1
|
Santos PKF, Kapheim KM. Convergent Evolution Associated with the Loss of Developmental Diapause May Promote Extended Lifespan in Bees. Genome Biol Evol 2024; 16:evae255. [PMID: 39579066 PMCID: PMC11632380 DOI: 10.1093/gbe/evae255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024] Open
Abstract
Diapause has long been proposed to play a significant role in the evolution of eusociality in Hymenoptera. Recent studies have shown that shifts in the diapause stage precede social evolution in wasps and bees; however, the genomic basis remains unknown. Given the overlap in molecular pathways that regulate diapause and lifespan, we hypothesized that the evolutionary loss of developmental diapause may lead to extended lifespan among adults, which is a prerequisite for the evolution of eusociality. To test whether the loss of prepupal diapause is followed by genomic changes associated with lifespan extension, we compared 27 bee genomes with or without prepupal diapause. Our results point to several potential mechanisms for lifespan extension in species lacking prepupal diapause, including the loss of the growth hormone PTTH and its receptor TORSO, along with convergent selection in genes known to regulate lifespan in animals. Specifically, we observed purifying selection of prolongevity genes and relaxed selection of antilongevity genes within the IIS/TOR pathway in species that have lost prepupal diapause. Changes in selection pressures on this pathway may lead to the evolution of new phenotypes, such as lifespan extension and altered responses to nutritional signals that are crucial for social evolution.
Collapse
Affiliation(s)
| | - Karen M Kapheim
- Department of Biology, Utah State University, Logan, UT 84322, USA
| |
Collapse
|
2
|
Berk Ş. Insulin and IGF-1 extend the lifespan of Caenorhabditis elegans by inhibiting insulin/insulin-like signaling and mTOR signaling pathways: C. elegans - Focused cancer research. Biochem Biophys Res Commun 2024; 729:150347. [PMID: 38976945 DOI: 10.1016/j.bbrc.2024.150347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/10/2024]
Abstract
The mutations in Caenorhabditis elegans (C. elegans) that extend lifespan slow down aging by interfering with several signaling pathways, including the insulin/IGF-1 signaling (IIS) pathway, AMP-activated protein kinase (AMPK), and mechanistic target of rapamycin (mTOR). The tumor suppressor pRb (retinoblastoma protein) is believed to be involved in almost all human cancers. Lin-35, the C. elegans orthologue of the tumor suppressor pRb, was included in the study to explore the effects of insulin and IGF-1 because it has been linked to cancer-related pRb function in mammals and exhibits a tumor suppressor effect by inhibiting mTOR or IIS signaling. According to our results, IGF-1 or insulin increased the lifespan of lin-35 worms compared to N2 worms by increasing fertilization efficiency, also causing a significant increase in body size. It was concluded that the expression of daf-2 and rsks-1 decreased after insulin or IGF-1 administration, thus extending the lifespan of C. elegans lin-35 worms through both IIS and mTOR-dependent mechanisms. This suggests that it was mediated by the combined effect of the TOR and IIS pathways. These results, especially obtained in cancer-associated mutant lin-35 worms, will be useful in elucidating the C. elegans cancer model in the future.
Collapse
Affiliation(s)
- Şeyda Berk
- Department of Molecular Biology and Genetics, Faculty of Science, Sivas Cumhuriyet University, Sivas, 58140, Turkey; Advanced Technology Research and Application Center (CUTAM), Sivas Cumhuriyet University, Sivas, 58140, Turkey.
| |
Collapse
|
3
|
Riordan R, Rong W, Yu Z, Ross G, Valerio J, Dimas-Muñoz J, Heredia V, Magnusson K, Galvan V, Perez VI. Effect of Nrf2 loss on senescence and cognition of tau-based P301S mice. GeroScience 2023; 45:1451-1469. [PMID: 36976489 PMCID: PMC10400516 DOI: 10.1007/s11357-023-00760-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/20/2023] [Indexed: 03/29/2023] Open
Abstract
Cellular senescence may contribute to chronic inflammation involved in the progression of age-related diseases such as Alzheimer's disease (AD), and its removal prevents cognitive impairment in a model of tauopathy. Nrf2, the major transcription factor for damage response pathways and regulators of inflammation, declines with age. Our previous work showed that silencing Nrf2 gives rise to premature senescence in cells and mice. Others have shown that Nrf2 ablation can exacerbate cognitive phenotypes of some AD models. In this study, we aimed to understand the relationship between Nrf2 elimination, senescence, and cognitive impairment in AD, by generating a mouse model expressing a mutant human tau transgene in an Nrf2 knockout (Nrf2KO) background. We assessed senescent cell burden and cognitive decline of P301S mice in the presence and absence of Nrf2. Lastly, we administered 4.5-month-long treatments with two senotherapeutic drugs to analyze their potential to prevent senescent cell burden and cognitive decline: the senolytic drugs dasatinib and quercetin (DQ) and the senomorphic drug rapamycin. Nrf2 loss accelerated the onset of hind-limb paralysis in P301S mice. At 8.5 months of age, P301S mice did not exhibit memory deficits, while P301S mice without Nrf2 were significantly impaired. However, markers of senescence were not elevated by Nrf2 ablation in any of tissues that we examined. Neither drug treatment improved cognitive performance, nor did it reduce expression of senescence markers in brains of P301S mice. Contrarily, rapamycin treatment at the doses used delayed spatial learning and led to a modest decrease in spatial memory. Taken together, our data suggests that the emergence of senescence may be causally associated with onset of cognitive decline in the P301S model, indicate that Nrf2 protects brain function in a model of AD through mechanisms that may include, but do not require the inhibition of senescence, and suggest possible limitations for DQ and rapamycin as therapies for AD.
Collapse
Affiliation(s)
- Ruben Riordan
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, 351 Linus Pauling Science Center, Corvallis, OR, 97331, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - Wang Rong
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, 351 Linus Pauling Science Center, Corvallis, OR, 97331, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - Zhen Yu
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, 351 Linus Pauling Science Center, Corvallis, OR, 97331, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - Grace Ross
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, 351 Linus Pauling Science Center, Corvallis, OR, 97331, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - Juno Valerio
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, 351 Linus Pauling Science Center, Corvallis, OR, 97331, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - Jovita Dimas-Muñoz
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, 351 Linus Pauling Science Center, Corvallis, OR, 97331, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - Valeria Heredia
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, 351 Linus Pauling Science Center, Corvallis, OR, 97331, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - Kathy Magnusson
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - Veronica Galvan
- Department of Biochemistry and Molecular Biology, Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 740 Stanton L. Young Bvd BMSB 821, Oklahoma City, OK, 73104, USA.
- Oklahoma City VA Medical Center, US Department of Veterans Affairs, Oklahoma City, OK, USA.
| | - Viviana I Perez
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, 351 Linus Pauling Science Center, Corvallis, OR, 97331, USA.
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA.
| |
Collapse
|
4
|
Steenwinkel TE, Hamre KK, Werner T. The use of non-model Drosophila species to study natural variation in TOR pathway signaling. PLoS One 2022; 17:e0270436. [PMID: 36137094 PMCID: PMC9499319 DOI: 10.1371/journal.pone.0270436] [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: 06/09/2022] [Accepted: 09/06/2022] [Indexed: 11/25/2022] Open
Abstract
Nutrition and growth are strongly linked, but not much is known about how nutrition leads to growth. To understand the connection between nutrition through the diet, growth, and proliferation, we need to study the phenotypes resulting from the activation and inhibition of central metabolic pathways. One of the most highly conserved metabolic pathways across eukaryotes is the Target of Rapamycin (TOR) pathway, whose primary role is to detect the availability of nutrients and to either induce or halt cellular growth. Here we used the model organism Drosophila melanogaster (D. mel.) and three non-model Drosophila species with different dietary needs, Drosophila guttifera (D. gut.), Drosophila deflecta (D. def.), and Drosophila tripunctata (D. tri.), to study the effects of dietary amino acid availability on fecundity and longevity. In addition, we inhibited the Target of Rapamycin (TOR) pathway, using rapamycin, to test how the inhibition interplays with the nutritional stimuli in these four fruit fly species. We hypothesized that the inhibition of the TOR pathway would reverse the phenotypes observed under conditions of overfeeding. Our results show that female fecundity increased with higher yeast availability in all four species but decreased in response to TOR inhibition. The longevity data were more varied: most species experienced an increase in median lifespan in both genders with an increase in yeast availability, while the lifespan of D. mel. females decreased. When exposed to the TOR inhibitor rapamycin, the life spans of most species decreased, except for D. tri, while we observed a major reduction in fecundity across all species. The obtained data can benefit future studies on the evolution of metabolism by showing the potential of using non-model species to track changes in metabolism. Particularly, our data show the possibility to use relatively closely related Drosophila species to gain insight on the evolution of TOR signaling.
Collapse
Affiliation(s)
- Tessa E. Steenwinkel
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan, United States of America
| | - Kailee K. Hamre
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan, United States of America
| | - Thomas Werner
- Department of Biological Sciences, Michigan Technological University, Houghton, Michigan, United States of America
- * E-mail:
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Dela Justina V, Miguez JSG, Priviero F, Sullivan JC, Giachini FR, Webb RC. Sex Differences in Molecular Mechanisms of Cardiovascular Aging. FRONTIERS IN AGING 2021; 2:725884. [PMID: 35822017 PMCID: PMC9261391 DOI: 10.3389/fragi.2021.725884] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease (CVD) is still the leading cause of illness and death in the Western world. Cardiovascular aging is a progressive modification occurring in cardiac and vascular morphology and physiology where increased endothelial dysfunction and arterial stiffness are observed, generally accompanied by increased systolic blood pressure and augmented pulse pressure. The effects of biological sex on cardiovascular pathophysiology have long been known. The incidence of hypertension is higher in men, and it increases in postmenopausal women. Premenopausal women are protected from CVD compared with age-matched men and this protective effect is lost with menopause, suggesting that sex-hormones influence blood pressure regulation. In parallel, the heart progressively remodels over the course of life and the pattern of cardiac remodeling also differs between the sexes. Lower autonomic tone, reduced baroreceptor response, and greater vascular function are observed in premenopausal women than men of similar age. However, postmenopausal women have stiffer arteries than their male counterparts. The biological mechanisms responsible for sex-related differences observed in cardiovascular aging are being unraveled over the last several decades. This review focuses on molecular mechanisms underlying the sex-differences of CVD in aging.
Collapse
Affiliation(s)
- Vanessa Dela Justina
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | | | - Fernanda Priviero
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC, United States
| | - Jennifer C. Sullivan
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Fernanda R. Giachini
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
- Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - R. Clinton Webb
- Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
8
|
Liu L, Guo P, Wang P, Zheng S, Qu Z, Liu N. The Review of Anti-aging Mechanism of Polyphenols on Caenorhabditis elegans. Front Bioeng Biotechnol 2021; 9:635768. [PMID: 34327192 PMCID: PMC8314386 DOI: 10.3389/fbioe.2021.635768] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 06/07/2021] [Indexed: 12/11/2022] Open
Abstract
Micronutrients extracted from natural plants or made by biological synthesis are widely used in anti-aging research and applications. Among more than 30 effective anti-aging substances, employing polyphenol organic compounds for modification or delaying of the aging process attracts great interest because of their distinct contribution in the prevention of degenerative diseases, such as cardiovascular disease and cancer. There is a profound potential for polyphenol extracts in the research of aging and the related diseases of the elderly. Previous studies have mainly focused on the properties of polyphenols implicated in free radical scavenging; however, the anti-oxidant effect cannot fully elaborate its biological functions, such as neuroprotection, Aβ protein production, ion channel coupling, and signal transduction pathways. Caenorhabditis elegans (C. elegans) has been considered as an ideal model organism for exploring the mechanism of anti-aging research and is broadly utilized in screening for natural bioactive substances. In this review, we have described the molecular mechanisms and pathways responsible for the slowdown of aging processes exerted by polyphenols. We also have discussed the possible mechanisms for their anti-oxidant and anti-aging properties in C. elegans from the perspective of different classifications of the specific polyphenols, such as flavonols, anthocyanins, flavan-3-ols, hydroxybenzoic acid, hydroxycinnamic acid, and stilbenes.
Collapse
Affiliation(s)
- Limin Liu
- College of Public Health, Zhengzhou University, Zhengzhou, China.,Institute of Chronic Disease Risks Assessment, School of Nursing and Health, Henan University, Kaifeng, China
| | - Peisen Guo
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Peixi Wang
- Institute of Chronic Disease Risks Assessment, School of Nursing and Health, Henan University, Kaifeng, China
| | - Shanqing Zheng
- School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Zhi Qu
- Institute of Chronic Disease Risks Assessment, School of Nursing and Health, Henan University, Kaifeng, China
| | - Nan Liu
- College of Public Health, Zhengzhou University, Zhengzhou, China.,Institute of Chronic Disease Risks Assessment, School of Nursing and Health, Henan University, Kaifeng, China.,Institute of Environment and Health, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
9
|
Gillsjö C, Karlsson S, Ståhl F, Eriksson I. Lifestyle's influence on community-dwelling older adults' health: A mixed-methods study design. Contemp Clin Trials Commun 2021; 21:100687. [PMID: 33385096 PMCID: PMC7770477 DOI: 10.1016/j.conctc.2020.100687] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 10/28/2020] [Accepted: 12/01/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Aging often involves health problems and difficulties, such as physical and psychological impairments, isolation, and loneliness, causing social and existential consequences. Studies have explored aging from different perspectives. However, few studies have examined healthy older adults' genetic backgrounds, lifestyles, and meaning in life separately or in combination. This study aims to describe how healthy older adults experience aging, health, lifestyles, and meaning in life and explore potential genetic correlations. METHODS AND DESIGN The project will comprise three main parts: a quantitative section featuring the development and testing of a lifestyle questionnaire, a quantitative genetic analysis, and a qualitative interview study. Participants will be community-dwelling, healthy, older adults between 70 and 95 years of age. A sample size of 800 older adults will be invited to participate in seminars in collaboration with the national Swedish association Active Seniors. Data will be collected through lifestyle questionnaire, DNA extracted from saliva samples, and interviews. Based on questionnaire responses, profile groups will be created and compared statistically with variations in genetic backgrounds, providing the basis for recruiting participants to the qualitative interviews. DISCUSSION This study's expected outcome will be to gain knowledge about variations in genetic backgrounds correlated with individual experiences regarding aging, health, and meaning in life. This knowledge can improve the understanding of motivations for healthy lifestyle changes. The results can reveal potential implications for individual prerequisites to healthy aging and how health-promoting aging and lifestyle counseling can be adjusted to meet individual needs.
Collapse
Affiliation(s)
- Catharina Gillsjö
- School of Health Sciences, University of Skövde, Skövde, Sweden
- College of Nursing, University of Rhode Island, Kingston, USA
| | - Sandra Karlsson
- Department of Natural Science and Biomedicine, School of Health and Welfare, Jönköping University, Jönköping, Sweden
| | - Fredrik Ståhl
- Faculty of Caring Science, Work Life and Social Welfare, University of Borås, Borås, Sweden
| | - Irene Eriksson
- School of Health Sciences, University of Skövde, Skövde, Sweden
| |
Collapse
|
10
|
Zajitschek F, Georgolopoulos G, Vourlou A, Ericsson M, Zajitschek SRK, Friberg U, Maklakov AA. Evolution Under Dietary Restriction Decouples Survival From Fecundity in Drosophila melanogaster Females. J Gerontol A Biol Sci Med Sci 2020; 74:1542-1548. [PMID: 29718269 DOI: 10.1093/gerona/gly070] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 04/10/2018] [Indexed: 01/17/2023] Open
Abstract
One of the key tenets of life-history theory is that reproduction and survival are linked and that they trade-off with each other. When dietary resources are limited, reduced reproduction with a concomitant increase in survival is commonly observed. It is often hypothesized that this dietary restriction effect results from strategically reduced investment in reproduction in favor of somatic maintenance to survive starvation periods until resources become plentiful again. We used experimental evolution to test this "waiting-for-the-good-times" hypothesis, which predicts that selection under sustained dietary restriction will favor increased investment in reproduction at the cost of survival because "good-times" never come. We assayed fecundity and survival of female Drosophila melanogaster fruit flies that had evolved for 50 generations on three different diets varying in protein content-low (classic dietary restriction diet), standard, and high-in a full-factorial design. High-diet females evolved overall increased fecundity but showed reduced survival on low and standard diets. Low-diet females evolved reduced survival on low diet without corresponding increase in reproduction. In general, there was little correspondence between the evolution of survival and fecundity across all dietary regimes. Our results contradict the hypothesis that resource reallocation between fecundity and somatic maintenance underpins life span extension under dietary restriction.
Collapse
Affiliation(s)
- Felix Zajitschek
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia.,Department of Animal Ecology, Evolutionary Biology Centre, Uppsala University, Sweden
| | | | - Anna Vourlou
- Department of Animal Ecology, Evolutionary Biology Centre, Uppsala University, Sweden
| | - Maja Ericsson
- Department of Animal Ecology, Evolutionary Biology Centre, Uppsala University, Sweden
| | - Susanne R K Zajitschek
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia.,Doñana Biological Station, EBD-CSIC, Seville, Spain.,Department of Evolutionary Biology, Evolutionary Biology Centre, Uppsala University, Sweden
| | - Urban Friberg
- Department of Evolutionary Biology, Evolutionary Biology Centre, Uppsala University, Sweden.,IFM Biology, AVIAN Behavioural, Genomics and Physiology Group, Linköping University, Sweden
| | - Alexei A Maklakov
- Department of Animal Ecology, Evolutionary Biology Centre, Uppsala University, Sweden.,School of Biological Sciences, Norwich Research Park, University of East Anglia, UK
| |
Collapse
|
11
|
Wang T, Wang Y, Liu L, Jiang Z, Li X, Tong R, He J, Shi J. Research progress on sirtuins family members and cell senescence. Eur J Med Chem 2020; 193:112207. [PMID: 32222662 DOI: 10.1016/j.ejmech.2020.112207] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 02/13/2020] [Accepted: 03/04/2020] [Indexed: 02/05/2023]
Abstract
Human aging is a phenomenon of gradual decline and loss of cell, tissue, organ and other functions under the action of external environment and internal factors. It is mainly related to genomic instability, telomere wear, mitochondrial dysfunction, protein balance disorder, antioxidant damage, microRNA expression disorder and so on. Sirtuins protein is a kind of deacetylase which can regulate cell metabolism and participate in a variety of cell physiological functions. It has been found that sirtuins family can prolong the lifespan of yeast. Sirtuins can inhibit human aging through many signaling pathways, including apoptosis signaling pathway, mTOR signaling pathway, sirtuins signaling pathway, AMPK signaling pathway, phosphatidylinositol 3 kinase (PI3K) signaling pathway and so on. Based on this, this paper reviews the action principle of anti-aging star members of sirtuins family Sirt1, Sirt3 and Sirt6 on anti-aging related signaling pathways and typical compounds, in order to provide ideas for the screening of anti-aging compounds of sirtuins family members.
Collapse
Affiliation(s)
- Ting Wang
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yujue Wang
- Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, China
| | - Li Liu
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Zhongliang Jiang
- Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Xingxing Li
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Rongsheng Tong
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jun He
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, 610041, China.
| | - Jianyou Shi
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
12
|
Lian S, Wang J, Zhang L, Xing Q, Hu N, Liu S, Dai X, Zhang F, Hu X, Bao Z, Wang S. Integration of Biochemical, Cellular, and Genetic Indicators for Understanding the Aging Process in a Bivalve Mollusk Chlamys farreri. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2019; 21:718-730. [PMID: 31392593 DOI: 10.1007/s10126-019-09917-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/24/2019] [Indexed: 06/10/2023]
Abstract
The major causal factors for the irreversible decline in physical vitality during organismal aging are postulated to be a chronic state of cellular redox imbalance, metabolic toxicity, and impaired energy homeostasis. We assessed whether the relevant enzyme activity, oxidative stress, and intracellular ATP might be causally involved in the aging of short-lived Chlamys farreri (life span 4~5 years). A total of eight related biochemical and cellular indicators were chosen for the subsequent analysis. All the indicators were measured in seven different tissues from scallops aged one to four years, and our data support that the aging of C. farreri is associated with attenuated tissue enzyme activity as well as a decreased metabolic rate. Through principal component analysis, we developed an integrated vigor index for each tissue for comprehensive age-related fitness evaluation. Remarkably, all tissue-integrated vigor indexes significantly declined with age, and the kidney was observed to be the most representative tissue. Further transcriptional profiling of the enzymatic genes provided additional detail on the molecular responses that may underlie the corresponding biochemical results. Moreover, these critical molecular responses may be attributed to the conserved hierarchical regulators, e.g., FOXO, AMPKs, mTOR, and IGF1R, which were identified as potentially novel markers for chronic fitness decline with age in bivalves. The present study provides a systematic approach that could potentially benefit the global assessment of the aging process in C. farreri and provide detailed evaluation of the biochemical, cellular, and genetic indicators that might be involved. This information may assist in a better understanding of bivalve adaptability and life span.
Collapse
Affiliation(s)
- Shanshan Lian
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Jing Wang
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
| | - Lingling Zhang
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Qiang Xing
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Naina Hu
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
| | - Sinuo Liu
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
| | - Xiaoting Dai
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
| | - Fengmei Zhang
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
| | - Xiaoli Hu
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Zhenmin Bao
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
| | - Shi Wang
- MOE Key Laboratory of Marine Genetics and Breeding, Ocean University of China, Qingdao, 266003, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| |
Collapse
|
13
|
Ozkurede U, Kala R, Johnson C, Shen Z, Miller RA, Garcia GG. Cap-independent mRNA translation is upregulated in long-lived endocrine mutant mice. J Mol Endocrinol 2019; 63:123-138. [PMID: 31357177 PMCID: PMC6691957 DOI: 10.1530/jme-19-0021] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 07/01/2019] [Indexed: 12/29/2022]
Abstract
It has been hypothesized that transcriptional changes associated with lower mTORC1 activity in mice with reduced levels of growth hormone and insulin-like growth factor 1 are responsible for the longer healthy lifespan of these mutant mice. Cell lines and tissues from these mice show alterations in the levels of many proteins that cannot be explained by corresponding changes in mRNAs. Such post-transcriptional modulation may be the result of preferential mRNA translation by the cap-independent translation of mRNA bearing the N6-methyl-adenosine (m6A) modification. The long-lived endocrine mutants - Snell dwarf, growth hormone receptor deletion and pregnancy-associated plasma protein-A knockout - all show increases in the N6-adenosine-methyltransferases (METTL3/14) that catalyze 6-methylation of adenosine (m6A) in the 5' UTR region of select mRNAs. In addition, these mice have elevated levels of YTH domain-containing protein 1 (YTHDF1), which recognizes m6A and promotes translation by a cap-independent mechanism. Consistently, multiple proteins that can be translated by the cap-independent mechanism are found to increase in these mice, including DNA repair and mitochondrial stress response proteins, without changes in corresponding mRNA levels. Lastly, a drug that augments cap-independent translation by inhibition of cap-dependent pathways (4EGI-1) was found to elevate levels of the same set of proteins and able to render cells resistant to several forms of in vitro stress. Augmented translation by cap-independent pathways facilitated by m6A modifications may contribute to the stress resistance and increased healthy longevity of mice with diminished GH and IGF-1 signals.
Collapse
Affiliation(s)
- Ulas Ozkurede
- Department of Pathology, Ann Arbor, Michigan 48109, USA
| | - Rishabh Kala
- Department of Pathology, Ann Arbor, Michigan 48109, USA
| | - Cameron Johnson
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan College of Literature, Science and the Arts, Ann Arbor, Michigan 48109, USA
| | - Ziqian Shen
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan College of Literature, Science and the Arts, Ann Arbor, Michigan 48109, USA
| | - Richard A. Miller
- Department of Pathology, Ann Arbor, Michigan 48109, USA
- University of Michigan Geriatrics Center, Ann Arbor, Michigan 48109, USA
| | | |
Collapse
|
14
|
Identification of protein kinase inhibitors to reprogram breast cancer cells. Cell Death Dis 2018; 9:915. [PMID: 30206213 PMCID: PMC6133942 DOI: 10.1038/s41419-018-1002-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 05/09/2018] [Accepted: 05/11/2018] [Indexed: 12/22/2022]
Abstract
Direct reversion of cancers into normal-like tissues is an ideal strategy for cancer treatment. Recent reports have showed that defined transcription factors can induce reprogramming of cancer cells into pluripotent stem cells, supporting this notion. Here, we have developed a reprogramming method that uses a conceptually unique strategy for breast cancer cell treatment. We have screened a kinase inhibitor library and found that Rho-associated protein kinase (ROCK) and mammalian target of rapamycin (mTOR) kinase inhibitors can substitute for all transcription factors to be sufficient to reprogram breast cancer cells into progenitor cells. Furthermore, ROCK–mTOR inhibitors could reprogram breast cancer cells to another terminal lineage-adipogenic cells. Genome-wide transcriptional analysis shows that the induced fat-like cells have a profile different from breast cancer cells and similar to that of normal adipocytes. In vitro and in vivo tumorigenesis assays have shown that induced fat-like cells lose proliferation and tumorigenicity. Moreover, reprogramming treatment with ROCK–mTOR inhibitors prevents breast cancer local recurrence in mice. Currently, ROCK–mTOR inhibitors are already used as antitumor drugs in patients, thus, this reprogramming strategy has significant potential to move rapidly toward clinical trials for breast cancer treatment.
Collapse
|
15
|
Zhou X, Sen I, Lin XX, Riedel CG. Regulation of Age-related Decline by Transcription Factors and Their Crosstalk with the Epigenome. Curr Genomics 2018; 19:464-482. [PMID: 30258277 PMCID: PMC6128382 DOI: 10.2174/1389202919666180503125850] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 04/20/2018] [Accepted: 04/30/2018] [Indexed: 12/16/2022] Open
Abstract
Aging is a complex phenomenon, where damage accumulation, increasing deregulation of biological pathways, and loss of cellular homeostasis lead to the decline of organismal functions over time. Interestingly, aging is not entirely a stochastic process and progressing at a constant rate, but it is subject to extensive regulation, in the hands of an elaborate and highly interconnected signaling network. This network can integrate a variety of aging-regulatory stimuli, i.e. fertility, nutrient availability, or diverse stresses, and relay them via signaling cascades into gene regulatory events - mostly of genes that confer stress resistance and thus help protect from damage accumulation and homeostasis loss. Transcription factors have long been perceived as the pivotal nodes in this network. Yet, it is well known that the epigenome substantially influences eukaryotic gene regulation, too. A growing body of work has recently underscored the importance of the epigenome also during aging, where it not only undergoes drastic age-dependent changes but also actively influences the aging process. In this review, we introduce the major signaling pathways that regulate age-related decline and discuss the synergy between transcriptional regulation and the epigenetic landscape.
Collapse
Affiliation(s)
| | | | | | - Christian G. Riedel
- Address correspondence to this author at the Integrated Cardio Metabolic Centre (ICMC), Department of Biosciences and Nutrition, Karolinska Institutet, Blickagången 6, Novum, 7 floor Huddinge, Stockholm 14157, Sweden; Tel: +46-736707008; E-mail:
| |
Collapse
|
16
|
Yuan J, Zhang F, Hallahan D, Zhang Z, He L, Wu LG, You M, Yang Q. Reprogramming glioblastoma multiforme cells into neurons by protein kinase inhibitors. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:181. [PMID: 30071868 PMCID: PMC6090992 DOI: 10.1186/s13046-018-0857-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 07/19/2018] [Indexed: 02/08/2023]
Abstract
Background Reprogramming of cancers into normal-like tissues is an innovative strategy for cancer treatment. Recent reports demonstrate that defined factors can reprogram cancer cells into pluripotent stem cells. Glioblastoma multiforme (GBM) is the most common and aggressive malignant brain tumor in humans. Despite multimodal therapy, the outcome for patients with GBM is still poor. Therefore, developing novel therapeutic strategy is a critical requirement. Methods We have developed a novel reprogramming method that uses a conceptually unique strategy for GBM treatment. We screened a kinase inhibitor library to find which candidate inhibitors under reprogramming condition can reprogram GBM cells into neurons. The induced neurons are identified whether functional and loss of tumorigenicity. Results We have found that mTOR and ROCK kinase inhibitors are sufficient to reprogram GBM cells into neural-like cells and “normal” neurons. The induced neurons expressed neuron-specific proteins, generated action potentials and neurotransmitter receptor-mediated currents. Genome-wide transcriptional analysis showed that the induced neurons had a profile different from GBM cells and were similar to that of control neurons induced by established methods. In vitro and in vivo tumorigenesis assays showed that induced neurons lost their proliferation ability and tumorigenicity. Moreover, reprogramming treatment with ROCK-mTOR inhibitors prevented GBM local recurrence in mice. Conclusion This study indicates that ROCK and mTOR inhibitors-based reprogramming treatment prevents GBM local recurrence. Currently ROCK-mTOR inhibitors are used as anti-tumor drugs in patients, so this reprogramming strategy has significant potential to move rapidly toward clinical trials. Electronic supplementary material The online version of this article (10.1186/s13046-018-0857-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jie Yuan
- Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, St. Louis, MO, 63108, USA.,Medical Center of Stomatology, the First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.,School of Stomatology, Jinan University, Guangzhou, 510630, China
| | - Fan Zhang
- Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, St. Louis, MO, 63108, USA
| | - Dennis Hallahan
- Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, St. Louis, MO, 63108, USA
| | - Zhen Zhang
- Synaptic Transmission Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, 20892, USA
| | - Liming He
- Synaptic Transmission Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, 20892, USA
| | - Ling-Gang Wu
- Synaptic Transmission Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, 20892, USA
| | - Meng You
- Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, St. Louis, MO, 63108, USA
| | - Qin Yang
- Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, St. Louis, MO, 63108, USA.
| |
Collapse
|
17
|
Jadiya P, Mir SS, Nazir A. Osmotic stress induced toxicity exacerbates Parkinson's associated effects via dysregulation of autophagy in transgenic C. elegans model. Cell Signal 2018; 45:71-80. [PMID: 29410282 DOI: 10.1016/j.cellsig.2018.01.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/30/2018] [Accepted: 01/31/2018] [Indexed: 02/07/2023]
Abstract
The accumulation of aggregate-prone proteins is a major representative of many neurological disorders, including Parkinson's disease (PD) wherein the cellular clearance mechanisms, such as the ubiquitin-proteasome and autophagy pathways are impaired. PD, known to be associated with multiple genetic and environmental factors, is characterized by the aggregation of α-synuclein protein and loss of dopaminergic neurons in midbrain. This disease is also associated with other cardiovascular ailments. Herein, we report our findings from studies on the effect of hyper and hypo-osmotic induced toxicity representing hyper and hypotensive condition as an extrinsic epigenetic factor towards modulation of Parkinsonism, using a genetic model Caenorhabditis elegans (C. elegans). Our studies showed that osmotic toxicity had an adverse effect on α-synuclein aggregation, autophagic puncta, lipid content and oxidative stress. Further, we figure that reduced autophagic activity may cause the inefficient clearance of α-synuclein aggregates in osmotic stress toxicity, thereby promoting α-synuclein deposition. Pharmacological induction of autophagy by spermidine proved to be a useful mechanism for protecting cells against the toxic effects of these proteins in such stress conditions. Our studies provide evidence that autophagy is required for the removal of aggregated proteins in these conditions. Studying specific autophagy pathways, we observe that the osmotic stress induced toxicity was largely associated with atg-7 and lgg-1 dependent autophagy pathway, brought together by involvement of mTOR pathway. This represents a unifying pathway to disease in hyper- and hypo-osmotic conditions within PD model of C. elegans.
Collapse
Affiliation(s)
- Pooja Jadiya
- Laboratory of Functional Genomics and Molecular Toxicology, Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226 031, UP, India
| | - Snober S Mir
- Department of Bioengineering, Integral University, Lucknow 226 026, India
| | - Aamir Nazir
- Laboratory of Functional Genomics and Molecular Toxicology, Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226 031, UP, India.
| |
Collapse
|
18
|
Das R, Melo JA, Thondamal M, Morton EA, Cornwell AB, Crick B, Kim JH, Swartz EW, Lamitina T, Douglas PM, Samuelson AV. The homeodomain-interacting protein kinase HPK-1 preserves protein homeostasis and longevity through master regulatory control of the HSF-1 chaperone network and TORC1-restricted autophagy in Caenorhabditis elegans. PLoS Genet 2017; 13:e1007038. [PMID: 29036198 PMCID: PMC5658188 DOI: 10.1371/journal.pgen.1007038] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 10/26/2017] [Accepted: 09/20/2017] [Indexed: 12/11/2022] Open
Abstract
An extensive proteostatic network comprised of molecular chaperones and protein clearance mechanisms functions collectively to preserve the integrity and resiliency of the proteome. The efficacy of this network deteriorates during aging, coinciding with many clinical manifestations, including protein aggregation diseases of the nervous system. A decline in proteostasis can be delayed through the activation of cytoprotective transcriptional responses, which are sensitive to environmental stress and internal metabolic and physiological cues. The homeodomain-interacting protein kinase (hipk) family members are conserved transcriptional co-factors that have been implicated in both genotoxic and metabolic stress responses from yeast to mammals. We demonstrate that constitutive expression of the sole Caenorhabditis elegans Hipk homolog, hpk-1, is sufficient to delay aging, preserve proteostasis, and promote stress resistance, while loss of hpk-1 is deleterious to these phenotypes. We show that HPK-1 preserves proteostasis and extends longevity through distinct but complementary genetic pathways defined by the heat shock transcription factor (HSF-1), and the target of rapamycin complex 1 (TORC1). We demonstrate that HPK-1 antagonizes sumoylation of HSF-1, a post-translational modification associated with reduced transcriptional activity in mammals. We show that inhibition of sumoylation by RNAi enhances HSF-1-dependent transcriptional induction of chaperones in response to heat shock. We find that hpk-1 is required for HSF-1 to induce molecular chaperones after thermal stress and enhances hormetic extension of longevity. We also show that HPK-1 is required in conjunction with HSF-1 for maintenance of proteostasis in the absence of thermal stress, protecting against the formation of polyglutamine (Q35::YFP) protein aggregates and associated locomotory toxicity. These functions of HPK-1/HSF-1 undergo rapid down-regulation once animals reach reproductive maturity. We show that HPK-1 fortifies proteostasis and extends longevity by an additional independent mechanism: induction of autophagy. HPK-1 is necessary for induction of autophagosome formation and autophagy gene expression in response to dietary restriction (DR) or inactivation of TORC1. The autophagy-stimulating transcription factors pha-4/FoxA and mxl-2/Mlx, but not hlh-30/TFEB or the nuclear hormone receptor nhr-62, are necessary for extended longevity resulting from HPK-1 overexpression. HPK-1 expression is itself induced by transcriptional mechanisms after nutritional stress, and post-transcriptional mechanisms in response to thermal stress. Collectively our results position HPK-1 at a central regulatory node upstream of the greater proteostatic network, acting at the transcriptional level by promoting protein folding via chaperone expression, and protein turnover via expression of autophagy genes. HPK-1 therefore provides a promising intervention point for pharmacological agents targeting the protein homeostasis system as a means of preserving robust longevity. Aging is the gradual and progressive decline of vitality. A hallmark of aging is the decay of protective mechanisms that normally preserve the robustness and resiliency of cells and tissues. Proteostasis is the term that applies specifically to those mechanisms that promote stability of the proteome, the collection of polypeptides that cells produce, by a combination of chaperone-assisted folding and degradation of misfolded or extraneous proteins. We have identified hpk-1 (encoding a homeodomain-interacting protein kinase) in the nematode C. elegans as an important transcriptional regulatory component of the proteostasis machinery. HPK-1 promotes proteostasis by linking two distinct mechanisms: first by stimulating chaperone gene expression via the heat shock transcription factor (HSF-1), and second by stimulating autophagy gene expression in opposition to the target of rapamycin (TOR) kinase signaling pathway. HPK-1 therefore provides an attractive target for interventions to preserve physiological resiliency during aging by preserving the overall health of the proteome.
Collapse
Affiliation(s)
- Ritika Das
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, New York, United States of America
- Department of Biology, University of Rochester, Rochester, New York, United States of America
| | - Justine A. Melo
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Manjunatha Thondamal
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Elizabeth A. Morton
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Adam B. Cornwell
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Beresford Crick
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Joung Heon Kim
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Elliot W. Swartz
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Todd Lamitina
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Peter M. Douglas
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Andrew V. Samuelson
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, New York, United States of America
- * E-mail:
| |
Collapse
|
19
|
Hughes PW. Between semelparity and iteroparity: Empirical evidence for a continuum of modes of parity. Ecol Evol 2017; 7:8232-8261. [PMID: 29075446 PMCID: PMC5648687 DOI: 10.1002/ece3.3341] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 07/24/2017] [Indexed: 01/19/2023] Open
Abstract
The number of times an organism reproduces (i.e., its mode of parity) is a fundamental life-history character, and evolutionary and ecological models that compare the relative fitnesses of different modes of parity are common in life-history theory and theoretical biology. Despite the success of mathematical models designed to compare intrinsic rates of increase (i.e., density-independent growth rates) between annual-semelparous and perennial-iteroparous reproductive schedules, there is widespread evidence that variation in reproductive allocation among semelparous and iteroparous organisms alike is continuous. This study reviews the ecological and molecular evidence for the continuity and plasticity of modes of parity-that is, the idea that annual-semelparous and perennial-iteroparous life histories are better understood as endpoints along a continuum of possible strategies. I conclude that parity should be understood as a continuum of different modes of parity, which differ by the degree to which they disperse or concentrate reproductive effort in time. I further argue that there are three main implications of this conclusion: (1) that seasonality should not be conflated with parity; (2) that mathematical models purporting to explain the general evolution of semelparous life histories from iteroparous ones (or vice versa) should not assume that organisms can only display either an annual-semelparous life history or a perennial-iteroparous one; and (3) that evolutionary ecologists should base explanations of how different life-history strategies evolve on the physiological or molecular basis of traits underlying different modes of parity.
Collapse
Affiliation(s)
- Patrick William Hughes
- Department of Plant Breeding and GeneticsMax Planck Institute for Plant Breeding ResearchKölnGermany
| |
Collapse
|
20
|
Ding AJ, Zheng SQ, Huang XB, Xing TK, Wu GS, Sun HY, Qi SH, Luo HR. Current Perspective in the Discovery of Anti-aging Agents from Natural Products. NATURAL PRODUCTS AND BIOPROSPECTING 2017; 7:335-404. [PMID: 28567542 PMCID: PMC5655361 DOI: 10.1007/s13659-017-0135-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 05/16/2017] [Indexed: 05/18/2023]
Abstract
Aging is a process characterized by accumulating degenerative damages, resulting in the death of an organism ultimately. The main goal of aging research is to develop therapies that delay age-related diseases in human. Since signaling pathways in aging of Caenorhabditis elegans (C. elegans), fruit flies and mice are evolutionarily conserved, compounds extending lifespan of them by intervening pathways of aging may be useful in treating age-related diseases in human. Natural products have special resource advantage and with few side effect. Recently, many compounds or extracts from natural products slowing aging and extending lifespan have been reported. Here we summarized these compounds or extracts and their mechanisms in increasing longevity of C. elegans or other species, and the prospect in developing anti-aging medicine from natural products.
Collapse
Affiliation(s)
- Ai-Jun Ding
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Shan-Qing Zheng
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Xiao-Bing Huang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Ti-Kun Xing
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Gui-Sheng Wu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
- Key Laboratory for Aging and Regenerative Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Hua-Ying Sun
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Shu-Hua Qi
- Guangdong Key Laboratory of Marine Material Medical, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301, Guangdong, China
| | - Huai-Rong Luo
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China.
- Key Laboratory for Aging and Regenerative Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming Institute of Botany, Chinese Academy of Sciences, 134 Lanhei Road, Kunming, 650201, Yunnan, China.
| |
Collapse
|
21
|
Somatic growth, aging, and longevity. NPJ Aging Mech Dis 2017; 3:14. [PMID: 28970944 PMCID: PMC5622030 DOI: 10.1038/s41514-017-0014-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 09/06/2017] [Accepted: 09/13/2017] [Indexed: 02/01/2023] Open
Abstract
Although larger species of animals typically live longer than smaller species, the relationship of body size to longevity within a species is generally opposite. The longevity advantage of smaller individuals can be considerable and is best documented in laboratory mice and in domestic dogs. Importantly, it appears to apply broadly, including humans. It is not known whether theses associations represent causal links between various developmental and physiological mechanisms affecting growth and/or aging. However, variations in growth hormone (GH) signaling are likely involved because GH is a key stimulator of somatic growth, and apparently also exerts various “pro-aging” effects. Mechanisms linking GH, somatic growth, adult body size, aging, and lifespan likely involve target of rapamycin (TOR), particularly one of its signaling complexes, mTORC1, as well as various adjustments in mitochondrial function, energy metabolism, thermogenesis, inflammation, and insulin signaling. Somatic growth, aging, and longevity are also influenced by a variety of hormonal and nutritional signals, and much work will be needed to answer the question of why smaller individuals may be likely to live longer.
Collapse
|
22
|
Approaches for extending human healthspan: from antioxidants to healthspan pharmacology. Essays Biochem 2017; 61:389-399. [PMID: 28698312 DOI: 10.1042/ebc20160091] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/16/2017] [Accepted: 05/17/2017] [Indexed: 02/07/2023]
Abstract
Dramatic increases in human lifespan and declining population growth are monumental achievements but these same achievements have also led to many societies today ageing at a faster rate than ever before. Extending healthy lifespan (healthspan) is a key translational challenge in this context. Disease-centric approaches to manage population ageing risk are adding years to life without adding health to these years. The growing consensus that ageing is driven by a limited number of interconnected processes suggests an alternative approach. Instead of viewing each age-dependent disease as the result of an independent chain of events, this approach recognizes that most age-dependent diseases depend on and are driven by a limited set of ageing processes. While the relative importance of each of these processes and the best intervention strategies targeting them are subjects of debate, there is increasing interest in providing preventative intervention options to healthy individuals even before overt age-dependent diseases manifest. Elevated oxidative damage is involved in the pathophysiology of most age-dependent diseases and markers of oxidative damage often increase with age in many organisms. However, correlation is not causation and, sadly, many intervention trials of supposed antioxidants have failed to extend healthspan and to prevent diseases. This does not, however, mean that reactive species (RS) and redox signalling are unimportant. Ultimately, the most effective antioxidants may not turn out to be the best geroprotective drugs, but effective geroprotective interventions might well turn out to also have excellent, if probably indirect, antioxidant efficacy.
Collapse
|
23
|
Heiss C, Spyridopoulos I, Haendeler J. Interventions to slow cardiovascular aging: Dietary restriction, drugs and novel molecules. Exp Gerontol 2017; 109:108-118. [PMID: 28658611 DOI: 10.1016/j.exger.2017.06.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 06/16/2017] [Accepted: 06/21/2017] [Indexed: 02/05/2023]
Abstract
Cardiovascular aging is a highly dynamic process. Despite the fact that cardiovascular function and structure change with age, they can still be modulated even in aged humans. The most prominent approaches to improve age-dependent vascular changes include dietary restriction and pharmacologic agents interacting with signaling pathways implicated in this context. These include inhibition of TOR, glycolysis, and GH/IGF-1, activation of sirtuins, and AMPK, as well as modulators of inflammation, epigenetic pathways, and telomeres. Promising nutritional approaches include Mediterranean diet and novel dietary bioactives including flavanols, anthocyanins, and lignins. Many plant bioactives improve cardiovascular parameters implied in vascular healthy aging including endothelial function, arterial stiffness, blood pressure, cholesterol, and glycemic control. However, the mechanism of action of most bioactives is not established and it remains to be elucidated whether they act as dietary restriction mimetics or via other modes of action. Even more importantly, whether these interventions can slow or even reverses components of cardiovascular aging itself and can increase healthspan or longevity in humans needs to be determined.
Collapse
Affiliation(s)
- Christian Heiss
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany.
| | - Ioakim Spyridopoulos
- Institute of Genetic Medicine, Medical Faculty, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK.
| | - Judith Haendeler
- Central Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty, University of Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany; IUF-Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, Germany.
| |
Collapse
|
24
|
Lushchak O, Strilbytska O, Piskovatska V, Storey KB, Koliada A, Vaiserman A. The role of the TOR pathway in mediating the link between nutrition and longevity. Mech Ageing Dev 2017; 164:127-138. [DOI: 10.1016/j.mad.2017.03.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 02/23/2017] [Accepted: 03/13/2017] [Indexed: 01/13/2023]
|
25
|
Moderate lifelong overexpression of tuberous sclerosis complex 1 (TSC1) improves health and survival in mice. Sci Rep 2017; 7:834. [PMID: 28400571 PMCID: PMC5429778 DOI: 10.1038/s41598-017-00970-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 03/10/2017] [Indexed: 12/16/2022] Open
Abstract
The tuberous sclerosis complex 1/2 (TSC1/2) is an endogenous regulator of the mechanistic target of rapamycin (mTOR). While mTOR has been shown to play an important role in health and aging, the role of TSC1/2 in aging has not been fully investigated. In the current study, a constitutive TSC1 transgenic (Tsc1tg) mouse model was generated and characterized. mTORC1 signaling was reduced in majority of the tissues, except the brain. In contrast, mTORC2 signaling was enhanced in Tsc1tg mice. Tsc1tg mice are more tolerant to exhaustive exercises and less susceptible to isoproterenol-induced cardiac hypertrophy at both young and advanced ages. Tsc1tg mice have less fibrosis and inflammation in aged as well as isoproterenol-challenged heart than age-matched wild type mice. The female Tsc1tg mice exhibit a higher fat to lean mass ratio at advanced ages than age-matched wild type mice. More importantly, the lifespan increased significantly in female Tsc1tg mice, but not in male Tsc1tg mice. Collectively, our data demonstrated that moderate increase of TSC1 expression can enhance overall health, particularly cardiovascular health, and improve survival in a gender-specific manner.
Collapse
|
26
|
Henne WM. Discovery and Roles of ER-Endolysosomal Contact Sites in Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 997:135-147. [PMID: 28815527 DOI: 10.1007/978-981-10-4567-7_10] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Inter-organelle membrane contact sites (MCSs) serve as unique microenvironments for the sensing and exchange of cellular metabolites and lipids. Though poorly defined, ER-endolysosomal contact sites are quickly becoming recognized as centers for inter-organelle lipid exchange and metabolic decision-making. Here, we review the discovery and current state of knowledge of ER-endolysosomal MCSs with particular focus on the molecular players that establish and/or utilize these contact sites in metabolism. We also discuss associations of ER-endolysosomal MCS-associated proteins in human disease, as well as the therapeutic promise these contact sites hold in modulating cellular physiology.
Collapse
Affiliation(s)
- William Mike Henne
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX, 75013, USA.
| |
Collapse
|
27
|
Ejzykowicz DE, Locken KM, Ruiz FJ, Manandhar SP, Olson DK, Gharakhanian E. Hygromycin B hypersensitive (hhy) mutants implicate an intact trans-Golgi and late endosome interface in efficient Tor1 vacuolar localization and TORC1 function. Curr Genet 2016; 63:531-551. [PMID: 27812735 DOI: 10.1007/s00294-016-0660-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 10/20/2016] [Accepted: 10/22/2016] [Indexed: 12/18/2022]
Abstract
Saccharomyces cerevisiae vacuoles are functionally analogous to mammalian lysosomes. Both also serve as physical platforms for Tor Complex 1 (TORC1) signal transduction, the master regulator of cellular growth and proliferation. Hygromycin B is a eukaryotic translation inhibitor. We recently reported on hygromycin B hypersensitive (hhy) mutants that fail to grow at subtranslation inhibitory concentrations of the drug and exhibit vacuolar defects (Banuelos et al. in Curr Genet 56:121-137, 2010). Here, we show that hhy phenotype is not due to increased sensitivity to translation inhibition and establish a super HHY (s-HHY) subgroup of genes comprised of ARF1, CHC1, DRS2, SAC1, VPS1, VPS34, VPS45, VPS52, and VPS54 that function exclusively or inclusively at trans-Golgi and late endosome interface. Live cell imaging of s-hhy mutants revealed that hygromycin B treatment disrupts vacuolar morphology and the localization of late endosome marker Pep12, but not that of late endosome-independent vacuolar SNARE Vam3. This, along with normal post-late endosome trafficking of the vital dye FM4-64, establishes that severe hypersensitivity to hygromycin B correlates specifically with compromised trans-Golgi and late endosome interface. We also show that Tor1p vacuolar localization and TORC1 anabolic functions, including growth promotion and phosphorylation of its direct substrate Sch9, are compromised in s-hhy mutants. Thus, an intact trans-Golgi and late endosome interface is a requisite for efficient Tor1 vacuolar localization and TORC1 function.
Collapse
Affiliation(s)
- Daniele E Ejzykowicz
- Department of Biological Sciences, California State University Long Beach, 1250 Bellflower Blvd, Long Beach, CA, 90840, USA
| | - Kristopher M Locken
- Department of Biological Sciences, California State University Long Beach, 1250 Bellflower Blvd, Long Beach, CA, 90840, USA
| | - Fiona J Ruiz
- Department of Biological Sciences, California State University Long Beach, 1250 Bellflower Blvd, Long Beach, CA, 90840, USA.,Department of Radiation Oncology, Cancer Biology Division, Washington University School of Medicine, Saint Louis, MO, 63108, USA
| | - Surya P Manandhar
- Department of Biological Sciences, California State University Long Beach, 1250 Bellflower Blvd, Long Beach, CA, 90840, USA
| | - Daniel K Olson
- Department of Biological Sciences, California State University Long Beach, 1250 Bellflower Blvd, Long Beach, CA, 90840, USA.,Inouye Center for Microbial Oceanography, Research and Education, University of Hawaii, Manoa, Honolulu, HI, 96822, USA
| | - Editte Gharakhanian
- Department of Biological Sciences, California State University Long Beach, 1250 Bellflower Blvd, Long Beach, CA, 90840, USA.
| |
Collapse
|
28
|
|
29
|
Pazoki-Toroudi H, Amani H, Ajami M, Nabavi SF, Braidy N, Kasi PD, Nabavi SM. Targeting mTOR signaling by polyphenols: A new therapeutic target for ageing. Ageing Res Rev 2016; 31:55-66. [PMID: 27453478 DOI: 10.1016/j.arr.2016.07.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 06/19/2016] [Accepted: 07/15/2016] [Indexed: 12/19/2022]
Abstract
Current ageing research is aimed not only at the promotion of longevity, but also at improving health span through the discovery and development of new therapeutic strategies by investigating molecular and cellular pathways involved in cellular senescence. Understanding the mechanism of action of polyphenolic compounds targeting mTOR (mechanistic target of rapamycin) and related pathways opens up new directions to revolutionize ways to slow down the onset and development of age-dependent degeneration. Herein, we will discuss the mechanisms by which polyphenols can delay the molecular pathogenesis of ageing via manipulation or more specifically inhibition of mTOR-signaling pathways. We will also discuss the implications of polyphenols in targeting mTOR and its related pathways on health life span extension and longevity..
Collapse
|
30
|
Abstract
Aging is an inevitable outcome of life, characterized by progressive decline in tissue and organ function and increased risk of mortality. Accumulating evidence links aging to genetic and epigenetic alterations. Given the reversible nature of epigenetic mechanisms, these pathways provide promising avenues for therapeutics against age-related decline and disease. In this review, we provide a comprehensive overview of epigenetic studies from invertebrate organisms, vertebrate models, tissues, and in vitro systems. We establish links between common operative aging pathways and hallmark chromatin signatures that can be used to identify "druggable" targets to counter human aging and age-related disease.
Collapse
Affiliation(s)
- Payel Sen
- Epigenetics Program, Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19130, USA
| | - Parisha P Shah
- Epigenetics Program, Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19130, USA
| | - Raffaella Nativio
- Epigenetics Program, Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19130, USA
| | - Shelley L Berger
- Epigenetics Program, Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19130, USA.
| |
Collapse
|
31
|
Abstract
Dietary restriction (DR), a moderate reduction in food intake, improves health during aging and extends life span across multiple species. Specific nutrients, rather than overall calories, mediate the effects of DR, with protein and specific amino acids (AAs) playing a key role. Modulations of single dietary AAs affect traits including growth, reproduction, physiology, health, and longevity in animals. Epidemiological data in humans also link the quality and quantity of dietary proteins to long-term health. Intricate nutrient-sensing pathways fine tune the metabolic responses to dietary AAs in a highly conserved manner. In turn, these metabolic responses can affect the onset of insulin resistance, obesity, neurodegenerative disease, and other age-related diseases. In this review we discuss how AA requirements are shaped and how ingested AAs regulate a spectrum of homeostatic processes. Finally, we highlight the resulting opportunity to develop nutritional strategies to improve human health during aging.
Collapse
Affiliation(s)
- George A Soultoukis
- Max Planck Institute for Biology of Ageing, Department of Biological Mechanisms of Ageing, Cologne 50931, Germany; ,
| | - Linda Partridge
- Max Planck Institute for Biology of Ageing, Department of Biological Mechanisms of Ageing, Cologne 50931, Germany; , .,Institute of Healthy Ageing and Department of Genetics, Evolution, and Environment, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
32
|
Wang Y, Yamada E, Zong H, Pessin JE. Fyn Activation of mTORC1 Stimulates the IRE1α-JNK Pathway, Leading to Cell Death. J Biol Chem 2015; 290:24772-83. [PMID: 26306048 DOI: 10.1074/jbc.m115.687020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Indexed: 12/25/2022] Open
Abstract
We previously reported that the skeletal muscle-specific overexpression of Fyn in mice resulted in a severe muscle wasting phenotype despite the activation of mTORC1 signaling. To investigate the bases for the loss of muscle fiber mass, we examined the relationship between Fyn activation of mTORC1, JNK, and endoplasmic reticulum stress. Overexpression of Fyn in skeletal muscle in vivo and in HEK293T cells in culture resulted in the activation of IRE1α and JNK, leading to increased cell death. Fyn synergized with the general endoplasmic reticulum stress inducer thapsigargin, resulting in the activation of IRE1α and further accelerated cell death. Moreover, inhibition of mTORC1 with rapamycin suppressed IRE1α activation and JNK phosphorylation, resulting in protecting cells against Fyn- and thapsigargin-induced cell death. Moreover, rapamycin treatment in vivo reduced the skeletal muscle IRE1α activation in the Fyn-overexpressing transgenic mice. Together, these data demonstrate the presence of a Fyn-induced endoplasmic reticulum stress that occurred, at least in part, through the activation of mTORC1, as well as subsequent activation of the IRE1α-JNK pathway driving cell death.
Collapse
Affiliation(s)
- Yichen Wang
- From the Departments of Molecular Pharmacology and
| | - Eijiro Yamada
- the Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan
| | - Haihong Zong
- Medicine, Albert Einstein College of Medicine, Bronx, New York 10461 and
| | - Jeffrey E Pessin
- From the Departments of Molecular Pharmacology and Medicine, Albert Einstein College of Medicine, Bronx, New York 10461 and
| |
Collapse
|
33
|
Chang GR, Chiu YS, Wu YY, Lin YC, Hou PH, Mao FC. Rapamycin impairs HPD-induced beneficial effects on glucose homeostasis. Br J Pharmacol 2015; 172:3793-804. [PMID: 25884889 PMCID: PMC4523336 DOI: 10.1111/bph.13168] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 03/08/2015] [Accepted: 04/07/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Rapamycin, which is used clinically to treat graft rejection, has also been proposed to have an effect on metabolic syndrome; however, very little information is available on its effects in lean animals/humans. The purpose of this study was to characterize further the effects of the continuous use of rapamycin on glucose homeostasis in lean C57BL6/J mice. EXPERIMENTAL APPROACH Mice were fed a high-protein diet (HPD) for 12 weeks to develop a lean model and then were treated daily with rapamycin for 5 weeks while remaining on a HPD. Metabolic parameters, endocrine profiles, glucose tolerance tests, insulin sensitivity index, the expression of the glucose transporter GLUT4 and chromium distribution were measured in vivo. KEY RESULTS Lower body weight gain as well as a decreased caloric intake, fat pads, fatty liver scores, adipocyte size and glucose tolerance test values were observed in HPD-fed mice compared with mice fed a high-fat or standard diet. Despite these beneficial effects, rapamycin-treated lean mice showed greater glucose intolerance, reduced insulin sensitivity, lower muscle GLUT4 expression and changes in chromium levels in tissues even with high insulin levels. CONCLUSION AND IMPLICATIONS Our findings demonstrate that continuous rapamycin administration may lead to the development of diabetes syndrome, as it was found to induce hyperglycaemia and glucose intolerance in a lean animal model.
Collapse
Affiliation(s)
- Geng-Ruei Chang
- Department of Veterinary Medicine, National Chung Hsing UniversityTaichung, Taiwan
- Division of Residual Control, Agricultural Chemicals and Toxic Substance Research Institute, Council of AgricultureTaichung, Taiwan
| | - Yi-Shin Chiu
- Department of Veterinary Medicine, National Chung Hsing UniversityTaichung, Taiwan
| | - Ying-Ying Wu
- Department of Veterinary Medicine, National Chung Hsing UniversityTaichung, Taiwan
| | - Yu-Chi Lin
- Department of Veterinary Medicine, National Chung Hsing UniversityTaichung, Taiwan
| | - Po-Hsun Hou
- Department of Veterinary Medicine, National Chung Hsing UniversityTaichung, Taiwan
- Department of Psychiatry, Taichung Veterans General HospitalTaichung, Taiwan
| | - Frank Chiahung Mao
- Department of Veterinary Medicine, National Chung Hsing UniversityTaichung, Taiwan
| |
Collapse
|
34
|
Abstract
SIGNIFICANCE The molecular mechanism of aging is still vigorously debated, although a general consensus exists that mitochondria are significantly involved in this process. However, the previously postulated role of mitochondrial-derived reactive oxygen species (ROS) as the damaging agents inducing functional loss in aging has fallen out of favor in the recent past. In this review, we critically examine the role of ROS in aging in the light of recent advances on the relationship between mitochondrial structure and function. RECENT ADVANCES The functional mitochondrial respiratory chain is now recognized as a reflection of the dynamic association of respiratory complexes in the form of supercomplexes (SCs). Besides providing kinetic advantage (channeling), SCs control ROS generation by the respiratory chain, thus providing a means to regulate ROS levels in the cell. Depending on their concentration, these ROS are either physiological signals essential for the life of the cell or toxic species that damage cell structure and functions. CRITICAL ISSUES We propose that under physiological conditions the dynamic nature of SCs reversibly controls the generation of ROS as signals involved in mitochondrial-nuclear communication. During aging, there is a progressive loss of control of ROS generation so that their production is irreversibly enhanced, inducing a vicious circle in which signaling is altered and structural damage takes place. FUTURE DIRECTIONS A better understanding on the forces affecting SC association would allow the manipulation of ROS generation, directing these species to their physiological signaling role.
Collapse
Affiliation(s)
- Maria Luisa Genova
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum-Università di Bologna , Bologna, Italy
| | - Giorgio Lenaz
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum-Università di Bologna , Bologna, Italy
| |
Collapse
|
35
|
Tower J. Mitochondrial maintenance failure in aging and role of sexual dimorphism. Arch Biochem Biophys 2015; 576:17-31. [PMID: 25447815 PMCID: PMC4409928 DOI: 10.1016/j.abb.2014.10.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 10/08/2014] [Accepted: 10/18/2014] [Indexed: 12/31/2022]
Abstract
Gene expression changes during aging are partly conserved across species, and suggest that oxidative stress, inflammation and proteotoxicity result from mitochondrial malfunction and abnormal mitochondrial-nuclear signaling. Mitochondrial maintenance failure may result from trade-offs between mitochondrial turnover versus growth and reproduction, sexual antagonistic pleiotropy and genetic conflicts resulting from uni-parental mitochondrial transmission, as well as mitochondrial and nuclear mutations and loss of epigenetic regulation. Aging phenotypes and interventions are often sex-specific, indicating that both male and female sexual differentiation promote mitochondrial failure and aging. Studies in mammals and invertebrates implicate autophagy, apoptosis, AKT, PARP, p53 and FOXO in mediating sex-specific differences in stress resistance and aging. The data support a model where the genes Sxl in Drosophila, sdc-2 in Caenorhabditis elegans, and Xist in mammals regulate mitochondrial maintenance across generations and in aging. Several interventions that increase life span cause a mitochondrial unfolded protein response (UPRmt), and UPRmt is also observed during normal aging, indicating hormesis. The UPRmt may increase life span by stimulating mitochondrial turnover through autophagy, and/or by inhibiting the production of hormones and toxic metabolites. The data suggest that metazoan life span interventions may act through a common hormesis mechanism involving liver UPRmt, mitochondrial maintenance and sexual differentiation.
Collapse
Affiliation(s)
- John Tower
- Molecular and Computational Biology Program, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089-2910, United States.
| |
Collapse
|
36
|
Schreiber KH, Ortiz D, Academia EC, Anies AC, Liao C, Kennedy BK. Rapamycin-mediated mTORC2 inhibition is determined by the relative expression of FK506-binding proteins. Aging Cell 2015; 14:265-73. [PMID: 25652038 PMCID: PMC4364838 DOI: 10.1111/acel.12313] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2014] [Indexed: 01/09/2023] Open
Abstract
The mechanism by which the drug rapamycin inhibits the mechanistic target of rapamycin (mTOR) is of intense interest because of its likely relevance in cancer biology, aging, and other age-related diseases. While rapamycin acutely and directly inhibits mTORC1, only chronic administration of rapamycin can inhibit mTORC2 in some, but not all, cell lines or tissues. The mechanism leading to cell specificity of mTORC2 inhibition by rapamycin is not understood and is especially important because many of the negative metabolic side effects of rapamycin, reported in mouse studies and human clinical trials, have been attributed recently to mTORC2 inhibition. Here, we identify the expression level of different FK506-binding proteins (FKBPs), primarily FKBP12 and FKBP51, as the key determinants for rapamycin-mediated inhibition of mTORC2. In support, enforced reduction of FKBP12 completely converts a cell line that is sensitive to mTORC2 inhibition to an insensitive cell line, and increased expression can enhance mTORC2 inhibition. Further reduction of FKBP12 in cell lines with already low FKBP12 levels completely blocks mTORC1 inhibition by rapamycin, indicating that relative FKBP12 levels are critical for both mTORC1 and mTORC2 inhibition, but at different levels. In contrast, reduction of FKBP51 renders cells more sensitive to mTORC2 inhibition. Our findings reveal that the expression of FKBP12 and FKBP51 is the rate limiting factor that determines the responsiveness of a cell line or tissue to rapamycin. These findings have implications for treating specific diseases, including neurodegeneration and cancer, as well as targeting aging in general.
Collapse
Affiliation(s)
| | - Denise Ortiz
- The Buck Institute for Research on Aging 8001 Redwood Blvd. Novato CA 94945 USA
| | | | - Arieanna C. Anies
- The Buck Institute for Research on Aging 8001 Redwood Blvd. Novato CA 94945 USA
| | - Chen‐Yu Liao
- The Buck Institute for Research on Aging 8001 Redwood Blvd. Novato CA 94945 USA
| | - Brian K. Kennedy
- The Buck Institute for Research on Aging 8001 Redwood Blvd. Novato CA 94945 USA
| |
Collapse
|
37
|
Dominick G, Berryman DE, List EO, Kopchick JJ, Li X, Miller RA, Garcia GG. Regulation of mTOR activity in Snell dwarf and GH receptor gene-disrupted mice. Endocrinology 2015; 156:565-75. [PMID: 25456069 PMCID: PMC4298324 DOI: 10.1210/en.2014-1690] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The involvement of mammalian target of rapamycin (mTOR) in lifespan control in invertebrates, calorie-restricted rodents, and extension of mouse lifespan by rapamycin have prompted speculation that diminished mTOR function may contribute to mammalian longevity in several settings. We show here that mTOR complex-1 (mTORC1) activity is indeed lower in liver, muscle, heart, and kidney tissue of Snell dwarf and global GH receptor (GHR) gene-disrupted mice (GHR-/-), consistent with previous studies. Surprisingly, activity of mTORC2 is higher in fasted Snell and GHR-/- than in littermate controls in all 4 tissues tested. Resupply of food enhanced mTORC1 activity in both controls and long-lived mutant mice but diminished mTORC2 activity only in the long-lived mice. Mice in which GHR has been disrupted only in the liver do not show extended lifespan and also fail to show the decline in mTORC1 and increase in mTORC2 seen in mice with global loss of GHR. The data suggest that the antiaging effects in the Snell dwarf and GHR-/- mice are accompanied by both a decline in mTORC1 in multiple organs and an increase in fasting levels of mTORC2. Neither the lifespan nor mTOR effects appear to be mediated by direct GH effects on liver or by the decline in plasma IGF-I, a shared trait in both global and liver-specific GHR-/- mice. Our data suggest that a more complex pattern of hormonal effects and intertissue interactions may be responsible for regulating both lifespan and mTORC2 function in these mouse models of delayed aging.
Collapse
Affiliation(s)
- Graham Dominick
- Department of Molecular, Cellular, and Developmental Biology (G.D.), University of Michigan College of Literature, Science and the Arts, Ann Arbor, Michigan 48109; Edison Biotechnology Institute (D.E.B., E.O.L., J.J.K.), Ohio University, Athens, Ohio 45701; Department of Pathology (X.L., R.A.M., G.G.G.), University of Michigan School of Medicine Ann Arbor, Michigan 48109; and University of Michigan Geriatrics Center (R.A.M.), Ann Arbor, Michigan 48109
| | | | | | | | | | | | | |
Collapse
|
38
|
He C, Tsuchiyama SK, Nguyen QT, Plyusnina EN, Terrill SR, Sahibzada S, Patel B, Faulkner AR, Shaposhnikov MV, Tian R, Tsuchiya M, Kaeberlein M, Moskalev AA, Kennedy BK, Polymenis M. Enhanced longevity by ibuprofen, conserved in multiple species, occurs in yeast through inhibition of tryptophan import. PLoS Genet 2014; 10:e1004860. [PMID: 25521617 PMCID: PMC4270464 DOI: 10.1371/journal.pgen.1004860] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 10/29/2014] [Indexed: 11/29/2022] Open
Abstract
The common non-steroidal anti-inflammatory drug ibuprofen has been associated with a reduced risk of some age-related pathologies. However, a general pro-longevity role for ibuprofen and its mechanistic basis remains unclear. Here we show that ibuprofen increased the lifespan of Saccharomyces cerevisiae, Caenorhabditis elegans and Drosophila melanogaster, indicative of conserved eukaryotic longevity effects. Studies in yeast indicate that ibuprofen destabilizes the Tat2p permease and inhibits tryptophan uptake. Loss of Tat2p increased replicative lifespan (RLS), but ibuprofen did not increase RLS when Tat2p was stabilized or in an already long-lived strain background impaired for aromatic amino acid uptake. Concomitant with lifespan extension, ibuprofen moderately reduced cell size at birth, leading to a delay in the G1 phase of the cell cycle. Similar changes in cell cycle progression were evident in a large dataset of replicatively long-lived yeast deletion strains. These results point to fundamental cell cycle signatures linked with longevity, implicate aromatic amino acid import in aging and identify a largely safe drug that extends lifespan across different kingdoms of life. Aging is the greatest risk factor for many diseases, which together account for the majority of global deaths and healthcare costs. Here we show that the common drug ibuprofen increases the lifespan of yeast, worms and flies, indicative of conserved longevity effects. In budding yeast, an excellent model of cellular longevity mechanisms, ibuprofen's pro-longevity action is independent of its known anti-inflammatory role. We show that the critical function of ibuprofen in longevity is to inhibit the uptake of aromatic amino acids, by destabilizing the high-affinity tryptophan permease. We further show that ibuprofen alters cell cycle progression. Mirroring the effects of ibuprofen, we found that most yeast long-lived mutants were also similarly affected in cell cycle progression. These findings identify a safe drug that extends the lifespan of divergent organisms and reveal fundamental cellular properties associated with longevity.
Collapse
Affiliation(s)
- Chong He
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Scott K. Tsuchiyama
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Quynh T. Nguyen
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, United States of America
| | - Ekaterina N. Plyusnina
- Institute of Biology of Komi Science Center of Ural Branch of RAS, Syktyvkar, Russia
- Syktyvkar State University, Syktyvkar, Russia
| | - Samuel R. Terrill
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, United States of America
| | - Sarah Sahibzada
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, United States of America
| | - Bhumil Patel
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Alena R. Faulkner
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Mikhail V. Shaposhnikov
- Institute of Biology of Komi Science Center of Ural Branch of RAS, Syktyvkar, Russia
- Syktyvkar State University, Syktyvkar, Russia
| | - Ruilin Tian
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Mitsuhiro Tsuchiya
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Matt Kaeberlein
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Alexey A. Moskalev
- Institute of Biology of Komi Science Center of Ural Branch of RAS, Syktyvkar, Russia
- Syktyvkar State University, Syktyvkar, Russia
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia
| | - Brian K. Kennedy
- Buck Institute for Research on Aging, Novato, California, United States of America
- * E-mail: (BKK); (MP)
| | - Michael Polymenis
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, United States of America
- * E-mail: (BKK); (MP)
| |
Collapse
|
39
|
Corella D, Ordovás JM. Aging and cardiovascular diseases: the role of gene-diet interactions. Ageing Res Rev 2014; 18:53-73. [PMID: 25159268 DOI: 10.1016/j.arr.2014.08.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 08/15/2014] [Accepted: 08/18/2014] [Indexed: 12/21/2022]
Abstract
In the study of longevity, increasing importance is being placed on the concept of healthy aging rather than considering the total number of years lived. Although the concept of healthy lifespan needs to be defined better, we know that cardiovascular diseases (CVDs) are the main age-related diseases. Thus, controlling risk factors will contribute to reducing their incidence, leading to healthy lifespan. CVDs are complex diseases influenced by numerous genetic and environmental factors. Numerous gene variants that are associated with a greater or lesser risk of the different types of CVD and of intermediate phenotypes (i.e., hypercholesterolemia, hypertension, diabetes) have been successfully identified. However, despite the close link between aging and CVD, studies analyzing the genes related to human longevity have not obtained consistent results and there has been little coincidence in the genes identified in both fields. The APOE gene stands out as an exception, given that it has been identified as being relevant in CVD and longevity. This review analyzes the genomic and epigenomic factors that may contribute to this, ranging from identifying longevity genes in model organisms to the importance of gene-diet interactions (outstanding among which is the case of the TCF7L2 gene).
Collapse
|
40
|
Colca JR, McDonald WG, Kletzien RF. Mitochondrial target of thiazolidinediones. Diabetes Obes Metab 2014; 16:1048-54. [PMID: 24774061 DOI: 10.1111/dom.12308] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 04/17/2014] [Accepted: 04/22/2014] [Indexed: 12/11/2022]
Abstract
Insulin-sensitizing thiazolidinediones exert a pleiotropic pharmacology with therapeutic potential in a number of disease states ranging from metabolic syndrome and diabetes to neurodegeneration and cancer. A growing understanding of their mechanism of action, working from the site of their binding in the mitochondrion, provides insight into the mechanism of action of the insulin sensitizers and the reasons for their pleiotropic pharmacology. This review helps to frame the direction of future work that should be helpful in setting a new direction for the discovery and development of new, more useful therapeutic agents for metabolic disease.
Collapse
Affiliation(s)
- J R Colca
- Metabolic Solutions Development Company, Kalamazoo, MI, USA
| | | | | |
Collapse
|
41
|
Hu J, Wei M, Mirzaei H, Madia F, Mirisola M, Amparo C, Chagoury S, Kennedy B, Longo VD. Tor-Sch9 deficiency activates catabolism of the ketone body-like acetic acid to promote trehalose accumulation and longevity. Aging Cell 2014; 13:457-67. [PMID: 24649827 PMCID: PMC4032597 DOI: 10.1111/acel.12202] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2013] [Indexed: 11/27/2022] Open
Abstract
In mammals, extended periods of fasting leads to the accumulation of blood ketone bodies including acetoacetate. Here we show that similar to the conversion of leucine to acetoacetate in fasting mammals, starvation conditions induced ketone body-like acetic acid generation from leucine in S. cerevisiae. Whereas wild-type and ras2Δ cells accumulated acetic acid, long-lived tor1Δ and sch9Δ mutants rapidly depleted it through a mitochondrial acetate CoA transferase-dependent mechanism, which was essential for lifespan extension. The sch9Δ-dependent utilization of acetic acid also required coenzyme Q biosynthetic genes and promoted the accumulation of intracellular trehalose. These results indicate that Tor-Sch9 deficiency extends longevity by switching cells to an alternative metabolic mode, in which acetic acid can be utilized for the storage of stress resistance carbon sources. These effects are reminiscent of those described for ketone bodies in fasting mammals and raise the possibility that the lifespan extension caused by Tor-S6K inhibition may also involve analogous metabolic changes in higher eukaryotes.
Collapse
Affiliation(s)
- Jia Hu
- Longevity Institute; Davis School of Gerontology; University of Southern California; Los Angeles CA 90089 USA
- Department of Biological Sciences; School of Dornsife College of Letters, Arts and Sciences; University of Southern California; Los Angeles CA 90089 USA
| | - Min Wei
- Longevity Institute; Davis School of Gerontology; University of Southern California; Los Angeles CA 90089 USA
| | - Hamed Mirzaei
- Longevity Institute; Davis School of Gerontology; University of Southern California; Los Angeles CA 90089 USA
| | - Federica Madia
- Longevity Institute; Davis School of Gerontology; University of Southern California; Los Angeles CA 90089 USA
| | - Mario Mirisola
- Longevity Institute; Davis School of Gerontology; University of Southern California; Los Angeles CA 90089 USA
- DiBiMeF; Universita’ di Palermo; 90133 Palermo Italy
| | - Camille Amparo
- Department of Biological Sciences; School of Dornsife College of Letters, Arts and Sciences; University of Southern California; Los Angeles CA 90089 USA
| | - Shawna Chagoury
- Department of Biological Sciences; School of Dornsife College of Letters, Arts and Sciences; University of Southern California; Los Angeles CA 90089 USA
| | - Brian Kennedy
- Buck Institute for Research on Aging; Novato CA 94945 USA
| | - Valter D. Longo
- Longevity Institute; Davis School of Gerontology; University of Southern California; Los Angeles CA 90089 USA
- Department of Biological Sciences; School of Dornsife College of Letters, Arts and Sciences; University of Southern California; Los Angeles CA 90089 USA
| |
Collapse
|
42
|
Snell TW, Johnston RK, Rabeneck B, Zipperer C, Teat S. Joint inhibition of TOR and JNK pathways interacts to extend the lifespan of Brachionus manjavacas (Rotifera). Exp Gerontol 2014; 52:55-69. [PMID: 24486130 PMCID: PMC3970784 DOI: 10.1016/j.exger.2014.01.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 01/17/2014] [Accepted: 01/20/2014] [Indexed: 12/30/2022]
Abstract
The TOR kinase pathway is central in modulating aging in a variety of animal models. The target of rapamycin (TOR) integrates a complex network of signals from growth conditions, nutrient availability, energy status, and physiological stresses and matches an organism's growth rate to the resource environment. Important remaining problems are the identification of the pathways that interact with TOR and their characterization as additive or synergistic. One of the most versatile stress sensors in metazoans is the Jun-N-terminal kinase (JNK) signaling pathway. JNK is an evolutionarily conserved stress-activated protein kinase that is induced by a range of stressors, including UV irradiation, reactive oxygen species, DNA damage, heat, and bacterial antigens. JNK is thought to interact with the TOR pathway, but its effects on TOR are poorly understood. We used the rotifer Brachionus manjavacas as a model animal to probe the regulation of TOR and JNK pathways and explore their interaction. The effect of various chemical inhibitors was examined in life table and stressor challenge experiments. A survey of 12 inhibitors revealed two, rapamycin and JNK inhibitor, that significantly extended lifespan of B. manjavacas. At 1 μM concentration, exposure to rapamycin or JNK inhibitor extended mean rotifer lifespan by 35% and maximum lifespan by 37%. Exposure to both rapamycin and JNK inhibitor simultaneously extended mean rotifer lifespan by 65% more than either alone. Exposure to a combination of rapamycin and JNK inhibitors conveyed greater protection to starvation, UV and osmotic stress than either inhibitor alone. RNAi knockdown of TOR and JNK gene expression was investigated for its ability to extend rotifer lifespan. RNAi knockdown of the TOR gene resulted in 29% extension of the mean lifespan compared to control and knockdown of the JNK gene resulted in 51% mean lifespan extension. In addition to the lifespan, we quantified mitochondria activity using the fluorescent marker MitoTracker and lysosome activity using LysoTracker. Treatment of rotifers with JNK inhibitor enhanced mitochondria activity nearly 3-fold, whereas rapamycin treatment had no significant effect. Treatment of rotifers with rapamycin or JNK inhibitor reduced lysosome activity in 1, 3 and 8 day old animals, but treatment with both inhibitors did not produce any additive effect. We conclude that inhibition of TOR and JNK pathways significantly extends the lifespan of B. manjavacas. These pathways interact so that inhibition of both simultaneously acts additively to extend rotifer lifespan more than the inhibition of either alone.
Collapse
Affiliation(s)
- Terry W Snell
- School of Biology, Georgia Institute of Technology, Atlanta, GA 30332-0230, USA.
| | - Rachel K Johnston
- School of Biology, Georgia Institute of Technology, Atlanta, GA 30332-0230, USA
| | - Brett Rabeneck
- School of Biology, Georgia Institute of Technology, Atlanta, GA 30332-0230, USA
| | - Cody Zipperer
- School of Biology, Georgia Institute of Technology, Atlanta, GA 30332-0230, USA
| | - Stephanie Teat
- School of Biology, Georgia Institute of Technology, Atlanta, GA 30332-0230, USA
| |
Collapse
|
43
|
Wei Y, Zhang YJ, Cai Y, Xu MH. The role of mitochondria in mTOR-regulated longevity. Biol Rev Camb Philos Soc 2014; 90:167-81. [PMID: 24673778 DOI: 10.1111/brv.12103] [Citation(s) in RCA: 272] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Revised: 02/07/2014] [Accepted: 02/27/2014] [Indexed: 11/27/2022]
Abstract
Several unbiased genome-wide RNA interference (RNAi) screens have pointed to mitochondrial metabolism as the major factor for lifespan regulation. However, conflicting data remain to be clarified concerning the mitochondrial free radical theory of aging (MFRTA). Recently, mTOR (mechanistic target of rapamycin) has been proposed to be the central regulator of aging although how mTOR modulates lifespan is poorly understood. Interestingly, mTOR has been shown to regulate many aspects of mitochondrial function, such as mitochondrial biogenesis, apoptosis, mitophagy and mitochondrial hormesis (mitohormesis) including the retrograde response and mitochondrial unfolded protein response (mito-UPR). Here we discuss the data linking mitochondrial metabolism to mTOR regulation of lifespan, suggesting that hormetic effects may be key to explaining some controversial results regarding the MFRTA. We also discuss the possibility that dysfunction of mitochondrial adaptive responses rather than free radicals per se contributes to the aging process.
Collapse
Affiliation(s)
- Yuehua Wei
- No.3 People's Hospital, School of Medicine, Shanghai Jiao Tong University, 280 Mohe Road, Shanghai, 201900, China
| | | | | | | |
Collapse
|
44
|
Hu F, Liu F. Targeting tissue-specific metabolic signaling pathways in aging: the promise and limitations. Protein Cell 2014; 5:21-35. [PMID: 24474199 PMCID: PMC3938851 DOI: 10.1007/s13238-013-0002-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 09/11/2013] [Indexed: 12/12/2022] Open
Abstract
It has been well established that most of the age-related diseases such as insulin resistance, type 2 diabetes, hypertension, cardiovascular disease, osteoporosis, and atherosclerosis are all closely related to metabolic dysfunction. On the other hand, interventions on metabolism such as calorie restriction or genetic manipulations of key metabolic signaling pathways such as the insulin and mTOR signaling pathways slow down the aging process and improve healthy aging. These findings raise an important question as to whether improving energy homeostasis by targeting certain metabolic signaling pathways in specific tissues could be an effective anti-aging strategy. With a more comprehensive understanding of the tissue-specific roles of distinct metabolic signaling pathways controlling energy homeostasis and the cross-talks between these pathways during aging may lead to the development of more effective therapeutic interventions not only for metabolic dysfunction but also for aging.
Collapse
Affiliation(s)
- Fang Hu
- Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | | |
Collapse
|
45
|
Labunskyy VM, Gerashchenko MV, Delaney JR, Kaya A, Kennedy BK, Kaeberlein M, Gladyshev VN. Lifespan extension conferred by endoplasmic reticulum secretory pathway deficiency requires induction of the unfolded protein response. PLoS Genet 2014; 10:e1004019. [PMID: 24391512 PMCID: PMC3879150 DOI: 10.1371/journal.pgen.1004019] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Accepted: 10/25/2013] [Indexed: 11/18/2022] Open
Abstract
Cells respond to accumulation of misfolded proteins in the endoplasmic reticulum (ER) by activating the unfolded protein response (UPR) signaling pathway. The UPR restores ER homeostasis by degrading misfolded proteins, inhibiting translation, and increasing expression of chaperones that enhance ER protein folding capacity. Although ER stress and protein aggregation have been implicated in aging, the role of UPR signaling in regulating lifespan remains unknown. Here we show that deletion of several UPR target genes significantly increases replicative lifespan in yeast. This extended lifespan depends on a functional ER stress sensor protein, Ire1p, and is associated with constitutive activation of upstream UPR signaling. We applied ribosome profiling coupled with next generation sequencing to quantitatively examine translational changes associated with increased UPR activity and identified a set of stress response factors up-regulated in the long-lived mutants. Besides known UPR targets, we uncovered up-regulation of components of the cell wall and genes involved in cell wall biogenesis that confer resistance to multiple stresses. These findings demonstrate that the UPR is an important determinant of lifespan that governs ER stress and identify a signaling network that couples stress resistance to longevity.
Collapse
Affiliation(s)
- Vyacheslav M. Labunskyy
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Maxim V. Gerashchenko
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Joe R. Delaney
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Alaattin Kaya
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Brian K. Kennedy
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Matt Kaeberlein
- Department of Pathology, University of Washington, Seattle, Washington, United States of America
| | - Vadim N. Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
46
|
Priebe S, Menzel U, Zarse K, Groth M, Platzer M, Ristow M, Guthke R. Extension of life span by impaired glucose metabolism in Caenorhabditis elegans is accompanied by structural rearrangements of the transcriptomic network. PLoS One 2013; 8:e77776. [PMID: 24204961 PMCID: PMC3813781 DOI: 10.1371/journal.pone.0077776] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 09/05/2013] [Indexed: 12/20/2022] Open
Abstract
Glucose restriction mimicked by feeding the roundworm Caenorhabditis elegans with 2-deoxy-D-glucose (DOG) - a glucose molecule that lacks the ability to undergo glycolysis - has been found to increase the life span of the nematodes considerably. To facilitate understanding of the molecular mechanisms behind this life extension, we analyzed transcriptomes of DOG-treated and untreated roundworms obtained by RNA-seq at different ages. We found that, depending on age, DOG changes the magnitude of the expression values of about 2 to 24 percent of the genes significantly, although our results reveal that the gross changes introduced by DOG are small compared to the age-induced changes. We found that 27 genes are constantly either up- or down-regulated by DOG over the whole life span, among them several members of the cytochrome P450 family. The monotonic change with age of the temporal expression patterns of the genes was investigated, leading to the result that 21 genes reverse their monotonic behaviour under impaired glycolysis. Put simply, the DOG-treatment reduces the gross transcriptional activity but increases the interconnectedness of gene expression. However, a detailed analysis of network parameters discloses that the introduced changes differ remarkably between individual signalling pathways. We found a reorganization of the hubs of the mTOR pathway when standard diet is replaced by DOG feeding. By constructing correlation based difference networks, we identified those signalling pathways that are most vigorously changed by impaired glycolysis. Taken together, we have found a number of genes and pathways that are potentially involved in the DOG-driven extension of life span of C. elegans. Furthermore, our results demonstrate how the network structure of ageing-relevant signalling pathways is reorganised under impaired glycolysis.
Collapse
Affiliation(s)
- Steffen Priebe
- Systems Biology and Bioinformatics Group, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knoell-Institute, Jena, Germany
- * E-mail:
| | - Uwe Menzel
- Systems Biology and Bioinformatics Group, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knoell-Institute, Jena, Germany
| | - Kim Zarse
- Department of Human Nutrition, Institute of Nutrition, University of Jena, Jena, Germany
| | - Marco Groth
- Genome Analysis Group, Leibniz Institute for Age Research - Fritz-Lipmann-Institute, Jena, Germany
| | - Matthias Platzer
- Genome Analysis Group, Leibniz Institute for Age Research - Fritz-Lipmann-Institute, Jena, Germany
| | - Michael Ristow
- Department of Human Nutrition, Institute of Nutrition, University of Jena, Jena, Germany
| | - Reinhard Guthke
- Systems Biology and Bioinformatics Group, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knoell-Institute, Jena, Germany
| |
Collapse
|
47
|
Annesley SJ, Chen S, Francione LM, Sanislav O, Chavan AJ, Farah C, De Piazza SW, Storey CL, Ilievska J, Fernando SG, Smith PK, Lay ST, Fisher PR. Dictyostelium, a microbial model for brain disease. Biochim Biophys Acta Gen Subj 2013; 1840:1413-32. [PMID: 24161926 DOI: 10.1016/j.bbagen.2013.10.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 10/05/2013] [Accepted: 10/10/2013] [Indexed: 12/25/2022]
Abstract
BACKGROUND Most neurodegenerative diseases are associated with mitochondrial dysfunction. In humans, mutations in mitochondrial genes result in a range of phenotypic outcomes which do not correlate well with the underlying genetic cause. Other neurodegenerative diseases are caused by mutations that affect the function and trafficking of lysosomes, endosomes and autophagosomes. Many of the complexities of these human diseases can be avoided by studying them in the simple eukaryotic model Dictyostelium discoideum. SCOPE OF REVIEW This review describes research using Dictyostelium to study cytopathological pathways underlying a variety of neurodegenerative diseases including mitochondrial, lysosomal and vesicle trafficking disorders. MAJOR CONCLUSIONS Generalised mitochondrial respiratory deficiencies in Dictyostelium produce a consistent pattern of defective phenotypes that are caused by chronic activation of a cellular energy sensor AMPK (AMP-activated protein kinase) and not ATP deficiency per se. Surprisingly, when individual subunits of Complex I are knocked out, both AMPK-dependent and AMPK-independent, subunit-specific phenotypes are observed. Many nonmitochondrial proteins associated with neurological disorders have homologues in Dictyostelium and are associated with the function and trafficking of lysosomes and endosomes. Conversely, some genes associated with neurodegenerative disorders do not have homologues in Dictyostelium and this provides a unique avenue for studying these mutated proteins in the absence of endogeneous protein. GENERAL SIGNIFICANCE Using the Dictyostelium model we have gained insights into the sublethal cytopathological pathways whose dysregulation contributes to phenotypic outcomes in neurodegenerative disease. This work is beginning to distinguish correlation, cause and effect in the complex network of cross talk between the various organelles involved. This article is part of a Special Issue entitled Frontiers of Mitochondrial Research.
Collapse
Affiliation(s)
- S J Annesley
- Department of Microbiology, La Trobe University, Plenty Rd., Bundoora, VIC, Australia, 3086
| | - S Chen
- Department of Microbiology, La Trobe University, Plenty Rd., Bundoora, VIC, Australia, 3086
| | - L M Francione
- Department of Microbiology, La Trobe University, Plenty Rd., Bundoora, VIC, Australia, 3086
| | - O Sanislav
- Department of Microbiology, La Trobe University, Plenty Rd., Bundoora, VIC, Australia, 3086
| | - A J Chavan
- Department of Microbiology, La Trobe University, Plenty Rd., Bundoora, VIC, Australia, 3086
| | - C Farah
- Department of Microbiology, La Trobe University, Plenty Rd., Bundoora, VIC, Australia, 3086
| | - S W De Piazza
- Department of Microbiology, La Trobe University, Plenty Rd., Bundoora, VIC, Australia, 3086
| | - C L Storey
- Department of Microbiology, La Trobe University, Plenty Rd., Bundoora, VIC, Australia, 3086
| | - J Ilievska
- Department of Microbiology, La Trobe University, Plenty Rd., Bundoora, VIC, Australia, 3086
| | - S G Fernando
- Department of Microbiology, La Trobe University, Plenty Rd., Bundoora, VIC, Australia, 3086
| | - P K Smith
- Department of Microbiology, La Trobe University, Plenty Rd., Bundoora, VIC, Australia, 3086
| | - S T Lay
- Department of Microbiology, La Trobe University, Plenty Rd., Bundoora, VIC, Australia, 3086
| | - P R Fisher
- Department of Microbiology, La Trobe University, Plenty Rd., Bundoora, VIC, Australia, 3086.
| |
Collapse
|
48
|
Targeting tissue-specific metabolic signaling pathways in aging: the promise and limitations. Protein Cell 2013. [DOI: 10.1007/s13238-013-3077-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
49
|
Rapamycin and mTOR inhibitors probably have therapeutic effects for post-operative cognitive dysfunction. Med Hypotheses 2013; 81:487-8. [DOI: 10.1016/j.mehy.2013.06.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Revised: 06/13/2013] [Accepted: 06/18/2013] [Indexed: 12/22/2022]
|
50
|
Anisimov VN, Bartke A. The key role of growth hormone-insulin-IGF-1 signaling in aging and cancer. Crit Rev Oncol Hematol 2013; 87:201-23. [PMID: 23434537 PMCID: PMC4095988 DOI: 10.1016/j.critrevonc.2013.01.005] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 12/22/2012] [Accepted: 01/18/2013] [Indexed: 12/14/2022] Open
Abstract
Studies in mammals have led to the suggestion that hyperglycemia and hyperinsulinemia are important factors in aging. GH/Insulin/insulin-like growth factor-1 (IGF-1) signaling molecules that have been linked to longevity include daf-2 and InR and their homologues in mammals, and inactivation of the corresponding genes increases lifespan in nematodes, fruit flies and mice. The life-prolonging effects of caloric restriction are likely related to decreasing IGF-1 levels. Evidence has emerged that antidiabetic drugs are promising candidates for both lifespan extension and prevention of cancer. Thus, antidiabetic drugs postpone spontaneous carcinogenesis in mice and rats, as well as chemical and radiation carcinogenesis in mice, rats and hamsters. Furthermore, metformin seems to decrease the risk for cancer in diabetic patients.
Collapse
Affiliation(s)
- Vladimir N Anisimov
- Department of Carcinogenesis and Oncogerontology, N.N. Petrov Research Institute of Oncology, St. Petersburg, Russia.
| | | |
Collapse
|