with hydroxyl groups, such as alcohols and cellosolves (0.5%-3%), reduced the amplitude of body bending, whereas solvents with ketone groups, such as ketones and acetate esters (0.5%-4%), increased it during undulatory locomotion. In addition, organic solvents caused changes in chemotaxis plasticity based on the association between starvation and chemical signals at concentrations lower than those that affect locomotion. This study describes a high-throughput method for acute chemical toxicity testing and provides new insights into behavioral responses to organic solvents that are toxic to humans and other animals.
Collapse
Affiliation(s)
- Masahiro Tomioka
- Division of Industrial Toxicology, Research Center for Chemical Information and Management, National Institute of Occupational Safety and Health, Kawasaki, Japan
| |
Collapse
|
5
|
Jiang WI, Cao Y, Xue Y, Ji Y, Winer BY, Chandra R, Zhang XF, Zhang M, Singhal NS, Pierce JT, Chen S, Ma DK. Suppressing APOE4-induced neural pathologies by targeting the VHL-HIF axis. Proc Natl Acad Sci U S A 2025; 122:e2417515122. [PMID: 39874294 PMCID: PMC11804744 DOI: 10.1073/pnas.2417515122] [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: 08/29/2024] [Accepted: 12/19/2024] [Indexed: 01/30/2025] Open
Abstract
The ε4 variant of human apolipoprotein E (APOE4) is a key genetic risk factor for neurodegeneration in Alzheimer's disease and elevated all-cause mortality in humans. Understanding the factors and mechanisms that can mitigate the harmful effects of APOE4 has significant implications. In this study, we find that inactivating the VHL-1 (Von Hippel-Lindau) protein can suppress mortality, neural and behavioral pathologies caused by transgenic human APOE4 in Caenorhabditis elegans. The protective effects of VHL-1 deletion are recapitulated by stabilized HIF-1 (hypoxia-inducible factor), a transcription factor degraded by VHL-1. HIF-1 activates a genetic program that safeguards against mitochondrial dysfunction, oxidative stress, proteostasis imbalance, and endolysosomal rupture-critical cellular events linked to neural pathologies and mortality. Furthermore, genetic inhibition of Vhl reduces cerebral vascular injury and synaptic lesions in APOE4 mice, suggesting an evolutionarily conserved mechanism. Thus, we identify the VHL-HIF axis as a potent modulator of APOE4-induced neural pathologies and propose that targeting this pathway in nonproliferative tissues may curb cellular damage, protect against neurodegeneration, and reduce tissue injuries and mortality.
Collapse
Affiliation(s)
- Wei I. Jiang
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA94158
| | - Yiming Cao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing210009, China
| | - Yue Xue
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing210009, China
| | - Yichun Ji
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing210009, China
| | - Benjamin Y. Winer
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA94158
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY10065
- HHMI, Chevy Chase, MD20815
| | - Rashmi Chandra
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA94158
| | - Xingyuan Fischer Zhang
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA94158
| | - Mengqi Zhang
- Department of Neurology, University of California San Francisco, San Francisco, CA94158
| | - Neel S. Singhal
- Department of Neurology, University of California San Francisco, San Francisco, CA94158
| | - Jonathan T. Pierce
- Department of Neuroscience, The Center for Learning and Memory, Waggoner Center for Alcohol and Addiction Research, Institute of Neuroscience, University of Texas at Austin, Austin, TX78712
| | - Song Chen
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing210009, China
| | - Dengke K. Ma
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA94158
- Department of Physiology, University of California San Francisco, San Francisco, CA94158
- Innovative Genomics Institute, University of California, Berkeley, CA94720
| |
Collapse
|
6
|
Ji Z, Wang B, Chandra R, Liu J, Yang S, Long Y, Egan M, L’Etoile N, Ma DK. Non-Visual Light Sensing Enhances Behavioral Memory and Drives Gene Expression in C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.634647. [PMID: 39975403 PMCID: PMC11838244 DOI: 10.1101/2025.01.27.634647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Visible light influences a range of physiological processes, yet how animals respond to it independently of the visual system remains largely unknown. Here, we uncover a previously undescribed light-induced transcriptional pathway that modulates behavioral plasticity in C. elegans, a roundworm without eyes. We demonstrate that ambient visible light or controlled-intensity visible-spectrum LED activates an effector gene cyp-14A5 in non-neuronal tissues through the bZIP transcription factors ZIP-2 and CEBP-2. Light induction of cyp-14A5 is more prominent at shorter wavelengths but is independent of the known blue light receptors LITE-1 and GUR-3 in C. elegans. This bZIP-dependent genetic pathway in non-neuronal tissues enhances behavioral adaptability and olfactory memory, suggesting a body-brain communication axis. Furthermore, we use the light-responsive cyp-14A5 promoter to drive ectopic gene expression, causing synthetic light-induced sleep and rapid aging phenotypes in C. elegans. These findings advance our understanding of light-responsive mechanisms outside the visual system and offer a new genetic tool for visible light-inducible gene expression in non-neuronal tissues.
Collapse
Affiliation(s)
- Zhijian Ji
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - Bingying Wang
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - Rashmi Chandra
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - Junqiang Liu
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - Supeng Yang
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
- Department of Molecular Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Yong Long
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Michael Egan
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
- Department of Molecular Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Noelle L’Etoile
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Dengke K. Ma
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
- Department of Physiology, University of California San Francisco, San Francisco, CA 94158, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
7
|
St Ange J, Weng Y, Kaletsky R, Stevenson ME, Moore RS, Zhou S, Murphy CT. Adult single-nucleus neuronal transcriptomes of insulin signaling mutants reveal regulators of behavior and learning. CELL GENOMICS 2024; 4:100720. [PMID: 39637862 DOI: 10.1016/j.xgen.2024.100720] [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: 02/22/2024] [Revised: 09/16/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024]
Abstract
Gene expression in individual neurons can change during development to adulthood and can have large effects on behavior. Additionally, the insulin/insulin-like signaling (IIS) pathway regulates many of the adult functions of Caenorhabditis elegans, including learning and memory, via transcriptional changes. We used the deep resolution of single-nucleus RNA sequencing to define the adult transcriptome of each neuron in wild-type and daf-2 mutants, revealing expression differences between L4 larval and adult neurons in chemoreceptors, synaptic genes, and learning/memory genes. We used these data to identify adult new AWC-specific regulators of chemosensory function that emerge upon adulthood. daf-2 gene expression changes correlate with improved cognitive functions, particularly in the AWC sensory neuron that controls learning and associative memory; behavioral assays of AWC-specific daf-2 genes revealed their roles in cognitive function. Combining technology and functional validation, we identified conserved genes that function in specific adult neurons to control behavior, including learning and memory.
Collapse
Affiliation(s)
- Jonathan St Ange
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Yifei Weng
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Rachel Kaletsky
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Morgan E Stevenson
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Rebecca S Moore
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Shiyi Zhou
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Coleen T Murphy
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
8
|
Beam TC, Bright M, Pearson AC, Dua I, Smith M, Dutta AK, Bhadra SC, Salman S, Strickler CN, Anderson CE, Peshkin L, Yampolsky LY. Short lifespan is one's fate, long lifespan is one's achievement: lessons from Daphnia. GeroScience 2024; 46:6361-6381. [PMID: 38900345 PMCID: PMC11493910 DOI: 10.1007/s11357-024-01244-7] [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: 04/30/2024] [Accepted: 06/05/2024] [Indexed: 06/21/2024] Open
Abstract
Studies of longevity rely on baseline life expectancy of reference genotypes measured in standardized conditions. Variation among labs, protocols, and genotypes makes longevity intervention studies difficult to compare. Furthermore, extending lifespan under suboptimal conditions or that of a short-lived genotype may be of a lesser theoretical and translational value than extending the maximal possible lifespan. Daphnia is becoming a model organism of choice for longevity research complementing data obtained on traditional models. In this study, we report longevity of several genotypes of a long-lived species D. magna under a variety of protocols, aiming to document the highest lifespan, factors reducing it, and parameters that change with age and correlate with longevity. Combining longevity data from 25 experiments across two labs, we report a strong intraspecific variation, moderate effects of group size and medium composition, and strong genotype-by-environment interactions with respect to food level. Specifically, short-lived genotypes show no caloric restriction (CR) effect, while long-lived ones expand their lifespan even further under CR. We find that the CR non-responsive clones show little correlation between longevity and two measures of lipid peroxidation. In contrast, the long-lived, CR-responsive clones show a positive correlation between longevity and lipid hydroperoxide abundance, and a negative correlation with MDA concentration. This indicates differences among genotypes in age-related accumulation and detoxification of LPO products and their effects on longevity. Our observations support the hypothesis that a long lifespan can be affected by CR and levels of oxidative damage, while genetically determined short lifespan remains short regardless.
Collapse
Affiliation(s)
- Thomas C Beam
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA
| | - Mchale Bright
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA
| | - Amelia C Pearson
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA
| | - Ishaan Dua
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA
| | - Meridith Smith
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA
| | - Ashit K Dutta
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA
| | - Shymal C Bhadra
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA
- Department of Biological Sciences, Purdue University Fort Wayne, Fort Wayne, IN, 46805, USA
| | - Saad Salman
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA
| | - Caleb N Strickler
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA
| | - Cora E Anderson
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Leonid Peshkin
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Lev Y Yampolsky
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, 37601, USA.
| |
Collapse
|
9
|
Cohen N, Rabinowitch I. Resolving transitions between distinct phases of memory consolidation at high resolution in Caenorhabditis elegans. iScience 2024; 27:111147. [PMID: 39524366 PMCID: PMC11547966 DOI: 10.1016/j.isci.2024.111147] [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: 05/30/2024] [Revised: 08/26/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Memory consolidation following learning is a dynamic and complex process comprising several transitions between distinct memory phases. Although memory consolidation has been studied extensively, it remains difficult to draw an integral description that can delimit the transition points between specific memory phases at the behavioral, neuronal, and genetic levels. To this end, we have developed a rapid and robust aversive conditioning protocol for the nematode worm Caenorhabditis elegans, tracing memory consolidation within the first hour post conditioning and then up to 18 h post conditioning. This made it possible to uncover time-dependent involvement of primary sensory neurons, transcription and translation processes, and diverse gene populations in memory consolidation. The change in neuronal valence was strong enough to induce second order conditioning, and was amenable to considerable modulation in specific mutant strains. Together, our work lends memory consolidation to detailed temporal and spatial analysis, advancing system-wide understanding of learning and memory.
Collapse
Affiliation(s)
- Netanel Cohen
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel
| | - Ithai Rabinowitch
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel
| |
Collapse
|
10
|
Wu P, Vandemeulebroucke L, Claeys M, Bert W, Braeckman BP. The Effect of Axenic Dietary Restriction on the Age-Related Changes in Caenorhabditis elegans. J Gerontol A Biol Sci Med Sci 2024; 79:glae205. [PMID: 39171522 DOI: 10.1093/gerona/glae205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Indexed: 08/23/2024] Open
Abstract
Axenic dietary restriction (ADR) is highly effective in extending lifespan of Caenorhabditis elegans, but its effects on healthspan improvement are less well characterized. Using transmission electron microscopy, morphometric analyses, and functional assays, we found ADR can preserve tissue ultrastructure, including the cuticle, epidermis, and intestinal lumen, and reduce age-associated pathologies like gonad degeneration, uterine tumor clusters, pharyngeal deterioration, and intestinal atrophy. However, there was no notable improvement in behavioral and functional metrics. Our results underscore that lifespan extension through ADR does not inherently translate to broad healthspan improvements.
Collapse
Affiliation(s)
- Ping Wu
- Laboratory of Aging Physiology and Molecular Evolution, Department of Biology, Ghent University, Ghent, Belgium
| | - Lieselot Vandemeulebroucke
- Laboratory of Aging Physiology and Molecular Evolution, Department of Biology, Ghent University, Ghent, Belgium
| | - Myriam Claeys
- Nematology Research Unit, Department of Biology, Ghent University, Ghent, Belgium
| | - Wim Bert
- Nematology Research Unit, Department of Biology, Ghent University, Ghent, Belgium
| | - Bart P Braeckman
- Laboratory of Aging Physiology and Molecular Evolution, Department of Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
11
|
Hayden AN, Brandel KL, Pietryk EW, Merlau PR, Vijayakumar P, Leptich EJ, Gaytan ES, Williams MI, Ni CW, Chao HT, Rosenfeld JA, Arey RN. Behavioral screening reveals a conserved residue in Y-Box RNA-binding protein required for associative learning and memory in C. elegans. PLoS Genet 2024; 20:e1011443. [PMID: 39423228 PMCID: PMC11524487 DOI: 10.1371/journal.pgen.1011443] [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: 05/17/2024] [Revised: 10/30/2024] [Accepted: 09/30/2024] [Indexed: 10/21/2024] Open
Abstract
RNA-binding proteins (RBPs) regulate translation and plasticity which are required for memory. RBP dysfunction has been linked to a range of neurological disorders where cognitive impairments are a key symptom. However, of the 2,000 RBPs in the human genome, many are uncharacterized with regards to neurological phenotypes. To address this, we used the model organism C. elegans to assess the role of 20 conserved RBPs in memory. We identified eight previously uncharacterized memory regulators, three of which are in the C. elegans Y-Box (CEY) RBP family. Of these, we determined that cey-1 is the closest ortholog to the mammalian Y-Box (YBX) RBPs. We found that CEY-1 is both necessary in the nervous system for memory ability and sufficient to promote memory. Leveraging human datasets, we found both copy number variation losses and single nucleotide variants in YBX1 and YBX3 in individuals with neurological symptoms. We identified one predicted deleterious YBX3 variant of unknown significance, p.Asn127Tyr, in two individuals with neurological symptoms. Introducing this variant into endogenous cey-1 locus caused memory deficits in the worm. We further generated two humanized worm lines expressing human YBX3 or YBX1 at the cey-1 locus to test evolutionary conservation of YBXs in memory and the potential functional significance of the p.Asn127Tyr variant. Both YBX1/3 can functionally replace cey-1, and introduction of p.Asn127Tyr into the humanized YBX3 locus caused memory deficits. Our study highlights the worm as a model to reveal memory regulators and identifies YBX dysfunction as a potential new source of rare neurological disease.
Collapse
Affiliation(s)
- Ashley N. Hayden
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, United States of America
| | - Katie L. Brandel
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, United States of America
| | - Edward W. Pietryk
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Paul R. Merlau
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, United States of America
| | - Priyadharshini Vijayakumar
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, United States of America
| | - Emily J. Leptich
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, United States of America
| | - Elizabeth S. Gaytan
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, United States of America
- Postbaccalaureate Research Education Program, Baylor College of Medicine, Houston, Texas, United States of America
| | - Meredith I. Williams
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, United States of America
| | - Connie W. Ni
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Neuroscience, Rice University, Houston, Texas, United States of America
| | - Hsiao-Tuan Chao
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pediatrics, Division of Neurology and Developmental Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
- Cain Pediatric Neurology Research Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas, United States of America
- McNair Medical Institute, The Robert and Janice McNair Foundation, Houston, Texas, United States of America
| | - Jill A. Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Baylor Genetics Laboratories, Houston, Texas, United States of America
| | - Rachel N. Arey
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
12
|
Sirwani N, Hedtke SM, Grant K, McColl G, Grant WN. Levels of Amyloid Beta ( Aβ) Expression in the Caenorhabditis elegans Neurons Influence the Onset and Severity of Neuronally Mediated Phenotypes. Cells 2024; 13:1598. [PMID: 39329779 PMCID: PMC11430350 DOI: 10.3390/cells13181598] [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: 08/16/2023] [Revised: 09/16/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024] Open
Abstract
A characteristic feature of Alzheimer's disease (AD) is the formation of neuronal extracellular senile plaques composed of aggregates of fibrillar amyloid β (Aβ) peptides, with the Aβ1-42 peptide being the most abundant species. These Aβ peptides have been proposed to contribute to the pathophysiology of the disease; however, there are few tools available to test this hypothesis directly. In particular, there are no data that establish a dose-response relationship between Aβ peptide expression level and disease. We have generated a panel of transgenic Caenorhabditis elegans strains expressing the human Aβ1-42 peptide under the control of promoter regions of two pan-neuronal expressed genes, snb-1 and rgef-1. Phenotypic data show strong age-related defects in motility, subtle changes in chemotaxis, reduced median and maximum lifespan, changes in health span indicators, and impaired learning. The Aβ1-42 expression level of these strains differed as a function of promoter identity and transgene copy number, and the timing and severity of phenotypes mediated by Aβ1-42 were strongly positively correlated with expression level. The pan-neuronal expression of varying levels of human Aβ1-42 in a nematode model provides a new tool to investigate the in vivo toxicity of neuronal Aβ expression and the molecular and cellular mechanisms underlying AD progression in the absence of endogenous Aβ peptides. More importantly, it allows direct quantitative testing of the dose-response relationship between neuronal Aβ peptide expression and disease for the first time. These strains may also be used to develop screens for novel therapeutics to treat Alzheimer's disease.
Collapse
Affiliation(s)
- Neha Sirwani
- Department of Environment and Genetics, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC 3086, Australia; (S.M.H.); (W.N.G.)
| | - Shannon M. Hedtke
- Department of Environment and Genetics, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC 3086, Australia; (S.M.H.); (W.N.G.)
| | - Kirsten Grant
- Department of Environment and Genetics, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC 3086, Australia; (S.M.H.); (W.N.G.)
| | - Gawain McColl
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia;
| | - Warwick N. Grant
- Department of Environment and Genetics, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC 3086, Australia; (S.M.H.); (W.N.G.)
| |
Collapse
|
13
|
Abstract
Behavioral plasticity allows animals to modulate their behavior based on experience and environmental conditions. Caenorhabditis elegans exhibits experience-dependent changes in its behavioral responses to various modalities of sensory cues, including odorants, salts, temperature, and mechanical stimulations. Most of these forms of behavioral plasticity, such as adaptation, habituation, associative learning, and imprinting, are shared with other animals. The C. elegans nervous system is considerably tractable for experimental studies-its function can be characterized and manipulated with molecular genetic methods, its activity can be visualized and analyzed with imaging approaches, and the connectivity of its relatively small number of neurons are well described. Therefore, C. elegans provides an opportunity to study molecular, neuronal, and circuit mechanisms underlying behavioral plasticity that are either conserved in other animals or unique to this species. These findings reveal insights into how the nervous system interacts with the environmental cues to generate behavioral changes with adaptive values.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Yuichi Iino
- Department of Biological Sciences, University of Tokyo, Tokyo 113-0032, Japan
| | - William R Schafer
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH, UK
- Department of Biology, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
14
|
Lai Y, Ay M, Hospital CD, Miller GW, Sarkar S. Seminar: Functional Exposomics and Mechanisms of Toxicity-Insights from Model Systems and NAMs. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:94201. [PMID: 39230330 PMCID: PMC11373422 DOI: 10.1289/ehp13120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/22/2024] [Accepted: 08/12/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND Significant progress has been made over the past decade in measuring the chemical components of the exposome, providing transformative population-scale frameworks in probing the etiologic link between environmental factors and disease phenotypes. While the analytical technologies continue to evolve with reams of data being generated, there is an opportunity to complement exposome-wide association studies (ExWAS) with functional analyses to advance etiologic search at organismal, cellular, and molecular levels. OBJECTIVES Exposomics is a transdisciplinary field aimed at enabling discovery-based analysis of the nongenetic factors that contribute to disease, including numerous environmental chemical stressors. While advances in exposure assessment are enhancing population-based discovery of exposome-wide effects and chemical exposure agents, functional screening and elucidation of biological effects of exposures represent the next logical step toward precision environmental health and medicine. In this work, we focus on the use, strategies, and prospects of alternative approaches and model systems to enhance the current human exposomics framework in biomarker search and causal understanding, spanning from bench-based nonmammalian organisms and cell culture to computational new approach methods (NAMs). DISCUSSION We visit the definition of the functional exposome and exposomics and discuss a need to leverage alternative models as opposed to mammalian animals for delineating exposome-wide health effects. Under the "three Rs" principle of reduction, replacement, and refinement, model systems such as roundworms, fruit flies, zebrafish, and induced pluripotent stem cells (iPSCs) are advantageous over mammals (e.g., rodents or higher vertebrates). These models are cost-effective, and cell-specific genetic manipulations in these models are easier and faster, compared to mammalian models. Meanwhile, in silico NAMs enhance hazard identification and risk assessment in humans by bridging the translational gaps between toxicology data and etiologic inference, as represented by in vitro to in vivo extrapolation (IVIVE) and integrated approaches to testing and assessment (IATA) under the adverse outcome pathway (AOP) framework. Together, these alternatives offer a strong toolbox to support functional exposomics to study toxicity and causal mediators underpinning exposure-disease links. https://doi.org/10.1289/EHP13120.
Collapse
Affiliation(s)
- Yunjia Lai
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Muhammet Ay
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Carolina Duarte Hospital
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Gary W. Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Souvarish Sarkar
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, USA
- Department of Neuroscience, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
15
|
Toraason E, Kaletsky R, Murphy C. In vivo neuron-specific expression of C. elegans reprogramming factor orthologs does not alleviate age-related cognitive decline. MICROPUBLICATION BIOLOGY 2024; 2024. [PMID: 39267613 PMCID: PMC11391276 DOI: 10.17912/micropub.biology.001304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024]
Abstract
Overexpression of the OSK(M) (Oct4, Sox2, Klf4, with or without cMyc) pluripotency factors have shown promise in rejuvenating the function of aged neurons. To test whether this intervention could also ameliorate age-associated cognitive decline, we used a doxycycline inducible system to overexpress the C. elegans OSK orthologs specifically in aging C. elegans neurons. We find that OSK does not improve short-term associative memory or extend lifespan and can further disrupt chemotaxis behavior. Taken together, our data suggest that OSK-mediated partial reprogramming may have deleterious effects on post-mitotic neurons that function in cognitive processes.
Collapse
Affiliation(s)
- Erik Toraason
- Lewis-Sigler Institute for Integrative Genomics, Princeton University
- Department of Molecular Biology, Princeton University
| | - Rachel Kaletsky
- Lewis-Sigler Institute for Integrative Genomics, Princeton University
- Department of Molecular Biology, Princeton University
| | - Coleen Murphy
- Department of Molecular Biology, Princeton University
- Lewis-Sigler Institute for Integrative Genomics, Princeton University
| |
Collapse
|
16
|
Xu K, Zhang J, Ma W, Wang Y, Chen B, Gao N, Pang J, Zhan H. Home-Based Shi's Knee Daoyin Exercise for Knee Osteoarthritis: A Randomized Controlled Pilot Trial. J Pain Res 2024; 17:2811-2822. [PMID: 39224147 PMCID: PMC11368107 DOI: 10.2147/jpr.s469176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Objective Shi's Knee Daoyin (SKD) exercise is a treatment derived from Traditional Chinese exercise (TCE) specifically designed for lower limb health care. This study aimed to assess the feasibility of conducting a randomized controlled trial to explore the effectiveness of SKD exercise in treating knee osteoarthritis (KOA). Methods Participants were randomized to receive Health Education (HE) or SKD exercise. The primary outcomes were feasibility and safety outcomes, including participant recruitment rate, retention rate, as well as adherence to intervention. The secondary outcomes included Visual Analogue Scale (VAS) scores for pain, the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) score, the 20-Meter Walk Test (20-MWT) and the 5-times Chair-Stand Test (5-CST). Results The results indicate that out of 89 individuals invited to participate in the study, 72 were eligible and agreed to participate, resulting in a recruitment rate of 80.9%. All participating patients completed the follow-up and were included in the analysis; no patients dropped out of the study due to adverse events. The secondary outcome measures showed that after twelve weeks of treatment, the VAS score, WOMAC total score, WOMAC pain score, WOMAC stiffness score, and WOMAC function score of patients in the HE group and SKD group all improved, but the improvement was more significant in the SKD group. The 20-MWT of SKD group after treatment was significantly shorter than before treatment (P<0.001); There was no significant difference in 20-MWT between the HE group and baseline after treatment. The performance of the two groups of patients improved in 5-CST, but there was no statistical difference between the two groups after treatment (P=2.439). Conclusion This study evaluated the feasibility and effectiveness of home-based SKD exercise intervention in alleviating symptoms in patients with symptomatic KOA, providing valuable information for designing an appropriate randomized controlled study.
Collapse
Affiliation(s)
- Kun Xu
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Traumatology & Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jiefan Zhang
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Traumatology & Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Wei Ma
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Traumatology & Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yongyu Wang
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Traumatology & Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Bo Chen
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Traumatology & Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Ningyang Gao
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Traumatology & Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jian Pang
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Traumatology & Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Hongsheng Zhan
- Shi’s Center of Orthopedics and Traumatology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Traumatology & Orthopedics, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
17
|
Hudgins AD, Zhou S, Arey RN, Rosenfeld MG, Murphy CT, Suh Y. A systems biology-based identification and in vivo functional screening of Alzheimer's disease risk genes reveal modulators of memory function. Neuron 2024; 112:2112-2129.e4. [PMID: 38692279 PMCID: PMC11223975 DOI: 10.1016/j.neuron.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 10/18/2023] [Accepted: 04/08/2024] [Indexed: 05/03/2024]
Abstract
Genome-wide association studies (GWASs) have uncovered over 75 genomic loci associated with risk for late-onset Alzheimer's disease (LOAD), but identification of the underlying causal genes remains challenging. Studies of induced pluripotent stem cell (iPSC)-derived neurons from LOAD patients have demonstrated the existence of neuronal cell-intrinsic functional defects. Here, we searched for genetic contributions to neuronal dysfunction in LOAD using an integrative systems approach that incorporated multi-evidence-based gene mapping and network-analysis-based prioritization. A systematic perturbation screening of candidate risk genes in Caenorhabditis elegans (C. elegans) revealed that neuronal knockdown of the LOAD risk gene orthologs vha-10 (ATP6V1G2), cmd-1 (CALM3), amph-1 (BIN1), ephx-1 (NGEF), and pho-5 (ACP2) alters short-/intermediate-term memory function, the cognitive domain affected earliest during LOAD progression. These results highlight the impact of LOAD risk genes on evolutionarily conserved memory function, as mediated through neuronal endosomal dysfunction, and identify new targets for further mechanistic interrogation.
Collapse
Affiliation(s)
- Adam D Hudgins
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Shiyi Zhou
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Rachel N Arey
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Michael G Rosenfeld
- Department of Medicine, School of Medicine, University of California, La Jolla, CA, USA; Howard Hughes Medical Institute, University of California, La Jolla, CA, USA
| | - Coleen T Murphy
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA; LSI Genomics, Princeton University, Princeton, NJ, USA.
| | - Yousin Suh
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA; Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
18
|
Weng Y, Zhou S, Morillo K, Kaletsky R, Lin S, Murphy CT. The neuron-specific IIS/FOXO transcriptome in aged animals reveals regulatory mechanisms of cognitive aging. eLife 2024; 13:RP95621. [PMID: 38922671 PMCID: PMC11208049 DOI: 10.7554/elife.95621] [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] [Indexed: 06/27/2024] Open
Abstract
Cognitive decline is a significant health concern in our aging society. Here, we used the model organism C. elegans to investigate the impact of the IIS/FOXO pathway on age-related cognitive decline. The daf-2 Insulin/IGF-1 receptor mutant exhibits a significant extension of learning and memory span with age compared to wild-type worms, an effect that is dependent on the DAF-16 transcription factor. To identify possible mechanisms by which aging daf-2 mutants maintain learning and memory with age while wild-type worms lose neuronal function, we carried out neuron-specific transcriptomic analysis in aged animals. We observed downregulation of neuronal genes and upregulation of transcriptional regulation genes in aging wild-type neurons. By contrast, IIS/FOXO pathway mutants exhibit distinct neuronal transcriptomic alterations in response to cognitive aging, including upregulation of stress response genes and downregulation of specific insulin signaling genes. We tested the roles of significantly transcriptionally-changed genes in regulating cognitive functions, identifying novel regulators of learning and memory. In addition to other mechanistic insights, a comparison of the aged vs young daf-2 neuronal transcriptome revealed that a new set of potentially neuroprotective genes is upregulated; instead of simply mimicking a young state, daf-2 may enhance neuronal resilience to accumulation of harm and take a more active approach to combat aging. These findings suggest a potential mechanism for regulating cognitive function with age and offer insights into novel therapeutic targets for age-related cognitive decline.
Collapse
Affiliation(s)
- Yifei Weng
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| | - Shiyi Zhou
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| | - Katherine Morillo
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| | - Rachel Kaletsky
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
- Princeton UniversityPrincetonUnited States
| | - Sarah Lin
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| | - Coleen T Murphy
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
- Princeton UniversityPrincetonUnited States
| |
Collapse
|
19
|
Weng Y, Murphy CT. Male-specific behavioral and transcriptomic changes in aging C. elegans neurons. iScience 2024; 27:109910. [PMID: 38783998 PMCID: PMC11111838 DOI: 10.1016/j.isci.2024.109910] [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: 12/18/2023] [Revised: 03/20/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Aging is a complex biological process with sexually dimorphic aspects. Although cognitive aging of Caenorhabditis elegans hermaphrodites has been studied, less is known about cognitive decline in males. We found that cognitive aging has both sex-shared and sex-dimorphic characteristics, and we identified neuron-specific age-associated sex-differential targets. In addition to sex-shared neuronal aging genes, males differentially downregulate mitochondrial metabolic genes and upregulate GPCR genes with age, while the X chromosome exhibits increased gene expression in hermaphrodites and altered dosage compensation complex expression with age, indicating possible X chromosome dysregulation that contributes to sexual dimorphism in cognitive aging. Finally, the sex-differentially expressed gene hrg-7, an aspartic-type endopeptidase, regulates male cognitive aging but does not affect hermaphrodites' behaviors. These results suggest that males and hermaphrodites exhibit different age-related neuronal changes. This study will strengthen our understanding of sex-specific vulnerability and resilience and identify pathways to target with treatments that could benefit both sexes.
Collapse
Affiliation(s)
- Yifei Weng
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
- LSI Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Coleen T. Murphy
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
- LSI Genomics, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
20
|
Hayden AN, Brandel KL, Merlau PR, Vijayakumar P, Leptich EJ, Pietryk EW, Gaytan ES, Ni CW, Chao HT, Rosenfeld JA, Arey RN. Behavioral screening of conserved RNA-binding proteins reveals CEY-1/YBX RNA-binding protein dysfunction leads to impairments in memory and cognition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.05.574402. [PMID: 38260399 PMCID: PMC10802296 DOI: 10.1101/2024.01.05.574402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
RNA-binding proteins (RBPs) regulate translation and plasticity which are required for memory. RBP dysfunction has been linked to a range of neurological disorders where cognitive impairments are a key symptom. However, of the 2,000 RBPs in the human genome, many are uncharacterized with regards to neurological phenotypes. To address this, we used the model organism C. elegans to assess the role of 20 conserved RBPs in memory. We identified eight previously uncharacterized memory regulators, three of which are in the C. elegans Y-Box (CEY) RBP family. Of these, we determined that cey-1 is the closest ortholog to the mammalian Y-Box (YBX) RBPs. We found that CEY-1 is both necessary in the nervous system for memory ability and sufficient to increase memory. Leveraging human datasets, we found both copy number variation losses and single nucleotide variants in YBX1 and YBX3 in individuals with neurological symptoms. We identified one predicted deleterious YBX3 variant of unknown significance, p.Asn127Tyr, in two individuals with neurological symptoms. Introducing this variant into endogenous cey-1 locus caused memory deficits in the worm. We further generated two humanized worm lines expressing human YBX3 or YBX1 at the cey-1 locus to test evolutionary conservation of YBXs in memory and the potential functional significance of the p.Asn127Tyr variant. Both YBX1/3 can functionally replace cey-1, and introduction of p.Asn127Tyr into the humanized YBX3 locus caused memory deficits. Our study highlights the worm as a model to reveal memory regulators and identifies YBX dysfunction as a potential new source of rare neurological disease.
Collapse
Affiliation(s)
- Ashley N Hayden
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
| | - Katie L Brandel
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
| | - Paul R Merlau
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
| | | | - Emily J Leptich
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
| | - Edward W Pietryk
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030
| | - Elizabeth S Gaytan
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
- Postbaccalaureate Research Education Program, Baylor College of Medicine, Houston, TX, 77030
| | - Connie W Ni
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
- Department of Neuroscience, Rice University, Houston, TX 77005
| | - Hsiao-Tuan Chao
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030
- Department of Pediatrics, Division of Neurology and Developmental Neuroscience, Baylor College of Medicine, Houston, TX, 77030
- Cain Pediatric Neurology Research Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, 77030
- McNair Medical Institute, The Robert and Janice McNair Foundation, Houston, TX, 77030
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030
- Baylor Genetics Laboratories, Houston, TX 77021
| | - Rachel N Arey
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030
| |
Collapse
|
21
|
Lissek T. Aging as a Consequence of the Adaptation-Maladaptation Dilemma. Adv Biol (Weinh) 2024; 8:e2300654. [PMID: 38299389 DOI: 10.1002/adbi.202300654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/11/2024] [Indexed: 02/02/2024]
Abstract
In aging, the organism is unable to counteract certain harmful influences over its lifetime which leads to progressive dysfunction and eventually death, thus delineating aging as one failed process of adaptation to a set of aging stimuli. A central problem in understanding aging is hence to explain why the organism cannot adapt to these aging stimuli. The adaptation-maladaptation theory of aging proposes that in aging adaptation processes such as adaptive transcription, epigenetic remodeling, and metabolic plasticity drive dysfunction themselves over time (maladaptation) and thereby cause aging-related disorders such as cancer and metabolic dysregulation. The central dilemma of aging is thus that the set of adaptation mechanisms that the body uses to deal with internal and external stressors acts as a stressor itself and cannot be effectively counteracted. The only available option for the organism to decrease maladaptation may be a program to progressively reduce the output of adaptive cascades (e.g., via genomic methylation) which then leads to reduced physiological adaptation capacity and syndromes like frailty, immunosenescence, and cognitive decline. The adaptation-maladaptation dilemma of aging entails that certain biological mechanisms can simultaneously protect against aging as well as drive aging. The key to longevity may lie in uncoupling adaptation from maladaptation.
Collapse
Affiliation(s)
- Thomas Lissek
- Interdisciplinary Center for Neurosciences, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany
| |
Collapse
|
22
|
Ange JS, Weng Y, Stevenson ME, Kaletsky R, Moore RS, Zhou S, Murphy CT. Adult Single-nucleus Neuronal Transcriptomes of Insulin Signaling Mutants Reveal Regulators of Behavior and Learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.07.579364. [PMID: 38370779 PMCID: PMC10871314 DOI: 10.1101/2024.02.07.579364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
The insulin/insulin-like signaling (IIS) pathway regulates many of C. elegans' adult functions, including learning and memory 1 . While whole-worm and tissue-specific transcriptomic analyses have identified IIS targets 2,3 , a higher-resolution single-cell approach is required to identify changes that confer neuron-specific improvements in the long-lived insulin receptor mutant, daf-2 . To understand how behaviors that are controlled by a small number of neurons change in daf-2 mutants, we used the deep resolution of single-nucleus RNA sequencing to define each neuron type's transcriptome in adult wild-type and daf-2 mutants. First, we found surprising differences between wild-type L4 larval neurons and young adult neurons in chemoreceptor expression, synaptic genes, and learning and memory genes. These Day 1 adult neuron transcriptomes allowed us to identify adult AWC-specific regulators of chemosensory function and to predict neuron-to-neuron peptide/receptor pairs. We then identified gene expression changes that correlate with daf-2's improved cognitive functions, particularly in the AWC sensory neuron that controls learning and associative memory 4 , and used behavioral assays to test their roles in cognitive function. Combining deep single-neuron transcriptomics, genetic manipulation, and behavioral analyses enabled us to identify genes that may function in a single adult neuron to control behavior, including conserved genes that function in learning and memory. One-Sentence Summary Single-nucleus sequencing of adult wild-type and daf-2 C. elegans neurons reveals functionally relevant transcriptional changes, including regulators of chemosensation, learning, and memory.
Collapse
|
23
|
Fabrizio P, Alcolei A, Solari F. Considering Caenorhabditis elegans Aging on a Temporal and Tissue Scale: The Case of Insulin/IGF-1 Signaling. Cells 2024; 13:288. [PMID: 38334680 PMCID: PMC10854721 DOI: 10.3390/cells13030288] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/24/2024] [Accepted: 01/31/2024] [Indexed: 02/10/2024] Open
Abstract
The aging process is inherently complex, involving multiple mechanisms that interact at different biological scales. The nematode Caenorhabditis elegans is a simple model organism that has played a pivotal role in aging research following the discovery of mutations extending lifespan. Longevity pathways identified in C. elegans were subsequently found to be conserved and regulate lifespan in multiple species. These pathways intersect with fundamental hallmarks of aging that include nutrient sensing, epigenetic alterations, proteostasis loss, and mitochondrial dysfunction. Here we summarize recent data obtained in C. elegans highlighting the importance of studying aging at both the tissue and temporal scale. We then focus on the neuromuscular system to illustrate the kinetics of changes that take place with age. We describe recently developed tools that enabled the dissection of the contribution of the insulin/IGF-1 receptor ortholog DAF-2 to the regulation of worm mobility in specific tissues and at different ages. We also discuss guidelines and potential pitfalls in the use of these new tools. We further highlight the opportunities that they present, especially when combined with recent transcriptomic data, to address and resolve the inherent complexity of aging. Understanding how different aging processes interact within and between tissues at different life stages could ultimately suggest potential intervention points for age-related diseases.
Collapse
Affiliation(s)
- Paola Fabrizio
- Laboratoire de Biologie et Modélisation de la Cellule, Ecole Normale Supérieure de Lyon, CNRS UMR5239, INSERM 1210, University Claude Bernard Lyon 1, 69364 Lyon, France;
| | - Allan Alcolei
- INMG, MeLiS, CNRS UMR 5284, INSERM U1314, University Claude Bernard Lyon 1, 69008 Lyon, France;
| | - Florence Solari
- INMG, MeLiS, CNRS UMR 5284, INSERM U1314, University Claude Bernard Lyon 1, 69008 Lyon, France;
| |
Collapse
|
24
|
Bellver-Sanchis A, Geng Q, Navarro G, Ávila-López PA, Companys-Alemany J, Marsal-García L, Larramona-Arcas R, Miró L, Perez-Bosque A, Ortuño-Sahagún D, Banerjee DR, Choudhary BS, Soriano FX, Poulard C, Pallàs M, Du HN, Griñán-Ferré C. G9a Inhibition Promotes Neuroprotection through GMFB Regulation in Alzheimer's Disease. Aging Dis 2024; 15:311-337. [PMID: 37307824 PMCID: PMC10796087 DOI: 10.14336/ad.2023.0424-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 04/24/2023] [Indexed: 06/14/2023] Open
Abstract
Epigenetic alterations are a fundamental pathological hallmark of Alzheimer's disease (AD). Herein, we show the upregulation of G9a and H3K9me2 in the brains of AD patients. Interestingly, treatment with a G9a inhibitor (G9ai) in SAMP8 mice reversed the high levels of H3K9me2 and rescued cognitive decline. A transcriptional profile analysis after G9ai treatment revealed increased gene expression of glia maturation factor β (GMFB) in SAMP8 mice. Besides, a H3K9me2 ChIP-seq analysis after G9a inhibition treatment showed the enrichment of gene promoters associated with neural functions. We observed the induction of neuronal plasticity and a reduction of neuroinflammation after G9ai treatment, and more strikingly, these neuroprotective effects were reverted by the pharmacological inhibition of GMFB in mice and cell cultures; this was also validated by the RNAi approach generating the knockdown of GMFB/Y507A.10 in Caenorhabditis elegans. Importantly, we present evidence that GMFB activity is controlled by G9a-mediated lysine methylation as well as we identified that G9a directly bound GMFB and catalyzed the methylation at lysine (K) 20 and K25 in vitro. Furthermore, we found that the neurodegenerative role of G9a as a GMFB suppressor would mainly rely on methylation of the K25 position of GMFB, and thus G9a pharmacological inhibition removes this methylation promoting neuroprotective effects. Then, our findings confirm an undescribed mechanism by which G9a inhibition acts at two levels, increasing GMFB and regulating its function to promote neuroprotective effects in age-related cognitive decline.
Collapse
Affiliation(s)
- Aina Bellver-Sanchis
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Qizhi Geng
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, RNA Institute, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Gemma Navarro
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
- Department Biochemistry and Physiology, Faculty of Pharmacy. Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Pedro A. Ávila-López
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.
| | - Júlia Companys-Alemany
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Laura Marsal-García
- Department of Biochemistry, McGill University, Montréal, Québec, Canada.
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, Québec, Canada.
| | - Raquel Larramona-Arcas
- Department of Cell Biology, Physiology, and Immunology, Celltec-UB, University of Barcelona, Barcelona, Spain, and Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain.
| | - Lluisa Miró
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació and Institut de Nutrició i Seguretat Alimentària, Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Anna Perez-Bosque
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació and Institut de Nutrició i Seguretat Alimentària, Universitat de Barcelona, 08028 Barcelona, Spain.
| | - Daniel Ortuño-Sahagún
- Laboratorio de Neuroinmunología Molecular, Instituto de Investigación de Ciencias Biomédicas (IICB) CUCS, Universidad de Guadalajara, Jalisco 44340, México.
| | | | - Bhanwar Singh Choudhary
- Department of Pharmacy, Central University of Rajasthan, Ajmer, Rajasthan, India.
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Mehsana, Gujarat, India.
| | - Francesc X Soriano
- Department of Cell Biology, Physiology, and Immunology, Celltec-UB, University of Barcelona, Barcelona, Spain, and Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain.
| | - Coralie Poulard
- Cancer Research Cancer Lyon, Université de Lyon, F-69000 Lyon, France.
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, F-69000 Lyon, France.
- CNRS UMR5286, Centre de Recherche en Cancérlogie de Lyon, F-69000 Lyon, France.
| | - Mercè Pallàs
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, 08028 Barcelona, Spain.
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| | - Hai-Ning Du
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, RNA Institute, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Christian Griñán-Ferré
- Department of Pharmacology and Therapeutic Chemistry, Institut de Neurociències-Universitat de Barcelona, 08028 Barcelona, Spain.
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
25
|
Yang Y, Arnold ML, Lange CM, Sun LH, Broussalian M, Doroodian S, Ebata H, Choy EH, Poon K, Moreno TM, Singh A, Driscoll M, Kumsta C, Hansen M. Autophagy protein ATG-16.2 and its WD40 domain mediate the beneficial effects of inhibiting early-acting autophagy genes in C. elegans neurons. NATURE AGING 2024; 4:198-212. [PMID: 38177330 PMCID: PMC11022750 DOI: 10.1038/s43587-023-00548-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 11/27/2023] [Indexed: 01/06/2024]
Abstract
While autophagy genes are required for lifespan of long-lived animals, their tissue-specific roles in aging remain unclear. Here, we inhibited autophagy genes in Caenorhabditis elegans neurons, and found that knockdown of early-acting autophagy genes, except atg-16.2, increased lifespan, and decreased neuronal PolyQ aggregates, independently of autophagosomal degradation. Neurons can secrete protein aggregates via vesicles called exophers. Inhibiting neuronal early-acting autophagy genes, except atg-16.2, increased exopher formation and exopher events extended lifespan, suggesting exophers promote organismal fitness. Lifespan extension, reduction in PolyQ aggregates and increase in exophers were absent in atg-16.2 null mutants, and restored by full-length ATG-16.2 expression in neurons, but not by ATG-16.2 lacking its WD40 domain, which mediates noncanonical functions in mammalian systems. We discovered a neuronal role for C. elegans ATG-16.2 and its WD40 domain in lifespan, proteostasis and exopher biogenesis. Our findings suggest noncanonical functions for select autophagy genes in both exopher formation and in aging.
Collapse
Affiliation(s)
- Yongzhi Yang
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- Scripps Research Institute, La Jolla, CA, USA
| | - Meghan Lee Arnold
- Rutgers, The State University of New Jersey, Nelson Biological Labs, Piscataway, NJ, USA
| | - Caitlin M Lange
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Ling-Hsuan Sun
- Buck Institute for Aging Research, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | | | | | | | - Elizabeth H Choy
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Karie Poon
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Tatiana M Moreno
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Anupama Singh
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Monica Driscoll
- Rutgers, The State University of New Jersey, Nelson Biological Labs, Piscataway, NJ, USA
| | - Caroline Kumsta
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| | - Malene Hansen
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
- Buck Institute for Aging Research, Novato, CA, USA.
| |
Collapse
|
26
|
McMillen A, Chew Y. Neural mechanisms of dopamine function in learning and memory in Caenorhabditis elegans. Neuronal Signal 2024; 8:NS20230057. [PMID: 38572143 PMCID: PMC10987485 DOI: 10.1042/ns20230057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/03/2023] [Accepted: 12/11/2023] [Indexed: 04/05/2024] Open
Abstract
Research into learning and memory over the past decades has revealed key neurotransmitters that regulate these processes, many of which are evolutionarily conserved across diverse species. The monoamine neurotransmitter dopamine is one example of this, with countless studies demonstrating its importance in regulating behavioural plasticity. However, dopaminergic neural networks in the mammalian brain consist of hundreds or thousands of neurons, and thus cannot be studied at the level of single neurons acting within defined neural circuits. The nematode Caenorhabditis elegans (C. elegans) has an experimentally tractable nervous system with a completely characterized synaptic connectome. This makes it an advantageous system to undertake mechanistic studies into how dopamine encodes lasting yet flexible behavioural plasticity in the nervous system. In this review, we synthesize the research to date exploring the importance of dopaminergic signalling in learning, memory formation, and forgetting, focusing on research in C. elegans. We also explore the potential for dopamine-specific fluorescent biosensors in C. elegans to visualize dopaminergic neural circuits during learning and memory formation in real-time. We propose that the use of these sensors in C. elegans, in combination with optogenetic and other light-based approaches, will further illuminate the detailed spatiotemporal requirements for encoding behavioural plasticity in an accessible experimental system. Understanding the key molecules and circuit mechanisms that regulate learning and forgetting in more compact invertebrate nervous systems may reveal new druggable targets for enhancing memory storage and delaying memory loss in bigger brains.
Collapse
Affiliation(s)
- Anna McMillen
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, 5042, South Australia, Australia
| | - Yee Lian Chew
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, 5042, South Australia, Australia
| |
Collapse
|
27
|
Kanakalatha RS, Thekkuveettil A. Insulin signaling in dopaminergic neurons regulates extended memory formation in Caenorhabditis elegans. J Neurosci Res 2024; 102:e25260. [PMID: 38284856 DOI: 10.1002/jnr.25260] [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: 05/20/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 01/30/2024]
Abstract
Insulin alters several brain functions, and perturbations in insulin levels could be a precipitating factor for Parkinson's disease, a disease associated with the degeneration of dopaminergic neurons. It is unclear whether insulin alters the dopamine signaling pathway and modulates learning and memory. In Caenorhabditis elegans, daf-2 insulin receptor mutants have extended memory when trained for olfactory adaptation. In this study, we show that the absence of daf-2 receptors in dopamine neurons results in this unusual learning behavior. Our results show that insulin function in memory is dopamine-dependent. In the absence of the daf-2 receptor, the calcium influx in dopamine neurons shows an altered pattern resulting in memory recall for an extended period. These results indicate that learning and memory involve insulin-dopamine crosstalk. Imbalances in this pathway result in changes in memory recall.
Collapse
Affiliation(s)
- Rasitha Santhosh Kanakalatha
- Division of Molecular Medicine, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, India
| | - Anoopkumar Thekkuveettil
- Division of Molecular Medicine, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, India
| |
Collapse
|
28
|
Lemieux GA, Yoo S, Lin L, Vohra M, Ashrafi K. The steroid hormone ADIOL promotes learning by reducing neural kynurenic acid levels. Genes Dev 2023; 37:998-1016. [PMID: 38092521 PMCID: PMC10760639 DOI: 10.1101/gad.350745.123] [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: 04/24/2023] [Accepted: 11/22/2023] [Indexed: 12/28/2023]
Abstract
Reductions in brain kynurenic acid levels, a neuroinhibitory metabolite, improve cognitive function in diverse organisms. Thus, modulation of kynurenic acid levels is thought to have therapeutic potential in a range of brain disorders. Here we report that the steroid 5-androstene 3β, 17β-diol (ADIOL) reduces kynurenic acid levels and promotes associative learning in Caenorhabditis elegans We identify the molecular mechanisms through which ADIOL links peripheral metabolic pathways to neural mechanisms of learning capacity. Moreover, we show that in aged animals, which normally experience rapid cognitive decline, ADIOL improves learning capacity. The molecular mechanisms that underlie the biosynthesis of ADIOL as well as those through which it promotes kynurenic acid reduction are conserved in mammals. Thus, rather than a minor intermediate in the production of sex steroids, ADIOL is an endogenous hormone that potently regulates learning capacity by causing reductions in neural kynurenic acid levels.
Collapse
Affiliation(s)
- George A Lemieux
- Department of Physiology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Shinja Yoo
- Department of Physiology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Lin Lin
- Department of Physiology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Mihir Vohra
- Department of Physiology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Kaveh Ashrafi
- Department of Physiology, University of California, San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
29
|
Kirchweger B, Zwirchmayr J, Grienke U, Rollinger JM. The role of Caenorhabditis elegans in the discovery of natural products for healthy aging. Nat Prod Rep 2023; 40:1849-1873. [PMID: 37585263 DOI: 10.1039/d3np00021d] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Covering: 2012 to 2023The human population is aging. Thus, the greatest risk factor for numerous diseases, such as diabetes, cancer and neurodegenerative disorders, is increasing worldwide. Age-related diseases do not typically occur in isolation, but as a result of multi-factorial causes, which in turn require holistic approaches to identify and decipher the mode of action of potential remedies. With the advent of C. elegans as the primary model organism for aging, researchers now have a powerful in vivo tool for identifying and studying agents that effect lifespan and health span. Natural products have been focal research subjects in this respect. This review article covers key developments of the last decade (2012-2023) that have led to the discovery of natural products with healthy aging properties in C. elegans. We (i) discuss the state of knowledge on the effects of natural products on worm aging including methods, assays and involved pathways; (ii) analyze the literature on natural compounds in terms of their molecular properties and the translatability of effects on mammals; (iii) examine the literature on multi-component mixtures with special attention to the studied organisms, extraction methods and efforts regarding the characterization of their chemical composition and their bioactive components. (iv) We further propose to combine small in vivo model organisms such as C. elegans and sophisticated analytical approaches ("wormomics") to guide the way to dissect complex natural products with anti-aging properties.
Collapse
Affiliation(s)
- Benjamin Kirchweger
- Division of Pharmacognosy, Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
| | - Julia Zwirchmayr
- Division of Pharmacognosy, Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
| | - Ulrike Grienke
- Division of Pharmacognosy, Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
| | - Judith M Rollinger
- Division of Pharmacognosy, Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
| |
Collapse
|
30
|
Stevenson ME, Bieri G, Kaletsky R, St Ange J, Remesal L, Pratt KJB, Zhou S, Weng Y, Murphy CT, Villeda SA. Neuronal activation of G αq EGL-30/GNAQ late in life rejuvenates cognition across species. Cell Rep 2023; 42:113151. [PMID: 37713310 PMCID: PMC10627507 DOI: 10.1016/j.celrep.2023.113151] [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: 06/16/2023] [Revised: 08/10/2023] [Accepted: 08/31/2023] [Indexed: 09/17/2023] Open
Abstract
Loss of cognitive function with age is devastating. EGL-30/GNAQ and Gαq signaling pathways are highly conserved between C. elegans and mammals, and murine Gnaq is enriched in hippocampal neurons and declines with age. We found that activation of EGL-30 in aged worms triples memory span, and GNAQ gain of function significantly improved memory in aged mice: GNAQ(gf) in hippocampal neurons of 24-month-old mice (equivalent to 70- to 80-year-old humans) rescued age-related impairments in well-being and memory. Single-nucleus RNA sequencing revealed increased expression of genes regulating synaptic function, axon guidance, and memory in GNAQ-treated mice, and worm orthologs of these genes were required for long-term memory extension in worms. These experiments demonstrate that C. elegans is a powerful model to identify mammalian regulators of memory, leading to the identification of a pathway that improves memory in extremely old mice. To our knowledge, this is the oldest age at which an intervention has improved age-related cognitive decline.
Collapse
Affiliation(s)
- Morgan E Stevenson
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Gregor Bieri
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA; Bakar Aging Research Institute, San Francisco, CA 94143, USA
| | - Rachel Kaletsky
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Jonathan St Ange
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - L Remesal
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA; Bakar Aging Research Institute, San Francisco, CA 94143, USA
| | - Karishma J B Pratt
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA; Bakar Aging Research Institute, San Francisco, CA 94143, USA
| | - Shiyi Zhou
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Yifei Weng
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Coleen T Murphy
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA.
| | - Saul A Villeda
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA; Bakar Aging Research Institute, San Francisco, CA 94143, USA.
| |
Collapse
|
31
|
Tang LTH, Lee GA, Cook SJ, Ho J, Potter CC, Bülow HE. Anatomical restructuring of a lateralized neural circuit during associative learning by asymmetric insulin signaling. Curr Biol 2023; 33:3835-3850.e6. [PMID: 37591249 PMCID: PMC10639090 DOI: 10.1016/j.cub.2023.07.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 08/19/2023]
Abstract
Studies of neuronal connectivity in model organisms, i.e., of their connectomes, have been instrumental in dissecting the structure-function relationship of nervous systems. However, the limited sample size of these studies has impeded analyses into how variation of connectivity across populations may influence circuit architecture and behavior. Moreover, little is known about how experiences induce changes in circuit architecture. Here, we show that an asymmetric salt-sensing circuit in the nematode Caenorhabditis elegans exhibits variation that predicts the animals' salt preferences and undergoes restructuring during salt associative learning. Naive worms memorize and prefer the salt concentration they experience in the presence of food through a left-biased neural network architecture. However, animals conditioned at elevated salt concentrations change this left-biased network to a right-biased network. This change in circuit architecture occurs through the addition of new synapses in response to asymmetric, paracrine insulin signaling. Therefore, experience-dependent changes in an animal's neural connectome are induced by insulin signaling and are fundamental to learning and behavior.
Collapse
Affiliation(s)
- Leo T H Tang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Garrett A Lee
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Steven J Cook
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jacquelin Ho
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Cassandra C Potter
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Hannes E Bülow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
32
|
Mangalath A, Thekkuveettil A. Olfactory imprinting enhances associative learning and memory in C. elegans. Biochem Biophys Res Commun 2023; 674:109-116. [PMID: 37419031 DOI: 10.1016/j.bbrc.2023.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/01/2023] [Indexed: 07/09/2023]
Abstract
Learning and memory are fundamental processes for an organism's normal physiological function. Learning can occur at any stage of the organism's physiological development. Imprinted memories formed during the early developmental stage, unlike learning and memory, can last a lifetime. It is not clear whether these two types of memories are interlinked. In this study, we investigated whether imprinted memory influences adult learning and memory in a C. elegans model system. We trained the worms for short-term (STAM) and long-term associated memory (LTAM) towards butanone (BT) after conditioning them for imprinted memory towards isoamyl alcohol (IAA). We observed that these worms had improved learning abilities. However, functional imaging revealed that the worms had a long-term depression in the firing pattern in the AIY interneuron, indicating that there were significant changes in neuronal excitation pattern after imprinting, which could explain the enhanced behavioural alterations in animals after imprinting.
Collapse
Affiliation(s)
- Aswathy Mangalath
- Division of Molecular Medicine, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, 695012, India
| | - Anoopkumar Thekkuveettil
- Division of Molecular Medicine, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, 695012, India.
| |
Collapse
|
33
|
Tomioka M, Umemura Y, Ueoka Y, Chin R, Katae K, Uchiyama C, Ike Y, Iino Y. Antagonistic regulation of salt and sugar chemotaxis plasticity by a single chemosensory neuron in Caenorhabditis elegans. PLoS Genet 2023; 19:e1010637. [PMID: 37669262 PMCID: PMC10503759 DOI: 10.1371/journal.pgen.1010637] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 09/15/2023] [Accepted: 08/20/2023] [Indexed: 09/07/2023] Open
Abstract
The nematode Caenorhabditis elegans memorizes various external chemicals, such as ions and odorants, during feeding. Here we find that C. elegans is attracted to the monosaccharides glucose and fructose after exposure to these monosaccharides in the presence of food; however, it avoids them without conditioning. The attraction to glucose requires a gustatory neuron called ASEL. ASEL activity increases when glucose concentration decreases. Optogenetic ASEL stimulation promotes forward movements; however, after glucose conditioning, it promotes turning, suggesting that after glucose conditioning, the behavioral output of ASEL activation switches toward glucose. We previously reported that chemotaxis toward sodium ion (Na+), which is sensed by ASEL, increases after Na+ conditioning in the presence of food. Interestingly, glucose conditioning decreases Na+ chemotaxis, and conversely, Na+ conditioning decreases glucose chemotaxis, suggesting the reciprocal inhibition of learned chemotaxis to distinct chemicals. The activation of PKC-1, an nPKC ε/η ortholog, in ASEL promotes glucose chemotaxis and decreases Na+ chemotaxis after glucose conditioning. Furthermore, genetic screening identified ENSA-1, an ortholog of the protein phosphatase inhibitor ARPP-16/19, which functions in parallel with PKC-1 in glucose-induced chemotactic learning toward distinct chemicals. These findings suggest that kinase-phosphatase signaling regulates the balance between learned behaviors based on glucose conditioning in ASEL, which might contribute to migration toward chemical compositions where the animals were previously fed.
Collapse
Affiliation(s)
- Masahiro Tomioka
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Yusuke Umemura
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Yutaro Ueoka
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Risshun Chin
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Keita Katae
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Chihiro Uchiyama
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Yasuaki Ike
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Yuichi Iino
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
34
|
Chandra R, Farah F, Muñoz-Lobato F, Bokka A, Benedetti KL, Brueggemann C, Saifuddin MFA, Miller JM, Li J, Chang E, Varshney A, Jimenez V, Baradwaj A, Nassif C, Alladin S, Andersen K, Garcia AJ, Bi V, Nordquist SK, Dunn RL, Garcia V, Tokalenko K, Soohoo E, Briseno F, Kaur S, Harris M, Guillen H, Byrd D, Fung B, Bykov AE, Odisho E, Tsujimoto B, Tran A, Duong A, Daigle KC, Paisner R, Zuazo CE, Lin C, Asundi A, Churgin MA, Fang-Yen C, Bremer M, Kato S, VanHoven MK, L'Étoile ND. Sleep is required to consolidate odor memory and remodel olfactory synapses. Cell 2023; 186:2911-2928.e20. [PMID: 37269832 PMCID: PMC10354834 DOI: 10.1016/j.cell.2023.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 02/02/2023] [Accepted: 05/05/2023] [Indexed: 06/05/2023]
Abstract
Animals with complex nervous systems demand sleep for memory consolidation and synaptic remodeling. Here, we show that, although the Caenorhabditis elegans nervous system has a limited number of neurons, sleep is necessary for both processes. In addition, it is unclear if, in any system, sleep collaborates with experience to alter synapses between specific neurons and whether this ultimately affects behavior. C. elegans neurons have defined connections and well-described contributions to behavior. We show that spaced odor-training and post-training sleep induce long-term memory. Memory consolidation, but not acquisition, requires a pair of interneurons, the AIYs, which play a role in odor-seeking behavior. In worms that consolidate memory, both sleep and odor conditioning are required to diminish inhibitory synaptic connections between the AWC chemosensory neurons and the AIYs. Thus, we demonstrate in a living organism that sleep is required for events immediately after training that drive memory consolidation and alter synaptic structures.
Collapse
Affiliation(s)
- Rashmi Chandra
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Fatima Farah
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Fernando Muñoz-Lobato
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Anirudh Bokka
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Kelli L Benedetti
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Chantal Brueggemann
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mashel Fatema A Saifuddin
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Julia M Miller
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Joy Li
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Eric Chang
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Aruna Varshney
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Vanessa Jimenez
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Anjana Baradwaj
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Cibelle Nassif
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Sara Alladin
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Kristine Andersen
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Angel J Garcia
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Veronica Bi
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Sarah K Nordquist
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Raymond L Dunn
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Vanessa Garcia
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Kateryna Tokalenko
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Emily Soohoo
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Fabiola Briseno
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Sukhdeep Kaur
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Malcolm Harris
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Hazel Guillen
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Decklin Byrd
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Brandon Fung
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Andrew E Bykov
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Emma Odisho
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Bryan Tsujimoto
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Alan Tran
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Alex Duong
- Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Kevin C Daigle
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Rebekka Paisner
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Carlos E Zuazo
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Christine Lin
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Aarati Asundi
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthew A Churgin
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher Fang-Yen
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Martina Bremer
- Department of Mathematics and Statistics, San José State University, San José, CA 95192, USA
| | - Saul Kato
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Miri K VanHoven
- Department of Biological Sciences, San José State University, San José, CA 95192, USA.
| | - Noëlle D L'Étoile
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
35
|
Lin TA, How CM, Yen PL, Liao VHC. Sulfate-modified nanosized polystyrene impairs memory by inhibiting ionotropic glutamate receptors and the cAMP-response element binding protein (CREB) pathway in Caenorhabditis elegans. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 875:162404. [PMID: 36868277 DOI: 10.1016/j.scitotenv.2023.162404] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/16/2023] [Accepted: 02/18/2023] [Indexed: 06/18/2023]
Abstract
Nanoplastic contamination is an emerging environmental concern worldwide. In particular, sulfate anionic surfactants often appear along with nanosized plastic particles in personal care products, suggesting that sulfate-modified nanosized polystyrene (S-NP) may occur, remain, and spread into the environment. However, whether S-NP adversely affects learning and memory is unknown. In this study, we used a positive butanone training protocol to evaluate the effects of S-NP exposure on short-term associative memory (STAM) and long-term associative memory (LTAM) in Caenorhabditis elegans. We observed that long-term S-NP exposure impairs both STAM and LTAM in C. elegans. We also observed that mutations in the glr-1, nmr-1, acy-1, unc-43, and crh-1 genes eliminated the STAM and LTAM impairment induced by S-NP, and the mRNA levels of these genes were also decreased upon S-NP exposure. These genes encode ionotropic glutamate receptors (iGluRs), cyclic adenosine monophosphate (cAMP)/Ca2+ signaling proteins, and cAMP-response element binding protein (CREB)/CRH-1 signaling proteins. Moreover, S-NP exposure inhibited the expression of the CREB-dependent LTAM genes nid-1, ptr-15, and unc-86. Our findings provide new insights into long-term S-NP exposure and the impairment of STAM and LTAM, which involve the highly conserved iGluRs and CRH-1/CREB signaling pathways.
Collapse
Affiliation(s)
- Ting-An Lin
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei 106, Taiwan
| | - Chun Ming How
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei 106, Taiwan
| | - Pei-Ling Yen
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei 106, Taiwan
| | - Vivian Hsiu-Chuan Liao
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei 106, Taiwan.
| |
Collapse
|
36
|
Higurashi S, Tsukada S, Aleogho BM, Park JH, Al-Hebri Y, Tanaka M, Nakano S, Mori I, Noma K. Bacterial diet affects the age-dependent decline of associative learning in Caenorhabditis elegans. eLife 2023; 12:81418. [PMID: 37252859 DOI: 10.7554/elife.81418] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 04/27/2023] [Indexed: 06/01/2023] Open
Abstract
The causality and mechanism of dietary effects on brain aging are still unclear due to the long time scales of aging. The nematode Caenorhabditis elegans has contributed to aging research because of its short lifespan and easy genetic manipulation. When fed the standard laboratory diet, Escherichia coli, C. elegans experiences an age-dependent decline in temperature-food associative learning, called thermotaxis. To address if diet affects this decline, we screened 35 lactic acid bacteria as alternative diet and found that animals maintained high thermotaxis ability when fed a clade of Lactobacilli enriched with heterofermentative bacteria. Among them, Lactobacillus reuteri maintained the thermotaxis of aged animals without affecting their lifespan and motility. The effect of Lb. reuteri depends on the DAF-16 transcription factor functioning in neurons. Furthermore, RNA sequencing analysis revealed that differentially expressed genes between aged animals fed different bacteria were enriched with DAF-16 targets. Our results demonstrate that diet can impact brain aging in a daf-16-dependent manner without changing the lifespan.
Collapse
Affiliation(s)
- Satoshi Higurashi
- Milk Science Research Institute, Megmilk Snow Brand Co. Ltd., Saitama, Japan
- Group of Nutritional Neuroscience, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Sachio Tsukada
- Milk Science Research Institute, Megmilk Snow Brand Co. Ltd., Saitama, Japan
- Group of Nutritional Neuroscience, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Binta Maria Aleogho
- Group of Nutritional Neuroscience, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
- Group of Molecular Neurobiology, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
- Group of Microbial Motility, Department of Biological Science, Division of Natural Science, Graduate school of Science, Nagoya University, Nagoya, Japan
| | - Joo Hyun Park
- Group of Nutritional Neuroscience, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Yana Al-Hebri
- Group of Nutritional Neuroscience, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Masaru Tanaka
- Milk Science Research Institute, Megmilk Snow Brand Co. Ltd., Saitama, Japan
- Group of Nutritional Neuroscience, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Shunji Nakano
- Group of Molecular Neurobiology, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Ikue Mori
- Group of Molecular Neurobiology, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Kentaro Noma
- Group of Nutritional Neuroscience, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
- Group of Molecular Neurobiology, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
- Group of Microbial Motility, Department of Biological Science, Division of Natural Science, Graduate school of Science, Nagoya University, Nagoya, Japan
| |
Collapse
|
37
|
Lev I, Zimmer M. When neurons split the load. eLife 2023; 12:87861. [PMID: 37140564 PMCID: PMC10159616 DOI: 10.7554/elife.87861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023] Open
Abstract
Various aspects of olfactory memory are represented as modulated responses across different classes of neurons in C. elegans.
Collapse
Affiliation(s)
- Itamar Lev
- Department of Neuroscience and Developmental Biology, Vienna Biocenter, University of Vienna, Vienna, Austria
| | - Manuel Zimmer
- Department of Neuroscience and Developmental Biology, Vienna Biocenter, University of Vienna, Vienna, Austria
- Research Institute of Molecular Pathology, Vienna Biocenter, Vienna, Austria
| |
Collapse
|
38
|
Pritz C, Itskovits E, Bokman E, Ruach R, Gritsenko V, Nelken T, Menasherof M, Azulay A, Zaslaver A. Principles for coding associative memories in a compact neural network. eLife 2023; 12:e74434. [PMID: 37140557 PMCID: PMC10159626 DOI: 10.7554/elife.74434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 03/08/2023] [Indexed: 05/05/2023] Open
Abstract
A major goal in neuroscience is to elucidate the principles by which memories are stored in a neural network. Here, we have systematically studied how four types of associative memories (short- and long-term memories, each as positive and negative associations) are encoded within the compact neural network of Caenorhabditis elegans worms. Interestingly, sensory neurons were primarily involved in coding short-term, but not long-term, memories, and individual sensory neurons could be assigned to coding either the conditioned stimulus or the experience valence (or both). Moreover, when considering the collective activity of the sensory neurons, the specific training experiences could be decoded. Interneurons integrated the modulated sensory inputs and a simple linear combination model identified the experience-specific modulated communication routes. The widely distributed memory suggests that integrated network plasticity, rather than changes to individual neurons, underlies the fine behavioral plasticity. This comprehensive study reveals basic memory-coding principles and highlights the central roles of sensory neurons in memory formation.
Collapse
Affiliation(s)
- Christian Pritz
- Department of Genetics, Silberman Institute for Life Sciences, Edmond J. Safra Campus, The Hebrew University of JerusalemJerusalemIsrael
| | - Eyal Itskovits
- Department of Genetics, Silberman Institute for Life Sciences, Edmond J. Safra Campus, The Hebrew University of JerusalemJerusalemIsrael
| | - Eduard Bokman
- Department of Genetics, Silberman Institute for Life Sciences, Edmond J. Safra Campus, The Hebrew University of JerusalemJerusalemIsrael
| | - Rotem Ruach
- Department of Genetics, Silberman Institute for Life Sciences, Edmond J. Safra Campus, The Hebrew University of JerusalemJerusalemIsrael
| | - Vladimir Gritsenko
- Department of Genetics, Silberman Institute for Life Sciences, Edmond J. Safra Campus, The Hebrew University of JerusalemJerusalemIsrael
| | - Tal Nelken
- Department of Genetics, Silberman Institute for Life Sciences, Edmond J. Safra Campus, The Hebrew University of JerusalemJerusalemIsrael
| | - Mai Menasherof
- Department of Genetics, Silberman Institute for Life Sciences, Edmond J. Safra Campus, The Hebrew University of JerusalemJerusalemIsrael
| | - Aharon Azulay
- Department of Genetics, Silberman Institute for Life Sciences, Edmond J. Safra Campus, The Hebrew University of JerusalemJerusalemIsrael
| | - Alon Zaslaver
- Department of Genetics, Silberman Institute for Life Sciences, Edmond J. Safra Campus, The Hebrew University of JerusalemJerusalemIsrael
| |
Collapse
|
39
|
Brandel-Ankrapp KL, Arey RN. Uncovering novel regulators of memory using C. elegans genetic and genomic analysis. Biochem Soc Trans 2023; 51:161-171. [PMID: 36744642 PMCID: PMC10518207 DOI: 10.1042/bst20220455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/20/2022] [Accepted: 01/19/2023] [Indexed: 02/07/2023]
Abstract
How organisms learn and encode memory is an outstanding question in neuroscience research. Specifically, how memories are acquired and consolidated at the level of molecular and gene pathways remains unclear. In addition, memory is disrupted in a wide variety of neurological disorders; therefore, discovering molecular regulators of memory may reveal therapeutic targets for these disorders. C. elegans are an excellent model to uncover molecular and genetic regulators of memory. Indeed, the nematode's invariant neuronal lineage, fully mapped genome, and conserved associative behaviors have allowed the development of a breadth of genetic and genomic tools to examine learning and memory. In this mini-review, we discuss novel and exciting genetic and genomic techniques used to examine molecular and genetic underpinnings of memory from the level of the whole-worm to tissue-specific and cell-type specific approaches with high spatiotemporal resolution.
Collapse
Affiliation(s)
- Katie L. Brandel-Ankrapp
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, U.S.A
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX 77030, U.S.A
| | - Rachel N. Arey
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX 77030, U.S.A
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, U.S.A
| |
Collapse
|
40
|
Tang LTH, Lee GA, Cook SJ, Ho J, Potter CC, Bülow HE. Restructuring of an asymmetric neural circuit during associative learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.12.523604. [PMID: 36711870 PMCID: PMC9882173 DOI: 10.1101/2023.01.12.523604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Asymmetric brain function is common across the animal kingdom and involved in language processing, and likely in learning and memory. What regulates asymmetric brain function remains elusive. Here, we show that the nematode Caenorhabditis elegans restructures an asymmetric salt sensing neural circuit during associative learning. Worms memorize and prefer the salt concentration at which they were raised in the presence of food through a left-biased network architecture. When conditioned at elevated salt concentrations, animals change the left-biased to a right-biased network, which explains the changed salt-seeking behavior. The changes in circuit architecture require new synapse formation induced through asymmetric, paracrine insulin-signaling. Therefore, experience-dependent changes in asymmetric network architecture rely on paracrine insulin signaling and are fundamental to learning and behavior.
Collapse
|
41
|
Targeting the "hallmarks of aging" to slow aging and treat age-related disease: fact or fiction? Mol Psychiatry 2023; 28:242-255. [PMID: 35840801 PMCID: PMC9812785 DOI: 10.1038/s41380-022-01680-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/20/2022] [Accepted: 06/27/2022] [Indexed: 01/09/2023]
Abstract
Aging is a major risk factor for a number of chronic diseases, including neurodegenerative and cerebrovascular disorders. Aging processes have therefore been discussed as potential targets for the development of novel and broadly effective preventatives or therapeutics for age-related diseases, including those affecting the brain. Mechanisms thought to contribute to aging have been summarized under the term the "hallmarks of aging" and include a loss of proteostasis, mitochondrial dysfunction, altered nutrient sensing, telomere attrition, genomic instability, cellular senescence, stem cell exhaustion, epigenetic alterations and altered intercellular communication. We here examine key claims about the "hallmarks of aging". Our analysis reveals important weaknesses that preclude strong and definitive conclusions concerning a possible role of these processes in shaping organismal aging rate. Significant ambiguity arises from the overreliance on lifespan as a proxy marker for aging, the use of models with unclear relevance for organismal aging, and the use of study designs that do not allow to properly estimate intervention effects on aging rate. We also discuss future research directions that should be taken to clarify if and to what extent putative aging regulators do in fact interact with aging. These include multidimensional analytical frameworks as well as designs that facilitate the proper assessment of intervention effects on aging rate.
Collapse
|
42
|
(-)- Gossypol Inhibition of Musashi-Mediated Forgetting Improves Memory and Age-Dependent Memory Decline in Caenorhabditis elegans. Mol Neurobiol 2023; 60:820-835. [PMID: 36378468 PMCID: PMC9849318 DOI: 10.1007/s12035-022-03116-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/03/2022] [Indexed: 11/17/2022]
Abstract
Musashi RNA-binding proteins (MSIs) retain a pivotal role in stem cell maintenance, tumorigenesis, and nervous system development. Recently, we showed in C. elegans that Musashi (MSI-1) actively promotes forgetting upon associative learning via a 3'UTR-dependent translational expression of the Arp2/3 actin branching complex. Here, we investigated the evolutionary conserved role of MSI proteins and the effect of their pharmacological inhibition on memory. Expression of human Musashi 1 (MSI1) and Musashi 2 (MSI2) under the endogenous Musashi promoter fully rescued the phenotype of msi-1(lf) worms. Furthermore, pharmacological inhibition of human MSI1 and MSI2 activity using (-)- gossypol resulted in improved memory retention, without causing locomotor, chemotactic, or learning deficits. No drug effect was observed in msi-1(lf) treated worms. Using Western blotting and confocal microscopy, we found no changes in MSI-1 protein abundance following (-)- gossypol treatment, suggesting that Musashi gene expression remains unaltered and that the compound exerts its inhibitory effect post-translationally. Additionally, (-)- gossypol suppressed the previously seen rescue of the msi-1(lf) phenotype in worms expressing human MSI1 specifically in the AVA neuron, indicating that (-)- gossypol can regulate the Musashi pathway in a memory-related neuronal circuit in worms. Finally, treating aged worms with (-)- gossypol reversed physiological age-dependent memory decline. Taken together, our findings indicate that pharmacological inhibition of Musashi might represent a promising approach for memory modulation.
Collapse
|
43
|
Aminzadeh-Gohari S, Kofler B, Herzog C. Dietary restriction in senolysis and prevention and treatment of disease. Crit Rev Food Sci Nutr 2022; 64:5242-5268. [PMID: 36484738 PMCID: PMC7616065 DOI: 10.1080/10408398.2022.2153355] [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] [Indexed: 12/13/2022]
Abstract
Aging represents a key risk factor for a plethora of diseases. Targeting detrimental processes which occur during aging, especially before onset of age-related disease, could provide drastic improvements in healthspan. There is increasing evidence that dietary restriction (DR), including caloric restriction, fasting, or fasting-mimicking diets, extend both lifespan and healthspan. This has sparked interest in the use of dietary regimens as a non-pharmacological means to slow aging and prevent disease. Here, we review the current evidence on the molecular mechanisms underlying DR-induced health improvements, including removal of senescent cells, metabolic reprogramming, and epigenetic rejuvenation.
Collapse
Affiliation(s)
- Sepideh Aminzadeh-Gohari
- Research Program for Receptor Biochemistry and Tumor Metabollism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria
- Research Institute for Biomedical Ageing, Universität Innsbruck, Innsbruck, Austria
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumor Metabollism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Chiara Herzog
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria
- Research Institute for Biomedical Ageing, Universität Innsbruck, Innsbruck, Austria
| |
Collapse
|
44
|
Dembitskaya Y, Piette C, Perez S, Berry H, Magistretti PJ, Venance L. Lactate supply overtakes glucose when neural computational and cognitive loads scale up. Proc Natl Acad Sci U S A 2022; 119:e2212004119. [PMID: 36375086 PMCID: PMC9704697 DOI: 10.1073/pnas.2212004119] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/10/2022] [Indexed: 07/23/2023] Open
Abstract
Neural computational power is determined by neuroenergetics, but how and which energy substrates are allocated to various forms of memory engram is unclear. To solve this question, we asked whether neuronal fueling by glucose or lactate scales differently upon increasing neural computation and cognitive loads. Here, using electrophysiology, two-photon imaging, cognitive tasks, and mathematical modeling, we show that both glucose and lactate are involved in engram formation, with lactate supporting long-term synaptic plasticity evoked by high-stimulation load activity patterns and high attentional load in cognitive tasks and glucose being sufficient for less demanding neural computation and learning tasks. Indeed, we show that lactate is mandatory for demanding neural computation, such as theta-burst stimulation, while glucose is sufficient for lighter forms of activity-dependent long-term potentiation (LTP), such as spike timing-dependent plasticity (STDP). We find that subtle variations of spike number or frequency in STDP are sufficient to shift the on-demand fueling from glucose to lactate. Finally, we demonstrate that lactate is necessary for a cognitive task requiring high attentional load, such as the object-in-place task, and for the corresponding in vivo hippocampal LTP expression but is not needed for a less demanding task, such as a simple novel object recognition. Overall, these results demonstrate that glucose and lactate metabolism are differentially engaged in neuronal fueling depending on the complexity of the activity-dependent plasticity and behavior.
Collapse
Affiliation(s)
- Yulia Dembitskaya
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Charlotte Piette
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Sylvie Perez
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Hugues Berry
- AIStroSight Lab, INRIA, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, 69603 Villeurbanne, France
- University of Lyon, LIRIS UMR5205, 69622 Villeurbanne, France
| | - Pierre J. Magistretti
- Biological and Environmental Sciences and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Saudi Arabia
- Brain Mind Institute, EPFL, 1015 Lausanne, Switzerland
| | - Laurent Venance
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, 75005 Paris, France
| |
Collapse
|
45
|
Cho I, Song HO, Ji HE, Yang S, Cho JH. BAM15 Relieves Neurodegeneration in Aged Caenorhabditis elegans and Extends Lifespan. Metabolites 2022; 12:1129. [PMID: 36422268 PMCID: PMC9698188 DOI: 10.3390/metabo12111129] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 09/01/2023] Open
Abstract
BAM15 was recently screened as a protonophore uncoupler specifically for the mitochondrial membrane but not the plasma membrane. It is equally as potent as FCCP, but less toxic. Previously, mitochondrial uncoupling via DNP alleviates neurodegeneration in the nematode Caenorhabditis elegans during aging. Therefore, we investigated whether BAM15 uncouplers could phenotypically and functionally reduce neuronal defects in aged nematodes. We observed green fluorescence protein-tagged mechanosensory neurons and performed touch and chemotaxis assays during aging. Wild-type animals treated with both 50 µM BAM15 and 10 µM DNP showed reduced mechanosensory neuronal defects during aging, which correlates with the maintenance of touch responses and short-term memory during aging. Uncoupler mutant ucp-4 also responded the same way as the wild-type, reducing neurodegeneration in 50 µM BAM15 and 10 µM DNP-treated animals compared to the DMSO control. These results suggest that 50 µM BAM15 alleviates neurodegeneration phenotypically and functionally in C. elegans during aging, potentially through mitochondrial uncoupling. In accordance with the preserved neuronal shape and function in aged C. elegans, 50 µM BAM15 extended the mean lifespan of both wild-type and ucp-4 mutants.
Collapse
Affiliation(s)
- Injeong Cho
- Department of Biology Education, College of Education, Chosun University, Gwangju 61452, Republic of Korea
| | - Hyun-Ok Song
- Department of Infection Biology, Wonkwang University School of Medicine, Iksan 54538, Republic of Korea
| | - Ha Eun Ji
- Department of Biology Education, College of Education, Chosun University, Gwangju 61452, Republic of Korea
| | - Sungtae Yang
- Department of Microbiology, School of Medicine, Chosun University, Gwangju 61452, Republic of Korea
| | - Jeong Hoon Cho
- Department of Biology Education, College of Education, Chosun University, Gwangju 61452, Republic of Korea
| |
Collapse
|
46
|
Pribadi AK, Chalasani SH. Fear conditioning in invertebrates. Front Behav Neurosci 2022; 16:1008818. [PMID: 36439964 PMCID: PMC9686301 DOI: 10.3389/fnbeh.2022.1008818] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/19/2022] [Indexed: 09/30/2023] Open
Abstract
Learning to identify and predict threats is a basic skill that allows animals to avoid harm. Studies in invertebrates like Aplysia californica, Drosophila melanogaster, and Caenorhabditis elegans have revealed that the basic mechanisms of learning and memory are conserved. We will summarize these studies and highlight the common pathways and mechanisms in invertebrate fear-associated behavioral changes. Fear conditioning studies utilizing electric shock in Aplysia and Drosophila have demonstrated that serotonin or dopamine are typically involved in relaying aversive stimuli, leading to changes in intracellular calcium levels and increased presynaptic neurotransmitter release and short-term changes in behavior. Long-term changes in behavior typically require multiple, spaced trials, and involve changes in gene expression. C. elegans studies have demonstrated these basic aversive learning principles as well; however, fear conditioning has yet to be explicitly demonstrated in this model due to stimulus choice. Because predator-prey relationships can be used to study learned fear in a naturalistic context, this review also summarizes what is known about predator-induced behaviors in these three organisms, and their potential applications for future investigations into fear conditioning.
Collapse
Affiliation(s)
- Amy K. Pribadi
- Biological Sciences Graduate Program, University of California, San Diego, La Jolla, San Diego, CA, United States
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Sreekanth H. Chalasani
- Biological Sciences Graduate Program, University of California, San Diego, La Jolla, San Diego, CA, United States
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, United States
| |
Collapse
|
47
|
McKay A, Costa EK, Chen J, Hu CK, Chen X, Bedbrook CN, Khondker RC, Thielvoldt M, Priya Singh P, Wyss-Coray T, Brunet A. An automated feeding system for the African killifish reveals the impact of diet on lifespan and allows scalable assessment of associative learning. eLife 2022; 11:e69008. [PMID: 36354233 PMCID: PMC9788828 DOI: 10.7554/elife.69008] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 11/09/2022] [Indexed: 11/11/2022] Open
Abstract
The African turquoise killifish is an exciting new vertebrate model for aging studies. A significant challenge for any model organism is the control over its diet in space and time. To address this challenge, we created an automated and networked fish feeding system. Our automated feeder is designed to be open-source, easily transferable, and built from widely available components. Compared to manual feeding, our automated system is highly precise and flexible. As a proof of concept for the feeding flexibility of these automated feeders, we define a favorable regimen for growth and fertility for the African killifish and a dietary restriction regimen where both feeding time and quantity are reduced. We show that this dietary restriction regimen extends lifespan in males (but not in females) and impacts the transcriptomes of killifish livers in a sex-specific manner. Moreover, combining our automated feeding system with a video camera, we establish a quantitative associative learning assay to provide an integrative measure of cognitive performance for the killifish. The ability to precisely control food delivery in the killifish opens new areas to assess lifespan and cognitive behavior dynamics and to screen for dietary interventions and drugs in a scalable manner previously impossible with traditional vertebrate model organisms.
Collapse
Affiliation(s)
- Andrew McKay
- Department of Genetics, Stanford UniversityStanfordUnited States
- Biology Graduate Program, Stanford UniversityStanfordUnited States
| | - Emma K Costa
- Department of Neurology and Neurological Sciences, Stanford UniversityStanfordUnited States
- Neurosciences Interdepartmental Program, Stanford University School of MedicineStanfordUnited States
| | - Jingxun Chen
- Department of Genetics, Stanford UniversityStanfordUnited States
| | - Chi-Kuo Hu
- Department of Genetics, Stanford UniversityStanfordUnited States
| | - Xiaoshan Chen
- Department of Genetics, Stanford UniversityStanfordUnited States
| | - Claire N Bedbrook
- Department of Genetics, Stanford UniversityStanfordUnited States
- Department of Bioengineering, Stanford UniversityStanfordUnited States
| | | | | | | | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford UniversityStanfordUnited States
- Glenn Laboratories for the Biology of Aging, Stanford UniversityStanfordUnited States
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordUnited States
| | - Anne Brunet
- Department of Genetics, Stanford UniversityStanfordUnited States
- Glenn Laboratories for the Biology of Aging, Stanford UniversityStanfordUnited States
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordUnited States
| |
Collapse
|
48
|
Loose JA, Amrit FRG, Patil T, Yanowitz JL, Ghazi A. Meiotic dysfunction accelerates somatic aging in Caenorhabditis elegans. Aging Cell 2022; 21:e13716. [PMID: 36176234 PMCID: PMC9649607 DOI: 10.1111/acel.13716] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 08/07/2022] [Accepted: 08/31/2022] [Indexed: 01/25/2023] Open
Abstract
An expanding body of evidence, from studies in model organisms to human clinical data, reveals that reproductive health influences organismal aging. However, the impact of germline integrity on somatic aging is poorly understood. Moreover, assessing the causal relationship of such an impact is challenging to address in human and vertebrate models. Here, we demonstrate that disruption of meiosis, a germline restricted process, shortened lifespan, impaired individual aspects of healthspan, and accelerated somatic aging in Caenorhabditis elegans. Young meiotic mutants exhibited transcriptional profiles that showed remarkable overlap with the transcriptomes of old worms and shared similarities with transcriptomes of aging human tissues as well. We found that meiosis dysfunction caused increased expression of functionally relevant longevity determinants whose inactivation enhanced the lifespan of normal animals. Further, meiotic mutants manifested destabilized protein homeostasis and enhanced proteasomal activity partially rescued the associated lifespan defects. Our study demonstrates a role for meiotic integrity in controlling somatic aging and reveals proteostasis control as a potential mechanism through which germline status impacts overall organismal health.
Collapse
Affiliation(s)
- Julia A. Loose
- Department of Pediatrics, John G. Rangos Sr. Research CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Francis R. G. Amrit
- Department of Pediatrics, John G. Rangos Sr. Research CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Thayjas Patil
- Department of Pediatrics, John G. Rangos Sr. Research CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Judith L. Yanowitz
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee‐Womens Research InstituteUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Arjumand Ghazi
- Department of Pediatrics, John G. Rangos Sr. Research CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA,Department of Developmental Biology, John G. Rangos Sr. Research CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA,Department of Cell Biology & PhysiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| |
Collapse
|
49
|
Mastrandreas P, Boglari C, Arnold A, Peter F, de Quervain DJF, Papassotiropoulos A, Stetak A. Phosphorylation of MSI-1 is implicated in the regulation of associative memory in Caenorhabditis elegans. PLoS Genet 2022; 18:e1010420. [PMID: 36223338 PMCID: PMC9555661 DOI: 10.1371/journal.pgen.1010420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 09/09/2022] [Indexed: 11/07/2022] Open
Abstract
The Musashi family of RNA-binding proteins controls several biological processes including stem cell maintenance, cell division and neural function. Previously, we demonstrated that the C. elegans Musashi ortholog, msi-1, regulates forgetting via translational repression of the Arp2/3 actin-branching complex. However, the mechanisms controlling MSI-1 activity during the regulation of forgetting are currently unknown. Here we investigated the effects of protein phosphorylation on MSI-1 activity. We showed that MSI-1 function is likely controlled by alterations of its activity rather than its expression levels. Furthermore, we found that MSI-1 is phosphorylated and using mass spectrometry we identified MSI-1 phosphorylation at three residues (T18, S19 and S34). CRISPR-based manipulations of MSI-1 phosphorylation sites revealed that phosphorylation is necessary for MSI-1 function in both short- and long-term aversive olfactory associative memory. Thus, our study provides insight into the mechanisms regulating memory-related MSI-1 activity and may facilitate the development of novel therapeutic approaches. Understanding neural circuits and molecular mechanisms underlying learning and memory are the major challenges of neuroscience. It is a generally accepted model that a learning event causes modification of synapses; strengthening some within a circuit and weakening others (termed “synaptic plasticity”). A plastic nervous system requires not only the ability to acquire and store but also to forget new inputs. While learning and memory is widely investigated, clear-cut evidence for mechanisms involved in forgetting is still sparse. Previously, we demonstrated the role of the protein Musashi (MSI-1) in the active regulation of forgetting in the nematode C. elegans. Here we investigated the role of protein modification (phosphorylation) as a possible regulatory mechanism of the MSI-1 protein activity. We found that MSI-1 protein is modified at different positions and all of these modifications at the protein level contribute to the correct activity of the protein leading to active forgetting of short and long-term memories.
Collapse
Affiliation(s)
- Pavlina Mastrandreas
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Molecular Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- Biozentrum, Life Sciences Training Facility, University of Basel, Basel, Switzerland
| | - Csaba Boglari
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Molecular Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- Biozentrum, Life Sciences Training Facility, University of Basel, Basel, Switzerland
| | - Andreas Arnold
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Molecular Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- Biozentrum, Life Sciences Training Facility, University of Basel, Basel, Switzerland
| | - Fabian Peter
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Molecular Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- Biozentrum, Life Sciences Training Facility, University of Basel, Basel, Switzerland
| | - Dominique J.-F. de Quervain
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Cognitive Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- University Psychiatric Clinics, University of Basel, Basel, Switzerland
| | - Andreas Papassotiropoulos
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Molecular Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- Biozentrum, Life Sciences Training Facility, University of Basel, Basel, Switzerland
- University Psychiatric Clinics, University of Basel, Basel, Switzerland
| | - Attila Stetak
- Transfaculty Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
- Division of Molecular Neuroscience, Department of Psychology, University of Basel, Basel, Switzerland
- Biozentrum, Life Sciences Training Facility, University of Basel, Basel, Switzerland
- University Psychiatric Clinics, University of Basel, Basel, Switzerland
- * E-mail:
| |
Collapse
|
50
|
Identification of a Hydroxygallic Acid Derivative, Zingibroside R1 and a Sterol Lipid as Potential Active Ingredients of Cuscuta chinensis Extract That Has Neuroprotective and Antioxidant Effects in Aged Caenorhabditis elegans. Nutrients 2022; 14:nu14194199. [PMID: 36235851 PMCID: PMC9570774 DOI: 10.3390/nu14194199] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/01/2022] [Accepted: 10/04/2022] [Indexed: 11/25/2022] Open
Abstract
We examined the effects of the extracts from two traditional Chinese medicine plants, Cuscuta chinensis and Eucommia ulmoides, on the healthspan of the model organism Caenorhabditis elegans. C. chinensis increased the short-term memory and the mechanosensory response of aged C. elegans. Furthermore, both extracts improved the resistance towards oxidative stress, and decreased the intracellular level of reactive oxygen species. Chemical analyses of the extracts revealed the presence of several bioactive compounds such as chlorogenic acid, cinnamic acid, and quercetin. A fraction from the C. chinensis extract enriched in zingibroside R1 improved the lifespan, the survival after heat stress, and the locomotion in a manner similar to the full C. chinensis extract. Thus, zingibroside R1 could be (partly) responsible for the observed health benefits of C. chinensis. Furthermore, a hydroxygallic acid derivative and the sterol lipid 4-alpha-formyl-stigmasta-7,24(241)-dien-3-beta-ol are abundantly present in the C. chinensis extract and its most bioactive fraction, but hardly in E. ulmoides, making them good candidates to explain the overall healthspan benefits of C. chinensis compared to the specific positive effects on stress resistance by E. ulmoides. Our findings highlight the overall anti-aging effects of C. chinensis in C. elegans and provide first hints about the components responsible for these effects.
Collapse
|