151
|
Huang M, Hong M, Hou X, Zhu C, Chen D, Chen X, Guang S, Feng X. H3K9me1/2 methylation limits the lifespan of daf-2 mutants in C. elegans. eLife 2022; 11:74812. [PMID: 36125117 PMCID: PMC9514849 DOI: 10.7554/elife.74812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 09/16/2022] [Indexed: 11/29/2022] Open
Abstract
Histone methylation plays crucial roles in the development, gene regulation, and maintenance of stem cell pluripotency in mammals. Recent work shows that histone methylation is associated with aging, yet the underlying mechanism remains unclear. In this work, we identified a class of putative histone 3 lysine 9 mono/dimethyltransferase genes (met-2, set-6, set-19, set-20, set-21, set-32, and set-33), mutations in which induce synergistic lifespan extension in the long-lived DAF-2 (insulin growth factor 1 [IGF-1] receptor) mutant in Caenorhabditis elegans. These putative histone methyltransferase plus daf-2 double mutants not only exhibited an average lifespan nearly three times that of wild-type animals and a maximal lifespan of approximately 100 days, but also significantly increased resistance to oxidative and heat stress. Synergistic lifespan extension depends on the transcription factor DAF-16 (FOXO). mRNA-seq experiments revealed that the mRNA levels of DAF-16 Class I genes, which are activated by DAF-16, were further elevated in the daf-2;set double mutants. Among these genes, tts-1, F35E8.7, ins-35, nhr-62, sod-3, asm-2, and Y39G8B.7 are required for the lifespan extension of the daf-2;set-21 double mutant. In addition, treating daf-2 animals with the H3K9me1/2 methyltransferase G9a inhibitor also extends lifespan and increases stress resistance. Therefore, investigation of DAF-2 and H3K9me1/2 deficiency-mediated synergistic longevity will contribute to a better understanding of the molecular mechanisms of aging and therapeutic applications.
Collapse
Affiliation(s)
- Meng Huang
- Department of Obstetrics and Gynecology, University of Science and Technology of China, Hefei, China
| | - Minjie Hong
- Department of Obstetrics and Gynecology, University of Science and Technology of China, Hefei, China
| | - Xinhao Hou
- Department of Obstetrics and Gynecology, University of Science and Technology of China, Hefei, China
| | - Chengming Zhu
- Department of Obstetrics and Gynecology, University of Science and Technology of China, Hefei, China
| | - Di Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Xiangyang Chen
- Department of Obstetrics and Gynecology, University of Science and Technology of China, Hefei, China
| | - Shouhong Guang
- Department of Obstetrics and Gynecology, University of Science and Technology of China, Hefei, China
| | - Xuezhu Feng
- Department of Obstetrics and Gynecology, University of Science and Technology of China, Hefei, China
| |
Collapse
|
152
|
Egan BM, Scharf A, Pohl F, Kornfeld K. Control of aging by the renin–angiotensin system: a review of C. elegans, Drosophila, and mammals. Front Pharmacol 2022; 13:938650. [PMID: 36188619 PMCID: PMC9518657 DOI: 10.3389/fphar.2022.938650] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
The free-living, non-parasitic nematode Caenorhabditis elegans is a premier model organism for the study of aging and longevity due to its short lifespan, powerful genetic tools, and conservation of fundamental mechanisms with mammals. Approximately 70 percent of human genes have homologs in C. elegans, including many that encode proteins in pathways that influence aging. Numerous genetic pathways have been identified in C. elegans that affect lifespan, including the dietary restriction pathway, the insulin/insulin-like growth factor (IGF) signaling pathway, and the disruption of components of the mitochondrial electron transport chain. C. elegans is also a powerful system for performing drug screens, and many lifespan-extending compounds have been reported; notably, several FDA-approved medications extend the lifespan in C. elegans, raising the possibility that they can also extend the lifespan in humans. The renin–angiotensin system (RAS) in mammals is an endocrine system that regulates blood pressure and a paracrine system that acts in a wide range of tissues to control physiological processes; it is a popular target for drugs that reduce blood pressure, including angiotensin-converting enzyme (ACE) inhibitors and angiotensin II receptor blockers (ARBs). Emerging evidence indicates that this system influences aging. In C. elegans, decreasing the activity of the ACE homolog acn-1 or treatment with the ACE-inhibitor Captopril significantly extends the lifespan. In Drosophila, treatment with ACE inhibitors extends the lifespan. In rodents, manipulating the RAS with genetic or pharmacological interventions can extend the lifespan. In humans, polymorphisms in the ACE gene are associated with extreme longevity. These results suggest the RAS plays a conserved role in controlling longevity. Here, we review studies of the RAS and aging, emphasizing the potential of C. elegans as a model for understanding the mechanism of lifespan control.
Collapse
Affiliation(s)
- Brian M. Egan
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States
| | - Andrea Scharf
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, United States
| | - Franziska Pohl
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Kerry Kornfeld
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States
- *Correspondence: Kerry Kornfeld,
| |
Collapse
|
153
|
Pattarachotanant N, Sornkaew N, Warayanon W, Rangsinth P, Sillapachaiyaporn C, Vongthip W, Chuchawankul S, Prasansuklab A, Tencomnao T. Aquilaria crassna Leaf Extract Ameliorates Glucose-Induced Neurotoxicity In Vitro and Improves Lifespan in Caenorhabditis elegans. Nutrients 2022; 14:nu14173668. [PMID: 36079924 PMCID: PMC9460374 DOI: 10.3390/nu14173668] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/19/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Hyperglycemia is one of the important causes of neurodegenerative disorders and aging. Aquilaria crassna Pierre ex Lec (AC) has been widely used to relieve various health ailments. However, the neuroprotective and anti-aging effects against high glucose induction have not been investigated. This study aimed to investigate the effects of hexane extract of AC leaves (ACH) in vitro using human neuroblastoma SH-SY5Y cells and in vivo using nematode Caenorhabditis elegans. SH-SY5Y cells and C. elegans were pre-exposed with high glucose, followed by ACH treatment. To investigate neuroprotective activities, neurite outgrowth and cell cycle progression were determined in SH-SY5Y cells. In addition, C. elegans was used to determine ACH effects on antioxidant activity, longevity, and healthspan. In addition, ACH phytochemicals were analyzed and the possible active compounds were identified using a molecular docking study. ACH exerted neuroprotective effects by inducing neurite outgrowth via upregulating growth-associated protein 43 and teneurin-4 expression and normalizing cell cycle progression through the regulation of cyclin D1 and SIRT1 expression. Furthermore, ACH prolonged lifespan, improved body size, body length, and brood size, and reduced intracellular ROS accumulation in high glucose-induced C. elegans via the activation of gene expression in the DAF-16/FoxO pathway. Finally, phytochemicals of ACH were analyzed and revealed that β-sitosterol and stigmasterol were the possible active constituents in inhibiting insulin-like growth factor 1 receptor (IGFR). The results of this study establish ACH as an alternative medicine to defend against high glucose effects on neurotoxicity and aging.
Collapse
Affiliation(s)
- Nattaporn Pattarachotanant
- Natural Products for Neuroprotection and Anti-Ageing (Neur-Age Natura) Research Unit, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Nilubon Sornkaew
- Natural Products for Neuroprotection and Anti-Ageing (Neur-Age Natura) Research Unit, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Watis Warayanon
- Natural Products for Neuroprotection and Anti-Ageing (Neur-Age Natura) Research Unit, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Panthakarn Rangsinth
- Department of Transfusion Medicine and Clinical Microbiology, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Chanin Sillapachaiyaporn
- Natural Products for Neuroprotection and Anti-Ageing (Neur-Age Natura) Research Unit, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Program in Clinical Biochemistry and Molecular Medicine, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Wudtipong Vongthip
- Natural Products for Neuroprotection and Anti-Ageing (Neur-Age Natura) Research Unit, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Program in Clinical Biochemistry and Molecular Medicine, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Siriporn Chuchawankul
- Department of Transfusion Medicine and Clinical Microbiology, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Anchalee Prasansuklab
- Natural Products for Neuroprotection and Anti-Ageing (Neur-Age Natura) Research Unit, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- College of Public Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-Ageing (Neur-Age Natura) Research Unit, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: ; Tel.: +66-2-218-1533
| |
Collapse
|
154
|
Li X, Shi X, McPherson M, Hager M, Garcia GG, Miller RA. Cap-independent translation of GPLD1 enhances markers of brain health in long-lived mutant and drug-treated mice. Aging Cell 2022; 21:e13685. [PMID: 35930768 PMCID: PMC9470888 DOI: 10.1111/acel.13685] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 06/20/2022] [Accepted: 07/08/2022] [Indexed: 01/25/2023] Open
Abstract
Glycosylphosphatidylinositol-specific phospholipase D1 (GPLD1) hydrolyzes inositol phosphate linkages in proteins anchored to the cell membrane. Mice overexpressing GPLD1 show enhanced neurogenesis and cognition. Snell dwarf (DW) and growth hormone receptor knockout (GKO) mice show delays in age-dependent cognitive decline. We hypothesized that augmented GPLD1 might contribute to retained cognitive function in these mice. We report that DW and GKO show higher GPLD1 levels in the liver and plasma. These mice also have elevated levels of hippocampal brain-derived neurotrophic factor (BDNF) and of doublecortin (DCX), suggesting a mechanism for maintenance of cognitive function at older ages. GPLD1 was not increased in the hippocampus of DW or GKO mice, suggesting that plasma GPLD1 increases elevated these brain proteins. Alteration of the liver and plasma GPLD1 was unaltered in mice with liver-specific GHR deletion, suggesting that the GH effect was not intrinsic to the liver. GPLD1 was also induced by caloric restriction and by each of four drugs that extend lifespan. The proteome of DW and GKO mice is molded by selective translation of mRNAs, involving cap-independent translation (CIT) of mRNAs marked by N6 methyladenosine. Because GPLD1 protein increases were independent of the mRNA level, we tested the idea that GPLD1 might be regulated by CIT. 4EGI-1, which enhances CIT, increased GPLD1 protein without changes in GPLD1 mRNA in cultured fibroblasts and mice. Furthermore, transgenic overexpression of YTHDF1, which promotes CIT by reading m6A signals, also led to increased GPLD1 protein, showing that elevation of GPLD1 reflects selective mRNA translation.
Collapse
Affiliation(s)
- Xinna Li
- Department of Pathology, School of MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Xiaofang Shi
- Department of Pathology, School of MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Madaline McPherson
- College of Literature, Sciences, & the ArtsUniversity of MichiganAnn ArborMichiganUSA
| | - Mary Hager
- College of Literature, Sciences, & the ArtsUniversity of MichiganAnn ArborMichiganUSA
| | - Gonzalo G. Garcia
- Department of Pathology, School of MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Richard A. Miller
- Department of Pathology, School of MedicineUniversity of MichiganAnn ArborMichiganUSA,University of Michigan Geriatrics CenterAnn ArborMichiganUSA
| |
Collapse
|
155
|
Booth LN, Shi C, Tantilert C, Yeo RW, Miklas JW, Hebestreit K, Hollenhorst CN, Maures TJ, Buckley MT, Murphy CT, Brunet A. Males induce premature demise of the opposite sex by multifaceted strategies. NATURE AGING 2022; 2:809-823. [PMID: 37118502 PMCID: PMC10154206 DOI: 10.1038/s43587-022-00276-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 08/03/2022] [Indexed: 04/30/2023]
Abstract
Interactions between the sexes negatively impact health in many species. In Caenorhabditis, males shorten the lifespan of the opposite sex-hermaphrodites or females. Here we use transcriptomic profiling and targeted screens to systematically uncover conserved genes involved in male-induced demise in C. elegans. Some genes (for example, delm-2, acbp-3), when knocked down, are specifically protective against male-induced demise. Others (for example, sri-40), when knocked down, extend lifespan with and without males, suggesting general mechanisms of protection. In contrast, many classical long-lived mutants are impacted more negatively than wild type by the presence of males, highlighting the importance of sexual environment for longevity. Interestingly, genes induced by males are triggered by specific male components (seminal fluid, sperm and pheromone), and manipulating these genes in combination in hermaphrodites induces stronger protection. One of these genes, the conserved ion channel delm-2, acts in the nervous system and intestine to regulate lipid metabolism. Our analysis reveals striking differences in longevity in single sex versus mixed sex environments and uncovers elaborate strategies elicited by sexual interactions that could extend to other species.
Collapse
Affiliation(s)
- Lauren N Booth
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Calico Life Sciences, South San Francisco, CA, USA
| | - Cheng Shi
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- LSI Genomics, Princeton University, Princeton, NJ, USA
| | - Cindy Tantilert
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Robin W Yeo
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jason W Miklas
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Katja Hebestreit
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Travis J Maures
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthew T Buckley
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Coleen T Murphy
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
- LSI Genomics, Princeton University, Princeton, NJ, USA.
| | - Anne Brunet
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Glenn Laboratories for the Biology of Aging and Stanford University, Stanford, CA, USA.
| |
Collapse
|
156
|
Donlon TA, Morris BJ, Masaki KH, Chen R, Davy PMC, Kallianpur KJ, Nakagawa K, Owens JB, Willcox DC, Allsopp RC, Willcox BJ. FOXO3, a Resilience Gene: Impact on Lifespan, Healthspan, and Deathspan. J Gerontol A Biol Sci Med Sci 2022; 77:1479-1484. [PMID: 35960854 PMCID: PMC9373965 DOI: 10.1093/gerona/glac132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Timothy A Donlon
- Center of Biomedical Research Excellence for Translational Research on Aging and Department of Research, Kuakini Medical Center, Honolulu, Hawaii, USA
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Brian J Morris
- Center of Biomedical Research Excellence for Translational Research on Aging and Department of Research, Kuakini Medical Center, Honolulu, Hawaii, USA
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
- School of Medical Sciences, University of Sydney, Sydney, New South Wales, Australia
| | - Kamal H Masaki
- Center of Biomedical Research Excellence for Translational Research on Aging and Department of Research, Kuakini Medical Center, Honolulu, Hawaii, USA
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Randi Chen
- Center of Biomedical Research Excellence for Translational Research on Aging and Department of Research, Kuakini Medical Center, Honolulu, Hawaii, USA
| | - Phillip M C Davy
- Center of Biomedical Research Excellence for Translational Research on Aging and Department of Research, Kuakini Medical Center, Honolulu, Hawaii, USA
- Institute for Biogenesis Research, University of Hawaii, Honolulu, Hawaii, USA
| | - Kalpana J Kallianpur
- Center of Biomedical Research Excellence for Translational Research on Aging and Department of Research, Kuakini Medical Center, Honolulu, Hawaii, USA
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Kazuma Nakagawa
- Center of Biomedical Research Excellence for Translational Research on Aging and Department of Research, Kuakini Medical Center, Honolulu, Hawaii, USA
- Neuroscience Institute, The Queen’s Medical Center, Honolulu, Hawaii, USA
- Department of Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Jesse B Owens
- Center of Biomedical Research Excellence for Translational Research on Aging and Department of Research, Kuakini Medical Center, Honolulu, Hawaii, USA
- Institute for Biogenesis Research, University of Hawaii, Honolulu, Hawaii, USA
| | - D Craig Willcox
- Center of Biomedical Research Excellence for Translational Research on Aging and Department of Research, Kuakini Medical Center, Honolulu, Hawaii, USA
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
- Department of Human Welfare, Okinawa International University, Ginowan, Okinawa, Japan
| | - Richard C Allsopp
- Center of Biomedical Research Excellence for Translational Research on Aging and Department of Research, Kuakini Medical Center, Honolulu, Hawaii, USA
- Institute for Biogenesis Research, University of Hawaii, Honolulu, Hawaii, USA
| | - Bradley J Willcox
- Center of Biomedical Research Excellence for Translational Research on Aging and Department of Research, Kuakini Medical Center, Honolulu, Hawaii, USA
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| |
Collapse
|
157
|
Chai CM, Park H, Sternberg PW. Brain-wide bidirectional neuropeptide modulation of individual neuron classes regulates a developmental decision. Curr Biol 2022; 32:3365-3373.e6. [PMID: 35679871 PMCID: PMC10588560 DOI: 10.1016/j.cub.2022.05.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 05/06/2022] [Accepted: 05/17/2022] [Indexed: 11/18/2022]
Abstract
Secreted neuromodulators, like biogenic amines and neuropeptides, can reconfigure circuit functions both locally and at a distance and establish global brain states that alter circuit outputs over prolonged timescales.1-3 Despite their diversity and ubiquitous presence, many studies on neuromodulation tend to focus on dissecting the function and site of action of individual neuropeptides. Here, we take a different approach by conducting a systems-level investigation of neuropeptide receptor signaling function and cell-type-specific distribution in the context of the Caenorhabditis elegans diapause entry developmental decision. C. elegans diapause entry is controlled by sensory perception of external factors and is regulated by neuropeptide signaling.4-8 We performed a comprehensive functional screen of neuropeptide receptor mutants for pheromone-induced diapause entry phenotypes and integrated these results with published C. elegans single-cell RNA-seq data to reveal that almost all neuron classes expressed at least one receptor with a role in diapause entry.9 Our receptor expression analysis also identified four highly modulated neural hubs with no previously reported roles in diapause entry that are distributed throughout the animal's body, possibly as a means of synchronizing the whole-organism transition into the appropriate larval morph. Furthermore, most neuron classes expressed unique neuropeptide receptor repertoires that have opposing effects on the diapause entry decision. We propose that brain-wide antagonistic neuropeptide modulation of individual neuron classes by distinct neuropeptide receptor subsets could serve as a strategy against overmodulation and that this motif might generalize to other decision-making paradigms in other organisms.
Collapse
Affiliation(s)
- Cynthia M Chai
- Division of Biology & Biological Engineering, California Institute of Technology, 1200 E California Boulevard, Pasadena, CA 91125, USA.
| | - Heenam Park
- Division of Biology & Biological Engineering, California Institute of Technology, 1200 E California Boulevard, Pasadena, CA 91125, USA
| | - Paul W Sternberg
- Division of Biology & Biological Engineering, California Institute of Technology, 1200 E California Boulevard, Pasadena, CA 91125, USA.
| |
Collapse
|
158
|
Meraş İ, Chotard L, Liontis T, Ratemi Z, Wiles B, Seo JH, Van Raamsdonk JM, Rocheleau CE. The Rab GTPase activating protein TBC-2 regulates endosomal localization of DAF-16 FOXO and lifespan. PLoS Genet 2022; 18:e1010328. [PMID: 35913999 PMCID: PMC9371356 DOI: 10.1371/journal.pgen.1010328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/11/2022] [Accepted: 07/06/2022] [Indexed: 12/02/2022] Open
Abstract
FOXO transcription factors have been shown to regulate longevity in model organisms and are associated with longevity in humans. To gain insight into how FOXO functions to increase lifespan, we examined the subcellular localization of DAF-16 in C. elegans. We show that DAF-16 is localized to endosomes and that this endosomal localization is increased by the insulin-IGF signaling (IIS) pathway. Endosomal localization of DAF-16 is modulated by endosomal trafficking proteins. Disruption of the Rab GTPase activating protein TBC-2 increases endosomal localization of DAF-16, while inhibition of TBC-2 targets, RAB-5 or RAB-7 GTPases, decreases endosomal localization of DAF-16. Importantly, the amount of DAF-16 that is localized to endosomes has functional consequences as increasing endosomal localization through mutations in tbc-2 reduced the lifespan of long-lived daf-2 IGFR mutants, depleted their fat stores, and DAF-16 target gene expression. Overall, this work identifies endosomal localization as a mechanism regulating DAF-16 FOXO, which is important for its functions in metabolism and aging. FOXO transcription factors have been shown to modulate lifespan in multiple model organisms and to be associated with longevity in humans. Here we describe a new localization of the C. elegans FOXO transcription factor, called DAF-16. We report that DAF-16 localizes to endosomes, membrane compartments internalized from the plasma membrane at the cell surface. We demonstrate that expansion of these endosome compartments by disruption of an endosomal regulator called TBC-2 results in increased localization of DAF-16 on endosomes at the expense of nuclear localization in the intestinal cells. This results in altered expression of DAF-16 target genes, reduced fat storage and decreased lifespan. These results demonstrate the importance of endosomal trafficking for proper localization of DAF-16 and suggest that the endosome is an important site of FOXO regulation. An intriguing possibility based on our results is that storage of FOXO on endosomes facilitates the mobilization of FOXO as a rapid response to environmental stress.
Collapse
Affiliation(s)
- İçten Meraş
- Department of Anatomy and Cell Biology, McGill University, Montreal, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McGill University, Montreal, Canada
- Metabolic Disorders and Complications Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Laëtitia Chotard
- Division of Endocrinology and Metabolism, Department of Medicine, McGill University, Montreal, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Canada
| | - Thomas Liontis
- Metabolic Disorders and Complications Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- Brain Repair and Integrative Neuroscience Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Zakaria Ratemi
- Metabolic Disorders and Complications Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Benjamin Wiles
- Metabolic Disorders and Complications Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Jung Hwa Seo
- Metabolic Disorders and Complications Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Jeremy M. Van Raamsdonk
- Metabolic Disorders and Complications Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- Brain Repair and Integrative Neuroscience Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Christian E. Rocheleau
- Department of Anatomy and Cell Biology, McGill University, Montreal, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McGill University, Montreal, Canada
- Metabolic Disorders and Complications Program, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Canada
- * E-mail:
| |
Collapse
|
159
|
Lazaro-Pena MI, Ward ZC, Yang S, Strohm A, Merrill AK, Soto CA, Samuelson AV. HSF-1: Guardian of the Proteome Through Integration of Longevity Signals to the Proteostatic Network. FRONTIERS IN AGING 2022; 3:861686. [PMID: 35874276 PMCID: PMC9304931 DOI: 10.3389/fragi.2022.861686] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 06/13/2022] [Indexed: 12/15/2022]
Abstract
Discoveries made in the nematode Caenorhabditis elegans revealed that aging is under genetic control. Since these transformative initial studies, C. elegans has become a premier model system for aging research. Critically, the genes, pathways, and processes that have fundamental roles in organismal aging are deeply conserved throughout evolution. This conservation has led to a wealth of knowledge regarding both the processes that influence aging and the identification of molecular and cellular hallmarks that play a causative role in the physiological decline of organisms. One key feature of age-associated decline is the failure of mechanisms that maintain proper function of the proteome (proteostasis). Here we highlight components of the proteostatic network that act to maintain the proteome and how this network integrates into major longevity signaling pathways. We focus in depth on the heat shock transcription factor 1 (HSF1), the central regulator of gene expression for proteins that maintain the cytosolic and nuclear proteomes, and a key effector of longevity signals.
Collapse
Affiliation(s)
- Maria I. Lazaro-Pena
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, United States
| | - Zachary C. Ward
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, United States
| | - Sifan Yang
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, United States
- Department of Biology, University of Rochester, Rochester, NY, United States
| | - Alexandra Strohm
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, United States
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, United States
- Toxicology Training Program, University of Rochester Medical Center, Rochester, NY, United States
| | - Alyssa K. Merrill
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, United States
- Toxicology Training Program, University of Rochester Medical Center, Rochester, NY, United States
| | - Celia A. Soto
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, United States
- Cell Biology of Disease Graduate Program, University of Rochester Medical Center, Rochester, NY, United States
| | - Andrew V. Samuelson
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, United States
- *Correspondence: Andrew V. Samuelson,
| |
Collapse
|
160
|
Roy C, Molin L, Alcolei A, Solyga M, Bonneau B, Vachon C, Bessereau JL, Solari F. DAF-2/insulin IGF-1 receptor regulates motility during aging by integrating opposite signaling from muscle and neuronal tissues. Aging Cell 2022; 21:e13660. [PMID: 35808897 PMCID: PMC9381905 DOI: 10.1111/acel.13660] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 05/27/2022] [Accepted: 06/05/2022] [Indexed: 12/11/2022] Open
Abstract
During aging, preservation of locomotion is generally considered an indicator of sustained good health, in elderlies and in animal models. In Caenorhabditis elegans, mutants of the insulin‐IGF‐1 receptor DAF2/IIRc represent a paradigm of healthy aging, as their increased lifespan is accompanied by a delay in age‐related loss of motility. Here, we investigated the DAF‐2/IIRc‐dependent relationship between longevity and motility using an auxin‐inducible degron to trigger tissue‐specific degradation of endogenous DAF‐2/IIRc. As previously reported, inactivation of DAF‐2/IIRc in neurons or intestine was sufficient to extend the lifespan of worms, whereas depletion in epidermis, germline, or muscle was not. However, neither intestinal nor neuronal depletion of DAF‐2/IIRc prevented the age‐related loss of motility. In 1‐day‐old adults, DAF‐2/IIRc depletion in neurons reduced motility in a DAF‐16/FOXO dependent manner, while muscle depletion had no effect. By contrast, DAF‐2 depletion in the muscle of middle‐age animals improved their motility independently of DAF‐16/FOXO but required UNC‐120/SRF. Yet, neuronal or muscle DAF‐2/IIRc depletion both preserved the mitochondria network in aging muscle. Overall, these results show that the motility pattern of daf‐2 mutants is determined by the sequential and opposing impact of neurons and muscle tissues and can be dissociated from the regulation of the lifespan. This work also provides the characterization of a versatile tool to analyze the tissue‐specific contribution of insulin‐like signaling in integrated phenotypes at the whole organism level.
Collapse
Affiliation(s)
- Charline Roy
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR5284, INSERMU1314, Institut NeuroMyoGène, MeLis, Lyon, France
| | - Laurent Molin
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR5284, INSERMU1314, Institut NeuroMyoGène, MeLis, Lyon, France
| | - Allan Alcolei
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR5284, INSERMU1314, Institut NeuroMyoGène, MeLis, Lyon, France
| | - Mathilde Solyga
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR5284, INSERMU1314, Institut NeuroMyoGène, MeLis, Lyon, France
| | - Benjamin Bonneau
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR5284, INSERMU1314, Institut NeuroMyoGène, MeLis, Lyon, France
| | - Camille Vachon
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR5284, INSERMU1314, Institut NeuroMyoGène, MeLis, Lyon, France
| | - Jean-Louis Bessereau
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR5284, INSERMU1314, Institut NeuroMyoGène, MeLis, Lyon, France
| | - Florence Solari
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR5284, INSERMU1314, Institut NeuroMyoGène, MeLis, Lyon, France
| |
Collapse
|
161
|
Dravecz N, Shaw T, Davies I, Brown C, Ormerod L, Vu G, Walker T, Taank T, Shirras AD, Broughton SJ. Reduced Insulin Signaling Targeted to Serotonergic Neurons but Not Other Neuronal Subtypes Extends Lifespan in Drosophila melanogaster. Front Aging Neurosci 2022; 14:893444. [PMID: 35865744 PMCID: PMC9294736 DOI: 10.3389/fnagi.2022.893444] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
Reduced Insulin/IGF-like signaling (IIS) plays an evolutionarily conserved role in improving longevity and some measures of health-span in model organisms. Recent studies, however, have found a disconnection between lifespan extension and behavioral health-span. We have previously shown that reduction of IIS in Drosophila neurons extends female lifespan but does not improve negative geotaxis senescence and has a detrimental effect on exploratory walking senescence in both sexes. We hypothesize that individual neuronal subtypes respond differently to IIS changes, thus the behavioral outcomes of pan-neuronal IIS reduction are the balance of positive, negative and neutral functional effects. In order to further understand how reduced IIS in neurons independently modulates lifespan and locomotor behavioral senescence we expressed a dominant negative Insulin receptor transgene selectively in individual neuronal subtypes and measured the effects on lifespan and two measures of locomotor senescence, negative geotaxis and exploratory walking. IIS reduction in cholinergic, GABAergic, dopaminergic, glutamatergic, and octopaminergic neurons was found to have either no affect or a detrimental effect on lifespan and locomotor senescence. However, reduction of IIS selectively in serotonergic neurons resulted in extension of lifespan in females with no effect on locomotor senescence. These data indicate that individual neuronal subtypes respond differently to IIS changes in the modulation of lifespan and locomotor senescence, and identify a specific role for the insulin receptor in serotonergic neurons in the modulation of lifespan.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Susan J. Broughton
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom
| |
Collapse
|
162
|
Kageyama N, Nose M, Ono M, Matsunaga Y, Iwasaki T, Kawano T. The FMRFamide-like peptide FLP-2 is involved in the modulation of larval development and adult lifespan by regulating the secretion of the insulin-like peptide INS-35 in Caenorhabditis elegans. Biosci Biotechnol Biochem 2022; 86:1231-1239. [PMID: 35786701 DOI: 10.1093/bbb/zbac108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/05/2022] [Indexed: 11/12/2022]
Abstract
In the animal kingdom, neuropeptides regulate diverse physiological functions. In invertebrates, FMRFamide and its related peptides, a family of neuropeptides, play an important role as neurotransmitters. The FMRFamide-like peptides (FLPs) are one of the most diverse neuropeptide families and are conserved in nematodes. Our screen for flp genes of the free-living soil nematode Caenorhabditis elegans revealed that the flp-2 gene is involved in larval development. The gene is also conserved in plant-parasitic root-knot nematodes. Our molecular genetic analyses of the C. elegans flp-2 gene demonstrated as follows: 1) the production and secretion of FLP-2, produced in the head neurons, are controlled by environmental factors (growth density and food); 2) the FLP-2 is involved in not only larval development but also adult lifespan by regulating the secretion of one of the insulin-like peptides INS-35, produced in the intestine. These findings provide new insight into the development of new nematicides.
Collapse
Affiliation(s)
- Natsumi Kageyama
- Department of Agricultural Science, Graduate School of Sustainability Science
| | - Masayo Nose
- Department of Agricultural Science, Graduate School of Sustainability Science
| | - Masahiro Ono
- Department of Bioresources Science, The United Graduate School of Agriculture, Tottori University, Tottori, Japan
| | | | - Takashi Iwasaki
- Department of Agricultural Science, Graduate School of Sustainability Science.,Department of Bioresources Science, The United Graduate School of Agriculture, Tottori University, Tottori, Japan
| | - Tsuyoshi Kawano
- Department of Agricultural Science, Graduate School of Sustainability Science.,Department of Bioresources Science, The United Graduate School of Agriculture, Tottori University, Tottori, Japan
| |
Collapse
|
163
|
Arampatzis AS, Tsave O, Kirchweger B, Zwirchmayr J, Papageorgiou VP, Rollinger JM, Assimopoulou AN. Expanding the Biological Properties of Alkannins and Shikonins: Their Impact on Adipogenesis and Life Expectancy in Nematodes. Front Pharmacol 2022; 13:909285. [PMID: 35754463 PMCID: PMC9216188 DOI: 10.3389/fphar.2022.909285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
Alkannin, shikonin (A/S) and their derivatives are naturally occurring hydroxynaphthoquinones biosynthesized in some species of the Boraginaceae family. These natural compounds have been extensively investigated for their biological properties over the last 40 years, demonstrating a plethora of activities, such as wound healing, regenerative, anti-inflammatory, antitumor, antimicrobial and antioxidant. This study aims to extend the current knowledge by investigating the effects of various A/S compounds on two model systems, namely on 3T3-L1 pre-adipocytes and the nematode Caenorhabditis elegans. The former constitutes an established in vitro model for investigating anti-obesity and insulin-mimetic properties, while the latter has been widely used as a model organism for studying fat accumulation, lifespan and the anthelmintic potential. A set of chemically well-defined A/S derivatives were screened for their effect on pre-adipocytes to assess cell toxicity, cell morphology, and cell differentiation. The differentiation of pre-adipocytes into mature adipocytes was examined upon treatment with A/S compounds in the presence/absence of insulin, aiming to establish a structure-activity relationship. The majority of A/S compounds induced cell proliferation at sub-micromolar concentrations. The ester derivatives exhibited higher IC50 values, and thus, proved to be less toxic to 3T3-L1 cells. The parent molecules, A and S tested at 1 μM resulted in a truncated differentiation with a reduced number of forming lipids, whereas compounds lacking the side chain hydroxyl group projected higher populations of mature adipocytes. In C. elegans mutant strain SS104, A/S enriched extracts were not able to inhibit the fat accumulation but resulted in a drastic shortage of survival. Thus, the set of A/S compounds were tested at 15 and 60 μg/ml in the wild-type strain N2 for their nematocidal activity, which is of relevance for the discovery of anthelmintic drugs. The most pronounced nematocidal activity was observed for naphthazarin and β,β-dimethyl-acryl-shikonin, followed by isovaleryl-shikonin. The latter 2 A/S esters were identified as the most abundant constituents in the mixture of A/S derivatives isolated from Alkanna tinctoria (L.) Tausch. Taken together, the findings show that the structural variations in the moiety of A/S compounds significantly impact the modulation of their biological activities in both model systems investigated in this study.
Collapse
Affiliation(s)
- Athanasios S Arampatzis
- Organic Chemistry Laboratory, School of Chemical Engineering, Aristotle University of Thessaloniki and Natural Products Research Centre of Excellence (NatPro-AUTH), Center for Interdisciplinary Research and Innovation of AUTh (CIRI-AUTh), Thessaloniki, Greece
| | - Olga Tsave
- Organic Chemistry Laboratory, School of Chemical Engineering, Aristotle University of Thessaloniki and Natural Products Research Centre of Excellence (NatPro-AUTH), Center for Interdisciplinary Research and Innovation of AUTh (CIRI-AUTh), Thessaloniki, Greece
| | - Benjamin Kirchweger
- Division of Pharmacognosy, Department of Pharmaceutical Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Julia Zwirchmayr
- Division of Pharmacognosy, Department of Pharmaceutical Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Vassilios P Papageorgiou
- Organic Chemistry Laboratory, School of Chemical Engineering, Aristotle University of Thessaloniki and Natural Products Research Centre of Excellence (NatPro-AUTH), Center for Interdisciplinary Research and Innovation of AUTh (CIRI-AUTh), Thessaloniki, Greece
| | - Judith M Rollinger
- Division of Pharmacognosy, Department of Pharmaceutical Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Andreana N Assimopoulou
- Organic Chemistry Laboratory, School of Chemical Engineering, Aristotle University of Thessaloniki and Natural Products Research Centre of Excellence (NatPro-AUTH), Center for Interdisciplinary Research and Innovation of AUTh (CIRI-AUTh), Thessaloniki, Greece
| |
Collapse
|
164
|
Naranjo-Galindo FJ, Ai R, Fang EF, Nilsen HL, SenGupta T. C. elegans as an Animal Model to Study the Intersection of DNA Repair, Aging and Neurodegeneration. FRONTIERS IN AGING 2022; 3:916118. [PMID: 35821838 PMCID: PMC9261396 DOI: 10.3389/fragi.2022.916118] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022]
Abstract
Since its introduction as a genetic model organism, Caenorhabditis elegans has yielded insights into the causes of aging. In addition, it has provided a molecular understanding of mechanisms of neurodegeneration, one of the devastating effects of aging. However, C. elegans has been less popular as an animal model to investigate DNA repair and genomic instability, which is a major hallmark of aging and also a cause of many rare neurological disorders. This article provides an overview of DNA repair pathways in C. elegans and the impact of DNA repair on aging hallmarks, such as mitochondrial dysfunction, telomere maintenance, and autophagy. In addition, we discuss how the combination of biological characteristics, new technical tools, and the potential of following precise phenotypic assays through a natural life-course make C. elegans an ideal model organism to study how DNA repair impact neurodegeneration in models of common age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Francisco José Naranjo-Galindo
- Department of Clinical Molecular Biology, University of Oslo, Oslo, Norway
- Section of Clinical Molecular Biology (EpiGen), Akershus University Hospital, Lørenskog, Norway
| | - Ruixue Ai
- Department of Clinical Molecular Biology, University of Oslo, Oslo, Norway
| | - Evandro Fei Fang
- Department of Clinical Molecular Biology, University of Oslo, Oslo, Norway
| | - Hilde Loge Nilsen
- Department of Clinical Molecular Biology, University of Oslo, Oslo, Norway
- Section of Clinical Molecular Biology (EpiGen), Akershus University Hospital, Lørenskog, Norway
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Tanima SenGupta
- Department of Clinical Molecular Biology, University of Oslo, Oslo, Norway
- Section of Clinical Molecular Biology (EpiGen), Akershus University Hospital, Lørenskog, Norway
| |
Collapse
|
165
|
Zegarra-Valdivia J, Fernandez AM, Martinez-Rachadell L, Herrero-Labrador R, Fernandes J, Torres Aleman I. Insulin and insulin-like growth factor-I receptors in astrocytes exert different effects on behavior and Alzheimer´s-like pathology. F1000Res 2022; 11:663. [PMID: 36636477 PMCID: PMC9823242.3 DOI: 10.12688/f1000research.121901.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Pleiotropic actions of insulin and insulin-like growth factor I (IGF-I) in the brain are context- and cell-dependent, but whether this holds for their receptors (insulin receptor (IR) and IGF-I receptor (IGF-IR), respectively), is less clear. Methods: We compared mice lacking IR or IGF-IR in glial fibrillary astrocytic protein (GFAP)-expressing astrocytes in a tamoxifen-regulated manner, to clarify their role in this type of glial cells, as the majority of data of their actions in brain have been obtained in neurons. Results: We observed that mice lacking IR in GFAP astrocytes (GFAP IR KO mice) develop mood disturbances and maintained intact cognition, while at the same time show greater pathology when cross-bred with APP/PS1 mice, a model of familial Alzheimer´s disease (AD). Conversely, mice lacking IGF-IR in GFAP astrocytes (GFAP-IGF-IR KO mice) show cognitive disturbances, maintained mood tone, and show control-dependent changes in AD-like pathology. Conclusions: These observations confirm that the role of IR and IGF-IR in the brain is cell-specific and context-dependent.
Collapse
Affiliation(s)
- Jonathan Zegarra-Valdivia
- Achucarro Basque Center for Neuroscience, Leioa, Bizkaia, 48940, Spain
- Cajal Institute, Madrid, 28002, Spain
- CIBERNED, Madrid, Spain
- Universidad Señor de Sipán, Chiclayo, Peru
| | | | | | | | - Jansen Fernandes
- Cajal Institute, Madrid, 28002, Spain
- Universidade Federal São Paulo, São Paulo, Brazil
| | - Ignacio Torres Aleman
- Achucarro Basque Center for Neuroscience, Leioa, Bizkaia, 48940, Spain
- CIBERNED, Madrid, Spain
- Ikerbasque Foundation for Science, Bilbao, Spain
| |
Collapse
|
166
|
Zegarra-Valdivia J, Fernandez AM, Martinez-Rachadell L, Herrero-Labrador R, Fernandes J, Torres Aleman I. Insulin and insulin-like growth factor-I receptors in astrocytes exert different effects on behavior and Alzheimer´s-like pathology. F1000Res 2022; 11:663. [PMID: 36636477 PMCID: PMC9823242 DOI: 10.12688/f1000research.121901.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/26/2022] [Indexed: 05/10/2025] Open
Abstract
Background: Pleiotropic actions of insulin and insulin-like growth factor I (IGF-I) in the brain are context- and cell-dependent, but whether this holds for their receptors (insulin receptor (IR) and IGF-I receptor (IGF-IR), respectively), is less clear. Methods: We compared mice lacking IR or IGF-IR in glial fibrillary astrocytic protein (GFAP)-expressing astrocytes in a tamoxifen-regulated manner, to clarify their role in this type of glial cells, as the majority of data of their actions in brain have been obtained in neurons. Results: We observed that mice lacking IR in GFAP astrocytes (GFAP IR KO mice) develop mood disturbances and maintained intact cognition, while at the same time show greater pathology when cross-bred with APP/PS1 mice, a model of familial Alzheimer´s disease (AD). Conversely, mice lacking IGF-IR in GFAP astrocytes (GFAP-IGF-IR KO mice) show cognitive disturbances, maintained mood tone, and show control-dependent changes in AD-like pathology. Conclusions: These observations confirm that the role of IR and IGF-IR in the brain is cell-specific and context-dependent.
Collapse
Affiliation(s)
- Jonathan Zegarra-Valdivia
- Achucarro Basque Center for Neuroscience, Leioa, Bizkaia, 48940, Spain
- Cajal Institute, Madrid, 28002, Spain
- CIBERNED, Madrid, Spain
- Universidad Señor de Sipán, Chiclayo, Peru
| | | | | | | | - Jansen Fernandes
- Cajal Institute, Madrid, 28002, Spain
- Universidade Federal São Paulo, São Paulo, Brazil
| | - Ignacio Torres Aleman
- Achucarro Basque Center for Neuroscience, Leioa, Bizkaia, 48940, Spain
- CIBERNED, Madrid, Spain
- Ikerbasque Foundation for Science, Bilbao, Spain
| |
Collapse
|
167
|
Zegarra-Valdivia J, Fernandez AM, Martinez-Rachadell L, Herrero-Labrador R, Fernandes J, Torres Aleman I. Insulin and insulin-like growth factor-I receptors in astrocytes exert different effects on behavior and Alzheimer´s-like pathology. F1000Res 2022; 11:663. [PMID: 36636477 PMCID: PMC9823242.2 DOI: 10.12688/f1000research.121901.2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/09/2022] [Indexed: 05/10/2025] Open
Abstract
Background: Pleiotropic actions of insulin and insulin-like growth factor I (IGF-I) in the brain are context- and cell-dependent, but whether this holds for their receptors (insulin receptor (IR) and IGF-I receptor (IGF-IR), respectively), is less clear. Methods: We compared mice lacking IR or IGF-IR in glial fibrillary astrocytic protein (GFAP)-expressing astrocytes in a tamoxifen-regulated manner, to clarify their role in this type of glial cells, as the majority of data of their actions in brain have been obtained in neurons. Results: We observed that mice lacking IR in GFAP astrocytes (GFAP IR KO mice) develop mood disturbances and maintained intact cognition, while at the same time show greater pathology when cross-bred with APP/PS1 mice, a model of familial Alzheimer´s disease (AD). Conversely, mice lacking IGF-IR in GFAP astrocytes (GFAP-IGF-IR KO mice) show cognitive disturbances, maintained mood tone, and show control-dependent changes in AD-like pathology. Conclusions: These observations confirm that the role of IR and IGF-IR in the brain is cell-specific and context-dependent.
Collapse
Affiliation(s)
- Jonathan Zegarra-Valdivia
- Achucarro Basque Center for Neuroscience, Leioa, Bizkaia, 48940, Spain
- Cajal Institute, Madrid, 28002, Spain
- CIBERNED, Madrid, Spain
- Universidad Señor de Sipán, Chiclayo, Peru
| | | | | | | | - Jansen Fernandes
- Cajal Institute, Madrid, 28002, Spain
- Universidade Federal São Paulo, São Paulo, Brazil
| | - Ignacio Torres Aleman
- Achucarro Basque Center for Neuroscience, Leioa, Bizkaia, 48940, Spain
- CIBERNED, Madrid, Spain
- Ikerbasque Foundation for Science, Bilbao, Spain
| |
Collapse
|
168
|
Dakic T, Jevdjovic T, Vujovic P, Mladenovic A. The Less We Eat, the Longer We Live: Can Caloric Restriction Help Us Become Centenarians? Int J Mol Sci 2022; 23:ijms23126546. [PMID: 35742989 PMCID: PMC9223351 DOI: 10.3390/ijms23126546] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 02/04/2023] Open
Abstract
Striving for longevity is neither a recent human desire nor a novel scientific field. The first article on this topic was published in 1838, when the average human life expectancy was approximately 40 years. Although nowadays people on average live almost as twice as long, we still (and perhaps more than ever) look for new ways to extend our lifespan. During this seemingly endless journey of discovering efficient methods to prolong life, humans were enthusiastic regarding several approaches, one of which is caloric restriction (CR). Where does CR, initially considered universally beneficial for extending both lifespan and health span, stand today? Does a lifelong decrease in food consumption represent one of the secrets of centenarians’ long and healthy life? Do we still believe that if we eat less, we will live longer? This review aims to summarize the current literature on CR as a potential life-prolonging intervention in humans and discusses metabolic pathways that underlie this effect.
Collapse
Affiliation(s)
- Tamara Dakic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia; (T.D.); (T.J.); (P.V.)
| | - Tanja Jevdjovic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia; (T.D.); (T.J.); (P.V.)
| | - Predrag Vujovic
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry “Ivan Djaja”, Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia; (T.D.); (T.J.); (P.V.)
| | - Aleksandra Mladenovic
- Department of Neurobiology, Institute for Biological Research “Sinisa Stankovic”—National Institute of Republic of Serbia, University of Belgrade, Bul.D. Stefana 142, 11000 Belgrade, Serbia
- Correspondence:
| |
Collapse
|
169
|
Peng G, Zhu C, Sun Q, Li J, Chen Y, Guo Y, Ji H, Yang F, Dong W. Testicular miRNAs and tsRNAs provide insight into gene regulation during overwintering and reproduction of Onychostoma macrolepis. FISH PHYSIOLOGY AND BIOCHEMISTRY 2022; 48:481-499. [PMID: 35595880 DOI: 10.1007/s10695-022-01078-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 04/19/2022] [Indexed: 06/15/2023]
Abstract
The late overwintering period and breeding period are two important developmental stages of testis in Onychostoma macrolepis. Small non-coding RNAs (sncRNAs) are well-known regulators of biological processes associated with numerous biological processes. This study aimed to elucidate the roles of four sncRNA classes (microRNAs [miRNAs], Piwi-interacting RNAs [piRNAs], tRNA-derived small RNAs [tsRNAs], and rRNA-derived small RNAs [rsRNAs]) across testes in the late overwintering period (in March) and breeding period (in June) by high-throughput sequencing. The testis of O. macrolepis displayed the highest levels of piRNAs and lowest levels of rsRNAs. Compared with miRNAs and tsRNAs in June, tsRNAs in March had a higher abundance, while miRNAs in March had a much lower abundance. Bioinformatics analysis identified 1,362 and 1,340 differentially expressed miRNAs and tsRNAs, respectively. Further analysis showed that miR-200-1, miR-143-1, tRFi-Lys-CTT-1, and tRFi-Glu-CTC-1 could play critical roles during the overwintering and breeding periods. Our findings provided an unprecedented insight to reveal the epigenetic mechanism underlying the overwintering and reproduction process of male O. macrolepis.
Collapse
Affiliation(s)
- Guofan Peng
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China
- Biology Research Centre of Qin Mountains Wildlife, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China
| | - Chao Zhu
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China
- Biology Research Centre of Qin Mountains Wildlife, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China
| | - Qingfang Sun
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China
| | - Jincan Li
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China
| | - Yining Chen
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China
| | - Yingjie Guo
- Biology Research Centre of Qin Mountains Wildlife, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China
- College of Forestry, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China
| | - Hong Ji
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China
| | - Fangxia Yang
- Biology Research Centre of Qin Mountains Wildlife, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China.
- College of Forestry, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China.
| | - Wuzi Dong
- College of Animal Science and Technology, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China.
- Biology Research Centre of Qin Mountains Wildlife, Northwest A&F University, No. 22 Xinong Road, Shaanxi, 712100, China.
| |
Collapse
|
170
|
Ollinger N, Neuhauser C, Schwarzinger B, Wallner M, Schwarzinger C, Blank‐Landeshammer B, Hager R, Sadova N, Drotarova I, Mathmann K, Karamouzi E, Panopoulos P, Rimbach G, Lüersen K, Weghuber J, Röhrl C. Anti-Hyperglycemic Effects of Oils and Extracts Derived from Sea Buckthorn - A Comprehensive Analysis Utilizing In Vitro and In Vivo Models. Mol Nutr Food Res 2022; 66:e2101133. [PMID: 35426970 PMCID: PMC9285508 DOI: 10.1002/mnfr.202101133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/14/2022] [Indexed: 12/15/2022]
Abstract
SCOPE Sea buckthorn (Hippophaes rhamnoides) is capable of ameliorating disturbed glucose metabolism in animal models and human subjects. Here, the effect of sea buckthorn oil as well as of extracts of fruits, leaves, and press cake on postprandial glucose metabolism is systematically investigated. METHODS AND RESULTS Sea buckthorn did neither exert decisive effects in an in vitro model of intestinal glucose absorption nor did it alter insulin secretion. However, sea buckthorn stimulates GLUT4 translocation to the plasma membrane comparable to insulin, indicative of increased glucose clearance from the circulation. Isorhamnetin is identified in all sea buckthorn samples investigated and is biologically active in triggering GLUT4 cell surface localization. Consistently, sea buckthorn products lower circulating glucose by ≈10% in a chick embryo model. Moreover, sea buckthorn products fully revert hyperglycemia in the nematode Caenorhabditis elegans while they are ineffective in Drosophila melanogaster under euglycemic conditions. CONCLUSION These data indicate that edible sea buckthorn products as well as by-products are promising resources for hypoglycemic nutrient supplements that increase cellular glucose clearance into target tissues.
Collapse
Affiliation(s)
- Nicole Ollinger
- FFoQSI – Austrian Competence Centre for Feed and Food QualitySafety & InnovationFFoQSI GmbHTechnopark 1DTulln3430Austria
| | - Cathrina Neuhauser
- University of Applied Sciences Upper AustriaStelzhamerstrasse 23Wels4600Austria
| | - Bettina Schwarzinger
- FFoQSI – Austrian Competence Centre for Feed and Food QualitySafety & InnovationFFoQSI GmbHTechnopark 1DTulln3430Austria
- University of Applied Sciences Upper AustriaStelzhamerstrasse 23Wels4600Austria
| | - Melanie Wallner
- FFoQSI – Austrian Competence Centre for Feed and Food QualitySafety & InnovationFFoQSI GmbHTechnopark 1DTulln3430Austria
| | - Clemens Schwarzinger
- Johannes Kepler UniversityInstitute for Chemical Technology of Organic MaterialsLinz4040Austria
| | - Bernhard Blank‐Landeshammer
- FFoQSI – Austrian Competence Centre for Feed and Food QualitySafety & InnovationFFoQSI GmbHTechnopark 1DTulln3430Austria
| | - Roland Hager
- University of Applied Sciences Upper AustriaStelzhamerstrasse 23Wels4600Austria
| | - Nadiia Sadova
- University of Applied Sciences Upper AustriaStelzhamerstrasse 23Wels4600Austria
| | - Ivana Drotarova
- University of Applied Sciences Upper AustriaStelzhamerstrasse 23Wels4600Austria
| | - Katrin Mathmann
- University of Applied Sciences Upper AustriaStelzhamerstrasse 23Wels4600Austria
| | - Eugenia Karamouzi
- European Research & Development Rezos Brands196 New National Road Patras‐AthensPatras26443Greece
| | - Panagiotis Panopoulos
- European Research & Development Rezos Brands196 New National Road Patras‐AthensPatras26443Greece
| | - Gerald Rimbach
- Institute of Human Nutrition and Food ScienceUniversity of KielHermann‐Rodewald‐Strasse 6Kiel24118Germany
| | - Kai Lüersen
- Institute of Human Nutrition and Food ScienceUniversity of KielHermann‐Rodewald‐Strasse 6Kiel24118Germany
| | - Julian Weghuber
- FFoQSI – Austrian Competence Centre for Feed and Food QualitySafety & InnovationFFoQSI GmbHTechnopark 1DTulln3430Austria
- University of Applied Sciences Upper AustriaStelzhamerstrasse 23Wels4600Austria
| | - Clemens Röhrl
- University of Applied Sciences Upper AustriaStelzhamerstrasse 23Wels4600Austria
| |
Collapse
|
171
|
The anti-aging activity of Lycium barbarum polysaccharide extracted by yeast fermentation: In vivo and in vitro studies. Int J Biol Macromol 2022; 209:2032-2041. [PMID: 35500780 DOI: 10.1016/j.ijbiomac.2022.04.184] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/24/2022] [Accepted: 04/25/2022] [Indexed: 01/04/2023]
Abstract
Lycium barbarum polysaccharide (LBP) is an important active substance in Lycium barbarum. In this study, LBP was extracted by the hot water method and yeast fermentation method to obtain products called LBP-W and LBP-Y, respectively. Both LBPs have a strong ability to scavenge DPPH, hydroxyl, and superoxide anion free radicals and have a total antioxidant capacity. Both LBPs prolong the lifespan of C. elegans under normal conditions, oxidative stress and heat stress and do not affect fertility, LBPs could prolong the lifespan of C. elegans by upregulating the expression of daf-16, sod-3 and hsp-16.2 genes, and LBP-Y is more efficacious. The molecular weight of the LBPs was characterized by gel permeation chromatography (GPC), and the results showed that LBP-Y is smaller and more uniform than LBP-W. The skin penetration experiment showed that the absorption effect of LBP-Y is better than that of LBP-W. These lines of evidence suggest that the yeast fermentation extraction of LBP produces better antioxidant and anti-aging effects than those obtained with the traditional hot water extraction, which is more suitable for obtaining raw materials with anti-aging functions that can potentially be used in the food and cosmetic industries.
Collapse
|
172
|
Handley A, Wu Q, Sherry T, Cornell R, Pocock R. Diet-responsive transcriptional regulation of insulin in a single neuron controls systemic metabolism. PLoS Biol 2022; 20:e3001655. [PMID: 35594303 PMCID: PMC9162364 DOI: 10.1371/journal.pbio.3001655] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 06/02/2022] [Accepted: 04/29/2022] [Indexed: 11/18/2022] Open
Abstract
Metabolic homeostasis is coordinated through a robust network of signaling pathways acting across all tissues. A key part of this network is insulin-like signaling, which is fundamental for surviving glucose stress. Here, we show that Caenorhabditis elegans fed excess dietary glucose reduce insulin-1 (INS-1) expression specifically in the BAG glutamatergic sensory neurons. We demonstrate that INS-1 expression in the BAG neurons is directly controlled by the transcription factor ETS-5, which is also down-regulated by glucose. We further find that INS-1 acts exclusively from the BAG neurons, and not other INS-1-expressing neurons, to systemically inhibit fat storage via the insulin-like receptor DAF-2. Together, these findings reveal an intertissue regulatory pathway where regulation of insulin expression in a specific neuron controls systemic metabolism in response to excess dietary glucose. Metabolic homeostasis is coordinated through a robust network of signaling pathways acting across all tissues. This study shows that Caenorhabditis elegans nematodes fed excess dietary glucose reduce the expression of insulin-1 specifically in the BAG glutamatergic sensory neurons, and that insulin-1 produced by these neurons systemically inhibits fat storage via the insulin-like receptor DAF-2.
Collapse
Affiliation(s)
- Ava Handley
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
- * E-mail: (AH); (RP)
| | - Qiuli Wu
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
- Key Laboratory of Developmental Genes and Human Diseases in Ministry of Education, Medical School of Southeast University, Nanjing, China
| | - Tessa Sherry
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| | - Rebecca Cornell
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| | - Roger Pocock
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
- * E-mail: (AH); (RP)
| |
Collapse
|
173
|
Nutrient-Response Pathways in Healthspan and Lifespan Regulation. Cells 2022; 11:cells11091568. [PMID: 35563873 PMCID: PMC9102925 DOI: 10.3390/cells11091568] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/02/2022] [Accepted: 05/05/2022] [Indexed: 02/01/2023] Open
Abstract
Cellular, small invertebrate and vertebrate models are a driving force in biogerontology studies. Using various models, such as yeasts, appropriate tissue culture cells, Drosophila, the nematode Caenorhabditis elegans and the mouse, has tremendously increased our knowledge around the relationship between diet, nutrient-response signaling pathways and lifespan regulation. In recent years, combinatorial drug treatments combined with mutagenesis, high-throughput screens, as well as multi-omics approaches, have provided unprecedented insights in cellular metabolism, development, differentiation, and aging. Scientists are, therefore, moving towards characterizing the fine architecture and cross-talks of growth and stress pathways towards identifying possible interventions that could lead to healthy aging and the amelioration of age-related diseases in humans. In this short review, we briefly examine recently uncovered knowledge around nutrient-response pathways, such as the Insulin Growth Factor (IGF) and the mechanistic Target of Rapamycin signaling pathways, as well as specific GWAS and some EWAS studies on lifespan and age-related disease that have enhanced our current understanding within the aging and biogerontology fields. We discuss what is learned from the rich and diverse generated data, as well as challenges and next frontiers in these scientific disciplines.
Collapse
|
174
|
Invited review: Unearthing the mechanisms of age-related neurodegenerative disease using Caenorhabditis elegans. Comp Biochem Physiol A Mol Integr Physiol 2022; 267:111166. [PMID: 35176489 DOI: 10.1016/j.cbpa.2022.111166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 12/11/2022]
Abstract
As human life expectancy increases, neurodegenerative diseases present a growing public health threat, for which there are currently few effective treatments. There is an urgent need to understand the molecular and genetic underpinnings of these disorders so new therapeutic targets can be identified. Here we present the argument that the simple nematode worm Caenorhabditis elegans is a powerful tool to rapidly study neurodegenerative disorders due to their short lifespan and vast array of genetic tools, which can be combined with characterization of conserved neuronal processes and behavior orthologous to those disrupted in human disease. We review how pre-existing C. elegans models provide insight into human neurological disease as well as an overview of current tools available to study neurodegenerative diseases in the worm, with an emphasis on genetics and behavior. We also discuss open questions that C. elegans may be particularly well suited for in future studies and how worms will be a valuable preclinical model to better understand these devastating neurological disorders.
Collapse
|
175
|
Nakai J, Chikamoto N, Fujimoto K, Totani Y, Hatakeyama D, Dyakonova VE, Ito E. Insulin and Memory in Invertebrates. Front Behav Neurosci 2022; 16:882932. [PMID: 35558436 PMCID: PMC9087806 DOI: 10.3389/fnbeh.2022.882932] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/04/2022] [Indexed: 11/17/2022] Open
Abstract
Insulin and insulin-like peptides (ILP) help to maintain glucose homeostasis, whereas insulin-like growth factor (IGF) promotes the growth and differentiation of cells in both vertebrates and invertebrates. It is sometimes difficult to distinguish between ILP and IGF in invertebrates, however, because in some cases ILP has the same function as IGF. In the present review, therefore, we refer to these peptides as ILP/IGF signaling (IIS) in invertebrates, and discuss the role of IIS in memory formation after classical conditioning in invertebrates. In the arthropod Drosophila melanogaster, IIS is involved in aversive olfactory memory, and in the nematode Caenorhabditis elegans, IIS controls appetitive/aversive response to NaCl depending on the duration of starvation. In the mollusk Lymnaea stagnalis, IIS has a critical role in conditioned taste aversion. Insulin in mammals is also known to play an important role in cognitive function, and many studies in humans have focused on insulin as a potential treatment for Alzheimer’s disease. Although analyses of tissue and cellular levels have progressed in mammals, the molecular mechanisms, such as transcriptional and translational levels, of IIS function in cognition have been far advanced in studies using invertebrates. We anticipate that the present review will help to pave the way for studying the effects of insulin, ILPs, and IGFs in cognitive function across phyla.
Collapse
Affiliation(s)
- Junko Nakai
- Department of Biology, Waseda University, Tokyo, Japan
| | | | | | - Yuki Totani
- Department of Biology, Waseda University, Tokyo, Japan
| | - Dai Hatakeyama
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan
| | - Varvara E. Dyakonova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - Etsuro Ito
- Department of Biology, Waseda University, Tokyo, Japan
- Graduate Institute of Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- *Correspondence: Etsuro Ito
| |
Collapse
|
176
|
Brown-Borg HM. Growth hormone, not IGF-1 is the key longevity regulator in mammals. J Gerontol A Biol Sci Med Sci 2022; 77:1719-1723. [PMID: 35436323 PMCID: PMC9434454 DOI: 10.1093/gerona/glac092] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Holly M Brown-Borg
- Department of Biomedical Sciences, University of North Dakota School of Medicine & Health Sciences, Grand Forks, ND
| |
Collapse
|
177
|
Molecular and cytological profiling of biological aging of mouse cochlear inner and outer hair cells. Cell Rep 2022; 39:110665. [PMID: 35417713 PMCID: PMC9069708 DOI: 10.1016/j.celrep.2022.110665] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/08/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022] Open
Abstract
Age-related hearing loss (ARHL) negatively impacts quality of life in the elderly population. The prevalent cause of ARHL is loss of mechanosensitive cochlear hair cells (HCs). The molecular and cellular mechanisms of HC degeneration remain poorly understood. Using RNA-seq transcriptomic analyses of inner and outer HCs isolated from young and aged mice, we show that HC aging is associated with changes in key molecular processes, including transcription, DNA damage, autophagy, and oxidative stress, as well as genes related to HC specialization. At the cellular level, HC aging is characterized by loss of stereocilia, shrinkage of HC soma, and reduction in outer HC mechanical properties, suggesting that functional decline in mechanotransduction and cochlear amplification precedes HC loss and contributes to ARHL. Our study reveals molecular and cytological profiles of aging HCs and identifies genes such as Sod1, Sirt6, Jund, and Cbx3 as biomarkers and potential therapeutic targets for ameliorating ARHL. Using RNA-seq, advanced imaging, and electrophysiology, Liu et al. reveal molecular and cytological profiles of aging cochlear hair cells. Their study also suggests that a functional decline in mechanotransduction and cochlear amplification precedes hair cell loss and contributes to age-related hearing loss.
Collapse
|
178
|
Wang Q, Li H, Zhang G, Chen X, Wang X. Itaconate prolongs the healthy lifespan by activating UPR mt in Caenorhabditis elegans. Eur J Pharmacol 2022; 923:174951. [PMID: 35405114 DOI: 10.1016/j.ejphar.2022.174951] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/29/2022] [Accepted: 04/05/2022] [Indexed: 11/18/2022]
Abstract
Itaconate (ItA), a byproduct of the Krebs cycle, has recently emerged as an anti-inflammatory metabolite for inhibiting the overactive immune response. In addition to its immunomodulatory and antimicrobial effects, ItA may have other therapeutic avenues. Herein, the effect of ItA on aging was explored in order to better establish the therapeutic potential of this promising metabolite. ItA extended the lifespan and enhanced the stress resistance of Caenorhabditis elegans (C. elegans), even at the doses of 0.01 and 0.1 μM. Moreover, the lifespan extension effect of ItA was pronounced even for the aged worms (days 7 and 9 post adult stage). Furthermore, ItA was found to extend the healthy longevity of C. elegans in a mitochondria-dependent manner. ItA protected the mitochondrial integrity, increased ATP content, and decreased the reactive oxygen species (ROS) in C. elegans. Mechanistic investigations showed that ItA specifically activated the mitochondrial unfolded protein response (UPRmt) in worms and significantly increased the expression of activating transcription factor associated with stress-1 (ATFS-1) that senses mitochondrial stress and communicates with the nucleus during the UPRmt. ItA extended the lifespan of C. elegans in an ATFS-1-dependent manner. In summary, this study elucidates the molecular mechanism by which ItA extends the healthy lifespan and highlights the importance of mitochondrial integrity in the intervention of aging.
Collapse
Affiliation(s)
- Qingqing Wang
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, Jilin, 130012, China
| | - Hongyuan Li
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Gangwei Zhang
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, Jilin, 130012, China
| | - Xiaoguang Chen
- School of Chemistry and Life Science, Changchun University of Technology, Changchun, Jilin, 130012, China; College of Humanities and information, Changchun University of Technology, Changchun, Jilin, 130122, China.
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China; Beijing National Laboratory for Molecular Sciences, Beijing, 100190, China.
| |
Collapse
|
179
|
Vegetables and Their Bioactive Compounds as Anti-Aging Drugs. Molecules 2022; 27:molecules27072316. [PMID: 35408714 PMCID: PMC9000296 DOI: 10.3390/molecules27072316] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 12/27/2022] Open
Abstract
Aging is a continuous process over time that is mainly related to natural alterations in mechanical–biological processes. This phenomenon is due to several factors, including the time and energy of biological processes. Aging can be attributed to biological factors such as oxidative stress, cell longevity, and stem cell senescence. Currently, aging is associated with several diseases, such as neurodegenerative diseases, cancer, and other diseases related to oxidative stress. In addition, certain natural molecules, including those derived from vegetables, have shown the ability to delay the aging process. Their effects are linked to different mechanisms of action, such as tissue regeneration and the activation of longevity and anti-senescence genes. The present work discusses the impact of vegetables, and bioactive compounds isolated from vegetables, against the physiological and pathological aging process and accompanying human diseases.
Collapse
|
180
|
Zhu A, Zheng F, Zhang W, Li L, Li Y, Hu H, Wu Y, Bao W, Li G, Wang Q, Li H. Oxidation and Antioxidation of Natural Products in the Model Organism Caenorhabditiselegans. Antioxidants (Basel) 2022; 11:antiox11040705. [PMID: 35453390 PMCID: PMC9029379 DOI: 10.3390/antiox11040705] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 12/15/2022] Open
Abstract
Natural products are small molecules naturally produced by multiple sources such as plants, animals, fungi, bacteria and archaea. They exert both beneficial and detrimental effects by modulating biological targets and pathways involved in oxidative stress and antioxidant response. Natural products’ oxidative or antioxidative properties are usually investigated in preclinical experimental models, including virtual computing simulations, cell and tissue cultures, rodent and nonhuman primate animal models, and human studies. Due to the renewal of the concept of experimental animals, especially the popularization of alternative 3R methods for reduction, replacement and refinement, many assessment experiments have been carried out in new alternative models. The model organism Caenorhabditis elegans has been used for medical research since Sydney Brenner revealed its genetics in 1974 and has been introduced into pharmacology and toxicology in the past two decades. The data from C. elegans have been satisfactorily correlated with traditional experimental models. In this review, we summarize the advantages of C. elegans in assessing oxidative and antioxidative properties of natural products and introduce methods to construct an oxidative damage model in C. elegans. The biomarkers and signaling pathways involved in the oxidative stress of C. elegans are summarized, as well as the oxidation and antioxidation in target organs of the muscle, nervous, digestive and reproductive systems. This review provides an overview of the oxidative and antioxidative properties of natural products based on the model organism C. elegans.
Collapse
Affiliation(s)
- An Zhu
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China; (Y.W.); (W.B.)
- Correspondence: (A.Z.); (G.L.); (Q.W.); (H.L.)
| | - Fuli Zheng
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350108, China; (F.Z.); (H.H.)
| | - Wenjing Zhang
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing 100013, China;
| | - Ludi Li
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China; (L.L.); (Y.L.)
| | - Yingzi Li
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China; (L.L.); (Y.L.)
| | - Hong Hu
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350108, China; (F.Z.); (H.H.)
| | - Yajiao Wu
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China; (Y.W.); (W.B.)
- Department of Pathogen Biology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
| | - Wenqiang Bao
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China; (Y.W.); (W.B.)
- Department of Pathogen Biology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
| | - Guojun Li
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing 100013, China;
- School of Public Health, Capital Medical University, Beijing 100069, China
- Correspondence: (A.Z.); (G.L.); (Q.W.); (H.L.)
| | - Qi Wang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China; (L.L.); (Y.L.)
- Key Laboratory of State Administration of Traditional Chinese Medicine for Compatibility Toxicology, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
- Correspondence: (A.Z.); (G.L.); (Q.W.); (H.L.)
| | - Huangyuan Li
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350108, China; (F.Z.); (H.H.)
- The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350108, China
- Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350108, China
- Correspondence: (A.Z.); (G.L.); (Q.W.); (H.L.)
| |
Collapse
|
181
|
Abstract
Biology of aging is an active and rapidly expanding area of biomedical research. Over the years, focus of work in this field has been gradually shifting from studying the effects and symptoms of aging to searching for mechanisms of the aging process. Progress of this work led to an additional shift from looking for "the mechanism" of aging and formulating the corresponding "theories of aging" to appreciation that aging represents a net result of multiple physiological changes and their intricate interactions. It was also shown that mechanisms of aging include nutrient-dependent signaling pathways which have been remarkably conserved in the course of the evolution. Another important development in this field is increased emphasis on searching for pharmacological and environmental interventions that can extend healthspan or influence other aspects of aging. Progress in understanding the key role of aging as a risk factor for chronic disease provides impetus for these studies. Data from the recent pandemic provided additional evidence for the impact of age on resilience. Progress of work in this area also was influenced by major analytical and technological advances, including greatly improved methods for the study of gene expression, protein, lipids, and metabolites profiles, enhanced ability to produce various genetic modifications and novel approaches to assessment of biological age. Progress in research on the biology of aging provides reasons for optimism about the chances that safe and widely applicable anti-aging interventions with significant benefits for both individual and public health will be developed in the not too distant future.
Collapse
Affiliation(s)
- Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge St., P. O. Box 19628, Springfield, IL, 62794-9628, USA.
| |
Collapse
|
182
|
Filina O, Demirbas B, Haagmans R, van Zon JS. Temporal scaling in C. elegans larval development. Proc Natl Acad Sci U S A 2022; 119:e2123110119. [PMID: 35263226 PMCID: PMC8931370 DOI: 10.1073/pnas.2123110119] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/03/2022] [Indexed: 12/02/2022] Open
Abstract
SignificanceAn enduring mystery of development is how its timing is controlled, particularly for development after birth, where timing is highly flexible and depends on environmental conditions, such as food availability and diet. We followed timing of cell- and organism-level events in individual Caenorhabditis elegans larvae developing from hatching to adulthood, uncovering widespread variations in event timing, both between isogenic individuals in the same environment and when changing conditions and genotypes. However, in almost all cases, we found that events occurred at the same time, when time was rescaled by the duration of development measured in each individual. This observation of "temporal scaling" poses strong constraints on models to explain timing of larval development.
Collapse
Affiliation(s)
- Olga Filina
- Department of Autonomous Matter, AMOLF, Amsterdam, 1098 XG The Netherlands
| | - Burak Demirbas
- Department of Autonomous Matter, AMOLF, Amsterdam, 1098 XG The Netherlands
| | - Rik Haagmans
- Department of Autonomous Matter, AMOLF, Amsterdam, 1098 XG The Netherlands
| | - Jeroen S. van Zon
- Department of Autonomous Matter, AMOLF, Amsterdam, 1098 XG The Netherlands
| |
Collapse
|
183
|
Walker RF. A Mechanistic Theory of Development-Aging Continuity in Humans and Other Mammals. Cells 2022; 11:cells11050917. [PMID: 35269539 PMCID: PMC8909351 DOI: 10.3390/cells11050917] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 12/29/2022] Open
Abstract
There is consensus among biogerontologists that aging occurs either as the result of a purposeful genome-based, evolved program or due to spontaneous, randomly occurring, maladaptive events. Neither concept has yet identified a specific mechanism to explain aging’s emergence and acceleration during mid-life and beyond. Presented herein is a novel, unifying mechanism with empirical evidence that describes how aging becomes continuous with development. It assumes that aging emerges from deterioration of a regulatory process that directs morphogenesis and morphostasis. The regulatory system consists of a genome-wide “backbone” within which its specific genes are differentially expressed by the local epigenetic landscapes of cells and tissues within which they reside, thereby explaining its holistic nature. Morphostasis evolved in humans to ensure the nurturing of dependent offspring during the first decade of young adulthood when peak parental vitality prevails in the absence of aging. The strict redundancy of each morphostasis regulatory cycle requires sensitive dependence upon initial conditions to avoid initiating deterministic chaos behavior. However, when natural selection declines as midlife approaches, persistent, progressive, and specific DNA damage and misrepair changes the initial conditions of the regulatory process, thereby compromising morphostasis regulatory redundancy, instigating chaos, initiating senescence, and accelerating aging thereafter.
Collapse
|
184
|
Binti S, Melinda RV, Joseph BB, Edeen PT, Miller SD, Fay DS. A life cycle alteration can correct molting defects in Caenorhabditis elegans. Dev Biol 2022; 483:143-156. [PMID: 35038442 PMCID: PMC8867747 DOI: 10.1016/j.ydbio.2022.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/21/2021] [Accepted: 01/03/2022] [Indexed: 11/28/2022]
Abstract
Molting is a widespread feature in the development of many invertebrates, including nematodes and arthropods. In Caenorhabditis elegans, the highly conserved protein kinases NEKL-2/NEK8/9 and NEKL-3/NEK6/7 (NEKLs) promote molting through their involvement in the uptake and intracellular trafficking of epidermal cargos. We found that the relative requirements for NEKL-2 and NEKL-3 differed at different life-cycle stages and under different environmental conditions. Most notably, the transition from the second to the third larval stage (L2→L3 molt) required a higher level of NEKL function than during several other life stages or when animals had experienced starvation at the L1 stage. Specifically, larvae that entered the pre-dauer L2d stage could escape molting defects when transiting to the (non-dauer) L3 stage. Consistent with this, mutations that promote entry into L2d suppressed nekl-associated molting defects, whereas mutations that inhibit L2d entry reduced starvation-mediated suppression. We further showed that loss or reduction of NEKL functions led to defects in the transcription of cyclically expressed molting genes, many of which are under the control of systemic steroid hormone regulation. Moreover, the timing and severity of these transcriptional defects correlated closely with the strength of nekl alleles and with their stage of arrest. Interestingly, transit through L2d rescued nekl-associated expression defects in suppressed worms, providing an example of how life-cycle decisions can impact subsequent developmental events. Given that NEKLs are implicated in the uptake of sterols by the epidermis, we propose that loss of NEKLs leads to a physiological reduction in steroid-hormone signaling and consequent defects in the transcription of genes required for molting.
Collapse
Affiliation(s)
- Shaonil Binti
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY, 82071, USA
| | - Rosa V Melinda
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY, 82071, USA
| | - Braveen B Joseph
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY, 82071, USA
| | - Phillip T Edeen
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY, 82071, USA
| | - Sam D Miller
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY, 82071, USA
| | - David S Fay
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY, 82071, USA.
| |
Collapse
|
185
|
Liu YJ, Gao AW, Smith RL, Janssens GE, Panneman DM, Jongejan A, van Weeghel M, Vaz FM, Silvestrini MJ, Lapierre LR, MacInnes AW, Houtkooper RH. Reduced ech-6 expression attenuates fat-induced lifespan shortening in C. elegans. Sci Rep 2022; 12:3350. [PMID: 35233004 PMCID: PMC8888598 DOI: 10.1038/s41598-022-07397-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 02/17/2022] [Indexed: 11/30/2022] Open
Abstract
Deregulated energy homeostasis represents a hallmark of aging and results from complex gene-by-environment interactions. Here, we discovered that reducing the expression of the gene ech-6 encoding enoyl-CoA hydratase remitted fat diet-induced deleterious effects on lifespan in Caenorhabditis elegans, while a basal expression of ech-6 was important for survival under normal dietary conditions. Lipidomics revealed that supplementation of fat in ech-6-silenced worms had marginal effects on lipid profiles, suggesting an alternative fat utilization for energy production. Transcriptomics further suggest a causal relation between the lysosomal pathway, energy production, and the longevity effect conferred by the interaction between ech-6 and fat diets. Indeed, enhancing energy production from endogenous fat by overexpressing lysosomal lipase lipl-4 recapitulated the lifespan effects of fat diets on ech-6-silenced worms. Collectively, these results suggest that the gene ech-6 is potential modulator of metabolic flexibility and may be a target for promoting metabolic health and longevity.
Collapse
Affiliation(s)
- Yasmine J Liu
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Arwen W Gao
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.,Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Reuben L Smith
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Georges E Janssens
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Daan M Panneman
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.,Radboud Center for Mitochondrial Medicine, Department of Pediatrics, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - Aldo Jongejan
- Bioinformatics Laboratory, Department of Epidemiology and Data Science, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, AZ, Amsterdam, The Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.,Core Facility Metabolomics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Frédéric M Vaz
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.,Core Facility Metabolomics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Melissa J Silvestrini
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, 02912, USA
| | - Louis R Lapierre
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, 02912, USA
| | - Alyson W MacInnes
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
186
|
Lefoulon E, McMullen JG, Stock SP. Transcriptomic Analysis of Steinernema Nematodes Highlights Metabolic Costs Associated to Xenorhabdus Endosymbiont Association and Rearing Conditions. Front Physiol 2022; 13:821845. [PMID: 35283769 PMCID: PMC8914265 DOI: 10.3389/fphys.2022.821845] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/01/2022] [Indexed: 12/13/2022] Open
Abstract
Entomopathogenic nematodes of the genus Steinernema have a mutualistic relationship with bacteria of the genus Xenorhabdus and together they form an antagonist partnership against their insect hosts. The nematodes (third-stage infective juveniles, or IJs) protect the bacteria from the external environmental stressors and vector them from one insect host to another. Xenorhabdus produce secondary metabolites and antimicrobial compounds inside the insect that protect the cadaver from soil saprobes and scavengers. The bacteria also become the nematodes’ food, allowing them to grow and reproduce. Despite these benefits, it is yet unclear what the potential metabolic costs for Steinernema IJs are relative to the maintenance and vectoring of Xenorhabdus. In this study, we performed a comparative dual RNA-seq analysis of IJs of two nematode-bacteria partnerships: Steinernema carpocapsae-Xenorhabdus nematophila and Steinernema. puntauvense-Xenorhbdus bovienii. For each association, three conditions were studied: (1) IJs reared in the insect (in vivo colonized), (2) colonized IJs reared on liver-kidney agar (in vitro colonized), and (3) IJs depleted by the bacteria reared on liver-kidney agar (in vitro aposymbiotic). Our study revealed the downregulation of numerous genes involved in metabolism pathways, such as carbohydrate, amino acid, and lipid metabolism when IJs were reared in vitro, both colonized and without the symbiont. This downregulation appears to impact the longevity pathway, with the involvement of glycogen and trehalose metabolism, as well as arginine metabolism. Additionally, a differential expression of the venom protein known to be secreted by the nematodes was observed when both Steinernema species were depleted of their symbiotic partners. These results suggest Steinernema IJs may have a mechanism to adapt their virulence in absence of their symbionts.
Collapse
Affiliation(s)
- Emilie Lefoulon
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, United States
| | - John G. McMullen
- Department of Biology, Indiana University, Bloomington, IN, United States
| | - S. Patricia Stock
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, United States
- College of Agriculture, California State University Chico, Chico, CA, United States
- *Correspondence: S. Patricia Stock,
| |
Collapse
|
187
|
Youssef FS, Sobeh M, Dmirieh M, Bogari HA, Koshak AE, Wink M, Ashour ML, Elhady SS. Metabolomics-Based Profiling of Clerodendrum speciosum (Lamiaceae) Leaves Using LC/ESI/MS-MS and In Vivo Evaluation of Its Antioxidant Activity Using Caenorhabditis elegans Model. Antioxidants (Basel) 2022; 11:antiox11020330. [PMID: 35204212 PMCID: PMC8868248 DOI: 10.3390/antiox11020330] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 02/04/2023] Open
Abstract
We investigated the antioxidant activity of the total methanol extract of C. speciosum leaves (CST), the ethyl acetate (CSE), and the remaining aqueous (CSR) fractions in vitro, in vivo using Caenorhabditis elegans model, and in silico. LC-ESI-MS/MS analysis was employed for metabolic profiling of CST. ADME/TOPAKT prediction was performed to determine the potential pharmacokinetic, pharmacodynamic, and toxicity properties of the major identified phytoconstituents. All examined samples showed considerable antioxidant activity where CST, CSE, and CSR displayed EC50 values of 27.1, 16.2, and 21.3 µg/mL, respectively, in 2,2-diphenyl-1-picrylhydrazyl (DPPH•) assay, whereas they showed 11.44, 16.27, and 12.16 Fe2+ equivalents/mg of sample, respectively, in ferric reducing antioxidant power (FRAP) assay. CST, CSE, and CSR displayed total phenolic content of 262, 326, and 289 mg GAE/g sample, respectively. In vivo antioxidant study revealed that CST at 150 μg/mL increased the survival rate of C. elegans by 71.88% compared to untreated group. Regarding intracellular reactive oxygen species (ROS), worms treated with 150 μg/mL of CSE exhibited 60.42% reduction of ROS compared to the untreated group. Quantitation of hsp-16.2/GFP expression in Caenorhabditis elegans showed that worms treated with 150 μg/mL of CSR exerted 40.43% reduction in fluorescence with respect to the untreated group. LC-ESI-MS/MS of CST revealed the presence of sixteen secondary metabolites belonging mainly to polyphenolics with phenyl propanoids constituting the major detected class. The in silico study showed that rosmarinic acid displayed the best fitting within the active sites of Daf-2 protein with considerable safety profile and limited pharmacokinetic and pharmacodynamic that could be slightly enhanced by certain treatment.
Collapse
Affiliation(s)
- Fadia S. Youssef
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Abbasia, Cairo 11566, Egypt
- Correspondence: (F.S.Y.); (M.L.A.)
| | - Mansour Sobeh
- AgroBioSciences, Mohammed VI Polytechnic University, Lot 660–Hay MoulayRachid, Ben-Guerir 43150, Morocco; (M.S.); (M.W.)
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany;
| | - Malak Dmirieh
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany;
| | - Hanin A. Bogari
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Abdulrahman E. Koshak
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.E.K.); (S.S.E.)
| | - Michael Wink
- AgroBioSciences, Mohammed VI Polytechnic University, Lot 660–Hay MoulayRachid, Ben-Guerir 43150, Morocco; (M.S.); (M.W.)
| | - Mohamed L. Ashour
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Abbasia, Cairo 11566, Egypt
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah 21442, Saudi Arabia
- Correspondence: (F.S.Y.); (M.L.A.)
| | - Sameh S. Elhady
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.E.K.); (S.S.E.)
| |
Collapse
|
188
|
Lee SK. "Cutting and Burning Guts" Nourish the Young Caenorhabditis elegans lyse their guts to produce nutritious yolk milk to feed larvae. Mol Cells 2022; 45:1-3. [PMID: 35114642 PMCID: PMC8819495 DOI: 10.14348/molcells.2021.5036] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/01/2021] [Accepted: 12/24/2021] [Indexed: 11/27/2022] Open
Affiliation(s)
- Sun-Kyung Lee
- Department of Life Sciences, Research Institute for Natural Sciences, College of Natural Sciences, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
189
|
Zečić A, Dhondt I, Braeckman BP. Accumulation of Glycogen and Upregulation of LEA-1 in C. elegans daf-2(e1370) Support Stress Resistance, Not Longevity. Cells 2022; 11:245. [PMID: 35053361 PMCID: PMC8773926 DOI: 10.3390/cells11020245] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/26/2021] [Accepted: 01/07/2022] [Indexed: 02/04/2023] Open
Abstract
DAF-16-dependent activation of a dauer-associated genetic program in the C. elegans insulin/IGF-1 daf-2(e1370) mutant leads to accumulation of large amounts of glycogen with concomitant upregulation of glycogen synthase, GSY-1. Glycogen is a major storage sugar in C. elegans that can be used as a short-term energy source for survival, and possibly as a reservoir for synthesis of a chemical chaperone trehalose. Its role in mitigating anoxia, osmotic and oxidative stress has been demonstrated previously. Furthermore, daf-2 mutants show increased abundance of the group 3 late embryogenesis abundant protein LEA-1, which has been found to act in synergy with trehalose to exert its protective role against desiccation and heat stress in vitro, and to be essential for desiccation tolerance in C. elegans dauer larvae. Here we demonstrate that accumulated glycogen is not required for daf-2 longevity, but specifically protects against hyperosmotic stress, and serves as an important energy source during starvation. Similarly, lea-1 does not act to support daf-2 longevity. Instead, it contributes to increased resistance of daf-2 mutants to heat, osmotic, and UV stress. In summary, our experimental results suggest that longevity and stress resistance can be uncoupled in IIS longevity mutants.
Collapse
Affiliation(s)
| | | | - Bart P. Braeckman
- Laboratory of Aging Physiology and Molecular Evolution, Department of Biology, Ghent University, K. L. Ledeganckstraat 35, B-9000 Ghent, Belgium; (A.Z.); (I.D.)
| |
Collapse
|
190
|
Schwartz EKC, Sosner EN, Desmond HE, Lum SJ, Sze JY, Mobbs CV. Serotonin and Dopamine Mimic Glucose-Induced Reinforcement in C. elegans: Potential Role of NSM Neurons and the Serotonin Subtype 4 Receptor. Front Physiol 2022; 12:783359. [PMID: 34987416 PMCID: PMC8721000 DOI: 10.3389/fphys.2021.783359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/22/2021] [Indexed: 12/17/2022] Open
Abstract
Food produces powerful reinforcement that can lead to overconsumption and likely contributes to the obesity epidemic. The present studies examined molecular mechanisms mediating food-induced reinforcement in the model system C. elegans. After a 1-h training session during which food (bacteria) is paired with the odorant butanone, odor preference for butanone robustly increased. Glucose mimicked this effect of bacteria. Glucose-induced odor preference was enhanced similarly by prior food withdrawal or blocking glucose metabolism in the presence of food. Food- and glucose-induced odor preference was mimicked by serotonin signaling through the serotonin type-4 (5-HT4) receptor. Dopamine (thought to act primarily through a D1-like receptor) facilitated, whereas the D2 agonist bromocriptine blocked, food- and glucose-induced odor preference. Furthermore, prior food withdrawal similarly influenced reward produced by serotonin, dopamine, or food, implying post-synaptic enhancement of sensitivity to serotonin and dopamine. These results suggest that glucose metabolism plays a key role in mediating both food-induced reinforcement and enhancement of that reinforcement by prior food withdrawal and implicate serotonergic signaling through 5-HT4 receptor in the re-enforcing properties of food.
Collapse
Affiliation(s)
- Elizabeth K C Schwartz
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Eitan N Sosner
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Hayley E Desmond
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Stephanie J Lum
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ji Ying Sze
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Charles V Mobbs
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
191
|
Bayer EA, Liberatore KM, Schneider JR, Schlesinger E, He Z, Birnbaum S, Wightman B. Insulin signaling and osmotic stress response regulate arousal and developmental progression of C. elegans at hatching. Genetics 2022; 220:iyab202. [PMID: 34788806 PMCID: PMC8733457 DOI: 10.1093/genetics/iyab202] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/03/2021] [Indexed: 12/29/2022] Open
Abstract
The progression of animal development from embryonic to juvenile life depends on the coordination of organism-wide responses with environmental conditions. We found that two transcription factors that function in interneuron differentiation in Caenorhabditis elegans, fax-1, and unc-42, are required for arousal and progression from embryogenesis to larval life by potentiating insulin signaling. The combination of mutations in either transcription factor and a mutation in daf-2 insulin receptor results in a novel perihatching arrest phenotype; embryos are fully developed but inactive, often remaining trapped within the eggshell, and fail to initiate pharyngeal pumping. This pathway is opposed by an osmotic sensory response pathway that promotes developmental arrest and a sleep state at the end of embryogenesis in response to elevated salt concentration. The quiescent state induced by loss of insulin signaling or by osmotic stress can be reversed by mutations in genes that are required for sleep. Therefore, countervailing signals regulate late embryonic arousal and developmental progression to larval life, mechanistically linking the two responses. Our findings demonstrate a role for insulin signaling in an arousal circuit, consistent with evidence that insulin-related regulation may function in control of sleep states in many animals. The opposing quiescent arrest state may serve as an adaptive response to the osmotic threat from high salinity environments.
Collapse
Affiliation(s)
- Emily A Bayer
- Biology Department, Muhlenberg College, Allentown, PA 18104, USA
| | | | | | - Evan Schlesinger
- Biology Department, Muhlenberg College, Allentown, PA 18104, USA
| | - Zhengying He
- Biology Department, Muhlenberg College, Allentown, PA 18104, USA
| | - Susanna Birnbaum
- Biology Department, Muhlenberg College, Allentown, PA 18104, USA
| | - Bruce Wightman
- Biology Department, Muhlenberg College, Allentown, PA 18104, USA
| |
Collapse
|
192
|
Green CL, Lamming DW, Fontana L. Molecular mechanisms of dietary restriction promoting health and longevity. Nat Rev Mol Cell Biol 2022; 23:56-73. [PMID: 34518687 PMCID: PMC8692439 DOI: 10.1038/s41580-021-00411-4] [Citation(s) in RCA: 366] [Impact Index Per Article: 122.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2021] [Indexed: 02/08/2023]
Abstract
Dietary restriction with adequate nutrition is the gold standard for delaying ageing and extending healthspan and lifespan in diverse species, including rodents and non-human primates. In this Review, we discuss the effects of dietary restriction in these mammalian model organisms and discuss accumulating data that suggest that dietary restriction results in many of the same physiological, metabolic and molecular changes responsible for the prevention of multiple ageing-associated diseases in humans. We further discuss how different forms of fasting, protein restriction and specific reductions in the levels of essential amino acids such as methionine and the branched-chain amino acids selectively impact the activity of AKT, FOXO, mTOR, nicotinamide adenine dinucleotide (NAD+), AMP-activated protein kinase (AMPK) and fibroblast growth factor 21 (FGF21), which are key components of some of the most important nutrient-sensing geroprotective signalling pathways that promote healthy longevity.
Collapse
Affiliation(s)
- Cara L Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Luigi Fontana
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.
- Department of Clinical and Experimental Sciences, Brescia University School of Medicine, Brescia, Italy.
| |
Collapse
|
193
|
McKay FM, McCoy CJ, Crooks B, Marks NJ, Maule AG, Atkinson LE, Mousley A. In silico analyses of neuropeptide-like protein (NLP) profiles in parasitic nematodes. Int J Parasitol 2022; 52:77-85. [PMID: 34450132 PMCID: PMC8764417 DOI: 10.1016/j.ijpara.2021.07.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/01/2021] [Accepted: 07/05/2021] [Indexed: 01/06/2023]
Abstract
Nematode parasite infections cause disease in humans and animals and threaten global food security by reducing productivity in livestock and crop farming. The escalation of anthelmintic resistance in economically important nematode parasites underscores the need for the identification of novel drug targets in these worms. Nematode neuropeptide signalling is an attractive system for chemotherapeutic exploitation, with neuropeptide G-protein coupled receptors (NP-GPCRs) representing the lead targets. In order to successfully validate NP-GPCRs for parasite control it is necessary to characterise their function and importance to nematode biology. This can be aided through identification of receptor activating ligand(s) via deorphanisation. Such efforts require the identification of all neuropeptide ligands within parasites. Here we mined the genomes of nine therapeutically relevant pathogenic nematodes to characterise the neuropeptide-like protein complements and demonstrate that: (i) parasitic nematodes possess a reduced complement of neuropeptide-like protein-encoding genes relative to Caenorhabditis elegans; (ii) parasite neuropeptide-like protein profiles are broadly conserved between nematode clades; (iii) five Ce-nlps are completely conserved across the nematode species examined; (iv) the extent and position of neuropeptide-like protein-motif conservation is variable; (v) novel RPamide-encoding genes are present in parasitic nematodes; (vi) novel Allatostatin-C-like peptide encoding genes are present in both C. elegans and parasitic nematodes; (vii) novel neuropeptide-like protein families are absent in C. elegans; and (viii) highly conserved nematode neuropeptide-like proteins are bioactive. These data highlight the complexity of nematode neuropeptide-like proteins and reveal the need for nomenclature revision in this diverse neuropeptide family. The identification of neuropeptide-like protein ligands, and characterisation of those with functional relevance, advance our understanding of neuropeptide signalling to support exploitation of the neuropeptidergic system as an anthelmintic target.
Collapse
Affiliation(s)
- Fiona M McKay
- Microbes & Pathogen Biology, The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast BT9 5DL, United Kingdom
| | - Ciaran J McCoy
- Microbes & Pathogen Biology, The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast BT9 5DL, United Kingdom
| | - Bethany Crooks
- Microbes & Pathogen Biology, The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast BT9 5DL, United Kingdom
| | - Nikki J Marks
- Microbes & Pathogen Biology, The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast BT9 5DL, United Kingdom
| | - Aaron G Maule
- Microbes & Pathogen Biology, The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast BT9 5DL, United Kingdom
| | - Louise E Atkinson
- Microbes & Pathogen Biology, The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast BT9 5DL, United Kingdom
| | - Angela Mousley
- Microbes & Pathogen Biology, The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast BT9 5DL, United Kingdom.
| |
Collapse
|
194
|
DeVito LM, Barzilai N, Cuervo AM, Niedernhofer LJ, Milman S, Levine M, Promislow D, Ferrucci L, Kuchel GA, Mannick J, Justice J, Gonzales MM, Kirkland JL, Cohen P, Campisi J. Extending human healthspan and longevity: a symposium report. Ann N Y Acad Sci 2022; 1507:70-83. [PMID: 34498278 PMCID: PMC10231756 DOI: 10.1111/nyas.14681] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 08/09/2021] [Indexed: 12/13/2022]
Abstract
For many years, it was believed that the aging process was inevitable and that age-related diseases could not be prevented or reversed. The geroscience hypothesis, however, posits that aging is, in fact, malleable and, by targeting the hallmarks of biological aging, it is indeed possible to alleviate age-related diseases and dysfunction and extend longevity. This field of geroscience thus aims to prevent the development of multiple disorders with age, thereby extending healthspan, with the reduction of morbidity toward the end of life. Experts in the field have made remarkable advancements in understanding the mechanisms underlying biological aging and identified ways to target aging pathways using both novel agents and repurposed therapies. While geroscience researchers currently face significant barriers in bringing therapies through clinical development, proof-of-concept studies, as well as early-stage clinical trials, are underway to assess the feasibility of drug evaluation and lay a regulatory foundation for future FDA approvals in the future.
Collapse
Affiliation(s)
| | - Nir Barzilai
- Albert Einstein College of Medicine, Bronx, New York
| | | | | | - Sofiya Milman
- Albert Einstein College of Medicine, Bronx, New York
| | | | | | - Luigi Ferrucci
- National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - George A Kuchel
- University of Connecticut School of Medicine, Farmington, Connecticut
| | | | - Jamie Justice
- Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Mitzi M Gonzales
- University of Texas Health Sciences Center San Antonio, San Antonio, Texas
| | | | - Pinchas Cohen
- USC Leonard Davis School of Gerontology, Los Angeles, California
| | - Judith Campisi
- The Buck Institute for Research on Aging, Novato, California
- Lawrence Berkeley National Laboratory, Berkley, California
| |
Collapse
|
195
|
Li J, Liu D, Li D, Guo Y, Du H, Cao Y. Phytochemical composition and anti-aging activity of n-butanol extract of Hedyotis diffusa in Caenorhabditis elegans. Chem Biodivers 2021; 19:e202100685. [PMID: 34935259 DOI: 10.1002/cbdv.202100685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/21/2021] [Indexed: 11/08/2022]
Abstract
Hedyotis diffusa Willd. ( H. diffusa ), a kind of traditional Chinese medicine, has been evaluated to potential display antioxidant and anti-aging effects in vitro experiments. In this work, we investigated the effects on lifespan and stress resistance of the N-butanol extract from H. diffusa (NHD) in vivo using a Caenorhabditis elegans ( C. elegans ) model. The phytochemicals of NHD were identified by UPLC-ESI-qTOF-MS/MS method. NHD-treated wild-type N2 worms showed an increase in survival time under both normal and stress conditions. Meanwhile, NHD promoted the healthspan of nematodes by stimulating growth and development, reducing the deposition of age pigment, increasing the activities of superoxide dismutase (SOD) and glutathione peroxidase dismutase (GSH-Px), and decreasing the level of ROS without impairing fertility. Moreover, the upregulating of the expression of daf-16 , gst-4 , sod-3 , hsp12.6 genes and the downregulating of the expression of daf-2 were involved in the NHD-mediated lifespan extension. Finally, the increasing of the expression of GST-4::GFP in CL2166 transgenic nematodes and the life-span-extending activity of NHD was completely abolished in daf-2 and daf-16 mutants further revealed that the potential roles for these genes in NHD-induced longevity in C. elegans . Collectively, our findings suggest that NHD may have an active effect in healthy aging and age-related diseases.
Collapse
Affiliation(s)
- Jing Li
- Hubei University of Chinese Medicine, college of pharmcy, Hongshan district, 16# West road Huangjiahu, Wuhan, CHINA
| | - Di Liu
- Hubei University of Chinese Medicine, college of pharmcy, Hongshan district, 16# West road Huangjiahu, Wuhan, CHINA
| | - Danqing Li
- Hubei University of Chinese Medicine, college of pharmcy, Hongshan district, 16# West road Huangjiahu, Wuhan, CHINA
| | - Yujie Guo
- Hubei University of Chinese Medicine, college of pharmcy, Hongshan district, 16# West road Huangjiahu, Wuhan, CHINA
| | - Hongzhi Du
- Hubei University of Chinese Medicine, college of pharmcy, Hongshan district, 16# West road Huangjiahu, Wuhan, CHINA
| | - Yan Cao
- Hubei University of Chinese Medicine, college of pharmacy, Hongshan district, 16# West road Huangjiahu, 430065, Wuhan, CHINA
| |
Collapse
|
196
|
Bartke A, Brown-Borg H. Mutations Affecting Mammalian Aging: GH and GHR vs IGF-1 and Insulin. Front Genet 2021; 12:667355. [PMID: 34899820 PMCID: PMC8652133 DOI: 10.3389/fgene.2021.667355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Holly Brown-Borg
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| |
Collapse
|
197
|
Zhao J, Yu J, Zhi Q, Yuan T, Lei X, Zeng K, Ming J. Anti-aging effects of the fermented anthocyanin extracts of purple sweet potato on Caenorhabditis elegans. Food Funct 2021; 12:12647-12658. [PMID: 34821891 DOI: 10.1039/d1fo02671b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Anthocyanins have anti-inflammatory, anticarcinogenic and antioxidant properties and anti-aging effects as well as potential application as pigments. The metabolism of anthocyanins in fermented food has attracted increasing attention. However, the effect of lactic acid bacteria (LAB) fermentation on its anti-aging activity remains mostly unknown. The current study aimed to investigate the compositions, antioxidant activities and anti-aging effect of fermented purple sweet potato anthocyanins (FSPA) on aging Caenorhabditis elegans compared to raw purple sweet potato anthocyanins (PSPA). Results showed that anthocyanins were degraded into more bioavailable phenolic acids by Weissella confusa fermentation. PSPA and FSPA can extend the lifespan of C. elegans by 26.7% and 37.5%, respectively, through improving the activity of antioxidant enzymes as well as decreasing MDA content, ROS levels and lipofuscin accumulation. Pretreatment of the worms with PSPA and FSPA induced their potential to resist to thermal tolerance and oxidative stress, and FSPA exerted a higher anti-stress effect than PSPA. Moreover, FSPA supplementation upregulated the mRNA expressions of genes daf-16, hsp-16.2, sir-2.1, skn-1 and sod-3 and downregulated the expression of daf-2 in the nematodes, whereas PSPA only induced the increase in the expressions of sir-2.1, skn-1 and sod-3. Overall, FSPA can improve stress resistance and extend the lifespan of C. elegans by both insulin/IGF-1 signaling pathway and dietary restriction pathway, providing a theoretical basis for the application of PSPA in fermented food as functional pigments.
Collapse
Affiliation(s)
- Jichun Zhao
- College of Food Science, Southwest University, Chongqing, 400715, People's Republic of China. .,Department of Food Science, Faculty of Science, University of Copenhagen, Frederiksberg 1958, Denmark
| | - Jie Yu
- College of Food Science, Southwest University, Chongqing, 400715, People's Republic of China.
| | - Qi Zhi
- College of Food Science, Southwest University, Chongqing, 400715, People's Republic of China.
| | - Tingting Yuan
- College of Food Science, Southwest University, Chongqing, 400715, People's Republic of China.
| | - Xiaojuan Lei
- College of Food Science, Southwest University, Chongqing, 400715, People's Republic of China.
| | - Kaifang Zeng
- College of Food Science, Southwest University, Chongqing, 400715, People's Republic of China. .,Research Center of Food Storage & Logistics, Southwest University, Chongqing 400715, People's Republic of China
| | - Jian Ming
- College of Food Science, Southwest University, Chongqing, 400715, People's Republic of China. .,Research Center of Food Storage & Logistics, Southwest University, Chongqing 400715, People's Republic of China
| |
Collapse
|
198
|
Das S, Min S, Prahlad V. Gene bookmarking by the heat shock transcription factor programs the insulin-like signaling pathway. Mol Cell 2021; 81:4843-4860.e8. [PMID: 34648748 PMCID: PMC8642288 DOI: 10.1016/j.molcel.2021.09.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/09/2021] [Accepted: 09/17/2021] [Indexed: 12/13/2022]
Abstract
Maternal stress can have long-lasting epigenetic effects on offspring. To examine how epigenetic changes are triggered by stress, we examined the effects of activating the universal stress-responsive heat shock transcription factor HSF-1 in the germline of Caenorhabditis elegans. We show that, when activated in germ cells, HSF-1 recruits MET-2, the putative histone 3 lysine 9 (H3K9) methyltransferase responsible for repressive H3K9me2 (H3K9 dimethyl) marks in chromatin, and negatively bookmarks the insulin receptor daf-2 and other HSF-1 target genes. Increased H3K9me2 at these genes persists in adult progeny and shifts their stress response strategy away from inducible chaperone expression as a mechanism to survive stress and instead rely on decreased insulin/insulin growth factor (IGF-1)-like signaling (IIS). Depending on the duration of maternal heat stress exposure, this epigenetic memory is inherited by the next generation. Thus, paradoxically, HSF-1 recruits the germline machinery normally responsible for erasing transcriptional memory but, instead, establishes a heritable epigenetic memory of prior stress exposure.
Collapse
Affiliation(s)
- Srijit Das
- Department of Biology, Aging Mind and Brain Initiative, 143 Biology Building, Iowa City, IA 52242-1324, USA
| | - Sehee Min
- Department of Biology, Aging Mind and Brain Initiative, 143 Biology Building, Iowa City, IA 52242-1324, USA
| | - Veena Prahlad
- Department of Biology, Aging Mind and Brain Initiative, 143 Biology Building, Iowa City, IA 52242-1324, USA; Department of Biology, 143 Biology Building, Iowa City, IA 52242-1324, USA; Iowa Neuroscience Institute, 169 Newton Road, 2312 Pappajohn Biomedical Discovery Building, Iowa City, IA 52242, USA.
| |
Collapse
|
199
|
Phosphorothioate-DNA bacterial diet reduces the ROS levels in C. elegans while improving locomotion and longevity. Commun Biol 2021; 4:1335. [PMID: 34824369 PMCID: PMC8617147 DOI: 10.1038/s42003-021-02863-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 11/05/2021] [Indexed: 11/20/2022] Open
Abstract
DNA phosphorothioation (PT) is widely distributed in the human gut microbiome. In this work, PT-diet effect on nematodes was studied with PT-bioengineering bacteria. We found that the ROS level decreased by about 20–50% and the age-related lipofuscin accumulation was reduced by 15–25%. Moreover, the PT-feeding worms were more active at all life periods, and more resistant to acute stressors. Intriguingly, their lifespans were prolonged by ~21.7%. Comparative RNA-seq analysis indicated that many gene expressions were dramatically regulated by PT-diet, such as cysteine-rich protein (scl-11/12/13), sulfur-related enzyme (cpr-2), longevity gene (jnk-1) and stress response (sod-3/5, gps-5/6, gst-18/20, hsp-12.6). Both the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis suggested that neuroactivity pathways were upregulated, while phosphoryl transfer and DNA-repair pathways were down-regulated in good-appetite young worms. The findings pave the way for pro-longevity of multicellular organisms by PT-bacterial interference. Qiang Huang et al. fed C. elegans with E. coli containing phosphorothioate (PT) DNA or a control strain and evaluated the impact on animal physiology. They observed that worms fed PT( + ) diets exhibited low reactive oxygen species, more active movement, and a longer lifespan compared to controls, suggesting that PT-DNA may have a positive effect on animal health.
Collapse
|
200
|
daf-16/FOXO blocks adult cell fate in Caenorhabditis elegans dauer larvae via lin-41/TRIM71. PLoS Genet 2021; 17:e1009881. [PMID: 34780472 PMCID: PMC8629381 DOI: 10.1371/journal.pgen.1009881] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/29/2021] [Accepted: 10/15/2021] [Indexed: 02/07/2023] Open
Abstract
Many tissue-specific stem cells maintain the ability to produce multiple cell types during long periods of non-division, or quiescence. FOXO transcription factors promote quiescence and stem cell maintenance, but the mechanisms by which FOXO proteins promote multipotency during quiescence are still emerging. The single FOXO ortholog in C. elegans, daf-16, promotes entry into a quiescent and stress-resistant larval stage called dauer in response to adverse environmental cues. During dauer, stem and progenitor cells maintain or re-establish multipotency to allow normal development to resume after dauer. We find that during dauer, daf-16/FOXO prevents epidermal stem cells (seam cells) from prematurely adopting differentiated, adult characteristics. In particular, dauer larvae that lack daf-16 misexpress collagens that are normally adult-enriched. Using col-19p::gfp as an adult cell fate marker, we find that all major daf-16 isoforms contribute to opposing col-19p::gfp expression during dauer. By contrast, daf-16(0) larvae that undergo non-dauer development do not misexpress col-19p::gfp. Adult cell fate and the timing of col-19p::gfp expression are regulated by the heterochronic gene network, including lin-41 and lin-29. lin-41 encodes an RNA-binding protein orthologous to LIN41/TRIM71 in mammals, and lin-29 encodes a conserved zinc finger transcription factor. In non-dauer development, lin-41 opposes adult cell fate by inhibiting the translation of lin-29, which directly activates col-19 transcription and promotes adult cell fate. We find that during dauer, lin-41 blocks col-19p::gfp expression, but surprisingly, lin-29 is not required in this context. Additionally, daf-16 promotes the expression of lin-41 in dauer larvae. The col-19p::gfp misexpression phenotype observed in dauer larvae with reduced daf-16 requires the downregulation of lin-41, but does not require lin-29. Taken together, this work demonstrates a novel role for daf-16/FOXO as a heterochronic gene that promotes expression of lin-41/TRIM71 to contribute to multipotent cell fate in a quiescent stem cell model. In adults and juveniles, tissue-specific stem cells divide as needed to replace cells that are lost due to injury or normal wear and tear. Many stem cells spend long periods of time in cellular quiescence, or non-division. During quiescence, stem cells remain multipotent, where they retain the ability to produce all cell types within their tissue. In this study, we define a new role for the FOXO protein DAF-16 in promoting multipotency during the quiescent C. elegans dauer larva stage. C. elegans larvae enter dauer midway through development in response to adverse environmental conditions. Epidermal stem cells are multipotent in C. elegans larvae but differentiate at adulthood, a process controlled by the “heterochronic” genes. We found that daf-16 blocks the expression of adult cell fate specifically in dauer larvae by promoting the expression of the heterochronic gene lin-41. lin-41 normally blocks adult fate by repressing the expression of another heterochronic gene, lin-29, but surprisingly, lin-29 is not needed for the expression of adult cell fate in this context. These findings may be relevant to mammals where the orthologs of daf-16 and lin-41 are important in stem cell maintenance and opposing differentiation.
Collapse
|