301
|
Buffenstein R, Lewis KN, Gibney PA, Narayan V, Grimes KM, Smith M, Lin TD, Brown-Borg HM. Probing Pedomorphy and Prolonged Lifespan in Naked Mole-Rats and Dwarf Mice. Physiology (Bethesda) 2020; 35:96-111. [DOI: 10.1152/physiol.00032.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Pedomorphy, maintenance of juvenile traits throughout life, is most pronounced in extraordinarily long-lived naked mole-rats. Many of these traits (e.g., slow growth rates, low hormone levels, and delayed sexual maturity) are shared with spontaneously mutated, long-lived dwarf mice. Although some youthful traits likely evolved as adaptations to subterranean habitats (e.g., thermolability), the nature of these intrinsic pedomorphic features may also contribute to their prolonged youthfulness, longevity, and healthspan.
Collapse
Affiliation(s)
| | | | - Patrick A. Gibney
- Calico Life Sciences LLC, South San Francisco, California
- Department of Food Science, College of Agriculture and Life Sciences, Stocking Hall, Cornell University, Ithaca, New York
| | - Vikram Narayan
- Calico Life Sciences LLC, South San Francisco, California
| | - Kelly M. Grimes
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Megan Smith
- Calico Life Sciences LLC, South San Francisco, California
| | - Tzuhua D. Lin
- Calico Life Sciences LLC, South San Francisco, California
| | - Holly M. Brown-Borg
- Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota
| |
Collapse
|
302
|
Martinez BA, Reis Rodrigues P, Nuñez Medina RM, Mondal P, Harrison NJ, Lone MA, Webster A, Gurkar AU, Grill B, Gill MS. An alternatively spliced, non-signaling insulin receptor modulates insulin sensitivity via insulin peptide sequestration in C. elegans. eLife 2020; 9:49917. [PMID: 32096469 PMCID: PMC7041946 DOI: 10.7554/elife.49917] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 01/10/2020] [Indexed: 01/05/2023] Open
Abstract
In the nematode C. elegans, insulin signaling regulates development and aging in response to the secretion of numerous insulin peptides. Here, we describe a novel, non-signaling isoform of the nematode insulin receptor (IR), DAF-2B, that modulates insulin signaling by sequestration of insulin peptides. DAF-2B arises via alternative splicing and retains the extracellular ligand binding domain but lacks the intracellular signaling domain. A daf-2b splicing reporter revealed active regulation of this transcript through development, particularly in the dauer larva, a diapause stage associated with longevity. CRISPR knock-in of mScarlet into the daf-2b genomic locus confirmed that DAF-2B is expressed in vivo and is likely secreted. Genetic studies indicate that DAF-2B influences dauer entry, dauer recovery and adult lifespan by altering insulin sensitivity according to the prevailing insulin milieu. Thus, in C. elegans alternative splicing at the daf-2 locus generates a truncated IR that fine-tunes insulin signaling in response to the environment.
Collapse
Affiliation(s)
- Bryan A Martinez
- Department of Molecular Medicine, The Scripps Research Institute - Scripps Florida, Jupiter, United States
| | - Pedro Reis Rodrigues
- Department of Molecular Medicine, The Scripps Research Institute - Scripps Florida, Jupiter, United States
| | - Ricardo M Nuñez Medina
- Department of Molecular Medicine, The Scripps Research Institute - Scripps Florida, Jupiter, United States
| | - Prosenjit Mondal
- Department of Molecular Medicine, The Scripps Research Institute - Scripps Florida, Jupiter, United States
| | - Neale J Harrison
- Department of Molecular Medicine, The Scripps Research Institute - Scripps Florida, Jupiter, United States
| | - Museer A Lone
- Department of Molecular Medicine, The Scripps Research Institute - Scripps Florida, Jupiter, United States
| | - Amanda Webster
- Department of Molecular Medicine, The Scripps Research Institute - Scripps Florida, Jupiter, United States
| | - Aditi U Gurkar
- Department of Molecular Medicine, The Scripps Research Institute - Scripps Florida, Jupiter, United States
| | - Brock Grill
- Department of Neuroscience, The Scripps Research Institute - Scripps Florida, Jupiter, United States
| | - Matthew S Gill
- Department of Molecular Medicine, The Scripps Research Institute - Scripps Florida, Jupiter, United States
| |
Collapse
|
303
|
Abstract
A truncated version of the only insulin receptor in C. elegans has been discovered.
Collapse
Affiliation(s)
- Coleen T Murphy
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
- LSI Genomics, Princeton UniversityPrincetonUnited States
| |
Collapse
|
304
|
Kowalczyk A, Partha R, Clark NL, Chikina M. Pan-mammalian analysis of molecular constraints underlying extended lifespan. eLife 2020; 9:e51089. [PMID: 32043462 PMCID: PMC7012612 DOI: 10.7554/elife.51089] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 01/14/2020] [Indexed: 12/23/2022] Open
Abstract
Although lifespan in mammals varies over 100-fold, the precise evolutionary mechanisms underlying variation in longevity remain unknown. Species-specific genetic changes have been observed in long-lived species including the naked mole-rat, bats, and the bowhead whale, but these adaptations do not generalize to other mammals. We present a novel method to identify associations between rates of protein evolution and continuous phenotypes across the entire mammalian phylogeny. Unlike previous analyses that focused on individual species, we treat absolute and relative longevity as quantitative traits and demonstrate that these lifespan traits affect the evolutionary constraint on hundreds of genes. Specifically, we find that genes related to cell cycle, DNA repair, cell death, the IGF1 pathway, and immunity are under increased evolutionary constraint in large and long-lived mammals. For mammals exceptionally long-lived for their body size, we find increased constraint in inflammation, DNA repair, and NFKB-related pathways. Strikingly, these pathways have considerable overlap with those that have been previously reported to have potentially adaptive changes in single-species studies, and thus would be expected to show decreased constraint in our analysis. This unexpected finding of increased constraint in many longevity-associated pathways underscores the power of our quantitative approach to detect patterns that generalize across the mammalian phylogeny.
Collapse
Affiliation(s)
- Amanda Kowalczyk
- Joint Carnegie Mellon University-University of Pittsburgh PhD Program in Computational BiologyPittsburghUnited States
- Department of Computational and Systems BiologyUniversity of PittsburghPittsburghUnited States
| | - Raghavendran Partha
- Joint Carnegie Mellon University-University of Pittsburgh PhD Program in Computational BiologyPittsburghUnited States
- Department of Computational and Systems BiologyUniversity of PittsburghPittsburghUnited States
| | - Nathan L Clark
- Department of Computational and Systems BiologyUniversity of PittsburghPittsburghUnited States
- Pittsburgh Center for Evolutionary Biology and MedicineUniversity of PittsburghPittsburghUnited States
- Department of Human GeneticsUniversity of UtahSalt Lake CityUnited States
| | - Maria Chikina
- Department of Computational and Systems BiologyUniversity of PittsburghPittsburghUnited States
| |
Collapse
|
305
|
Poeschla M, Valenzano DR. The turquoise killifish: a genetically tractable model for the study of aging. ACTA ACUST UNITED AC 2020; 223:223/Suppl_1/jeb209296. [PMID: 32034047 DOI: 10.1242/jeb.209296] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Lifespan is a remarkably diverse trait in nature, ranging from just hours in adult mayflies to hundreds of years in the Greenland shark and quahog clam. Great disparities in lifespan are often observed even among somewhat closely related species; for example, in the laboratory, wild-derived strains of the common house mouse have a maximum observed lifespan of approximately 6 years, while a similarly sized rodent, the naked mole rat, can live for over 30 years. Comparative biology of aging across the tree of life provides a tremendous opportunity for understanding the molecular and genetic basis underlying lifespan and aging. However, a lack of molecular and laboratory tools has limited the ability of researchers to take full advantage of the incredible diversity of aging phenotypes in nature. Recent developments in genomic technology have made it increasingly possible to study non-canonical model organisms for aging. One promising new genetic model organism amenable to a range of experimental interventions is the turquoise killifish (Nothobranchius furzeri). This fish species has a naturally short lifespan and undergoes a wide range of aging-related transformations. These fish have a fully sequenced genome and transcriptome, and killifish embryos are accessible to transgenesis and genome editing. Furthermore, different killifish species and populations show striking differences in lifespan, providing the opportunity for comparative analysis of aging. This Review introduces the natural life history of the turquoise killifish, its emerging applicability as an aging model system, the genetic tools that have been developed to study aging for this species and a summary of recent studies facilitated by these new tools.
Collapse
Affiliation(s)
- Michael Poeschla
- Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany.,CECAD, University of Cologne, 50931 Cologne, Germany
| | - Dario R Valenzano
- Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany .,CECAD, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
306
|
Rashid S, Pho KB, Mesbahi H, MacNeil LT. Nutrient Sensing and Response Drive Developmental Progression in Caenorhabditis elegans. Bioessays 2020; 42:e1900194. [PMID: 32003906 DOI: 10.1002/bies.201900194] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/22/2019] [Indexed: 12/18/2022]
Abstract
In response to nutrient limitation, many animals, including Caenorhabditis elegans, slow or arrest their development. This process requires mechanisms that sense essential nutrients and induce appropriate responses. When faced with nutrient limitation, C. elegans can induce both short and long-term survival strategies, including larval arrest, decreased developmental rate, and dauer formation. To select the most advantageous strategy, information from many different sensors must be integrated into signaling pathways, including target of rapamycin (TOR) and insulin, that regulate developmental progression. Here, how nutrient information is sensed and integrated into developmental decisions that determine developmental rate and progression in C. elegans is reviewed.
Collapse
Affiliation(s)
- Sabih Rashid
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, L8S 4K1, Ontario, Canada
| | - Kim B Pho
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, L8S 4K1, Ontario, Canada
| | - Hiva Mesbahi
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, L8S 4K1, Ontario, Canada
| | - Lesley T MacNeil
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, L8S 4K1, Ontario, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, L8S 4K1, Ontario, Canada.,Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, L8S 4K1, Ontario, Canada
| |
Collapse
|
307
|
Goya ME, Xue F, Sampedro-Torres-Quevedo C, Arnaouteli S, Riquelme-Dominguez L, Romanowski A, Brydon J, Ball KL, Stanley-Wall NR, Doitsidou M. Probiotic Bacillus subtilis Protects against α-Synuclein Aggregation in C. elegans. Cell Rep 2020; 30:367-380.e7. [PMID: 31940482 PMCID: PMC6963774 DOI: 10.1016/j.celrep.2019.12.078] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/23/2019] [Accepted: 12/19/2019] [Indexed: 12/13/2022] Open
Abstract
Recent discoveries have implicated the gut microbiome in the progression and severity of Parkinson's disease; however, how gut bacteria affect such neurodegenerative disorders remains unclear. Here, we report that the Bacillus subtilis probiotic strain PXN21 inhibits α-synuclein aggregation and clears preformed aggregates in an established Caenorhabditis elegans model of synucleinopathy. This protection is seen in young and aging animals and is partly mediated by DAF-16. Multiple B. subtilis strains trigger the protective effect via both spores and vegetative cells, partly due to a biofilm formation in the gut of the worms and the release of bacterial metabolites. We identify several host metabolic pathways differentially regulated in response to probiotic exposure, including sphingolipid metabolism. We further demonstrate functional roles of the sphingolipid metabolism genes lagr-1, asm-3, and sptl-3 in the anti-aggregation effect. Our findings provide a basis for exploring the disease-modifying potential of B. subtilis as a dietary supplement.
Collapse
Affiliation(s)
- María Eugenia Goya
- University of Edinburgh, Centre for Discovery Brain Sciences, Edinburgh, Scotland
| | - Feng Xue
- University of Edinburgh, Centre for Discovery Brain Sciences, Edinburgh, Scotland
| | | | | | | | - Andrés Romanowski
- University of Edinburgh, School of Biological Sciences, Edinburgh, Scotland
| | - Jack Brydon
- University of Edinburgh, Institute of Genetics & Molecular Medicine, Edinburgh, Scotland
| | - Kathryn L Ball
- University of Edinburgh, Institute of Genetics & Molecular Medicine, Edinburgh, Scotland
| | | | - Maria Doitsidou
- University of Edinburgh, Centre for Discovery Brain Sciences, Edinburgh, Scotland.
| |
Collapse
|
308
|
DAF-16/FOXO requires Protein Phosphatase 4 to initiate transcription of stress resistance and longevity promoting genes. Nat Commun 2020; 11:138. [PMID: 31919361 PMCID: PMC6952425 DOI: 10.1038/s41467-019-13931-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 12/09/2019] [Indexed: 12/21/2022] Open
Abstract
In C. elegans, the conserved transcription factor DAF-16/FOXO is a powerful aging regulator, relaying dire conditions into expression of stress resistance and longevity promoting genes. For some of these functions, including low insulin/IGF signaling (IIS), DAF-16 depends on the protein SMK-1/SMEK, but how SMK-1 exerts this role has remained unknown. We show that SMK-1 functions as part of a specific Protein Phosphatase 4 complex (PP4SMK-1). Loss of PP4SMK-1 hinders transcriptional initiation at several DAF-16-activated genes, predominantly by impairing RNA polymerase II recruitment to their promoters. Search for the relevant substrate of PP4SMK-1 by phosphoproteomics identified the conserved transcriptional regulator SPT-5/SUPT5H, whose knockdown phenocopies the loss of PP4SMK-1. Phosphoregulation of SPT-5 is known to control transcriptional events such as elongation and termination. Here we also show that transcription initiating events are influenced by the phosphorylation status of SPT-5, particularly at DAF-16 target genes where transcriptional initiation appears rate limiting, rendering PP4SMK-1 crucial for many of DAF-16’s physiological roles. The transcription factor DAF-16/FOXO mediates a wide variety of aging-preventive responses by driving the expression of stress resistance and longevity promoting genes. Here the authors show that transcriptional initiation at many DAF-16/FOXO target genes requires the dephosphorylation of SPT-5 by Protein Phosphatase 4.
Collapse
|
309
|
DAF-16/FoxO in Caenorhabditis elegans and Its Role in Metabolic Remodeling. Cells 2020; 9:cells9010109. [PMID: 31906434 PMCID: PMC7017163 DOI: 10.3390/cells9010109] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/30/2019] [Accepted: 12/31/2019] [Indexed: 12/31/2022] Open
Abstract
DAF-16, the only forkhead box transcription factors class O (FoxO) homolog in Caenorhabditis elegans, integrates signals from upstream pathways to elicit transcriptional changes in many genes involved in aging, development, stress, metabolism, and immunity. The major regulator of DAF-16 activity is the insulin/insulin-like growth factor 1 (IGF-1) signaling (IIS) pathway, reduction of which leads to lifespan extension in worms, flies, mice, and humans. In C. elegans daf-2 mutants, reduced IIS leads to a heterochronic activation of a dauer survival program during adulthood. This program includes elevated antioxidant defense and a metabolic shift toward accumulation of carbohydrates (i.e., trehalose and glycogen) and triglycerides, and activation of the glyoxylate shunt, which could allow fat-to-carbohydrate conversion. The longevity of daf-2 mutants seems to be partially supported by endogenous trehalose, a nonreducing disaccharide that mammals cannot synthesize, which points toward considerable differences in downstream mechanisms by which IIS regulates aging in distinct groups.
Collapse
|
310
|
Turner MJ, Cox JK, Spellman AC, Stahl C, Bavari S. Avoidance behavior independent of innate-immune signaling seen in Caenorhabditis elegans challenged with Bacillus anthracis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 102:103453. [PMID: 31326564 DOI: 10.1016/j.dci.2019.103453] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 07/16/2019] [Accepted: 07/16/2019] [Indexed: 06/10/2023]
Abstract
Small organisms, like the nematode C. elegans, are emerging as insightful models in which to study host/pathogen interactions and the evolving interplay between host defenses and microbial offenses. In C. elegans the innate immune response has been shown to be connected to the DAF-2 insulin/insulin-like growth factor 1 (IGF-1) signal pathway, a critical transduction pathway that mediates stress response in the worms via the DAF-16 FOXO/forkhead transcription factor. Our studies of the C. elegans' phenotypes that are associated with behavioral innate immune response (avoidance behavior) and IGF-1 signaling perturbations (lifespan effects) led us to question the cause of the avoidance behavior observed when C. elegans are challenged with B. anthracis. While worms indeed avoid B. anthracis, and this behavior seems to be partly tied to IGF-1 signaling, the bacteria have neither nematocidal nor visible pathogenic effects on the worms. In fact, worms fed B. anthracis alone exhibit extended lifespans. We demonstrate that the extended lifespan phenotype seen in worms fed B. anthracis is likely the result of calorie restriction, and that worms do not eat B. anthracis even when avoidance behaviors have been suppressed. We further demonstrate a large time lag between the onset of avoidance behavior (which occurs upon contact with B. anthracis), and the induction of IGF-1 signaling (which occurs much later) in worms fed B. anthracis. Taken together, our data demonstrate behavioral avoidance that does not appear to be linked to a measurable immune response. We propose that, in some situations, avoidance behaviors categorized as immunological might be more accurately described as broad foraging behaviors induced in worms presented with a non-preferred food choice, or with a food choice that is either difficult or impossible for the worms to ingest.
Collapse
Affiliation(s)
- Michael J Turner
- School of Natural Sciences and Mathematics, Department of Science, Mount St. Mary's University, 16300 Old Emmitsburg Rd, Emmitsburg, MD, 21727, USA; Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Fort Detrick, Frederick, MD, 21702, USA.
| | - Justin K Cox
- School of Natural Sciences and Mathematics, Department of Science, Mount St. Mary's University, 16300 Old Emmitsburg Rd, Emmitsburg, MD, 21727, USA.
| | - Anthony C Spellman
- School of Natural Sciences and Mathematics, Department of Science, Mount St. Mary's University, 16300 Old Emmitsburg Rd, Emmitsburg, MD, 21727, USA.
| | - Craig Stahl
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Fort Detrick, Frederick, MD, 21702, USA.
| | - Sina Bavari
- Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Fort Detrick, Frederick, MD, 21702, USA.
| |
Collapse
|
311
|
Sarfstein R, Yeheskel A, Sinai-Livne T, Pasmanik-Chor M, Werner H. Systems Analysis of Insulin and IGF1 Receptors Networks in Breast Cancer Cells Identifies Commonalities and Divergences in Expression Patterns. Front Endocrinol (Lausanne) 2020; 11:435. [PMID: 32733384 PMCID: PMC7359857 DOI: 10.3389/fendo.2020.00435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 06/02/2020] [Indexed: 11/13/2022] Open
Abstract
Insulin and insulin-like growth factor-1 (IGF1), acting respectively via the insulin (INSR) and IGF1 (IGF1R) receptors, play key developmental and metabolic roles throughout life. In addition, both signaling pathways fulfill important roles in cancer initiation and progression. The present study was aimed at identifying mechanistic differences between INSR and IGF1R using a recently developed bioinformatics tool, the Biological Network Simulator (BioNSi). This application allows to import and merge multiple pathways and interaction information from the KEGG database into a single network representation. The BioNsi network simulation tool allowed us to exploit the availability of gene expression data derived from breast cancer cell lines with specific disruptions of the INSR or IGF1R genes in order to investigate potential differences in protein expression that might be linked to biological attributes of the specific receptor networks. Modeling-generated information was corroborated by experimental and biological assays. BioNSi analyses revealed that the expression of 75 and 71 genes changed during simulation of IGF1R-KD and INSR-KD, compared to control cells, respectively. Out of 16 proteins that BioNSi analysis was based on, validated by Western blotting, nine were shown to be involved in DNA repair, eight in cell cycle checkpoints, six in proliferation, eight in apoptosis, seven in oxidative stress, six in cell migration, two in energy homeostasis, and three in senescence. Taken together, analyses identified a number of commonalities and, most importantly, dissimilarities between the IGF1R and INSR pathways that might help explain the basis for the biological differences between these networks.
Collapse
MESH Headings
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Gene Regulatory Networks
- Humans
- Receptor, IGF Type 1/antagonists & inhibitors
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Receptor, Insulin/antagonists & inhibitors
- Receptor, Insulin/genetics
- Receptor, Insulin/metabolism
- Systems Analysis
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Rive Sarfstein
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adva Yeheskel
- Bioinformatics Unit, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tali Sinai-Livne
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Metsada Pasmanik-Chor
- Bioinformatics Unit, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- *Correspondence: Metsada Pasmanik-Chor
| | - Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Yoran Institute for Human Genome Research, Tel Aviv University, Tel Aviv, Israel
- Haim Werner
| |
Collapse
|
312
|
FUNATO H. Forward genetic approach for behavioral neuroscience using animal models. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2020; 96:10-31. [PMID: 31932526 PMCID: PMC6974404 DOI: 10.2183/pjab.96.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/18/2019] [Indexed: 06/10/2023]
Abstract
Forward genetics is a powerful approach to understand the molecular basis of animal behaviors. Fruit flies were the first animal to which this genetic approach was applied systematically and have provided major discoveries on behaviors including sexual, learning, circadian, and sleep-like behaviors. The development of different classes of model organism such as nematodes, zebrafish, and mice has enabled genetic research to be conducted using more-suitable organisms. The unprecedented success of forward genetic approaches was the identification of the transcription-translation negative feedback loop composed of clock genes as a fundamental and conserved mechanism of circadian rhythm. This approach has now expanded to sleep/wakefulness in mice. A conventional strategy such as dominant and recessive screenings can be modified with advances in DNA sequencing and genome editing technologies.
Collapse
Affiliation(s)
- Hiromasa FUNATO
- Department of Anatomy, Faculty of Medicine, Toho University, Tokyo, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
313
|
Wu T, Duan F, Yang W, Liu H, Caballero A, Fernandes de Abreu DA, Dar AR, Alcedo J, Ch'ng Q, Butcher RA, Zhang Y. Pheromones Modulate Learning by Regulating the Balanced Signals of Two Insulin-like Peptides. Neuron 2019; 104:1095-1109.e5. [PMID: 31676170 PMCID: PMC7009321 DOI: 10.1016/j.neuron.2019.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 08/09/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023]
Abstract
Social environment modulates learning through unknown mechanisms. Here, we report that a pheromone mixture that signals overcrowding inhibits C. elegans from learning to avoid pathogenic bacteria. We find that learning depends on the balanced signaling of two insulin-like peptides (ILPs), INS-16 and INS-4, which act respectively in the pheromone-sensing neuron ADL and the bacteria-sensing neuron AWA. Pheromone exposure inhibits learning by disrupting this balance: it activates ADL and increases expression of ins-16, and this cellular effect reduces AWA activity and AWA-expressed ins-4. The activities of the sensory neurons are required for learning and the expression of the ILPs. Interestingly, pheromones also promote the ingestion of pathogenic bacteria while increasing resistance to the pathogen. Thus, the balance of the ILP signals integrates social information into the learning process as part of a coordinated adaptive response that allows consumption of harmful food during times of high population density.
Collapse
Affiliation(s)
- Taihong Wu
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Fengyun Duan
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Wenxing Yang
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - He Liu
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Antonio Caballero
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Diana Andrea Fernandes de Abreu
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Abdul Rouf Dar
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| | - Joy Alcedo
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| | - QueeLim Ch'ng
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Rebecca A Butcher
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| | - Yun Zhang
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Center for Brain Science, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
314
|
Li X, Itani OA, Haataja L, Dumas KJ, Yang J, Cha J, Flibotte S, Shih HJ, Delaney CE, Xu J, Qi L, Arvan P, Liu M, Hu PJ. Requirement for translocon-associated protein (TRAP) α in insulin biogenesis. SCIENCE ADVANCES 2019; 5:eaax0292. [PMID: 31840061 PMCID: PMC6892615 DOI: 10.1126/sciadv.aax0292] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 10/07/2019] [Indexed: 05/26/2023]
Abstract
The mechanistic basis for the biogenesis of peptide hormones and growth factors is poorly understood. Here, we show that the conserved endoplasmic reticulum membrane translocon-associated protein α (TRAPα), also known as signal sequence receptor 1, plays a critical role in the biosynthesis of insulin. Genetic analysis in the nematode Caenorhabditis elegans and biochemical studies in pancreatic β cells reveal that TRAPα deletion impairs preproinsulin translocation while unexpectedly disrupting distal steps in insulin biogenesis including proinsulin processing and secretion. The association of common intronic single-nucleotide variants in the human TRAPα gene with susceptibility to type 2 diabetes and pancreatic β cell dysfunction suggests that impairment of preproinsulin translocation and proinsulin trafficking may contribute to the pathogenesis of type 2 diabetes.
Collapse
Affiliation(s)
- Xin Li
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Omar A. Itani
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Leena Haataja
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Kathleen J. Dumas
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jing Yang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Jeeyeon Cha
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Stephane Flibotte
- Departments of Zoology and Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hung-Jen Shih
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Colin E. Delaney
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jialu Xu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Ling Qi
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Peter Arvan
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Patrick J. Hu
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
315
|
Marks ND, Winter AD, Gu HY, Maitland K, Gillan V, Ambroz M, Martinelli A, Laing R, MacLellan R, Towne J, Roberts B, Hanks E, Devaney E, Britton C. Profiling microRNAs through development of the parasitic nematode Haemonchus identifies nematode-specific miRNAs that suppress larval development. Sci Rep 2019; 9:17594. [PMID: 31772378 PMCID: PMC6879476 DOI: 10.1038/s41598-019-54154-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/04/2019] [Indexed: 02/05/2023] Open
Abstract
Parasitic nematodes transition between dramatically different free-living and parasitic stages, with correctly timed development and migration crucial to successful completion of their lifecycle. However little is known of the mechanisms controlling these transitions. microRNAs (miRNAs) negatively regulate gene expression post-transcriptionally and regulate development of diverse organisms. Here we used microarrays to determine the expression profile of miRNAs through development and in gut tissue of the pathogenic nematode Haemonchus contortus. Two miRNAs, mir-228 and mir-235, were enriched in infective L3 larvae, an arrested stage analogous to Caenorhabditis elegans dauer larvae. We hypothesized that these miRNAs may suppress development and maintain arrest. Consistent with this, inhibitors of these miRNAs promoted H. contortus development from L3 to L4 stage, while genetic deletion of C. elegans homologous miRNAs reduced dauer arrest. Epistasis studies with C. elegans daf-2 mutants showed that mir-228 and mir-235 synergise with FOXO transcription factor DAF-16 in the insulin signaling pathway. Target prediction suggests that these miRNAs suppress metabolic and transcription factor activity required for development. Our results provide novel insight into the expression and functions of specific miRNAs in regulating nematode development and identify miRNAs and their target genes as potential therapeutic targets to limit parasite survival within the host.
Collapse
Affiliation(s)
- Neil D Marks
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
| | - Alan D Winter
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
- West of Scotland Genetic Services, Level 2B, Laboratory Medicine, Queen Elizabeth University Hospital, Govan Road, Glasgow, G51 4TF, UK
| | - Henry Y Gu
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
| | - Kirsty Maitland
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
| | - Victoria Gillan
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
| | - Martin Ambroz
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Hradec Kralove, Czech Republic
| | - Axel Martinelli
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, N20 W10, Kita-ku, Sapporo, Japan
| | - Roz Laing
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
| | - Rachel MacLellan
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
| | - Jessica Towne
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
| | - Brett Roberts
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University Avenue, Glasgow, G12 8QQ, UK
| | - Eve Hanks
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK
| | - Eileen Devaney
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK.
| | - Collette Britton
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden Road, Glasgow, G61 1QH, UK.
| |
Collapse
|
316
|
Exploring Target Genes Involved in the Effect of Quercetin on the Response to Oxidative Stress in Caenorhabditis elegans. Antioxidants (Basel) 2019; 8:antiox8120585. [PMID: 31775265 PMCID: PMC6943653 DOI: 10.3390/antiox8120585] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 01/09/2023] Open
Abstract
Quercetin is one the most abundant flavonoids in the human diet. Although it is well known that quercetin exhibits a range of biological activities, the mechanisms behind these activities remain unresolved. The aim of this work is to progress in the knowledge of the molecular mechanisms involved in the biological effects of quercetin using Caenorhabditis elegans as a model organism. With this aim, the nematode has been used to explore the ability of this flavonoid to modulate the insulin/insulin-like growth factor 1(IGF-1) signaling pathway (IIS) and the expression of some genes related to stress response. Different methodological approaches have been used, i.e., assays in knockout mutant worms, gene expression assessment by RT-qPCR, and C. elegans transgenic strains expressing green fluorescent protein (GFP) reporters. The results showed that the improvement of the oxidative stress resistance of C. elegans induced by quercetin could be explained, at least in part, by the modulation of the insulin signaling pathway, involving genes age-1, akt-1, akt-2, daf-18, sgk-1, daf-2, and skn-1. However, this effect could be independent of the transcription factors DAF-16 and HSF-1 that regulate this pathway. Moreover, quercetin was also able to increase expression of hsp-16.2 in aged worms. This observation could be of particular interest to explain the effects of enhanced lifespan and greater resistance to stress induced by quercetin in C. elegans, since the expression of many heat shock proteins diminishes in aging worms.
Collapse
|
317
|
Horowitz LB, Brandt JP, Ringstad N. Repression of an activity-dependent autocrine insulin signal is required for sensory neuron development in C. elegans. Development 2019; 146:dev.182873. [PMID: 31628111 DOI: 10.1242/dev.182873] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/14/2019] [Indexed: 11/20/2022]
Abstract
Nervous system development is instructed by genetic programs and refined by distinct mechanisms that couple neural activity to gene expression. How these processes are integrated remains poorly understood. Here, we report that the regulated release of insulin-like peptides (ILPs) during development of the Caenorhabditis elegans nervous system accomplishes such an integration. We find that the p38 MAP kinase PMK-3, which is required for the differentiation of chemosensory BAG neurons, limits an ILP signal that represses expression of a BAG neuron fate. ILPs are released from BAGs themselves in an activity-dependent manner during development, indicating that ILPs constitute an autocrine signal that regulates the differentiation of BAG neurons. Expression of a specialized neuronal fate is, therefore, coordinately regulated by a genetic program that sets levels of ILP expression during development, and by neural activity, which regulates ILP release. Autocrine signals of this kind might have general and conserved functions as integrators of deterministic genetic programs with activity-dependent mechanisms during neurodevelopment.
Collapse
Affiliation(s)
- Lauren Bayer Horowitz
- Skirball Institute of Biomolecular Medicine, Helen L. and Martin S. Kimmel Center for Biology and Medicine, Department of Cell Biology, Neuroscience Institute, NYU School of Medicine, New York, NY 10016, USA
| | - Julia P Brandt
- Skirball Institute of Biomolecular Medicine, Helen L. and Martin S. Kimmel Center for Biology and Medicine, Department of Cell Biology, Neuroscience Institute, NYU School of Medicine, New York, NY 10016, USA
| | - Niels Ringstad
- Skirball Institute of Biomolecular Medicine, Helen L. and Martin S. Kimmel Center for Biology and Medicine, Department of Cell Biology, Neuroscience Institute, NYU School of Medicine, New York, NY 10016, USA
| |
Collapse
|
318
|
Kruempel JC, Howington MB, Leiser SF. Computational tools for geroscience. TRANSLATIONAL MEDICINE OF AGING 2019; 3:132-143. [PMID: 33241167 PMCID: PMC7685266 DOI: 10.1016/j.tma.2019.11.004] [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] [Indexed: 11/30/2022] Open
Abstract
The rapid progress of the past three decades has led the geroscience field near a point where human interventions in aging are plausible. Advances across scientific areas, such as high throughput "-omics" approaches, have led to an exponentially increasing quantity of data available for biogerontologists. To best translate the lifespan and healthspan extending interventions discovered by basic scientists into preventative medicine, it is imperative that the current data are comprehensively utilized to generate testable hypotheses about translational interventions. Building a translational pipeline for geroscience will require both systematic efforts to identify interventions that extend healthspan across taxa and diagnostics that can identify patients who may benefit from interventions prior to the onset of an age-related morbidity. Databases and computational tools that organize and analyze both the wealth of information available on basic biogerontology research and clinical data on aging populations will be critical in developing such a pipeline. Here, we review the current landscape of databases and computational resources available for translational aging research. We discuss key platforms and tools available for aging research, with a focus on how each tool can be used in concert with hypothesis driven experiments to move closer to human interventions in aging.
Collapse
Affiliation(s)
- Joseph C.P. Kruempel
- Molecular & Integrative Physiology Department, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Marshall B. Howington
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Scott F. Leiser
- Molecular & Integrative Physiology Department, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
319
|
Kim J, Kang Y, Choi D, Cho Y, Cho S, Choi H, Kim H. The natural phytochemical trans‐communic acid inhibits cellular senescence and pigmentation through FoxO3a activation. Exp Dermatol 2019; 28:1270-1278. [DOI: 10.1111/exd.14025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/11/2019] [Accepted: 08/01/2019] [Indexed: 12/21/2022]
Affiliation(s)
| | | | - Dong‐hwa Choi
- Biocenter, Gyeonggido Business & Science Accelerator Suwon Korea
| | | | | | | | | |
Collapse
|
320
|
Blackwell TK, Sewell AK, Wu Z, Han M. TOR Signaling in Caenorhabditis elegans Development, Metabolism, and Aging. Genetics 2019; 213:329-360. [PMID: 31594908 PMCID: PMC6781902 DOI: 10.1534/genetics.119.302504] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 07/18/2019] [Indexed: 12/30/2022] Open
Abstract
The Target of Rapamycin (TOR or mTOR) is a serine/threonine kinase that regulates growth, development, and behaviors by modulating protein synthesis, autophagy, and multiple other cellular processes in response to changes in nutrients and other cues. Over recent years, TOR has been studied intensively in mammalian cell culture and genetic systems because of its importance in growth, metabolism, cancer, and aging. Through its advantages for unbiased, and high-throughput, genetic and in vivo studies, Caenorhabditis elegans has made major contributions to our understanding of TOR biology. Genetic analyses in the worm have revealed unexpected aspects of TOR functions and regulation, and have the potential to further expand our understanding of how growth and metabolic regulation influence development. In the aging field, C. elegans has played a leading role in revealing the promise of TOR inhibition as a strategy for extending life span, and identifying mechanisms that function upstream and downstream of TOR to influence aging. Here, we review the state of the TOR field in C. elegans, and focus on what we have learned about its functions in development, metabolism, and aging. We discuss knowledge gaps, including the potential pitfalls in translating findings back and forth across organisms, but also describe how TOR is important for C. elegans biology, and how C. elegans work has developed paradigms of great importance for the broader TOR field.
Collapse
Affiliation(s)
- T Keith Blackwell
- Research Division, Joslin Diabetes Center, Department of Genetics, Harvard Medical School, Harvard Stem Cell Institute, Boston, Massachusetts
| | - Aileen K Sewell
- Department of MCDB, University of Colorado at Boulder, and
- Howard Hughes Medical Institute, Boulder, Colorado
| | - Ziyun Wu
- Research Division, Joslin Diabetes Center, Department of Genetics, Harvard Medical School, Harvard Stem Cell Institute, Boston, Massachusetts
| | - Min Han
- Department of MCDB, University of Colorado at Boulder, and
- Howard Hughes Medical Institute, Boulder, Colorado
| |
Collapse
|
321
|
Reynolds JA. Noncoding RNA Regulation of Dormant States in Evolutionarily Diverse Animals. THE BIOLOGICAL BULLETIN 2019; 237:192-209. [PMID: 31714856 DOI: 10.1086/705484] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Dormancy is evolutionarily widespread and can take many forms, including diapause, dauer formation, estivation, and hibernation. Each type of dormancy is characterized by distinct features; but accumulating evidence suggests that each is regulated by some common processes, often referred to as a common "toolkit" of regulatory mechanisms, that likely include noncoding RNAs that regulate gene expression. Noncoding RNAs, especially microRNAs, are well-known regulators of biological processes associated with numerous dormancy-related processes, including cell cycle progression, cell growth and proliferation, developmental timing, metabolism, and environmental stress tolerance. This review provides a summary of our current understanding of noncoding RNAs and their involvement in regulating dormancy.
Collapse
|
322
|
Bareja A, Lee DE, White JP. Maximizing Longevity and Healthspan: Multiple Approaches All Converging on Autophagy. Front Cell Dev Biol 2019; 7:183. [PMID: 31555646 PMCID: PMC6742954 DOI: 10.3389/fcell.2019.00183] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 08/20/2019] [Indexed: 12/13/2022] Open
Abstract
Our understanding of the molecular basis of aging has greatly increased over the past few decades. In this review, we provide an overview of the key signaling pathways associated with aging, and whose modulation has been shown to extend lifespan in a range of model organisms. We also describe how these pathways converge onto autophagy, a catabolic process that functions to recycle dysfunctional cellular material and maintains energy homeostasis. Finally, we consider various approaches of therapeutically modulating these longevity pathways, highlighting exercise as a potent geroprotector.
Collapse
Affiliation(s)
- Akshay Bareja
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, United States
| | - David E Lee
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, United States
| | - James P White
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, United States.,Division of Hematology, Department of Medicine, Duke University School of Medicine, Durham, NC, United States.,Duke Center for the Study of Aging and Human Development, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
323
|
West MD, Sternberg H, Labat I, Janus J, Chapman KB, Malik NN, de Grey ADNJ, Larocca D. Toward a unified theory of aging and regeneration. Regen Med 2019; 14:867-886. [DOI: 10.2217/rme-2019-0062] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Growing evidence supports the antagonistic pleiotropy theory of mammalian aging. Accordingly, changes in gene expression following the pluripotency transition, and subsequent transitions such as the embryonic–fetal transition, while providing tumor suppressive and antiviral survival benefits also result in a loss of regenerative potential leading to age-related fibrosis and degenerative diseases. However, reprogramming somatic cells to pluripotency demonstrates the possibility of restoring telomerase and embryonic regeneration pathways and thus reversing the age-related decline in regenerative capacity. A unified model of aging and loss of regenerative potential is emerging that may ultimately be translated into new therapeutic approaches for establishing induced tissue regeneration and modulation of the embryo-onco phenotype of cancer.
Collapse
Affiliation(s)
| | | | - Ivan Labat
- AgeX Therapeutics, Inc., Alameda, CA 94501, USA
| | | | | | - Nafees N Malik
- AgeX Therapeutics, Inc., Alameda, CA 94501, USA
- Juvenescence Ltd, London, UK
| | - Aubrey DNJ de Grey
- AgeX Therapeutics, Inc., Alameda, CA 94501, USA
- SENS Research Foundation, Mountain View, CA 94041, USA
| | | |
Collapse
|
324
|
Mitochondria in the signaling pathways that control longevity and health span. Ageing Res Rev 2019; 54:100940. [PMID: 31415807 PMCID: PMC7479635 DOI: 10.1016/j.arr.2019.100940] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/09/2019] [Accepted: 08/06/2019] [Indexed: 12/26/2022]
Abstract
Genetic and pharmacological intervention studies have identified evolutionarily conserved and functionally interconnected networks of cellular energy homeostasis, nutrient-sensing, and genome damage response signaling pathways, as prominent regulators of longevity and health span in various species. Mitochondria are the primary sites of ATP production and are key players in several other important cellular processes. Mitochondrial dysfunction diminishes tissue and organ functional performance and is a commonly considered feature of the aging process. Here we review the evidence that through reciprocal and multilevel functional interactions, mitochondria are implicated in the lifespan modulation function of these pathways, which altogether constitute a highly dynamic and complex system that controls the aging process. An important characteristic of these pathways is their extensive crosstalk and apparent malleability to modification by non-invasive pharmacological, dietary, and lifestyle interventions, with promising effects on lifespan and health span in animal models and potentially also in humans.
Collapse
|
325
|
Ackley BD. Behavior: Should I Stay or Should I Go? Curr Biol 2019; 29:R842-R844. [DOI: 10.1016/j.cub.2019.07.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
326
|
Yang WH, Chen CY, Wang KL, Kwok HL, Stern A, Lo SJ, Yang HC. Reflex and habituation behavior of Caenorhabditis elegans assessed by a mechanical vibration system and image analysis. J Neurosci Methods 2019; 328:108415. [PMID: 31470028 DOI: 10.1016/j.jneumeth.2019.108415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 08/26/2019] [Accepted: 08/26/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND The nematode Caenorhabditis elegans is an emerging invertebrate animal model for investigating neuronal functions in behavioral assays. C. elegans mechanosensation was characterized by the use of a constant mechanical stimulation transmitter followed by quantitative imaging. NEW METHOD C. elegans reflex and habituation behaviors were characterized by mechanical vibration followed by image analysis. A custom-designed system consists of an aluminum alloy Petri dish holder frame coupled with a mechanical vibration buzzer delivering adjustable pulsed vibration to an agar plate. The basal and evoked movements of C. elegans were recorded by a microscopic digital camera followed by quantitative analysis using microscopic imaging software. RESULTS Application of the platform in C. elegans was demonstrated with three proof-of-concept experiments: (1) Evaluation of the reflex response stimulated by tapping and mechanical vibration with a mechano-sensation defective mutant. (2) Comparison of the reflex response stimulated by mechanical vibration between wild type and aging mutants. (3) Assessment of the efficacy of the mechanical vibration system on long-term memory for habituation. COMPARISON WITH EXISTING METHODS Conventional C. elegans mechanosensation techniques depend on stimulation either by manually touching a single animal or tapping the Petri dish followed by scoring via visual observation from the examiner. The mechanical vibration method has greater capacity compared to conventional methods which are labor-intensive, have low throughput and lack quantifiable parameters. CONCLUSIONS The mechanical vibration system followed by image analysis is a convenient and integrated platform for investigatingC. elegans reflex and habituation in aging and neural behavioral assays.
Collapse
Affiliation(s)
- Wan-Hua Yang
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu, Taiwan; Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Hsinchu Branch, Hsinchu, Taiwan
| | - Chia-Yi Chen
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | | | - Hong Luen Kwok
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu, Taiwan
| | - Arnold Stern
- New York University School of Medicine, New York, USA
| | - Szecheng J Lo
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hung-Chi Yang
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu, Taiwan.
| |
Collapse
|
327
|
Peng Y, Sun Q, Gao R, Park Y. AAK-2 and SKN-1 Are Involved in Chicoric-Acid-Induced Lifespan Extension in Caenorhabditis elegans. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:9178-9186. [PMID: 30835107 DOI: 10.1021/acs.jafc.9b00705] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Chicoric acid is a dicaffeoyl ester with many bioactivities, including antioxidation, antidiabetes, and anti-inflammation. A previous study reported that chicoric acid extended the lifespan in Caenorhabditis elegans; however, the mechanism behind the effect of chicoric acid on the extended lifespan remains unknown. Consistent with the previous report, chicoric acid (25 and 50 μM) extended the maximum lifespan compared to the control (17.5 ± 3.3 and 15.6 ± 5%, respectively; p < 0.001 for both). The declines of the pumping rate and locomotive activity, two indicators of aging, were delayed by chicoric acid. Moreover, chicoric acid enhanced resistance to oxidative stress in C. elegans. It was further determined that the extended lifespan by chicoric acid was in part via aak-2 [a homologue of adenosine monophosphate (AMP)-activated protein kinase] and skn-1 (a homologue of nuclear factor erythroid 2-related factor 2). The current findings suggest that chicoric acid has the potential to be used as an anti-aging bioactive compound.
Collapse
Affiliation(s)
- Ye Peng
- School of Food and Biological Engineering , Jiangsu University , 301 Xuefu Road , Zhenjiang , Jiangsu 212013 , People's Republic of China
- Department of Food Science , University of Massachusetts, Amherst , 102 Holdsworth Way , Amherst , Massachusetts 01003 , United States
| | - Quancai Sun
- School of Food and Biological Engineering , Jiangsu University , 301 Xuefu Road , Zhenjiang , Jiangsu 212013 , People's Republic of China
| | - Ruichang Gao
- School of Food and Biological Engineering , Jiangsu University , 301 Xuefu Road , Zhenjiang , Jiangsu 212013 , People's Republic of China
| | - Yeonhwa Park
- Department of Food Science , University of Massachusetts, Amherst , 102 Holdsworth Way , Amherst , Massachusetts 01003 , United States
| |
Collapse
|
328
|
Dall KB, Færgeman NJ. Metabolic regulation of lifespan from a C. elegans perspective. GENES & NUTRITION 2019; 14:25. [PMID: 31428207 PMCID: PMC6694653 DOI: 10.1186/s12263-019-0650-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023]
Abstract
Decline of cellular functions especially cognitive is a major deficit that arises with age in humans. Harnessing the strengths of small and genetic tractable model systems has revealed key conserved regulatory biochemical and signaling pathways that control aging. Here, we review some of the key signaling and biochemical pathways that coordinate aging processes with special emphasis on Caenorhabditis elegans as a model system and discuss how nutrients and metabolites can regulate lifespan by coordinating signaling and epigenetic programs. We focus on central nutrient-sensing pathways such as mTOR and insulin/insulin-like growth factor signaling and key transcription factors including the conserved basic helix-loop-helix transcription factor HLH-30/TFEB.
Collapse
Affiliation(s)
- Kathrine B. Dall
- Department of Biochemistry and Molecular Biology, Villum Center for Bioanalytical Sciences, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Nils J. Færgeman
- Department of Biochemistry and Molecular Biology, Villum Center for Bioanalytical Sciences, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| |
Collapse
|
329
|
Jia R, Zhang J, Jia K. Neuroendocrine regulation of fat metabolism by autophagy gene atg-18 in C. elegans dauer larvae. FEBS Open Bio 2019; 9:1623-1631. [PMID: 31368651 PMCID: PMC6722879 DOI: 10.1002/2211-5463.12708] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/10/2019] [Accepted: 07/31/2019] [Indexed: 12/21/2022] Open
Abstract
In environments with limited food and high population density, Caenorhabditis elegans larvae may enter the dauer stage, in which metabolism is shifted to fat accumulation to allow larvae to survive for months without food. Mutations in the insulin‐like receptor gene daf‐2 force C. elegans to constitutively form dauer larva at higher temperature. It has been reported that autophagy is required for fat accumulation in daf‐2 dauer larva. However, the mechanism underlying this process remains unknown. Here, we report that autophagy gene atg‐18 acts in a cell nonautonomous manner in neurons and intestinal cells to mediate the influence of daf‐2 signaling on fat metabolism. Moreover, ATG‐18 in chemosensory neurons plays a vital role in this metabolic process. Finally, we report that neuronal ATG‐18 functions through neurotransmitters to control fat storage in daf‐2 dauers, which suggests an essential role of autophagy in the neuroendocrine regulation of fat metabolism by insulin‐like signaling.
Collapse
Affiliation(s)
- Ray Jia
- Department of Biological Sciences, Florida Atlantic University, Jupiter, FL, USA
| | - Jiuli Zhang
- Department of Biological Sciences, Florida Atlantic University, Jupiter, FL, USA
| | - Kailiang Jia
- Department of Biological Sciences, Florida Atlantic University, Jupiter, FL, USA
| |
Collapse
|
330
|
Lin YR, Parikh H, Park Y. Stress resistance and lifespan enhanced by downregulation of antimicrobial peptide genes in the Imd pathway. Aging (Albany NY) 2019; 10:622-631. [PMID: 29677000 PMCID: PMC5940113 DOI: 10.18632/aging.101417] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 04/10/2018] [Indexed: 12/22/2022]
Abstract
Biological behaviors and longevity of ectothermic animals are remarkably influenced by ambient temperature. Development at 18°C significantly enhances the stress resistance of adult flies with more accumulation of nutrients (especially fat) in the body than development at 25°C. Gene expression analysis between the flies developed at 18°C and 25°C revealed that the Immune deficiency (Imd) pathway, including the downstream antimicrobial peptides (AMPs), is downregulated in the flies developed at 18°C. When hypomorphic imd mutant flies with reduced AMP expressions were developed at 25°C, they showed induced stress resistance with higher fat content in the body similar to the wild-type flies developed at 18°C. However, severe hypomorphic imd mutants could not enhance stress resistance due to the downregulation of another downstream JNK pathway that expresses stress tolerance genes. Interestingly, the downregulation of AMP genes, itself, extended lifespan with increased stress resistance. Especially, fat body-specific downregulation of Imd AMP genes exhibited a longer lifespan with higher heat resistance. The fat body is known to function in metabolic homeostasis, stress tolerance, growth, and longevity in Drosophila. Here, we provide the first evidence that mild downregulation of the Imd pathway with AMP genes increases fat content, stress resistance, and lifespan in adult flies.
Collapse
Affiliation(s)
- Yuh-Ru Lin
- Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark, NJ 07103, USA.,Present address: Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Hardik Parikh
- Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark, NJ 07103, USA.,Present address: Institute of Ophthalmology and Visual Science, Rutgers, State University of New Jersey, Newark, NJ 07103, USA
| | - Yongkyu Park
- Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
331
|
Maruzs T, Simon-Vecsei Z, Kiss V, Csizmadia T, Juhász G. On the Fly: Recent Progress on Autophagy and Aging in Drosophila. Front Cell Dev Biol 2019; 7:140. [PMID: 31396511 PMCID: PMC6667644 DOI: 10.3389/fcell.2019.00140] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/09/2019] [Indexed: 01/03/2023] Open
Abstract
Autophagy ensures the lysosome-mediated breakdown and recycling of self-material, as it not only degrades obsolete or damaged intracellular constituents but also provides building blocks for biosynthetic and energy producing reactions. Studies in animal models including Drosophila revealed that autophagy defects lead to the rapid decline of neuromuscular function, neurodegeneration, sensitivity to stress (such as starvation or oxidative damage), and stem cell loss. Of note, recently identified human Atg gene mutations cause similar symptoms including ataxia and mental retardation. Physiologically, autophagic degradation (flux) is known to decrease during aging, and this defect likely contributes to the development of such age-associated diseases. Many manipulations that extend lifespan (including dietary restriction, reduced TOR kinase signaling, exercise or treatment with various anti-aging substances) require autophagy for their beneficial effect on longevity, pointing to the key role of this housekeeping process. Importantly, genetic (e.g., Atg8a overexpression in either neurons or muscle) or pharmacological (e.g., feeding rapamycin or spermidine to animals) promotion of autophagy has been successfully used to extend lifespan in Drosophila, suggesting that this intracellular degradation pathway can rejuvenate cells and organisms. In this review, we highlight key discoveries and recent progress in understanding the relationship of autophagy and aging in Drosophila.
Collapse
Affiliation(s)
- Tamás Maruzs
- Institute of Genetics, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Zsófia Simon-Vecsei
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, Hungary
| | - Viktória Kiss
- Institute of Genetics, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Tamás Csizmadia
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, Hungary
| | - Gábor Juhász
- Institute of Genetics, Biological Research Centre of the Hungarian Academy of Sciences, Szeged, Hungary.,Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
332
|
Nagashima T, Iino Y, Tomioka M. DAF-16/FOXO promotes taste avoidance learning independently of axonal insulin-like signaling. PLoS Genet 2019; 15:e1008297. [PMID: 31323047 PMCID: PMC6668909 DOI: 10.1371/journal.pgen.1008297] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 07/31/2019] [Accepted: 07/08/2019] [Indexed: 12/12/2022] Open
Abstract
The avoidance of starvation is critical for the survival of most organisms, thus animals change behavior based on past nutritional conditions. Insulin signaling is important for nutritional state-dependent behavioral plasticity, yet the underlying regulatory mechanism at the cellular level remains unclear. Previous studies showed that insulin-like signaling is required for taste avoidance learning, in which the nematode Caenorhabditis elegans avoids salt concentrations encountered under starvation conditions. DAF-2c, a splice isoform of the DAF-2 insulin receptor, functions in the axon of the ASER sensory neuron, which senses changes in salt concentrations. In addition, mutants of a major downstream factor of DAF-2, the forkhead transcription factor O (FOXO) homolog DAF-16, show defects in taste avoidance learning. Interestingly, the defect of the daf-2 mutant is not suppressed by daf-16 mutations in the learning, unlike those in other phenomena, such as longevity and development. Here we show that multiple DAF-16 isoforms function in ASER. By epistasis analysis using a DAF-2c isoform-specific mutant and an activated form of DAF-16, we found that DAF-16 acts in the nucleus in parallel with the DAF-2c-dependent pathway in the axon, indicating that insulin-like signaling acts both in the cell body and axon of a single neuron, ASER. Starvation conditioning induces nuclear translocation of DAF-16 in ASER and degradation of DAF-16 before starvation conditioning causes defects in taste avoidance learning. Forced nuclear localization of DAF-16 in ASER biased chemotaxis towards lower salt concentrtions and this effect required the Gq/PKC pathway and neuropeptide processing enzymes. These data imply that DAF-16/FOXO transmits starvation signals and modulates neuropeptide transmission in the learning. Animals change behavior based on remembered experiences of hunger and appetite. Signaling by insulin and insulin-like peptides in the nervous system plays key roles in behavioral responses to hunger and satiety. In C. elegans, insulin-like signaling in the gustatory sensory neuron ASER regulates learned avoidance of salt concentrations experienced during fasting, which we call taste avoidance learning. DAF-2c, an isoform of the insulin receptor homolog, is localized to the axon of ASER and regulates taste avoidance learning. Here, we show that DAF-16, the forkhead transcription factor O (FOXO) homolog, translocates into the nucleus of ASER during fasting and promotes taste avoidance learning. DAF-16 is negatively regulated by insulin-like signaling independently of axonal DAF-2c signaling. This dual function of insulin-like signaling in the cell body and the axon ensures dynamic changes in behavioral responses after experience of hunger. By genetic analyses using constitutively nuclear-translocated DAF-16, we show that DAF-16 in ASER regulates taste avoidance learning via modulating neuropeptide signaling in the nervous system, which is reminiscent of the function of FOXO in the hypothalamus in the regulation of food-seeking behavior in mammals.
Collapse
Affiliation(s)
- Takashi Nagashima
- 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
| | - Masahiro Tomioka
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- * E-mail:
| |
Collapse
|
333
|
Zymolytic Grain Extract (ZGE) Significantly Extends the Lifespan and Enhances the Environmental Stress Resistance of Caenorhabditis elegans. Int J Mol Sci 2019; 20:ijms20143489. [PMID: 31315221 PMCID: PMC6678847 DOI: 10.3390/ijms20143489] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/11/2019] [Accepted: 07/12/2019] [Indexed: 12/16/2022] Open
Abstract
Many reports have shown that grains play an important role in our daily lives and can provide energy and nutrients to protect us from various diseases, and they are considered to be indispensable parts of our lives. It has been reported that some constituents in grains could exert functional effects against HIV infections and multiple cancers. Zymolytic grain can produce some new useful molecules and thus support the cell nutrients in the human body. In this study, the effects of zymolytic grain extract (ZGE) supernatants on the changes of nematode indicators were investigated, including lifespan, self-brood size, and body length in environmental conditions (temperature, ultraviolet radiation or 5-fluoro-2′-deoxyuridine (FUDR) stimuli). We found that, compared to the control group, the ZGE supernatant-feeding group could prolong the lifespan of nematodes under normal conditions. More importantly, ZGE supernatants could improve the ability of nematodes to resist stress. When the concentration of FUDR was 400 or 50 μM, the ZGE supernatant-feeding group could prolong lifespan by an average of 38.4% compared to the control group, and the eggs of the ZGE supernatant-feeding group could hatch and develop into adults. These results indicated that ZGE could protect C. elegans from external stress and thus prolong their lifespan and improve the physiological state of nematodes. Therefore, ZGE supernatant has potential to be used as a nutritional product in antioxidant and anti-aging research.
Collapse
|
334
|
Miller DL. There Are Worms in My Aging Research! J Gerontol A Biol Sci Med Sci 2019; 74:1170-1172. [PMID: 31056635 DOI: 10.1093/gerona/glz106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Indexed: 11/13/2022] Open
Affiliation(s)
- Dana L Miller
- Department of Biochemistry, University of Washington School of Medicine, Seattle
| |
Collapse
|
335
|
Taormina G, Ferrante F, Vieni S, Grassi N, Russo A, Mirisola MG. Longevity: Lesson from Model Organisms. Genes (Basel) 2019; 10:genes10070518. [PMID: 31324014 PMCID: PMC6678192 DOI: 10.3390/genes10070518] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/31/2022] Open
Abstract
Research on longevity and healthy aging promises to increase our lifespan and decrease the burden of degenerative diseases with important social and economic effects. Many aging theories have been proposed, and important aging pathways have been discovered. Model organisms have had a crucial role in this process because of their short lifespan, cheap maintenance, and manipulation possibilities. Yeasts, worms, fruit flies, or mammalian models such as mice, monkeys, and recently, dogs, have helped shed light on aging processes. Genes and molecular mechanisms that were found to be critical in simple eukaryotic cells and species have been confirmed in humans mainly by the functional analysis of mammalian orthologues. Here, we review conserved aging mechanisms discovered in different model systems that are implicated in human longevity as well and that could be the target of anti-aging interventions in human.
Collapse
Affiliation(s)
- Giusi Taormina
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università di Palermo, Via del Vespro 129, 90100 Palermo, Italy
| | - Federica Ferrante
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università di Palermo, Via del Vespro 129, 90100 Palermo, Italy
| | - Salvatore Vieni
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università di Palermo, Via del Vespro 129, 90100 Palermo, Italy
| | - Nello Grassi
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università di Palermo, Via del Vespro 129, 90100 Palermo, Italy
| | - Antonio Russo
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università di Palermo, Via del Vespro 129, 90100 Palermo, Italy
| | - Mario G Mirisola
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università di Palermo, Via del Vespro 129, 90100 Palermo, Italy.
| |
Collapse
|
336
|
Su L, Li H, Huang C, Zhao T, Zhang Y, Ba X, Li Z, Zhang Y, Huang B, Lu J, Zhao Y, Li X. Muscle-Specific Histone H3K36 Dimethyltransferase SET-18 Shortens Lifespan of Caenorhabditis elegans by Repressing daf-16a Expression. Cell Rep 2019. [PMID: 29514099 DOI: 10.1016/j.celrep.2018.02.029] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Mounting evidence shows that histone methylation, a typical epigenetic mark, is crucial for gene expression regulation during aging. Decreased trimethylation of Lys 36 on histone H3 (H3K36me3) in worms and yeast is reported to shorten lifespan. The function of H3K36me2 in aging remains unclear. In this study, we identified Caenorhabditis elegans SET-18 as a histone H3K36 dimethyltransferase. SET-18 deletion extended lifespan and increased oxidative stress resistance, dependent on daf-16 activity in the insulin/IGF pathway. In set-18 mutants, transcription of daf-16 isoform a (daf-16a) was specifically upregulated. Accordingly, a decrease in H3K36me2 on daf-16a promoter was observed. Muscle-specific expression of SET-18 increased in aged worms (day 7 and day 11), attributable to elevation of global H3K36me2 and inhibition of daf-16a expression. Consequently, longevity was shortened. These findings suggested that chromatic repression mediated by tissue-specific H3K36 dimethyltransferase might be detrimental to lifespan and may have implications in human age-related diseases.
Collapse
Affiliation(s)
- Liangping Su
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun 130024, China
| | - Hongyuan Li
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun 130024, China
| | - Cheng Huang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun 130024, China
| | - Tingting Zhao
- Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China
| | - Yongjun Zhang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun 130024, China
| | - Xueqing Ba
- Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China
| | - Zhongwei Li
- Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China
| | - Yu Zhang
- Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China
| | - Baiqu Huang
- Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China
| | - Jun Lu
- Institute of Genetics and Cytology, Northeast Normal University, Changchun 130024, China.
| | - Yanmei Zhao
- Key Laboratory of RNA Biology, Beijing Key Laboratory of Noncoding RNA, Institute of Biophysics, CAS Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing 100101, China.
| | - Xiaoxue Li
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal University, Changchun 130024, China.
| |
Collapse
|
337
|
Yasuda K, Sakamoto K. Oxytocin promotes heat stress tolerance via insulin signals in Caenorhabditis elegans. Biosci Biotechnol Biochem 2019; 83:1858-1866. [PMID: 31198094 DOI: 10.1080/09168451.2019.1630253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Oxytocin, has various physiological functions that have been well studied and many that remain unknown. Here, we aimed to determine new physiological functions of oxytocin using Caenorhabditis elegans. Oxytocin treatment promoted the restoration of movement after heat stress and enhanced the viability under heat stress. However, oxytocin had no effect on the life span and only little effect on the oxidative stress tolerance. In contrast, oxytocin treatment didn't promote the restoration of movement or enhance the viability of deficient mutants of ntr-1/2, which is the gene encoding the oxytocin receptor. In addition, for mutants of daf-16, daf-2, tax-4, and some insulin-like peptides, the heat stress tolerance effect by oxytocin was canceled. Furthermore, oxytocin increased the expression levels of the DAF-16 target genes. Our results suggest that oxytocin treatment promoted the heat stress tolerance of C. elegans via the insulin/IGF-1 signaling pathway.
Collapse
Affiliation(s)
- Kensuke Yasuda
- Graduate School of Life and Environmental Sciences, University of Tsukuba , Tsukuba , Japan
| | - Kazuichi Sakamoto
- Graduate School of Life and Environmental Sciences, University of Tsukuba , Tsukuba , Japan
| |
Collapse
|
338
|
Niedernhofer LJ, Gurkar AU, Wang Y, Vijg J, Hoeijmakers JHJ, Robbins PD. Nuclear Genomic Instability and Aging. Annu Rev Biochem 2019; 87:295-322. [PMID: 29925262 DOI: 10.1146/annurev-biochem-062917-012239] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The nuclear genome decays as organisms age. Numerous studies demonstrate that the burden of several classes of DNA lesions is greater in older mammals than in young mammals. More challenging is proving this is a cause rather than a consequence of aging. The DNA damage theory of aging, which argues that genomic instability plays a causal role in aging, has recently gained momentum. Support for this theory stems partly from progeroid syndromes in which inherited defects in DNA repair increase the burden of DNA damage leading to accelerated aging of one or more organs. Additionally, growing evidence shows that DNA damage accrual triggers cellular senescence and metabolic changes that promote a decline in tissue function and increased susceptibility to age-related diseases. Here, we examine multiple lines of evidence correlating nuclear DNA damage with aging. We then consider how, mechanistically, nuclear genotoxic stress could promote aging. We conclude that the evidence, in toto, supports a role for DNA damage as a nidus of aging.
Collapse
Affiliation(s)
- Laura J Niedernhofer
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute Florida, Jupiter, Florida 33458, USA;
| | - Aditi U Gurkar
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute Florida, Jupiter, Florida 33458, USA; .,Department of Medicine, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Yinsheng Wang
- Department of Chemistry, University of California, Riverside, California 92521, USA
| | - Jan Vijg
- Department of Genetics, Albert Einstein College of Medicine, Michael F. Price Center, Bronx, New York 10461, USA
| | - Jan H J Hoeijmakers
- Department of Molecular Genetics, Erasmus University Medical Center, 3015 CE Rotterdam, The Netherlands
| | - Paul D Robbins
- Department of Molecular Medicine and the Center on Aging, The Scripps Research Institute Florida, Jupiter, Florida 33458, USA;
| |
Collapse
|
339
|
Kadekar P, Roy R. AMPK regulates germline stem cell quiescence and integrity through an endogenous small RNA pathway. PLoS Biol 2019; 17:e3000309. [PMID: 31166944 PMCID: PMC6576793 DOI: 10.1371/journal.pbio.3000309] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 06/17/2019] [Accepted: 05/16/2019] [Indexed: 01/07/2023] Open
Abstract
During suboptimal growth conditions, Caenorhabditis elegans larvae undergo a global developmental arrest called "dauer." During this stage, the germline stem cells (GSCs) become quiescent in an AMP-activated Protein Kinase (AMPK)-dependent manner, and in the absence of AMPK, the GSCs overproliferate and lose their reproductive capacity, leading to sterility when mutant animals resume normal growth. These defects correlate with the altered abundance and distribution of a number of chromatin modifications, all of which can be corrected by disabling components of the endogenous small RNA pathway, suggesting that AMPK regulates germ cell integrity by targeting an RNA interference (RNAi)-like pathway during dauer. The expression of AMPK in somatic cells restores all the germline defects, potentially through the transmission of small RNAs. Our findings place AMPK at a pivotal position linking energy stress detected in the soma to a consequent endogenous small RNA-mediated adaptation in germline gene expression, thereby challenging the "permeability" of the Weismann barrier.
Collapse
Affiliation(s)
- Pratik Kadekar
- Department of Biology, McGill University, Montreal, Quebec, Canada
| | - Richard Roy
- Department of Biology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
340
|
Chen W, Wang J, Shi J, Yang X, Yang P, Wang N, Yang S, Xie T, Yang H, Zhang M, Wang H, Fei J. Longevity Effect of Liuwei Dihuang in Both Caenorhabditis Elegans and Aged Mice. Aging Dis 2019; 10:578-591. [PMID: 31165002 PMCID: PMC6538212 DOI: 10.14336/ad.2018.0604] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 06/04/2018] [Indexed: 01/20/2023] Open
Abstract
Liuwei Dihuang (LWDH), a famous traditional Chinese medicine, is widely used in the clinical treatment of aging-related diseases in China. However, its pharmacological mechanisms are not clear. In the present study, we evaluated the lifespan extension effect of LWDH in C. elegans and mice and revealed its underlying mechanisms. The results showed that LWDH significantly extended the lifespan of C. elegans in a dose-dependent manner. LWDH also conferred protection to nematodes against oxidative stress and reduced their fat storage. Genetics analysis and microarray data showed that the longevity effect of LWDH was attributed to the regulation of the innate immune response, proteolysis, lipid metabolism, and the oxidation-reduction process and was dependent on daf-16. Among the six herbs in the formula, Radix Rehmanniae Preparata and Fructus Macrocarpii contributed most to the longevity effect of this medicine, while the other four components had a synergistic effect on the longevity effect of the prescription. The lack of any single herb reduced the efficacy of the complete formula. LWDH also extended the lifespan and reduced both the weight and oxidant stress status in aged mice. Taken together, these results suggested that LWDH might function in a multi-target manner to extend the lifespan in both C. elegans and aged mice, and the best effect was achieved with the complete formula.
Collapse
Affiliation(s)
- Weidong Chen
- 1School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Jinzeng Wang
- 1School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Jiahao Shi
- 1School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Xu Yang
- 3School of Medicine, Tongji University, Shanghai 200092, China
| | - Ping Yang
- 4Shanghai Engineering Research Center for Model Organisms, SRMOC/SMOC, Shanghai 201203, China
| | - Ning Wang
- 1School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Sai Yang
- 1School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Tianpei Xie
- 5Standard Testing Lab (Shanghai) Co., Ltd., Pudong, Shanghai 201203, China
| | - Hua Yang
- 1School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Mengjie Zhang
- 1School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Haiyun Wang
- 1School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Jian Fei
- 1School of Life Science and Technology, Tongji University, Shanghai 200092, China.,2Shenqi Institute for Ethnomedicine, Tongji University, Shanghai 200092, China
| |
Collapse
|
341
|
Nagy D, Cusumano P, Andreatta G, Anduaga AM, Hermann-Luibl C, Reinhard N, Gesto J, Wegener C, Mazzotta G, Rosato E, Kyriacou CP, Helfrich-Förster C, Costa R. Peptidergic signaling from clock neurons regulates reproductive dormancy in Drosophila melanogaster. PLoS Genet 2019; 15:e1008158. [PMID: 31194738 PMCID: PMC6592559 DOI: 10.1371/journal.pgen.1008158] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 06/25/2019] [Accepted: 04/25/2019] [Indexed: 11/18/2022] Open
Abstract
With the approach of winter, many insects switch to an alternative protective developmental program called diapause. Drosophila melanogaster females overwinter as adults by inducing a reproductive arrest that is characterized by inhibition of ovarian development at previtellogenic stages. The insulin producing cells (IPCs) are key regulators of this process, since they produce and release insulin-like peptides that act as diapause-antagonizing hormones. Here we show that in D. melanogaster two neuropeptides, Pigment Dispersing Factor (PDF) and short Neuropeptide F (sNPF) inhibit reproductive arrest, likely through modulation of the IPCs. In particular, genetic manipulations of the PDF-expressing neurons, which include the sNPF-producing small ventral Lateral Neurons (s-LNvs), modulated the levels of reproductive dormancy, suggesting the involvement of both neuropeptides. We expressed a genetically encoded cAMP sensor in the IPCs and challenged brain explants with synthetic PDF and sNPF. Bath applications of both neuropeptides increased cAMP levels in the IPCs, even more so when they were applied together, suggesting a synergistic effect. Bath application of sNPF additionally increased Ca2+ levels in the IPCs. Our results indicate that PDF and sNPF inhibit reproductive dormancy by maintaining the IPCs in an active state.
Collapse
Affiliation(s)
- Dóra Nagy
- Department of Biology, University of Padova, Padova, Italy
| | - Paola Cusumano
- Department of Biology, University of Padova, Padova, Italy
| | | | - Ane Martin Anduaga
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Christiane Hermann-Luibl
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Nils Reinhard
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - João Gesto
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Christian Wegener
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | | | - Ezio Rosato
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Charalambos P. Kyriacou
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Charlotte Helfrich-Förster
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Rodolfo Costa
- Department of Biology, University of Padova, Padova, Italy
| |
Collapse
|
342
|
Lai WF, Lin M, Wong WT. Tackling Aging by Using miRNA as a Target and a Tool. Trends Mol Med 2019; 25:673-684. [PMID: 31126873 DOI: 10.1016/j.molmed.2019.04.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/12/2019] [Accepted: 04/17/2019] [Indexed: 12/15/2022]
Abstract
miRNA is a class of short noncoding RNA that regulates gene expression at the post-transcriptional level. Evidence of age-associated changes in miRNA expression has been collected in models ranging from nematodes to humans; however, there has been little discussion of how to turn our knowledge of miRNA biology into antiaging therapy. This opinion article provides a snapshot of our current understanding of the roles of miRNA in modulating the aging process. We discuss major chemical techniques for modifying the miRNA structure as well as developing delivery systems for intervention. Finally, technical needs to be met for bench-to-clinic translation of miRNA-based interventions are highlighted for future research.
Collapse
Affiliation(s)
- Wing-Fu Lai
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China; Health Science Centre, Shenzhen University, Shenzhen, China.
| | - Marie Lin
- Health Science Centre, Shenzhen University, Shenzhen, China
| | - Wing-Tak Wong
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| |
Collapse
|
343
|
Pheromones and Nutritional Signals Regulate the Developmental Reliance on let-7 Family MicroRNAs in C. elegans. Curr Biol 2019; 29:1735-1745.e4. [PMID: 31104929 DOI: 10.1016/j.cub.2019.04.034] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/03/2019] [Accepted: 04/11/2019] [Indexed: 12/31/2022]
Abstract
Adverse environmental conditions can affect rates of animal developmental progression and lead to temporary developmental quiescence (diapause), exemplified by the dauer larva stage of the nematode Caenorhabditis elegans (C. elegans). Remarkably, patterns of cell division and temporal cell-fate progression in C. elegans larvae are not affected by changes in developmental trajectory. However, the underlying physiological and gene regulatory mechanisms that ensure robust developmental patterning despite substantial plasticity in developmental progression are largely unknown. Here, we report that diapause-inducing pheromones correct heterochronic developmental cell lineage defects caused by insufficient expression of let-7 family microRNAs in C. elegans. Moreover, two conserved endocrine signaling pathways, DAF-7/TGF-β and DAF-2/Insulin, that confer on the larva diapause and non-diapause alternative developmental trajectories interact with the nuclear hormone receptor, DAF-12, to initiate and regulate a rewiring of the genetic circuitry controlling temporal cell fates. This rewiring includes engagement of certain heterochronic genes, lin-46, lin-4, and nhl-2, that are previously associated with an altered genetic program in post-diapause animals, in combination with a novel ligand-independent DAF-12 activity, to downregulate the critical let-7 family target Hunchback-like-1 (HBL-1). Our results show how pheromone or endocrine signaling pathways can coordinately regulate both developmental progression and cell-fate transitions in C. elegans larvae under stress so that the developmental schedule of cell fates remains unaffected by changes in developmental trajectory.
Collapse
|
344
|
Fields BD, Nguyen SC, Nir G, Kennedy S. A multiplexed DNA FISH strategy for assessing genome architecture in Caenorhabditis elegans. eLife 2019; 8:42823. [PMID: 31084706 PMCID: PMC6516958 DOI: 10.7554/elife.42823] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 04/02/2019] [Indexed: 02/06/2023] Open
Abstract
Eukaryotic DNA is highly organized within nuclei and this organization is important for genome function. Fluorescent in situ hybridization (FISH) approaches allow 3D architectures of genomes to be visualized. Scalable FISH technologies, which can be applied to whole animals, are needed to help unravel how genomic architecture regulates, or is regulated by, gene expression during development, growth, reproduction, and aging. Here, we describe a multiplexed DNA FISH Oligopaint library that targets the entire Caenorhabditis elegans genome at chromosome, three megabase, and 500 kb scales. We describe a hybridization strategy that provides flexibility to DNA FISH experiments by coupling a single primary probe synthesis reaction to dye conjugated detection oligos via bridge oligos, eliminating the time and cost typically associated with labeling probe sets for individual experiments. The approach allows visualization of genome organization at varying scales in all/most cells across all stages of development in an intact animal model system. DNA contains the instructions needed to build and maintain a living organism. How DNA is physically arranged inside a cell is not random, and DNA organization is important because it can affect, for example, which genes are active, and which are not. Researchers often use a technique called “fluorescence in situ hybridization” (or FISH for short) to study how DNA is organized in cells. FISH tethers fluorescent molecules to defined sections of DNA, making those sections glow under the right wavelength of light. It is possible to collect images of the fluorescent DNA regions under a microscope to see where they are in relation to each other and to the rest of the cell. Fields, Nguyen et al. have now created a new library of FISH molecules that can be used to analyze the DNA of a microscopic worm known as Caenorhabditis elegans – a model organism that is widely used to study genetics, animal development, and cell biology. The library can be used to visualize the worm’s whole genome at different scales. The library enables accurate and reliable investigations of how DNA is organized inside C. elegans, including in intact worms, meaning it also offers the first chance to study DNA organization in a whole organism through all stages of its life cycle. This new resource could help to reveal the relationships between DNA organization, cell specialization and gene activity in different cells at different stages of development. This could help to clarify the relationships between physical DNA organization and biological change. This design strategy behind this whole genome library should also be adaptable for similar studies in other animal species.
Collapse
Affiliation(s)
- Brandon D Fields
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, United States.,Department of Genetics, Harvard Medical School, Boston, United States
| | - Son C Nguyen
- Department of Genetics, Harvard Medical School, Boston, United States
| | - Guy Nir
- Department of Genetics, Harvard Medical School, Boston, United States
| | - Scott Kennedy
- Department of Genetics, Harvard Medical School, Boston, United States
| |
Collapse
|
345
|
Tanisawa K, Hirose N, Arai Y, Shimokata H, Yamada Y, Kawai H, Kojima M, Obuchi S, Hirano H, Suzuki H, Fujiwara Y, Taniguchi Y, Shinkai S, Ihara K, Sugaya M, Higuchi M, Arai T, Mori S, Sawabe M, Sato N, Muramatsu M, Tanaka M. Inverse Association Between Height-Increasing Alleles and Extreme Longevity in Japanese Women. J Gerontol A Biol Sci Med Sci 2019; 73:588-595. [PMID: 28958036 DOI: 10.1093/gerona/glx155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 08/16/2017] [Indexed: 01/07/2023] Open
Abstract
Growth hormone (GH)/insulin-like growth factor-1 (IGF-1)/insulin signaling is one of the most plausible biological pathways regulating aging and longevity. Previous studies have demonstrated that several single nucleotide polymorphisms (SNPs) in the GH/IGF-1/insulin signaling-associated genes influence both longevity and adult height, suggesting the possibility of a shared genetic architecture between longevity and height. We therefore examined the relationship between 30 height-associated SNPs and extreme longevity in a Japanese population consisting of 428 centenarians and 4,026 younger controls. We confirmed that height-increasing genetic scores (HGSs) constructed based on 30 SNPs were significantly associated with height in the controls (p = 6.95 × 10-23). HGS was significantly and inversely associated with extreme longevity in women (p = .011), but not in men, although no SNPs were significantly associated with extreme longevity after Bonferroni correction. The odds ratio for extreme longevity in the lowest HGS group (≤27) and the second lowest HGS group (28-30) relative to the highest HGS group (≥37) was 1.71 (p = .056) and 1.69 (p = .034), respectively, for women. In conclusion, the present study demonstrated an inverse association between height-increasing alleles with extreme longevity in Japanese women, providing novel insight into the genetic architecture of longevity and aging.
Collapse
Affiliation(s)
- Kumpei Tanisawa
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo, Japan.,Department of Molecular Gerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan.,Faculty of Sport Sciences, Waseda University, Tokorozawa, Japan.,Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Nobuyoshi Hirose
- Center for Supercentenarian Research, Keio University School of Medicine, Tokyo, Japan
| | - Yasumichi Arai
- Center for Supercentenarian Research, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Shimokata
- Section of Longitudinal Study of Aging, National Institute for Longevity Sciences (NILS-LSA), National Center for Geriatrics and Gerontology, Obu, Japan.,Graduate School of Nutritional Sciences, Nagoya University of Arts and Sciences, Nisshin, Japan
| | - Yoshiji Yamada
- Department of Human Functional Genomics, Advanced Science Research Promotion Center, Mie University, Tsu, Japan
| | - Hisashi Kawai
- Human Care Research Team, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Motonaga Kojima
- Human Care Research Team, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Shuichi Obuchi
- Human Care Research Team, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Hirohiko Hirano
- Research Team for Promoting Independence of the Elderly, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Hiroyuki Suzuki
- Research Team for Social Participation and Community Health, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Yoshinori Fujiwara
- Research Team for Social Participation and Community Health, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Yu Taniguchi
- Research Team for Social Participation and Community Health, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Shoji Shinkai
- Research Team for Social Participation and Community Health, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Kazushige Ihara
- Department of Public Health, Toho University School of Medicine, Tokyo, Japan
| | - Maki Sugaya
- Department of Molecular Gerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Mitsuru Higuchi
- Faculty of Sport Sciences, Waseda University, Tokorozawa, Japan.,Institute of Advanced Active Aging Research, Waseda University, Tokorozawa, Japan
| | - Tomio Arai
- Department of Pathology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Seijiro Mori
- Center for Promotion of Clinical Investigation, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Motoji Sawabe
- Section of Molecular Pathology, Graduate School of Health Care Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Noriko Sato
- Department of Molecular Epidemiology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masaaki Muramatsu
- Department of Molecular Epidemiology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masashi Tanaka
- Department of Clinical Laboratory, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| |
Collapse
|
346
|
Griffin EF, Yan X, Caldwell KA, Caldwell GA. Distinct functional roles of Vps41-mediated neuroprotection in Alzheimer's and Parkinson's disease models of neurodegeneration. Hum Mol Genet 2019; 27:4176-4193. [PMID: 30508205 DOI: 10.1093/hmg/ddy308] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 08/21/2018] [Indexed: 12/25/2022] Open
Abstract
Commonalities and, in some cases, pathological overlap between neurodegenerative diseases have led to speculation that targeting of underlying mechanisms might be of potentially shared therapeutic benefit. Alzheimer's disease is characterized by the formation of plaques, composed primarily of the amyloid-β 1-42 (Aβ) peptide in the brain, resulting in neurodegeneration. Previously, we have shown that overexpression of the lysosomal-trafficking protein, human Vps41 (hVps41), is neuroprotective in a transgenic worm model of Parkinson's disease, wherein progressive dopaminergic neurodegeneration is induced by α-synuclein overexpression. Here, we report the results of a systematic comparison of hVps41-mediated neuroprotection between α-synuclein and Aβ in transgenic nematode models of Caenorhabditis elegans. Our results indicate that an ARF-like GTPase gene product, ARL-8, mitigates endocytic Aβ neurodegeneration in a VPS-41-dependent manner, rather than through RAB-7 and AP3 as with α-synuclein. Furthermore, the neuroprotective effect of ARL-8 or hVps41 appears to be dependent on their colocalization and the activity of ARL-8. Additionally, we demonstrate that the LC3 orthologue, LGG-2, plays a critical role in Aβ toxicity with ARL-8. Further analysis of functional effectors of Aβ protein processing via the lysosomal pathway will assist in the elucidation of the underlying mechanism involving VPS-41-mediated neuroprotection. These results reveal functional distinctions in the intracellular management of neurotoxic proteins that serve to better inform the path for development of therapeutic interventions to halt neurodegeneration.
Collapse
Affiliation(s)
- Edward F Griffin
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | | | - Kim A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, USA.,Departments of Neurology and Neurobiology, Center for Neurodegeneration and Experimental Therapeutics, Nathan Shock Center for Research on the Basic Biology of Aging, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Guy A Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, USA.,Departments of Neurology and Neurobiology, Center for Neurodegeneration and Experimental Therapeutics, Nathan Shock Center for Research on the Basic Biology of Aging, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| |
Collapse
|
347
|
The C. elegans intestine: organogenesis, digestion, and physiology. Cell Tissue Res 2019; 377:383-396. [DOI: 10.1007/s00441-019-03036-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 04/12/2019] [Indexed: 12/16/2022]
|
348
|
Zečić A, Dhondt I, Braeckman BP. The nutritional requirements of Caenorhabditis elegans. GENES AND NUTRITION 2019; 14:15. [PMID: 31080524 PMCID: PMC6501307 DOI: 10.1186/s12263-019-0637-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 04/10/2019] [Indexed: 12/14/2022]
Abstract
Animals require sufficient intake of a variety of nutrients to support their development, somatic maintenance and reproduction. An adequate diet provides cell building blocks, chemical energy to drive cellular processes and essential nutrients that cannot be synthesised by the animal, or at least not in the required amounts. Dietary requirements of nematodes, including Caenorhabditis elegans have been extensively studied with the major aim to develop a chemically defined axenic medium that would support their growth and reproduction. At the same time, these studies helped elucidating important aspects of nutrition-related biochemistry and metabolism as well as the establishment of C. elegans as a powerful model in studying evolutionarily conserved pathways, and the influence of the diet on health.
Collapse
Affiliation(s)
- Aleksandra Zečić
- Department of Biology, Laboratory of Aging Physiology and Molecular Evolution, Ghent University, 9000 Ghent, Belgium
| | - Ineke Dhondt
- Department of Biology, Laboratory of Aging Physiology and Molecular Evolution, Ghent University, 9000 Ghent, Belgium
| | - Bart P Braeckman
- Department of Biology, Laboratory of Aging Physiology and Molecular Evolution, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
349
|
Ng LF, Ng LT, van Breugel M, Halliwell B, Gruber J. Mitochondrial DNA Damage Does Not Determine C. elegans Lifespan. Front Genet 2019; 10:311. [PMID: 31031801 PMCID: PMC6473201 DOI: 10.3389/fgene.2019.00311] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 03/21/2019] [Indexed: 02/02/2023] Open
Abstract
The mitochondrial free radical theory of aging (mFRTA) proposes that accumulation of oxidative damage to macromolecules in mitochondria is a causative mechanism for aging. Accumulation of mitochondrial DNA (mtDNA) damage may be of particular interest in this context. While there is evidence for age-dependent accumulation of mtDNA damage, there have been only a limited number of investigations into mtDNA damage as a determinant of longevity. This lack of quantitative data regarding mtDNA damage is predominantly due to a lack of reliable assays to measure mtDNA damage. Here, we report adaptation of a quantitative real-time polymerase chain reaction (qRT-PCR) assay for the detection of sequence-specific mtDNA damage in C. elegans and apply this method to investigate the role of mtDNA damage in the aging of nematodes. We compare damage levels in old and young animals and also between wild-type animals and long-lived mutant strains or strains with modifications in ROS detoxification or production rates. We confirm an age-dependent increase in mtDNA damage levels in C. elegans but found that there is no simple relationship between mtDNA damage and lifespan. MtDNA damage levels were high in some mutants with long lifespan (and vice versa). We next investigated mtDNA damage, lifespan and healthspan effects in nematode subjected to exogenously elevated damage (UV- or γ-radiation induced). We, again, observed a complex relationship between damage and lifespan in such animals. Despite causing a significant elevation in mtDNA damage, γ-radiation did not shorten the lifespan of nematodes at any of the doses tested. When mtDNA damage levels were elevated significantly using UV-radiation, nematodes did suffer from shorter lifespan at the higher end of exposure tested. However, surprisingly, we also found hormetic lifespan and healthspan benefits in nematodes treated with intermediate doses of UV-radiation, despite the fact that mtDNA damage in these animals was also significantly elevated. Our results suggest that within a wide physiological range, the level of mtDNA damage does not control lifespan in C. elegans.
Collapse
Affiliation(s)
- Li Fang Ng
- Ageing Research Laboratory, Science Division, Yale-NUS College, Singapore, Singapore
| | - Li Theng Ng
- Ageing Research Laboratory, Science Division, Yale-NUS College, Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Michiel van Breugel
- Environmental Science Laboratory, Science Division, Yale-NUS College, Singapore, Singapore
| | - Barry Halliwell
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jan Gruber
- Ageing Research Laboratory, Science Division, Yale-NUS College, Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
350
|
Kumar S, Egan BM, Kocsisova Z, Schneider DL, Murphy JT, Diwan A, Kornfeld K. Lifespan Extension in C. elegans Caused by Bacterial Colonization of the Intestine and Subsequent Activation of an Innate Immune Response. Dev Cell 2019; 49:100-117.e6. [PMID: 30965033 PMCID: PMC6946027 DOI: 10.1016/j.devcel.2019.03.010] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 02/04/2019] [Accepted: 03/12/2019] [Indexed: 10/27/2022]
Abstract
Mechanisms that control aging are important yet poorly defined. To discover longevity control genes, we performed a forward genetic screen for delayed reproductive aging in C. elegans. Here, we show that am117 is a nonsense mutation in the phm-2 gene, which encodes a protein homologous to human scaffold attachment factor B. phm-2(lf) mutant worms have an abnormal pharynx grinder, which allows live bacteria to accumulate in the intestine. This defect shortens lifespan on highly pathogenic bacteria but extends lifespan and health span on the standard E. coli diet by activating innate immunity pathways that lead to bacterial avoidance behavior and dietary restriction. eat-2(lf) mutants displayed a similar phenotype, indicating accumulation of live bacteria also triggers extended longevity in this mutant. The analysis of phm-2 elucidates connections between pathogen response and aging by defining a mechanism of longevity extension in C. elegans-bacterial colonization, innate immune activation, and bacterial avoidance behavior.
Collapse
Affiliation(s)
- Sandeep Kumar
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Division of Bone & Mineral Diseases, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brian M Egan
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zuzana Kocsisova
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daniel L Schneider
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John T Murphy
- Center for Cardiovascular Research and Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Abhinav Diwan
- Center for Cardiovascular Research and Division of Cardiology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kerry Kornfeld
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|