1
|
Li Y, Xiang L, Qi J. Procyanidin A1 from Peanut Skin Exerts Anti-Aging Effects and Attenuates Senescence via Antioxidative Stress and Autophagy Induction. Antioxidants (Basel) 2025; 14:322. [PMID: 40227314 PMCID: PMC11939485 DOI: 10.3390/antiox14030322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/05/2025] [Accepted: 03/05/2025] [Indexed: 04/15/2025] Open
Abstract
The aging population is steadily increasing, with aging and age-related diseases serving as major risk factors for morbidity, mortality, and economic burden. Peanuts, known as the "longevity nut" in China, have been shown to offer various health benefits, with peanut skin extract (PSE) emerging as a key compound of interest. This study investigates the bioactive compound in PSE with anti-aging potential and explores its underlying mechanisms of action. Procyanidin A1 (PC A1) was isolated from PSE, guided by the K6001 yeast replicative lifespan model. PC A1 prolonged the replicative lifespan of yeast and the yeast-like chronological lifespan of PC12 cells. To further confirm its anti-aging effect, cellular senescence, a hallmark of aging, was assessed. In senescent cells induced by etoposide (Etop), PC A1 alleviated senescence by reducing ROS levels, decreasing the percentage of senescent cells, and restoring proliferative capacity. Transcriptomics analysis revealed that PC A1 induced apoptosis, reduced senescence-associated secretory phenotype (SASP) factors, and modulated the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway. The antioxidative capacity of PC A1 was also evaluated, showing enhanced resistance to oxidative stress in PC12 cells by reducing reactive oxygen species (ROS) and malondialdehyde (MDA) levels and increasing superoxide dismutase (SOD) activity. Moreover, PC A1 induced autophagy, as evidenced by an increase in fluorescence-labeled autophagic compartments and confirmation via Western blot analysis of autophagy-related proteins. In addition, the treatment of an autophagy inhibitor abolished the antioxidative stress and senescence-alleviating effects of PC A1. These findings reveal that PC A1 extended lifespans and alleviated cellular senescence by enhancing oxidative stress resistance and inducing autophagy, positioning it as a promising candidate for further exploration as a geroprotective agent.
Collapse
Affiliation(s)
| | | | - Jianhua Qi
- College of Pharmaceutical Sciences, Zhejiang University, Yu Hang Tang Road 866, Hangzhou 310058, China; (Y.L.); (L.X.)
| |
Collapse
|
2
|
Wang X, Zhu Y, Liu H, Wang X, Zhang H, Chen X. Nitazoxanide alleviates experimental pulmonary fibrosis by inhibiting the development of cellular senescence. Life Sci 2025; 361:123302. [PMID: 39662775 DOI: 10.1016/j.lfs.2024.123302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/26/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease characterized by irreversible lung scarring with a poor prognosis. Emerging evidence has revealed that IPF is an aging-related disease, and the development of cellular senescence plays a pivotal role in persistent remodeling and fibrotic scarring, acting as a key mechanism in the pathophysiology of IPF. Exploring therapeutic strategies for modulating cellular senescence can provide crucial insights into unraveling IPF processes. Here, we have identified Nitazoxanide (NTZ), an FDA-approved antiprotozoal agent, has specific effects on inhibiting cellular senescence development. In the bleomycin and D-galactose-induced senescence model, NTZ effectively inhibits senescence associated-β-gal staining and preserves cell proliferation ability. We also found that NTZ effectively impedes senescence progression in the bleomycin-induced pulmonary fibrosis model, while mitigating the release of senescence-associated secretory phenotype and alleviating pulmonary fibrosis. The anti-senescence effect of NTZ is mechanistically dependent on the preservation of nuclear SIRT1 expression. We observed that PI3K induces a WIPI1-mediated nucleophagic degradation of SIRT1, while NTZ effectively inhibits PI3K and suppresses WIPI1 expression, thereby maintaining SIRT1 expression in the nucleus and exerting its anti-senescence function. Collectively, our research has shown that NTZ can inhibit PI3K in senescence progression, leading to the inhibition of WIPI1-mediated SIRT1 nucleophagic degradation. As a result, NTZ alleviates fibrosis by inhibiting senescence development.
Collapse
Affiliation(s)
- Xianzhe Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao
| | - Yanyan Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao
| | - Huilin Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao
| | - Xiangchuan Wang
- Biological Imaging and Stem Cell Core, Faculty of Health Sciences, University of Macau, Taipa, Macao
| | - Hongjie Zhang
- Biological Imaging and Stem Cell Core, Faculty of Health Sciences, University of Macau, Taipa, Macao
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao.
| |
Collapse
|
3
|
Moka MK, George M, Sriram DK. Advancing Longevity: Exploring Antiaging Pharmaceuticals in Contemporary Clinical Trials Amid Aging Dynamics. Rejuvenation Res 2024; 27:220-233. [PMID: 39162996 DOI: 10.1089/rej.2024.0040] [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] [Indexed: 08/21/2024] Open
Abstract
Aging is an inevitable biological process that significantly impacts human health, leading to a decline in cellular function and an increase in cellular damage. This study elucidates the burgeoning potential of antiaging pharmaceuticals in mitigating the thriving burden of chronic conditions linked to advancing age. It underscores the pivotal role of these pharmacotherapeutic agents in fostering longevity free from debilitating age-related afflictions, notably cardiovascular disorders, neoplastic processes, and neurodegenerative pathologies. While commendable strides have been made evident in preclinical models, it is crucial to thoroughly investigate their effectiveness and safety in human groups. In addition, ethical concerns about fair access, societal impacts, and careful resource distribution are significant in discussions about developing and using antiaging medications. By approaching the development and utilization of antiaging medications with diligence and foresight, we can strive toward a future where individuals can enjoy extended lifespans free from the debilitating effects of age-related ailments.
Collapse
Affiliation(s)
| | - Melvin George
- Department of Clinical Research, Hindu Mission Hospital, Tambaram, India
| | - D K Sriram
- Department of Diabetology and Endocrinology, Hindu Mission Hospital, Tambaram, India
| |
Collapse
|
4
|
Bearden AA, Stewart EM, Casher CC, Shaddix MA, Nobles AC, Mockett RJ. Effects of Target of Rapamycin and Phosphatidylinositol 3-Kinase Inhibitors and Other Autophagy-Related Supplements on Life Span in y w Male Drosophila melanogaster. Int J Mol Sci 2024; 25:11504. [PMID: 39519056 PMCID: PMC11547029 DOI: 10.3390/ijms252111504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/10/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024] Open
Abstract
Various dietary supplements have been shown to extend the life span of Drosophila melanogaster, including several that promote autophagy, such as rapamycin and spermidine. The goal of the study presented here was to test numerous additional potential anti-aging supplements, primarily inhibitors of the target of rapamycin (TOR) and/or phosphatidylinositol 3-kinase (PI3K). Using a single, comparatively long-lived y w test strain, screening was performed in male flies supplemented either throughout adulthood or, in a few cases, beginning in middle or late adult life, with concentrations spanning 4-6 orders of magnitude in most cases. Supplementation with PP242 and deferiprone, an iron chelator, beginning in late adult life had no positive effect on life span. Lifelong supplementation with Ku-0063794, LY294002, PX-866-17OH, Torin2 and WYE-28 had no effect at any dose. Rapamycin, spermidine and wortmannin all had significant life-shortening effects at the highest doses tested. AZD8055, PI-103 hydrochloride and WYE-132 yielded slight beneficial effects at 1-2 doses, but only 100 nM AZD8055 was confirmed to have a minor (1.3%) effect in a replicate experiment, which was encompassed by other control groups within the same study. These compounds had no effect on fly fecundity (egg laying) or fertility (development of progeny to adulthood), but equivalent high doses of rapamycin abolished fertility. The solvent DMSO had no significant effect on life span at the concentrations used to solubilize most compounds in the fly medium, but it drastically curtailed both survival and fertility at higher concentrations. 2-Hydroxypropyl-β-cyclodextrin also failed to extend the life span when provided throughout adulthood or beginning in mid-adult life. Collectively, the results suggest that inhibition of the TOR/PI3K pathway and autophagy through dietary intervention is not a straightforward anti-aging strategy in Drosophila and that further extension of life is difficult in comparatively long-lived flies.
Collapse
Affiliation(s)
| | | | | | | | | | - Robin J. Mockett
- Department of Biomedical Sciences, University of South Alabama, Mobile, AL 36688-0002, USA; (A.A.B.); (E.M.S.); (C.C.C.); (M.A.S.); (A.C.N.)
| |
Collapse
|
5
|
Ohtsuka H, Shimasaki T, Aiba H. Low-Molecular Weight Compounds that Extend the Chronological Lifespan of Yeasts, Saccharomyces cerevisiae, and Schizosaccharomyces pombe. Adv Biol (Weinh) 2024; 8:e2400138. [PMID: 38616173 DOI: 10.1002/adbi.202400138] [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: 03/11/2024] [Revised: 04/04/2024] [Indexed: 04/16/2024]
Abstract
Yeast is an excellent model organism for research for regulating aging and lifespan, and the studies have made many contributions to date, including identifying various factors and signaling pathways related to aging and lifespan. More than 20 years have passed since molecular biological perspectives are adopted in this research field, and intracellular factors and signal pathways that control aging and lifespan have evolutionarily conserved from yeast to mammals. Furthermore, these findings have been applied to control the aging and lifespan of various model organisms by adjustment of the nutritional environment, genetic manipulation, and drug treatment using low-molecular weight compounds. Among these, drug treatment is easier than the other methods, and research into drugs that regulate aging and lifespan is consequently expected to become more active. Chronological lifespan, a definition of yeast lifespan, refers to the survival period of a cell population under nondividing conditions. Herein, low-molecular weight compounds are summarized that extend the chronological lifespan of Saccharomyces cerevisiae and Schizosaccharomyces pombe, along with their intracellular functions. The low-molecular weight compounds are also discussed that extend the lifespan of other model organisms. Compounds that have so far only been studied in yeast may soon extend lifespan in other organisms.
Collapse
Affiliation(s)
- Hokuto Ohtsuka
- Laboratory of Molecular Microbiology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Aichi, Japan
| | - Takafumi Shimasaki
- Laboratory of Molecular Microbiology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Aichi, Japan
| | - Hirofumi Aiba
- Laboratory of Molecular Microbiology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Aichi, Japan
| |
Collapse
|
6
|
Moskalev AA. Potential Geroprotectors - From Bench to Clinic. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:1732-1738. [PMID: 38105194 DOI: 10.1134/s0006297923110056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/20/2023] [Accepted: 09/20/2023] [Indexed: 12/19/2023]
Abstract
Geroprotectors are substances that slow down aging process and can be used for prevention of age-related diseases. Geroprotectors can improve functioning of various organ systems and enhance their homeostatic capabilities. We have developed a system of criteria for geroprotectors and proposed their classification based on the mechanisms of their action on the aging processes. Geroprotectors are required to reduce mortality, improve human aging biomarkers, have minimal side effects, and enhance quality of life. Additionally, there are approaches based on combining geroprotectors targeted to different targets and mechanisms of aging to achieve maximum effectiveness. Currently, numerous preclinical studies are being conducted to identify new molecular targets and develop new approaches to extend healthy aging, although the number of clinical trials is limited. Geroprotectors have the potential to become a new class of preventive medicines as they prevent onset of certain diseases or slow down their progression.
Collapse
Affiliation(s)
- Alexey A Moskalev
- Institute of Biogerontology, Lobachevsky University, Nizhny Novgorod, 603950, Russia.
| |
Collapse
|
7
|
Parkhitko AA, Filine E, Tatar M. Combinatorial interventions in aging. NATURE AGING 2023; 3:1187-1200. [PMID: 37783817 PMCID: PMC11194689 DOI: 10.1038/s43587-023-00489-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 08/15/2023] [Indexed: 10/04/2023]
Abstract
Insight on the underlying mechanisms of aging will advance our ability to extend healthspan, treat age-related pathology and improve quality of life. Multiple genetic and pharmacological manipulations extend longevity in different species, yet monotherapy may be relatively inefficient, and we have limited data on the effect of combined interventions. Here we summarize interactions between age-related pathways and discuss strategies to simultaneously retard these in different organisms. In some cases, combined manipulations additively increase their impact on common hallmarks of aging and lifespan, suggesting they quantitatively participate within the same pathway. In other cases, interactions affect different hallmarks, suggesting their joint manipulation may independently maximize their effects on lifespan and healthy aging. While most interaction studies have been conducted with invertebrates and show varying levels of translatability, the conservation of pro-longevity pathways offers an opportunity to identify 'druggable' targets relevant to multiple human age-associated pathologies.
Collapse
Affiliation(s)
- Andrey A Parkhitko
- Aging Institute of UPMC and the University of Pittsburgh, Pittsburgh, PA, USA.
| | - Elizabeth Filine
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Marc Tatar
- Department of Ecology, Evolution and Organismal Biology, Brown University, Providence, RI, USA.
| |
Collapse
|
8
|
Compound combinations targeting longevity: Challenges and perspectives. Ageing Res Rev 2023; 85:101851. [PMID: 36642188 DOI: 10.1016/j.arr.2023.101851] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/05/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023]
Abstract
Aging is one of the world's greatest concerns, requiring urgent, effective, large-scale interventions to decrease the number of late-life chronic diseases and improve human healthspan. Anti-aging drug therapy is one of the most promising strategies to combat the effects of aging. However, most geroprotective compounds are known to successfully affect only a few aging-related targets. Given this, there is a great biological rationale for the use of combinations of anti-aging interventions. In this review, we characterize the various types of compound combinations used to modulate lifespan, discuss the existing evidence on their role in life extension, and present some key points about current challenges and future prospects for the development of combination drug anti-aging therapy.
Collapse
|
9
|
Banerjee A, Malayaperumal S, Pathak S. Healthy Aging: Perseverance to Well Being. Endocr Metab Immune Disord Drug Targets 2023; 23:1245-1247. [PMID: 36959136 DOI: 10.2174/1871530323666230320120903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 01/11/2023] [Accepted: 01/18/2023] [Indexed: 03/25/2023]
Affiliation(s)
- Antara Banerjee
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Chennai, 603 103, India
| | - Sarubala Malayaperumal
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Chennai, 603 103, India
| | - Surajit Pathak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Chennai, 603 103, India
| |
Collapse
|
10
|
Therapeutic Antiaging Strategies. Biomedicines 2022; 10:biomedicines10102515. [PMID: 36289777 PMCID: PMC9599338 DOI: 10.3390/biomedicines10102515] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/21/2022] [Accepted: 09/24/2022] [Indexed: 11/17/2022] Open
Abstract
Aging constitutes progressive physiological changes in an organism. These changes alter the normal biological functions, such as the ability to manage metabolic stress, and eventually lead to cellular senescence. The process itself is characterized by nine hallmarks: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication. These hallmarks are risk factors for pathologies, such as cardiovascular diseases, neurodegenerative diseases, and cancer. Emerging evidence has been focused on examining the genetic pathways and biological processes in organisms surrounding these nine hallmarks. From here, the therapeutic approaches can be addressed in hopes of slowing the progression of aging. In this review, data have been collected on the hallmarks and their relative contributions to aging and supplemented with in vitro and in vivo antiaging research experiments. It is the intention of this article to highlight the most important antiaging strategies that researchers have proposed, including preventive measures, systemic therapeutic agents, and invasive procedures, that will promote healthy aging and increase human life expectancy with decreased side effects.
Collapse
|
11
|
Shaposhnikov MV, Guvatova ZG, Zemskaya NV, Koval LA, Schegoleva EV, Gorbunova AA, Golubev DA, Pakshina NR, Ulyasheva NS, Solovev IA, Bobrovskikh MA, Gruntenko NE, Menshanov PN, Krasnov GS, Kudryavseva AV, Moskalev AA. Molecular mechanisms of exceptional lifespan increase of Drosophila melanogaster with different genotypes after combinations of pro-longevity interventions. Commun Biol 2022; 5:566. [PMID: 35681084 PMCID: PMC9184560 DOI: 10.1038/s42003-022-03524-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 05/24/2022] [Indexed: 12/13/2022] Open
Abstract
Aging is one of the global challenges of our time. The search for new anti-aging interventions is also an issue of great actuality. We report on the success of Drosophila melanogaster lifespan extension under the combined influence of dietary restriction, co-administration of berberine, fucoxanthin, and rapamycin, photodeprivation, and low-temperature conditions up to 185 days in w1118 strain and up to 213 days in long-lived E(z)/w mutants. The trade-off was found between longevity and locomotion. The transcriptome analysis showed an impact of epigenetic alterations, lipid metabolism, cellular respiration, nutrient sensing, immune response, and autophagy in the registered effect. The lifespan of fruit flies can be extended up to 213 days under specialized conditions.
Collapse
|
12
|
McIntyre RL, Liu YJ, Hu M, Morris BJ, Willcox BJ, Donlon TA, Houtkooper RH, Janssens GE. Pharmaceutical and nutraceutical activation of FOXO3 for healthy longevity. Ageing Res Rev 2022; 78:101621. [PMID: 35421606 DOI: 10.1016/j.arr.2022.101621] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/10/2022] [Accepted: 04/07/2022] [Indexed: 12/12/2022]
Abstract
Life expectancy has increased substantially over the last 150 years. Yet this means that now most people also spend a greater length of time suffering from various age-associated diseases. As such, delaying age-related functional decline and extending healthspan, the period of active older years free from disease and disability, is an overarching objective of current aging research. Geroprotectors, compounds that target pathways that causally influence aging, are increasingly recognized as a means to extend healthspan in the aging population. Meanwhile, FOXO3 has emerged as a geroprotective gene intricately involved in aging and healthspan. FOXO3 genetic variants are linked to human longevity, reduced disease risks, and even self-reported health. Therefore, identification of FOXO3-activating compounds represents one of the most direct candidate approaches to extending healthspan in aging humans. In this work, we review compounds that activate FOXO3, or influence healthspan or lifespan in a FOXO3-dependent manner. These compounds can be classified as pharmaceuticals, including PI3K/AKT inhibitors and AMPK activators, antidepressants and antipsychotics, muscle relaxants, and HDAC inhibitors, or as nutraceuticals, including primary metabolites involved in cell growth and sustenance, and secondary metabolites including extracts, polyphenols, terpenoids, and other purified natural compounds. The compounds documented here provide a basis and resource for further research and development, with the ultimate goal of promoting healthy longevity in humans.
Collapse
Affiliation(s)
- Rebecca L McIntyre
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Yasmine J Liu
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Man Hu
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Brian J Morris
- School of Medical Sciences, University of Sydney, Sydney, NSW, Australia; Department of Research, Kuakini Medical Center, Honolulu, HI, USA; Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Bradley J Willcox
- Department of Research, Kuakini Medical Center, Honolulu, HI, USA; Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Timothy A Donlon
- Department of Research, Kuakini Medical Center, Honolulu, HI, USA; Department of Cell and Molecular Biology and Department of Pathology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Georges E Janssens
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
13
|
Moskalev A, Guvatova Z, Lopes IDA, Beckett CW, Kennedy BK, De Magalhaes JP, Makarov AA. Targeting aging mechanisms: pharmacological perspectives. Trends Endocrinol Metab 2022; 33:266-280. [PMID: 35183431 DOI: 10.1016/j.tem.2022.01.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/18/2022] [Accepted: 01/22/2022] [Indexed: 12/12/2022]
Abstract
Geroprotectors slow down aging and promote healthy longevity in model animals. Although hundreds of compounds have been shown to extend the life of laboratory model organisms, clinical studies on potential geroprotectors are exceedingly rare, especially in healthy elders. This review aims to classify potential geroprotectors based on the mechanisms by which they influence aging. These pharmacological interventions can be classified into the following groups: those that prevent oxidation; proteostasis regulators; suppressors of genomic instability; epigenetic drugs; those that preserve mitochondrial function; inhibitors of aging-associated signaling pathways; hormetins; senolytics/senostatics; anti-inflammatory drugs; antifibrotic agents; neurotrophic factors; factors preventing the impairment of barrier function; immunomodulators; and prebiotics, metabiotics, and enterosorbents.
Collapse
Affiliation(s)
- Alexey Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia; Institute of Biology of the Federal Research Center of Komi Science Center, Ural Branch of the Russian Academy of Sciences, 28 Kommunisticheskaya Street, Syktyvkar 167982, Russia.
| | - Zulfiya Guvatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| | - Ines De Almeida Lopes
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Charles W Beckett
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Brian K Kennedy
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Centre for Healthy Longevity, National University Health System, Singapore; Singapore Institute of Clinical Sciences, A*STAR, Singapore
| | - Joao Pedro De Magalhaes
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK.
| | - Alexander A Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia.
| |
Collapse
|
14
|
Zhang Y, Zhang J, Wang S. The Role of Rapamycin in Healthspan Extension via the Delay of Organ Aging. Ageing Res Rev 2021; 70:101376. [PMID: 34089901 DOI: 10.1016/j.arr.2021.101376] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 05/07/2021] [Accepted: 05/30/2021] [Indexed: 12/17/2022]
Abstract
Aging can not only shorten a healthy lifespan, but can also lead to multi-organ dysfunction and failure. Anti-aging is a complex and worldwide conundrum for eliminating the various pathologies of senility. The past decade has seen great progress in the understanding of the aging-associated signaling pathways and their application for developing anti-aging approaches. Currently, some drugs can improve quality of life. The activation of mammalian target of rapamycin (mTOR) signaling is one of the core and detrimental mechanisms related to aging; rapamycin can reduce the rate of aging, improve age-related diseases by inhibiting the mTOR pathway, and prolong lifespan and healthspan effectively. However, the current evidence for rapamycin in lifespan extension and organ aging is fragmented and scattered. In this review, we summarize the efficacy and safety of rapamycin in prolonging a healthy lifespan by systematically alleviating aging in multiple organ systems, i.e., the nervous, urinary, digestive, circulatory, motor, respiratory, endocrine, reproductive, integumentary and immune systems, to provide a theoretical basis for the future clinical application of rapamycin in anti-aging.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jinjin Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; National Clinical Research Center for Obstetrical and Gynecological Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
15
|
Platonova EY, Shaposhnikov MV, Lee HY, Lee JH, Min KJ, Moskalev A. Black chokeberry (Aronia melanocarpa) extracts in terms of geroprotector criteria. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.06.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
16
|
Shaposhnikov MV, Zemskaya NV, Koval LA, Schegoleva EV, Yakovleva DV, Ulyasheva NS, Gorbunova AA, Minnikhanova NR, Moskalev AA. Geroprotective potential of genetic and pharmacological interventions to endogenous hydrogen sulfide synthesis in Drosophila melanogaster. Biogerontology 2021; 22:197-214. [PMID: 33544267 DOI: 10.1007/s10522-021-09911-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/22/2021] [Indexed: 12/11/2022]
Abstract
Endogenous hydrogen sulfide (H2S) is a gasotransmitter with a wide range of physiological functions. Aging is accompanied by disruption of H2S homeostasis, therefore, interventions to the processes of H2S metabolism to maintain its balance may have geroprotective potential. Here we demonstrated the additive geroprotective effect of combined genetic and pharmacological interventions to the hydrogen sulfide biosynthesis system by overexpression of cystathionine-β-synthase and cystathionine-γ-lyase genes and treatment with precursors of H2S synthesis cysteine (Cys) and N-acetyl-L-cysteine (NAC). The obtained results suggest that additive effects of genetic and pharmacological interventions to H2S metabolism may be associated with the complex interaction between beneficial action of H2S production and prevention of adverse effects of excess H2S production by Cys and NAC treatment.
Collapse
Affiliation(s)
- Mikhail V Shaposhnikov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russian Federation.,Institute of Biology of Komi Science Center, Ural Branch of RAS, 167982, Syktyvkar, Russian Federation
| | - Nadezhda V Zemskaya
- Institute of Biology of Komi Science Center, Ural Branch of RAS, 167982, Syktyvkar, Russian Federation
| | - Liubov A Koval
- Institute of Biology of Komi Science Center, Ural Branch of RAS, 167982, Syktyvkar, Russian Federation
| | - Eugenia V Schegoleva
- Institute of Biology of Komi Science Center, Ural Branch of RAS, 167982, Syktyvkar, Russian Federation
| | - Daria V Yakovleva
- Institute of Biology of Komi Science Center, Ural Branch of RAS, 167982, Syktyvkar, Russian Federation
| | - Natalia S Ulyasheva
- Institute of Biology of Komi Science Center, Ural Branch of RAS, 167982, Syktyvkar, Russian Federation
| | - Anastasia A Gorbunova
- Institute of Biology of Komi Science Center, Ural Branch of RAS, 167982, Syktyvkar, Russian Federation
| | - Natalya R Minnikhanova
- Institute of Biology of Komi Science Center, Ural Branch of RAS, 167982, Syktyvkar, Russian Federation
| | - Alexey A Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russian Federation. .,Institute of Biology of Komi Science Center, Ural Branch of RAS, 167982, Syktyvkar, Russian Federation.
| |
Collapse
|
17
|
Moskalev A, Stambler I, Caruso C. Innate and Adaptive Immunity in Aging and Longevity: The Foundation of Resilience. Aging Dis 2020; 11:1363-1373. [PMID: 33269094 PMCID: PMC7673842 DOI: 10.14336/ad.2020.0603] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/03/2020] [Indexed: 12/15/2022] Open
Abstract
The interrelation of the processes of immunity and senescence now receives an unprecedented emphasis during the COVID-19 pandemic, which brings to the fore the critical need to combat immunosenescence and improve the immune function and resilience of older persons. Here we review the historical origins and the current state of the science of innate and adaptive immunity in aging and longevity. From the modern point of view, innate and adaptive immunity are not only affected by aging but also are important parts of its underlying mechanisms. Excessive levels or activity of antimicrobial peptides, C-reactive protein, complement system, TLR/NF-κB, cGAS/STING/IFN 1,3 and AGEs/RAGE pathways, myeloid cells and NLRP3 inflammasome, declined levels of NK cells in innate immunity, thymus involution and decreased amount of naive T-cells in adaptive immunity, are biomarkers of aging and predisposition factors for cellular senescence and aging-related pathologies. Long-living species, human centenarians, and women are characterized by less inflamm-aging and decelerated immunosenescence. Despite recent progress in understanding, the harmonious theory of immunosenescence is still developing. Geroprotectors targeting these mechanisms are just emerging and are comprehensively discussed in this article.
Collapse
Affiliation(s)
- Alexey Moskalev
- Institute of Biology of FRC of Komi Scientific Center of Ural Branch of Russian Academy of Sciences, Syktyvkar, 167982, Russia.
| | - Ilia Stambler
- Vetek (Seniority), The Movement for Longevity and Quality of Life, Israel.
| | - Calogero Caruso
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| |
Collapse
|
18
|
Garratt M. Why do sexes differ in lifespan extension? Sex-specific pathways of aging and underlying mechanisms for dimorphic responses. ACTA ACUST UNITED AC 2020. [DOI: 10.3233/nha-190067] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Males and females typically have different lifespans and frequently differ in their responses to anti-aging interventions. These sex-specific responses are documented in mice and Drosophila species, in addition to other organisms where interventions have been tested. While the prevalence of sex-specific responses to anti-aging interventions is now recognised, the underlying causes remain poorly understood. This review first summarises the main pathways and interventions that lead to sex-specific lifespan responses, including the growth-hormone/insulin-like growth factor 1 (GH-IGF1) axis, mechanistic target of rapamycin (mTOR) signalling, and nutritional and pharmacological interventions. After summarising current evidence, several different potential causes for sex-specific responses are discussed. These include sex-differences in xenobiotic metabolism, differing disease susceptibility, sex-specific hormone production and chromosomes, and the relative importance of different signalling pathways in the control of male and female life-history. Understanding why sex-differences in lifespan-extension occur should provide a greater understanding of the mechanisms that regulate the aging process in each sex, and will be crucial for understanding the full implications of these treatments if they are translated to humans.
Collapse
Affiliation(s)
- Michael Garratt
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
19
|
Transcriptomics-Based Screening Identifies Pharmacological Inhibition of Hsp90 as a Means to Defer Aging. Cell Rep 2020; 27:467-480.e6. [PMID: 30970250 PMCID: PMC6459000 DOI: 10.1016/j.celrep.2019.03.044] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/31/2019] [Accepted: 03/13/2019] [Indexed: 12/24/2022] Open
Abstract
Aging strongly influences human morbidity and mortality. Thus, aging-preventive compounds could greatly improve our health and lifespan. Here we screened for such compounds, known as geroprotectors, employing the power of transcriptomics to predict biological age. Using age-stratified human tissue transcriptomes and machine learning, we generated age classifiers and applied these to transcriptomic changes induced by 1,309 different compounds in human cells, ranking these compounds by their ability to induce a “youthful” transcriptional state. Testing the top candidates in C. elegans, we identified two Hsp90 inhibitors, monorden and tanespimycin, which extended the animals’ lifespan and improved their health. Hsp90 inhibition induces expression of heat shock proteins known to improve protein homeostasis. Consistently, monorden treatment improved the survival of C. elegans under proteotoxic stress, and its benefits depended on the cytosolic unfolded protein response-inducing transcription factor HSF-1. Taken together, our method represents an innovative geroprotector screening approach and was able to identify a class that acts by improving protein homeostasis. Transcriptome-based age classifiers can distinguish young versus old tissues Application of age classifiers to drug-induced transcriptomes finds geroprotectors Validation of geroprotectors in C. elegans highlights Hsp90 inhibitors Hsp90 inhibitors act through HSF-1 to improve health and extend lifespan
Collapse
|
20
|
Bakula D, Ablasser A, Aguzzi A, Antebi A, Barzilai N, Bittner MI, Jensen MB, Calkhoven CF, Chen D, de Grey AD, Feige JN, Georgievskaya A, Gladyshev VN, Golato T, Gudkov AV, Hoppe T, Kaeberlein M, Katajisto P, Kennedy BK, Lal U, Martin-Villalba A, Moskalev AA, Ozerov I, Petr MA, Reason, Rubinsztein DC, Tyshkovskiy A, Vanhaelen Q, Zhavoronkov A, Scheibye-Knudsen M. Latest advances in aging research and drug discovery. Aging (Albany NY) 2019; 11:9971-9981. [PMID: 31770722 PMCID: PMC6914421 DOI: 10.18632/aging.102487] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 11/09/2019] [Indexed: 12/19/2022]
Abstract
An increasing aging population poses a significant challenge to societies worldwide. A better understanding of the molecular, cellular, organ, tissue, physiological, psychological, and even sociological changes that occur with aging is needed in order to treat age-associated diseases. The field of aging research is rapidly expanding with multiple advances transpiring in many previously disconnected areas. Several major pharmaceutical, biotechnology, and consumer companies made aging research a priority and are building internal expertise, integrating aging research into traditional business models and exploring new go-to-market strategies. Many of these efforts are spearheaded by the latest advances in artificial intelligence, namely deep learning, including generative and reinforcement learning. To facilitate these trends, the Center for Healthy Aging at the University of Copenhagen and Insilico Medicine are building a community of Key Opinion Leaders (KOLs) in these areas and launched the annual conference series titled "Aging Research and Drug Discovery (ARDD)" held in the capital of the pharmaceutical industry, Basel, Switzerland (www.agingpharma.org). This ARDD collection contains summaries from the 6th annual meeting that explored aging mechanisms and new interventions in age-associated diseases. The 7th annual ARDD exhibition will transpire 2nd-4th of September, 2020, in Basel.
Collapse
Affiliation(s)
- Daniela Bakula
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Ablasser
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Adam Antebi
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Nir Barzilai
- Department of Medicine, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | - Cornelis F. Calkhoven
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, AD Groningen, The Netherlands
| | - Danica Chen
- Program in Metabolic Biology, Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| | | | - Jerome N. Feige
- Nestlé Research, EPFL Innovation Park, Lausanne, Switzerland
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | | | - Vadim N. Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Andrei V. Gudkov
- Roswell Park Comprehensive Cancer Center and Genome Protection, Inc., Buffalo, NY 14203, USA
| | - Thorsten Hoppe
- Institute for Genetics and CECAD Research Center, University of Cologne, Cologne, Germany
| | - Matt Kaeberlein
- Department of Pathology, School of Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Pekka Katajisto
- Department of Biosciences and Nutrition, Center for Innovative Medicine, Karolinska Institutet, Huddinge, Sweden
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Brian K. Kennedy
- Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University Singapore, Singapore
- Centre for Healthy Ageing, National University Healthy System, Singapore
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Unmesh Lal
- Frost and Sullivan, Frankfurt am Main, Germany
| | | | - Alexey A. Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Institute of Biology of Komi Science Center of Ural Branch of RAS, Syktyvkar, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Ivan Ozerov
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Rockville, MD 20850, USA
| | - Michael A. Petr
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Reason
- Repair Biotechnologies, Inc., Syracuse, NY 13210, USA
| | - David C. Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, The Keith Peters Building, Cambridge CB2 0XY, UK
- UK Dementia Research Institute, The Keith Peters Building, Cambridge Biomedical Campus, Cambridge, UK
| | - Alexander Tyshkovskiy
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia
| | - Quentin Vanhaelen
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Rockville, MD 20850, USA
| | - Alex Zhavoronkov
- Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Rockville, MD 20850, USA
| | - Morten Scheibye-Knudsen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
21
|
Dakik P, McAuley M, Chancharoen M, Mitrofanova D, Lozano Rodriguez ME, Baratang Junio JA, Lutchman V, Cortes B, Simard É, Titorenko VI. Pairwise combinations of chemical compounds that delay yeast chronological aging through different signaling pathways display synergistic effects on the extent of aging delay. Oncotarget 2019; 10:313-338. [PMID: 30719227 PMCID: PMC6349451 DOI: 10.18632/oncotarget.26553] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 12/20/2018] [Indexed: 01/08/2023] Open
Abstract
We have recently discovered six plant extracts that delay yeast chronological aging. Most of them affect different nodes, edges and modules of an evolutionarily conserved network of longevity regulation that integrates certain signaling pathways and protein kinases; this network is also under control of such aging-delaying chemical compounds as spermidine and resveratrol. We have previously shown that, if a strain carrying an aging-delaying single-gene mutation affecting a certain node, edge or module of the network is exposed to some of the six plant extracts, the mutation and the plant extract enhance aging-delaying efficiencies of each other so that their combination has a synergistic effect on the extent of aging delay. We therefore hypothesized that a pairwise combination of two aging-delaying plant extracts or a combination of one of these plant extracts and spermidine or resveratrol may have a synergistic effect on the extent of aging delay only if each component of this combination targets a different element of the network. To test our hypothesis, we assessed longevity-extending efficiencies of all possible pairwise combinations of the six plant extracts or of one of them and spermidine or resveratrol in chronologically aging yeast. In support of our hypothesis, we show that only pairwise combinations of naturally-occurring chemical compounds that slow aging through different nodes, edges and modules of the network delay aging in a synergistic manner.
Collapse
Affiliation(s)
- Pamela Dakik
- Department of Biology, Concordia University, Montreal, Quebec, Canada
| | - Mélissa McAuley
- Department of Biology, Concordia University, Montreal, Quebec, Canada
| | | | - Darya Mitrofanova
- Department of Biology, Concordia University, Montreal, Quebec, Canada
| | | | | | - Vicky Lutchman
- Department of Biology, Concordia University, Montreal, Quebec, Canada
| | - Berly Cortes
- Department of Biology, Concordia University, Montreal, Quebec, Canada
| | - Éric Simard
- Idunn Technologies Inc., Rosemere, Quebec, Canada
| | | |
Collapse
|
22
|
Ma S, Avanesov AS, Porter E, Lee BC, Mariotti M, Zemskaya N, Guigo R, Moskalev AA, Gladyshev VN. Comparative transcriptomics across 14 Drosophila species reveals signatures of longevity. Aging Cell 2018; 17:e12740. [PMID: 29671950 PMCID: PMC6052463 DOI: 10.1111/acel.12740] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2018] [Indexed: 12/14/2022] Open
Abstract
Lifespan varies dramatically among species, but the biological basis is not well understood. Previous studies in model organisms revealed the importance of nutrient sensing, mTOR, NAD/sirtuins, and insulin/IGF1 signaling in lifespan control. By studying life-history traits and transcriptomes of 14 Drosophila species differing more than sixfold in lifespan, we explored expression divergence and identified genes and processes that correlate with longevity. These longevity signatures suggested that longer-lived flies upregulate fatty acid metabolism, downregulate neuronal system development and activin signaling, and alter dynamics of RNA splicing. Interestingly, these gene expression patterns resembled those of flies under dietary restriction and several other lifespan-extending interventions, although on the individual gene level, there was no significant overlap with genes previously reported to have lifespan-extension effects. We experimentally tested the lifespan regulation potential of several candidate genes and found no consistent effects, suggesting that individual genes generally do not explain the observed longevity patterns. Instead, it appears that lifespan regulation across species is modulated by complex relationships at the system level represented by global gene expression.
Collapse
Affiliation(s)
- Siming Ma
- Division of GeneticsDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
- Genome Institute of SingaporeA*STARSingapore CitySingapore
| | - Andrei S. Avanesov
- Division of GeneticsDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Emily Porter
- Division of GeneticsDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Byung Cheon Lee
- Division of GeneticsDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
- College of Life Sciences and BiotechnologyKorea UniversitySeoulSouth Korea
| | - Marco Mariotti
- Division of GeneticsDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
- Bioinformatics and Genomics ProgramCentre for Genomic Regulation and Universitat Pompeu FabraBarcelonaSpain
| | - Nadezhda Zemskaya
- Institute of BiologyKomi Science CenterRussian Academy of SciencesSyktyvkarRussia
| | - Roderic Guigo
- Bioinformatics and Genomics ProgramCentre for Genomic Regulation and Universitat Pompeu FabraBarcelonaSpain
| | - Alexey A. Moskalev
- Institute of BiologyKomi Science CenterRussian Academy of SciencesSyktyvkarRussia
- Moscow Institute of Physics and TechnologyDolgoprudny, Moscow RegionRussia
- Engelhardt Institute of Molecular BiologyRussian Academy of SciencesMoscowRussia
| | - Vadim N. Gladyshev
- Division of GeneticsDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
- Belozersky Institute of Physico‐Chemical BiologyMoscow State UniversityMoscowRussia
| |
Collapse
|
23
|
Abstract
Inhibitors of mTOR, including clinically available rapalogs such as rapamycin (Sirolimus) and Everolimus, are gerosuppressants, which suppress cellular senescence. Rapamycin slows aging and extends life span in a variety of species from worm to mammals. Rapalogs can prevent age-related diseases, including cancer, atherosclerosis, obesity, neurodegeneration and retinopathy and potentially rejuvenate stem cells, immunity and metabolism. Here, I further suggest how rapamycin can be combined with metformin, inhibitors of angiotensin II signaling (Losartan, Lisinopril), statins (simvastatin, atorvastatin), propranolol, aspirin and a PDE5 inhibitor. Rational combinations of these drugs with physical exercise and an anti-aging diet (Koschei formula) can maximize their anti-aging effects and decrease side effects.
Collapse
|
24
|
Kudryavtseva A, Krasnov G, Lipatova A, Alekseev B, Maganova F, Shaposhnikov M, Fedorova M, Snezhkina A, Moskalev A. Effects of Abies sibirica terpenes on cancer- and aging-associated pathways in human cells. Oncotarget 2018; 7:83744-83754. [PMID: 27888805 PMCID: PMC5347801 DOI: 10.18632/oncotarget.13467] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 10/19/2016] [Indexed: 12/31/2022] Open
Abstract
A large number of terpenoids exhibit potential geroprotector and anti-cancer properties. Here, we studied whole transcriptomic effects of Abisil, the extract of fir (Abies sibirica) terpenes, on normal and cancer cell lines. We used early passaged and senescent none-immortalized fibroblasts as cellular aging models. It was revealed that in normal fibroblasts, terpenes induced genes of stress response, apoptosis regulation and tissue regeneration. The restoration of the expression level of some prolongevity genes after fir extract treatment was shown in old cells. In Caco-2 and AsPC-1 cancer cell lines, Abisil induced expression of both onco-suppressors (members of GADD45, DUSP, and DDIT gene families), and proto-oncogenes (c-Myc, c-Jun, EGR and others). Thus, the study demonstrates the potential anti-aging and anti-cancer effects of Abisil on senescent and cancer cell lines.
Collapse
Affiliation(s)
- Anna Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.,National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, 125284, Russia
| | - George Krasnov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Anastasiya Lipatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Boris Alekseev
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, 125284, Russia
| | | | - Mikhail Shaposhnikov
- Institute of Biology of Komi Science Center of Ural Branch of RAS, Syktyvkar, 167982, Russia
| | - Maria Fedorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Anastasiya Snezhkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Alexey Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.,Institute of Biology of Komi Science Center of Ural Branch of RAS, Syktyvkar, 167982, Russia.,Moscow Institute of Physics and Technology, Dolgoprudny, 141700, Russia
| |
Collapse
|
25
|
Abstract
Rapamycin slows organismal aging and delays age-related diseases, extending lifespan in numerous species. In cells, rapamycin and other rapalogs such as everolimus suppress geroconversion from quiescence to senescence. Rapamycin inhibits some, but not all, activities of mTOR. Recently we and others demonstrated that pan-mTOR inhibitors, known also as dual mTORC1/C2 inhibitors, suppress senescent phenotype. As a continuation of these studies, here we investigated in detail a panel of pan-mTOR inhibitors, to determine their optimal gerosuppressive concentrations. During geroconversion, cells become hypertrophic and flat, accumulate lysosomes (SA-beta-Gal staining) and lipids (Oil Red staining) and lose their re-proliferative potential (RPP). We determined optimal gerosuppressive concentrations: Torin1 (30 nM), Torin 2 (30 nM), AZD8055 (100 nM), PP242 (300 nM), both KU-006379 and GSK1059615 (1000 nM). These agents decreased senescence-associated hypertrophy with IC50s: 20, 18, 15, 200 and 400 nM, respectively. Preservation of RPP by pan-mTOR inhibitors was associated with inhibition of the pS6K/pS6 axis. Inhibition of rapamycin-insensitive functions of mTOR further contributed to anti-hypertrophic and cytostatic effects. Torin 1 and PP242 were more "rapamycin-like" than Torin 2 and AZD8055. Pan-mTOR inhibitors were superior to rapamycin in suppressing hypertrophy, senescent morphology, Oil Red O staining and in increasing so-called "chronological life span (CLS)". We suggest that, at doses lower than anti-cancer concentrations, pan-mTOR inhibitors can be developed as anti-aging drugs.
Collapse
|
26
|
Aliper A, Jellen L, Cortese F, Artemov A, Karpinsky-Semper D, Moskalev A, Swick AG, Zhavoronkov A. Towards natural mimetics of metformin and rapamycin. Aging (Albany NY) 2017; 9:2245-2268. [PMID: 29165314 PMCID: PMC5723685 DOI: 10.18632/aging.101319] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 11/02/2017] [Indexed: 12/14/2022]
Abstract
Aging is now at the forefront of major challenges faced globally, creating an immediate need for safe, widescale interventions to reduce the burden of chronic disease and extend human healthspan. Metformin and rapamycin are two FDA-approved mTOR inhibitors proposed for this purpose, exhibiting significant anti-cancer and anti-aging properties beyond their current clinical applications. However, each faces issues with approval for off-label, prophylactic use due to adverse effects. Here, we initiate an effort to identify nutraceuticals-safer, naturally-occurring compounds-that mimic the anti-aging effects of metformin and rapamycin without adverse effects. We applied several bioinformatic approaches and deep learning methods to the Library of Integrated Network-based Cellular Signatures (LINCS) dataset to map the gene- and pathway-level signatures of metformin and rapamycin and screen for matches among over 800 natural compounds. We then predicted the safety of each compound with an ensemble of deep neural network classifiers. The analysis revealed many novel candidate metformin and rapamycin mimetics, including allantoin and ginsenoside (metformin), epigallocatechin gallate and isoliquiritigenin (rapamycin), and withaferin A (both). Four relatively unexplored compounds also scored well with rapamycin. This work revealed promising candidates for future experimental validation while demonstrating the applications of powerful screening methods for this and similar endeavors.
Collapse
Affiliation(s)
- Alexander Aliper
- Insilico Medicine, Inc, Research Department, Baltimore, MD 21218, USA
| | - Leslie Jellen
- Insilico Medicine, Inc, Research Department, Baltimore, MD 21218, USA
| | - Franco Cortese
- Biogerontology Research Foundation, Research Department, Oxford, United Kingdom
- Department of Biomedical and Molecular Science, Queen's University School of Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Artem Artemov
- Insilico Medicine, Inc, Research Department, Baltimore, MD 21218, USA
| | | | - Alexey Moskalev
- Laboratory of Molecular Radiobiology and Gerontology, Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences, Syktyvkar, 167982, Russia
| | | | - Alex Zhavoronkov
- Insilico Medicine, Inc, Research Department, Baltimore, MD 21218, USA
- Biogerontology Research Foundation, Research Department, Oxford, United Kingdom
| |
Collapse
|
27
|
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
|
28
|
Moskalev A, Chernyagina E, Kudryavtseva A, Shaposhnikov M. Geroprotectors: A Unified Concept and Screening Approaches. Aging Dis 2017; 8:354-363. [PMID: 28580190 PMCID: PMC5440114 DOI: 10.14336/ad.2016.1022] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 10/22/2016] [Indexed: 12/20/2022] Open
Abstract
Although the geroprotectors discovery is a new biomedicine trend and more than 200 compounds can slow aging and increase the lifespan of the model organism, there are still no geroprotectors on the market. The reasons may be partly related to the lack of a unified concept of geroprotector, accepted by the scientific community. Such concept as a system of criteria for geroprotector identification and classification can form a basis for an analytical model of anti-aging drugs, help to consolidate the efforts of various research initiatives in this area and compare their results. Here, we review the existing classification and characteristics of geroprotectors based on their effect on the survival of a group of individuals or pharmaceutics classes, according to the proposed mechanism of their geroprotective action or theories of aging. After discussing advantages and disadvantages of these approaches, we offer a new concept based on the maintenance of homeostatic capacity because aging can be considered as exponential shrinkage of homeostatic capacity leading to the onset of age-related diseases and death. Besides, we review the most promising current screening approaches to finding new geroprotectors. Establishing the classification of existing geroprotectors based on physiology and current understanding of the nature of aging is essential for putting the existing knowledge into a single system. This system could be useful to formulate standards for finding and creating new geroprotectors. Standardization, in turn, would allow easier comparison and combination of experimental data obtained by different research groups.
Collapse
Affiliation(s)
- Alexey Moskalev
- 1Laboratory of postgenomic studies, Engelhardt Institute of Molecular Biology of Russian Academy of Sciences, Moscow, 119991, Russia.,2Laboratory of genetics of aging and longevity, Moscow Institute of Physics and Technology, Dolgoprudny, 141700, Russia.,3Laboratory of molecular radiobiology and gerontology, Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences, Syktyvkar, 167982, Russia
| | - Elizaveta Chernyagina
- 2Laboratory of genetics of aging and longevity, Moscow Institute of Physics and Technology, Dolgoprudny, 141700, Russia
| | - Anna Kudryavtseva
- 1Laboratory of postgenomic studies, Engelhardt Institute of Molecular Biology of Russian Academy of Sciences, Moscow, 119991, Russia
| | - Mikhail Shaposhnikov
- 3Laboratory of molecular radiobiology and gerontology, Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences, Syktyvkar, 167982, Russia
| |
Collapse
|
29
|
Cheng L, Zhang S, Hu Y. BLAT2DOLite: An Online System for Identifying Significant Relationships between Genetic Sequences and Diseases. PLoS One 2016; 11:e0157274. [PMID: 27315278 PMCID: PMC4912091 DOI: 10.1371/journal.pone.0157274] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/26/2016] [Indexed: 11/18/2022] Open
Abstract
The significantly related diseases of sequences could play an important role in understanding the functions of these sequences. In this paper, we introduced BLAT2DOLite, an online system for annotating human genes and diseases and identifying the significant relationships between sequences and diseases. Currently, BLAT2DOLite integrates Entrez Gene database and Disease Ontology Lite (DOLite), which contain loci of gene and relationships between genes and diseases. It utilizes hypergeometric test to calculate P-values between genes and diseases of DOLite. The system can be accessed from: http://123.59.132.21:8080/BLAT2DOLite. The corresponding web service is described in: http://123.59.132.21:8080/BLAT2DOLite/BLAT2DOLiteIDMappingPort?wsdl.
Collapse
Affiliation(s)
- Liang Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, PR China
- * E-mail: (LC); (YH)
| | - Shuo Zhang
- School of Management, Harbin University of Commerce, Harbin 150028, PR China
| | - Yang Hu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, PR China
- * E-mail: (LC); (YH)
| |
Collapse
|
30
|
Moskalev A, Chernyagina E, Tsvetkov V, Fedintsev A, Shaposhnikov M, Krut'ko V, Zhavoronkov A, Kennedy BK. Developing criteria for evaluation of geroprotectors as a key stage toward translation to the clinic. Aging Cell 2016; 15:407-15. [PMID: 26970234 PMCID: PMC4854916 DOI: 10.1111/acel.12463] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2016] [Indexed: 01/15/2023] Open
Abstract
In the coming decades, a massive shift in the aging segment of the population will have major social and economic consequences around the world. One way to offset this increase is to expedite the development of geroprotectors, substances that slow aging, repair age‐associated damage and extend healthy lifespan, or healthspan. While over 200 geroprotectors are now reported in model organisms and some are in human use for specific disease indications, the path toward determining whether they affect aging in humans remains obscure. Translation to the clinic is hampered by multiple issues including absence of a common set of criteria to define, select, and classify these substances, given the complexity of the aging process and their enormous diversity in mechanism of action. Translational research efforts would benefit from the formation of a scientific consensus on the following: the definition of ‘geroprotector’, the selection criteria for geroprotectors, a comprehensive classification system, and an analytical model. Here, we review current approaches to selection and put forth our own suggested selection criteria. Standardizing selection of geroprotectors will streamline discovery and analysis of new candidates, saving time and cost involved in translation to clinic.
Collapse
Affiliation(s)
- Alexey Moskalev
- Engelhardt Institute of Molecular Biology of Russian Academy of Sciences Moscow 119991 Russia
- Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences Syktyvkar 167982 Russia
- Moscow Institute of Physics and Technology Dolgoprudny 141700 Russia
| | | | - Vasily Tsvetkov
- Moscow Institute of Physics and Technology Dolgoprudny 141700 Russia
- The Research Institute for Translational Medicine Pirogov Russian National Research Medical University Moscow 117997 Russia
| | - Alexander Fedintsev
- Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences Syktyvkar 167982 Russia
| | - Mikhail Shaposhnikov
- Engelhardt Institute of Molecular Biology of Russian Academy of Sciences Moscow 119991 Russia
| | - Vyacheslav Krut'ko
- Institute for Systems Analysis Russian Academy of Sciences Moscow 117312 Russia
| | - Alex Zhavoronkov
- Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences Syktyvkar 167982 Russia
- D. Rogachev FRC Center for Pediatric Hematology, Oncology and Immunology Samory Machela 1 Moscow 117997 Russia
- The Biogerontology Research Foundation 2354 Chynoweth House, Trevissome Park, Blackwater, Truro Cornwall TR4 8UN UK
| | | |
Collapse
|
31
|
Danilov A, Shaposhnikov M, Shevchenko O, Zemskaya N, Zhavoronkov A, Moskalev A. Influence of non-steroidal anti-inflammatory drugs on Drosophila melanogaster longevity. Oncotarget 2016; 6:19428-44. [PMID: 26305987 PMCID: PMC4637296 DOI: 10.18632/oncotarget.5118] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 07/17/2015] [Indexed: 02/07/2023] Open
Abstract
Most age-related diseases and aging itself are associated with chronic inflammation. Thus pharmacological inhibition of inflammatory processes may be effective antiaging strategy. In this study we demonstrated that treatment of Drosophila melanogaster with 10 non-steroidal anti-inflammatory drugs (NSAIDs: CAY10404, aspirin, APHS, SC-560, NS-398, SC-58125, valeroyl salicylate, trans-resveratrol, valdecoxib, licofelone) leads to extension of lifespan, delays age-dependent decline of locomotor activity and increases stress resistance. The effect of the lifespan increase was associated with decrease of fecundity. Depending on the concentration, NSAIDs demonstrated both anti- and pro-oxidant properties in Drosophila tissues. However, we failed to identify clear correlation between antioxidant properties of NSAIDs and their pro-longevity effects. The lifespan extending effects of APHS, SC-58125, valeroyl salicylate, trans-resveratrol, valdecoxib, and licofelone were more pronounced in males, valdecoxib and aspirin - in females. We demonstrated that lifespan extension effect of NSAIDs was abolished in flies with defective genes involved in Pkh2-ypk1-lem3-tat2 pathway.
Collapse
Affiliation(s)
- Anton Danilov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Institute of Biology of Komi Science Center of Ural Branch of RAS, Syktyvkar, Russia
| | - Mikhail Shaposhnikov
- Institute of Biology of Komi Science Center of Ural Branch of RAS, Syktyvkar, Russia.,Syktyvkar State University, Syktyvkar, Russia
| | - Oksana Shevchenko
- Institute of Biology of Komi Science Center of Ural Branch of RAS, Syktyvkar, Russia
| | - Nadezhda Zemskaya
- Institute of Biology of Komi Science Center of Ural Branch of RAS, Syktyvkar, Russia
| | - Alex Zhavoronkov
- Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Alexey Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,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, Dolgoprudny, Russia
| |
Collapse
|
32
|
Caulin AF, Graham TA, Wang LS, Maley CC. Solutions to Peto's paradox revealed by mathematical modelling and cross-species cancer gene analysis. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0222. [PMID: 26056366 PMCID: PMC4581027 DOI: 10.1098/rstb.2014.0222] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Whales have 1000-fold more cells than humans and mice have 1000-fold fewer; however, cancer risk across species does not increase with the number of somatic cells and the lifespan of the organism. This observation is known as Peto's paradox. How much would evolution have to change the parameters of somatic evolution in order to equalize the cancer risk between species that differ by orders of magnitude in size? Analysis of previously published models of colorectal cancer suggests that a two- to three-fold decrease in the mutation rate or stem cell division rate is enough to reduce a whale's cancer risk to that of a human. Similarly, the addition of one to two required tumour-suppressor gene mutations would also be sufficient. We surveyed mammalian genomes and did not find a positive correlation of tumour-suppressor genes with increasing body mass and longevity. However, we found evidence of the amplification of TP53 in elephants, MAL in horses and FBXO31 in microbats, which might explain Peto's paradox in those species. Exploring parameters that evolution may have fine-tuned in large, long-lived organisms will help guide future experiments to reveal the underlying biology responsible for Peto's paradox and guide cancer prevention in humans.
Collapse
Affiliation(s)
- Aleah F Caulin
- Genomics and Computational Biology Graduate Program, University of Pennsylvania, Philadelphia, PA 19103, USA
| | - Trevor A Graham
- Evolution and Cancer Laboratory, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Li-San Wang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19103, USA
| | - Carlo C Maley
- Biodesign Institute, School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA Center for Evolution and Cancer, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
33
|
Noce A, Canale MP, Capria A, Rovella V, Tesauro M, Splendiani G, Annicchiarico-Petruzzelli M, Manzuoli M, Simonetti G, Di Daniele N. Coronary artery calcifications predict long term cardiovascular events in non diabetic Caucasian hemodialysis patients. Aging (Albany NY) 2016; 7:269-79. [PMID: 26131456 PMCID: PMC4429091 DOI: 10.18632/aging.100740] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Vascular calcifications are frequent in chronic renal disease and are associated to significant cardiovascular morbidity and mortality. The long term predictive value of coronary artery calcifications detected by multi-layer spiral computed tomography for major cardiovascular events was evaluated in non-diabetic Caucasian patients on maintenance hemodialysis free of clinical cardiovascular disease. Two-hundred and five patients on maintenance hemodialysis were enrolled into this observational, prospective cohort study. Patients underwent a single cardiac multi-layer spiral computed tomography. Calcium load was quantified and patients grouped according to the Agatston score: group 1 (Agatston score: 0), group 2 (Agatston score 1-400), group 3 (Agatston score 401-1000) and group 4 (Agatston score >1000). Follow-up was longer than seven years. Primary endpoint was death from a major cardiovascular event. Actuarial survival was calculated separately in the four groups with Kaplan-Meier method. Patients who died from causes other than cardiovascular disease and transplanted patients were censored. The “log rank” test was employed to compare survival curves. One-hundred two patients (49.7%) died for a major cardiovascular event during the follow-up period. Seven-year actuarial survival was more than 90% for groups 1 and 2, but failed to about 50% for group 3 and to <10% for group 4. Hence, Agatston score >400 predicts a significantly higher cardiovascular mortality compared with Agatston score <400 (p<0.0001); furthermore, serum Parathyroid hormone levels > 300 pg/l were associated to a lower survival (p < 0.05). Extended coronary artery calcifications detected by cardiac multi-layer spiral computed tomography, strongly predicted long term cardiovascular mortality in non-diabetic Caucasian patients on maintenance hemodialysis. Moreover, it was not related to conventional indices of atherosclerosis, but to other non-traditional risk factors, as serum Parathyroid hormone levels. A full cost-benefit analysis is however necessary to justify a widespread use of cardiac multi-layer spiral computed tomography in clinical practice.
Collapse
|
34
|
Lashmanova E, Proshkina E, Zhikrivetskaya S, Shevchenko O, Marusich E, Leonov S, Melerzanov A, Zhavoronkov A, Moskalev A. Fucoxanthin increases lifespan of Drosophila melanogaster and Caenorhabditis elegans. Pharmacol Res 2015; 100:228-41. [DOI: 10.1016/j.phrs.2015.08.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 08/07/2015] [Accepted: 08/13/2015] [Indexed: 12/19/2022]
|
35
|
Abstract
The most physiological type of cell cycle arrest - namely, contact inhibition in dense culture - is the least densely studied. Despite cell cycle arrest, confluent cells do not become senescent. We recently described that mTOR (target of rapamycin) is inactive in contact-inhibited cells. Therefore, conversion from reversible arrest to senescence (geroconversion) is suppressed. I this Perspective, we further extended the gerosuppression model. While causing senescence in regular cell density, etoposide failed to cause senescence in contact-inhibited cells. A transient reactivation of mTOR favored geroconversion in etoposide-treated confluent cells. Like p21, p16 did not cause senescence in high cell density. We discuss that suppression of geroconversion in confluent and contact-inhibited cultures mimics gerosuppression in the organism. We confirmed that levels of p-S6 were low in murine tissues in the organism compared with mouse embryonic fibroblasts in cell culture, whereas p-Akt was reciprocally high in the organism.
Collapse
Affiliation(s)
- Olga V Leontieva
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Elms and Carlson Streets, Buffalo, NY 14263, USA
| | - Mikhail V Blagosklonny
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Elms and Carlson Streets, Buffalo, NY 14263, USA
| |
Collapse
|
36
|
Abstract
Here we discuss the latest progress in development of some kinase inhibitors such as inhibitors of c-MET, LIM and Bcr-Abl kinases. Importantly, many oncogenic kinases signal via the mTOR pathway, suggesting a common target for drug combinations.
Collapse
|
37
|
Zhavoronkov A, Buzdin AA, Garazha AV, Borisov NM, Moskalev AA. Signaling pathway cloud regulation for in silico screening and ranking of the potential geroprotective drugs. Front Genet 2014; 5:49. [PMID: 24624136 PMCID: PMC3940060 DOI: 10.3389/fgene.2014.00049] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 02/16/2014] [Indexed: 12/24/2022] Open
Abstract
The major challenges of aging research include absence of the comprehensive set of aging biomarkers, the time it takes to evaluate the effects of various interventions on longevity in humans and the difficulty extrapolating the results from model organisms to humans. To address these challenges we propose the in silico method for screening and ranking the possible geroprotectors followed by the high-throughput in vivo and in vitro validation. The proposed method evaluates the changes in the collection of activated or suppressed signaling pathways involved in aging and longevity, termed signaling pathway cloud, constructed using the gene expression data and epigenetic profiles of young and old patients' tissues. The possible interventions are selected and rated according to their ability to regulate age-related changes and minimize differences in the signaling pathway cloud. While many algorithmic solutions to simulating the induction of the old into young metabolic profiles in silico are possible, this flexible and scalable approach may potentially be used to predict the efficacy of the many drugs that may extend human longevity before conducting pre-clinical work and expensive clinical trials.
Collapse
Affiliation(s)
- Alex Zhavoronkov
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology Dolgoprudny, Russia ; First Oncology Research and Advisory Center Moscow, Russia ; The Biogerontology Research Foundation London, UK ; Department of Experimental and Molecular Medicine, D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology Moscow, Russia
| | - Anton A Buzdin
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology Dolgoprudny, Russia ; First Oncology Research and Advisory Center Moscow, Russia ; Department of Experimental and Molecular Medicine, D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology Moscow, Russia ; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences Moscow, Russia
| | - Andrey V Garazha
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology Dolgoprudny, Russia ; First Oncology Research and Advisory Center Moscow, Russia ; Department of Experimental and Molecular Medicine, D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology Moscow, Russia ; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences Moscow, Russia
| | - Nikolay M Borisov
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology Dolgoprudny, Russia ; First Oncology Research and Advisory Center Moscow, Russia ; Burnasyan Federal Medical Biophysical Center Moscow, Russia
| | - Alexey A Moskalev
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology Dolgoprudny, Russia ; Department of Ecology, Syktyvkar State University Syktyvkar, Russia ; Laboratory of Molecular Radiobiology and Gerontology, Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences Syktyvkar, Russia
| |
Collapse
|
38
|
Abstract
Recent groundbreaking discoveries have revealed that IGF-1, Ras, MEK, AMPK, TSC1/2, FOXO, PI3K, mTOR, S6K, and NFκB are involved in the aging process. This is remarkable because the same signaling molecules, oncoproteins and tumor suppressors, are well-known targets for cancer therapy. Furthermore, anti-cancer drugs aimed at some of these targets have been already developed. This arsenal could be potentially employed for anti-aging interventions (given that similar signaling molecules are involved in both cancer and aging). In cancer, intrinsic and acquired resistance, tumor heterogeneity, adaptation, and genetic instability of cancer cells all hinder cancer-directed therapy. But for anti-aging applications, these hurdles are irrelevant. For example, since anti-aging interventions should be aimed at normal postmitotic cells, no selection for resistance is expected. At low doses, certain agents may decelerate aging and age-related diseases. Importantly, deceleration of aging can in turn postpone cancer, which is an age-related disease.
Collapse
|
39
|
Blagosklonny MV. Immunosuppressants in cancer prevention and therapy. Oncoimmunology 2013; 2:e26961. [PMID: 24575379 PMCID: PMC3926869 DOI: 10.4161/onci.26961] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 10/25/2013] [Accepted: 10/25/2013] [Indexed: 12/13/2022] Open
Abstract
Rapalogs such as rapamycin (sirolimus), everolimus, temserolimus, and deforolimus are indicated for the treatment of some malignancies. Rapamycin is the most effective cancer-preventive agent currently known, at least in mice, dramatically delaying carcinogenesis in both normal and cancer-prone murine strains. In addition, rapamycin and everolimus decrease the risk of cancer in patients receiving these drugs in the context of immunosuppressive regimens. In general, the main concern about the use of immunosuppressants in humans is an increased risk of cancer. Given that rapalogs are useful in cancer prevention and therapy, should they be viewed as immunosuppressants or immunostimulators? Or should we reconsider the role of immunity in cancer altogether? In addition to its anti-viral, anti-inflammatory, anti-angiogenic and anti-proliferative effects, rapamycin operates as a gerosuppressant, meaning that it inhibits the cellular conversion to a senescent state (the so-called geroconversion), a fundamental process involved in aging and age-related pathologies including cancer.
Collapse
|