51
|
Protective effect of Vigna unguiculata extract against aging and neurodegeneration. Aging (Albany NY) 2020; 12:19785-19808. [PMID: 33024055 PMCID: PMC7732273 DOI: 10.18632/aging.104069] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023]
Abstract
Aging and age-related neurodegeneration are among the major challenges in modern medicine because of the progressive increase in the number of elderly in the world population. Nutrition, which has important long-term consequences for health, is an important way to prevent diseases and achieve healthy aging. The beneficial effects of Vigna unguiculata on metabolic disorders have been widely documented. Here, we show that an aqueous extract of V. unguiculata beans delays senescence both in Saccharomyces cerevisiae and Drosophila melanogaster, in a Snf1/AMPK-dependent manner. Consistently, an increased expression of FOXO, SIRT1, NOTCH and heme oxygenase (HO) genes, already known to be required for the longevity extension in D. melanogaster, is also shown. Preventing α-synuclein self-assembly is one of the most promising approaches for the treatment of Parkinson's disease (PD), for which aging is a risk factor. In vitro aggregation of α-synuclein, its toxicity and membrane localization in yeast and neuroblastoma cells are strongly decreased in the presence of bean extract. In a Caenorhabditis elegans model of PD, V. unguiculata extract substantially reduces the number of the age-dependent degeneration of the cephalic dopaminergic neurons. Our findings support the role of V. unguiculata beans as a functional food in age-related disorders.
Collapse
|
52
|
Xie K, Kapetanou M, Sidiropoulou K, Bano D, Gonos ES, Djordjevic AM, Ehninger D. Signaling pathways of dietary energy restriction and metabolism on brain physiology and in age-related neurodegenerative diseases. Mech Ageing Dev 2020; 192:111364. [PMID: 32991920 DOI: 10.1016/j.mad.2020.111364] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/17/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
Several laboratory animal models have shown that dietary energy restriction (ER) can promote longevity and improve various health aspects in old age. However, whether the entire spectrum of ER-induced short- and long-term physiological and metabolic adaptions is translatable to humans remains to be determined. In this review article, we present recent evidence towards the elucidation of the impact of ER on brain physiology and in age-related neurodegenerative diseases. We also discuss modulatory influences of ER on metabolism and overall on human health, limitations of current experimental designs as well as future perspectives for ER trials in humans. Finally, we summarize signaling pathways and processes known to be affected by both aging and ER with a special emphasis on the link between ER and cellular proteostasis.
Collapse
Affiliation(s)
- Kan Xie
- Molecular and Cellular Cognition Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany
| | - Marianna Kapetanou
- Institute of Chemical Biology, National Hellenic Research Foundation, 48 Vassileos Constantinou Ave., Athens, 11635, Greece
| | | | - Daniele Bano
- Aging and Neurodegeneration Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany
| | - Efstathios S Gonos
- Institute of Chemical Biology, National Hellenic Research Foundation, 48 Vassileos Constantinou Ave., Athens, 11635, Greece
| | - Aleksandra Mladenovic Djordjevic
- Department of Neurobiology, Institute for Biological Research 'Sinisa Stankovic', University of Belgrade, National Institute of Republic of Serbia, Boulevard Despota Stefana 142, 11000 Belgrade, Serbia
| | - Dan Ehninger
- Molecular and Cellular Cognition Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany.
| |
Collapse
|
53
|
Jia F, Diao P, Wang X, Hu X, Kimura T, Nakamuta M, Nakamura I, Shirotori S, Sato Y, Moriya K, Koike K, Gonzalez FJ, Nakayama J, Aoyama T, Tanaka N. Dietary Restriction Suppresses Steatosis-Associated Hepatic Tumorigenesis in Hepatitis C Virus Core Gene Transgenic Mice. Liver Cancer 2020; 9:529-548. [PMID: 33083279 PMCID: PMC7548900 DOI: 10.1159/000508308] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/24/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND AND AIMS Dietary restriction (DR) is a preventive strategy for obesity, metabolic syndrome, cardiovascular disease, and diabetes. Although an interconnection between obesity, metabolic syndrome, fatty liver, and hepatocellular carcinoma has been documented, the mechanism and impact of DR on steatosis-derived hepatocarcinogenesis are not fully understood. This study aimed to evaluate whether DR can prevent hepatic tumorigenesis. METHODS Male hepatitis C virus core gene transgenic (HCVcpTg) mice that develop spontaneous age-dependent insulin resistance, hepatic steatosis, and ensuing liver tumor development without apparent hepatic fibrosis, were fed with either a control diet ad libitum (control group) or 70% of the same control diet (DR group) for 15 months, and liver phenotypes were investigated. RESULTS DR significantly reduced the number and volume of liver tumors. DR attenuated hepatic oxidative and endoplasmic reticulum stress and markedly suppressed nuclear factor-κB, signal transducer and activator of transcription 3 (STAT3) and STAT5, and phosphorylation of extracellular signal-regulated kinase, leading to downregulation of several pro-oncogenic mediators, such as cyclin D1. Serum insulin and insulin-like growth factor 1 levels, as well as hepatic expression of insulin receptor substrate 1/2, phosphatidylinositol-3 kinase, and serine/threonine-protein kinase AKT, were downregulated by DR. A transcriptome analysis revealed that STAT3 signaling and lipogenesis were the most suppressed hepatocarcinogenic pathways affected by DR. Additionally, DR stimulated autophagy and p62/sequestosome 1 degradation, enhanced phosphorylation of AMP-activated protein kinase α, increased fibroblast growth factor 21 expression, and attenuated expression of senescence-associated secretory phenotypes. CONCLUSION DR suppressed steatosis-associated hepatic tumorigenesis in HCVcpTg mice, mainly due to attenuation of pathways involved in inflammation, cellular stress, cell proliferation, insulin signaling, and senescence. These findings support the notion that persistent 30% reduction of daily food intake is beneficial for preventing steatosis-associated hepatocarcinogenesis caused by HCV core protein.
Collapse
Affiliation(s)
- Fangping Jia
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, Japan
| | - Pan Diao
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, Japan
| | - Xiaojing Wang
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, Japan,Department of Gastroenterology, Lishui Hospital, Zhejiang University School of Medicine, Lishui, China
| | - Xiao Hu
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, Japan,Department of Pathophysiology, Hebei Medical University, Shijiazhuang, China
| | - Takefumi Kimura
- Department of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Makoto Nakamuta
- Department of Gastroenterology, Kyushu Medical Center, Fukuoka, Japan
| | - Ibuki Nakamura
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, Japan
| | - Saki Shirotori
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yoshiko Sato
- Department of Molecular Pathology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Kyoji Moriya
- Department of Infection Control and Prevention, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
| | - Frank J. Gonzalez
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jun Nakayama
- Department of Molecular Pathology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Toshifumi Aoyama
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, Japan
| | - Naoki Tanaka
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, Japan,Research Center for Social Systems, Shinshu University, Matsumoto, Japan,*Naoki Tanaka, Department of Metabolic Regulation, Shinshu University School of Medicine, Asahi 3-1-1, Matsumoto 390-8621 (Japan),
| |
Collapse
|
54
|
Dahan L, Rampon C, Florian C. Age-related memory decline, dysfunction of the hippocampus and therapeutic opportunities. Prog Neuropsychopharmacol Biol Psychiatry 2020; 102:109943. [PMID: 32298784 DOI: 10.1016/j.pnpbp.2020.109943] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 04/01/2020] [Accepted: 04/01/2020] [Indexed: 12/13/2022]
Abstract
While the aging of the population is a sign of progress for societies, it also carries its load of negative aspects. Among them, cognitive decline and in particular memory loss is a common feature of non-pathological aging. Autobiographical memories, which rely on the hippocampus, are a primary target of age-related cognitive decline. Here, focusing on the neurobiological mechanisms of memory formation and storage, we describe how hippocampal functions are altered across time in non-pathological mammalian brains. Several hallmarks of aging have been well described over the last decades; among them, we consider altered synaptic communication and plasticity, reduction of adult neurogenesis and epigenetic alterations. Building on the neurobiological processes of cognitive aging that have been identified to date, we review some of the strategies based on lifestyle manupulation allowing to address age-related cognitive deficits.
Collapse
Affiliation(s)
- Lionel Dahan
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, Toulouse Cedex 9, France
| | - Claire Rampon
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, Toulouse Cedex 9, France
| | - Cédrick Florian
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, Toulouse Cedex 9, France.
| |
Collapse
|
55
|
Speakman JR. Why does caloric restriction increase life and healthspan? The 'clean cupboards' hypothesis. Natl Sci Rev 2020; 7:1153-1156. [PMID: 34692140 PMCID: PMC8288867 DOI: 10.1093/nsr/nwaa078] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The disposable soma hypothesis explanation of the effects of caloric restriction (CR) on lifespan fails to explain why CR generates negative impacts alongside the positive effects and does not work in all species. I propose here a novel idea called the clean cupboards hypothesis which overcomes these problems.
Collapse
Affiliation(s)
- John R Speakman
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, China
- Institute of Biological and Environmental Sciences, University of Aberdeen, UK
- CAS Center of Excellence in Animal Evolution and Genetics, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| |
Collapse
|
56
|
Martel J, Ojcius DM, Wu CY, Peng HH, Voisin L, Perfettini JL, Ko YF, Young JD. Emerging use of senolytics and senomorphics against aging and chronic diseases. Med Res Rev 2020; 40:2114-2131. [PMID: 32578904 DOI: 10.1002/med.21702] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 06/04/2020] [Accepted: 06/12/2020] [Indexed: 12/20/2022]
Abstract
Senescence is a state of cell cycle arrest that plays an important role in embryogenesis, wound healing and protection against cancer. Senescent cells also accumulate during aging and contribute to the development of age-related disorders and chronic diseases, such as atherosclerosis, type 2 diabetes, osteoarthritis, idiopathic pulmonary fibrosis, and liver disease. Molecules that induce apoptosis of senescent cells, such as dasatinib, quercetin, and fisetin, produce health benefits and extend lifespan in animal models. We describe here the mechanism of action of senolytics and senomorphics, many of which are derived from plants and fungi. We also discuss the possibility of using such compounds to delay aging and treat chronic diseases in humans.
Collapse
Affiliation(s)
- Jan Martel
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan, Republic of China.,Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan, Republic of China
| | - David M Ojcius
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan, Republic of China.,Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan, Republic of China.,Department of Biomedical Sciences, Arthur Dugoni School of Dentistry, University of the Pacific, San Francisco, California
| | - Cheng-Yeu Wu
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan, Republic of China.,Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan, Republic of China.,Research Center of Bacterial Pathogenesis, Chang Gung University, Taoyuan, Taiwan, Republic of China
| | - Hsin-Hsin Peng
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan, Republic of China.,Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan, Republic of China.,Laboratory Animal Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan, Republic of China
| | - Laurent Voisin
- Institut Gustave Roussy, INSERM U1030, Université Paris-Sud, Villejuif, France
| | - Jean-Luc Perfettini
- Department of Biomedical Sciences, Arthur Dugoni School of Dentistry, University of the Pacific, San Francisco, California.,Institut Gustave Roussy, INSERM U1030, Université Paris-Sud, Villejuif, France
| | - Yun-Fei Ko
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan, Republic of China.,Chang Gung Biotechnology Corporation, Taipei, Taiwan, Republic of China.,Biochemical Engineering Research Center, Ming Chi University of Technology, New Taipei City, Taiwan, Republic of China
| | - John D Young
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan, Republic of China.,Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan, Republic of China.,Chang Gung Biotechnology Corporation, Taipei, Taiwan, Republic of China.,Biochemical Engineering Research Center, Ming Chi University of Technology, New Taipei City, Taiwan, Republic of China
| |
Collapse
|
57
|
Arun P, Rossetti F, Wilder DM, Sajja S, Van Albert SA, Wang Y, Gist ID, Long JB. Blast Exposure Leads to Accelerated Cellular Senescence in the Rat Brain. Front Neurol 2020; 11:438. [PMID: 32508743 PMCID: PMC7253679 DOI: 10.3389/fneur.2020.00438] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/24/2020] [Indexed: 12/22/2022] Open
Abstract
Blast-induced traumatic brain injury (bTBI) is one of the major causes of persistent disabilities in Service Members, and a history of bTBI has been identified as a primary risk factor for developing age-associated neurodegenerative diseases. Clinical observations of several military blast casualties have revealed a rapid age-related loss of white matter integrity in the brain. In the present study, we have tested the effect of single and tightly coupled repeated blasts on cellular senescence in the rat brain. Isoflurane-anesthetized rats were exposed to either a single or 2 closely coupled blasts in an advanced blast simulator. Rats were euthanized and brains were collected at 24 h, 1 month and 1 year post-blast to determine senescence-associated-β-galactosidase (SA-β-gal) activity in the cells using senescence marker stain. Single and repeated blast exposures resulted in significantly increased senescence marker staining in several neuroanatomical structures, including cortex, auditory cortex, dorsal lateral thalamic nucleus, geniculate nucleus, superior colliculus, ventral thalamic nucleus and hippocampus. In general, the increases in SA-β-gal activity were more pronounced at 1 month than at 24 h or 1 year post-blast and were also greater after repeated than single blast exposures. Real-time quantitative RT-PCR analysis revealed decreased levels of mRNA for senescence marker protein-30 (SMP-30) and increased mRNA levels for p21 (cyclin dependent kinase inhibitor 1A, CDKN1A), two other related protein markers of cellular senescence. The increased senescence observed in some of these affected brain structures may be implicated in several long-term sequelae after exposure to blast, including memory disruptions and impairments in movement, auditory and ocular functions.
Collapse
Affiliation(s)
- Peethambaran Arun
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Franco Rossetti
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Donna M Wilder
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Sujith Sajja
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Stephen A Van Albert
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Ying Wang
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Irene D Gist
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Joseph B Long
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| |
Collapse
|
58
|
Basisty N, Kale A, Patel S, Campisi J, Schilling B. The power of proteomics to monitor senescence-associated secretory phenotypes and beyond: toward clinical applications. Expert Rev Proteomics 2020; 17:297-308. [PMID: 32425074 DOI: 10.1080/14789450.2020.1766976] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Cellular senescence is a rapidly growing field with potential relevance for the treatment of multiple human diseases. In the last decade, cellular senescence and the senescence-associated secretory phenotype (SASP) have emerged as central drivers of aging and many chronic diseases, including cancer, neurodegeneration, heart disease and osteoarthritis. Major efforts are underway to develop drugs that selectively eliminate senescent cells (senolytics) or alter the SASP (senomorphics) to treat age-related diseases in humans. The translation of senescence-targeting therapies into humans is still in early stages. Nonetheless, it is clear that proteomic approaches will facilitate the discovery of important SASP proteins, development of senescence- and SASP-derived biomarkers, and identification of therapeutic targets for senolytic and senomorphic drugs. AREAS COVERED We review recent proteomic studies of cellular senescence and their translational relevance and, particularly, characterization of the secretory phenotype and preclinical development of biomarkers (from 2008-2020, PubMed). We focus on emerging areas, such as the heterogeneity of senescent cells and the SASP, extracellular vesicles released by senescent cells, and validating biomarkers of aging in vivo. EXPERT OPINION Proteomic and multi-omic approaches will be important for the development of senescence-based biomarkers to facilitate and monitor future therapeutic interventions that target senescent cells.
Collapse
Affiliation(s)
- Nathan Basisty
- Buck Institute for Research on Aging, Novato , California, USA
| | - Abhijit Kale
- Buck Institute for Research on Aging, Novato , California, USA
| | - Sandip Patel
- Buck Institute for Research on Aging, Novato , California, USA
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato , California, USA.,Lawrence Berkeley National Laboratory, University of California , Berkeley, USA
| | | |
Collapse
|
59
|
Lubberts S, Meijer C, Demaria M, Gietema JA. Early ageing after cytotoxic treatment for testicular cancer and cellular senescence: Time to act. Crit Rev Oncol Hematol 2020; 151:102963. [PMID: 32446180 DOI: 10.1016/j.critrevonc.2020.102963] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/14/2020] [Accepted: 04/14/2020] [Indexed: 12/15/2022] Open
Abstract
Treatment of testicular cancer (TC) has an exceptionally high success rate compared to other cancer types; even in case of metastasized disease, 80-90 % of TC patients can be cured. Consequently, attention has been drawn to a potential downside of this treatment success: late adverse treatment effects such as the accelerated development of otherwise age-associated features like cardiovascular disease and second malignancies. Underlying mechanisms are poorly understood. Emerging data suggest that cytotoxic treatment induces cellular senescence, resulting in secretion of inflammatory factors contributing to this early ageing phenotype. Molecular and cellular characterization of this early ageing will enhance understanding the pathogenesis of TC treatment-induced morbidity and contribute to better recognition and prevention of late effects. In this review, we describe clinical manifestations of the early ageing phenotype among TC survivors, and subsequently focus on potential underlying mechanisms. We discuss the clinical implications and describe perspectives for future research and intervention strategies.
Collapse
Affiliation(s)
- Sjoukje Lubberts
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Coby Meijer
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Marco Demaria
- European Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Jourik A Gietema
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, The Netherlands.
| |
Collapse
|
60
|
Mechanisms of Lifespan Regulation by Calorie Restriction and Intermittent Fasting in Model Organisms. Nutrients 2020; 12:nu12041194. [PMID: 32344591 PMCID: PMC7230387 DOI: 10.3390/nu12041194] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/19/2020] [Accepted: 04/22/2020] [Indexed: 12/18/2022] Open
Abstract
Genetic and pharmacological interventions have successfully extended healthspan and lifespan in animals, but their genetic interventions are not appropriate options for human applications and pharmacological intervention needs more solid clinical evidence. Consequently, dietary manipulations are the only practical and probable strategies to promote health and longevity in humans. Caloric restriction (CR), reduction of calorie intake to a level that does not compromise overall health, has been considered as being one of the most promising dietary interventions to extend lifespan in humans. Although it is straightforward, continuous reduction of calorie or food intake is not easy to practice in real lives of humans. Recently, fasting-related interventions such as intermittent fasting (IF) and time-restricted feeding (TRF) have emerged as alternatives of CR. Here, we review the history of CR and fasting-related strategies in animal models, discuss the molecular mechanisms underlying these interventions, and propose future directions that can fill the missing gaps in the current understanding of these dietary interventions. CR and fasting appear to extend lifespan by both partially overlapping common mechanisms such as the target of rapamycin (TOR) pathway and circadian clock, and distinct independent mechanisms that remain to be discovered. We propose that a systems approach combining global transcriptomic, metabolomic, and proteomic analyses followed by genetic perturbation studies targeting multiple candidate pathways will allow us to better understand how CR and fasting interact with each other to promote longevity.
Collapse
|
61
|
Abstract
Life expectancy has increased substantially over the last few decades, leading to a worldwide increase in the prevalence and burden of aging-associated diseases. Recent evidence has proven that cellular senescence contributes substantially to the development of these disorders. Cellular senescence is a state of cell cycle arrest with suppressed apoptosis and concomitant secretion of multiple bioactive factors (the senescence-associated secretory phenotype-SASP) that plays a physiological role in embryonic development and healing processes. However, DNA damage and oxidative stress that occur during aging cause the accumulation of senescent cells, which through their SASP bring about deleterious effects on multiple organ and systemic functions. Ablation of senescent cells through genetic or pharmacological means leads to improved life span and health span in animal models, and preliminary evidence suggests it may also have a positive impact on human health. Thus, strategies to reduce or eliminate the burden of senescent cells or their products have the potential to impact multiple clinical outcomes with a single intervention. In this review, we touch upon the basics of cell senescence and summarize the current state of development of therapies against cell senescence for human use.
Collapse
|
62
|
|
63
|
Wang ZN, Su RN, Yang BY, Yang KX, Yang LF, Yan Y, Chen ZG. Potential Role of Cellular Senescence in Asthma. Front Cell Dev Biol 2020; 8:59. [PMID: 32117985 PMCID: PMC7026390 DOI: 10.3389/fcell.2020.00059] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/22/2020] [Indexed: 12/18/2022] Open
Abstract
Cellular senescence is a complicated process featured by irreversible cell cycle arrest and senescence-associated secreted phenotype (SASP), resulting in accumulation of senescent cells, and low-grade inflammation. Cellular senescence not only occurs during the natural aging of normal cells, but also can be accelerated by various pathological factors. Cumulative studies have shown the role of cellular senescence in the pathogenesis of chronic lung diseases including chronic obstructive pulmonary diseases (COPD) and idiopathic pulmonary fibrosis (IPF) by promoting airway inflammation and airway remodeling. Recently, great interest has been raised in the involvement of cellular senescence in asthma. Limited but valuable data has indicated accelerating cellular senescence in asthma. This review will compile current findings regarding the underlying relationship between cellular senescence and asthma, mainly through discussing the potential mechanisms of cellular senescence in asthma, the impact of senescent cells on the pathobiology of asthma, and the efficiency and feasibility of using anti-aging therapies in asthmatic patients.
Collapse
Affiliation(s)
- Zhao-Ni Wang
- Department of Pediatrics, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ruo-Nan Su
- Department of Pediatrics, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bi-Yuan Yang
- Department of Pediatrics, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ke-Xin Yang
- Department of Pediatrics, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li-Fen Yang
- Department of Pediatrics, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan Yan
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.,Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Zhuang-Gui Chen
- Department of Pediatrics, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
64
|
Pifferi F, Aujard F. Caloric restriction, longevity and aging: Recent contributions from human and non-human primate studies. Prog Neuropsychopharmacol Biol Psychiatry 2019; 95:109702. [PMID: 31325469 DOI: 10.1016/j.pnpbp.2019.109702] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/03/2019] [Accepted: 07/16/2019] [Indexed: 01/14/2023]
Abstract
The health benefits of chronic caloric restriction (CR) resulting in lifespan extension are well established in many species and has been recently demonstrated also in non-human primates, but its effects in humans remain to be proven on a long-term basis. CR might be a very efficient anti-aging strategy but its definition and limits must be well understood before envisaging to apply it to human. In this review, we first report and compare the recently issued CR studies in non-human primates and humans and then try to understand what an optimal caloric intake is. In a last part, we will discuss the pertinence of using CR as an anti-aging strategy with respect to the risks of frailty and obesity.
Collapse
Affiliation(s)
- Fabien Pifferi
- UMR CNRS/MNHN 7179, Mécanismes Adaptatifs et Evolution, 1 Avenue du Petit Château, 91800 Brunoy, France.
| | - Fabienne Aujard
- UMR CNRS/MNHN 7179, Mécanismes Adaptatifs et Evolution, 1 Avenue du Petit Château, 91800 Brunoy, France
| |
Collapse
|
65
|
Li N, Liu F, Yang P, Xiong F, Yu Q, Li J, Zhou Z, Zhang S, Wang CY. Aging and stress induced β cell senescence and its implication in diabetes development. Aging (Albany NY) 2019; 11:9947-9959. [PMID: 31721726 PMCID: PMC6874445 DOI: 10.18632/aging.102432] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 10/28/2019] [Indexed: 01/10/2023]
Abstract
Cellular senescence is a well-established defensive mechanism for tumor suppression, and is also proposed to play a crucial role in embryonic development, wound repair, aging and age-related diseases. Senescent cell is characterized by the marked morphological changes and active metabolism along with a distinctive senescence associated secretion phenotype (SASP). Cellular senescence is triggered by multiple endogenous and exogenous stressors, which collectively induce three types of senescence. It is believed that senescence represents a programmed phenomenon to facilitate β cell functional maturation and, therefore, senescence has been suggested to be involved in β cell regeneration, insulin secretion and diabetes development. Nevertheless, despite past extensive studies, the exact impact of senescence on β cell viability, regeneration and functionality, and its relevance to the development of diabetes are yet to be fully addressed. In this review, we will summarize the recent progress in β cell senescence, through which we intend to spark more instructive discussion and perspective with regard to the mechanisms underlying β cell senescence and their links to the pathogenesis of diabetes and the development of therapeutic strategies.
Collapse
Affiliation(s)
- Na Li
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Furong Liu
- Department of Dermatology, The People's Hospital of Shishou City, Shishou, Hubei, China
| | - Ping Yang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Xiong
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qilin Yu
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinxiu Li
- Shenzhen Third People's Hospital, Longgang District, Shenzhen, Guangdong, China
| | - Zhiguang Zhou
- Diabetes Center, The Second Xiangya Hospital, Institute of Metabolism and Endocrinology, Central South University, Changsha, China
| | - Shu Zhang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong-Yi Wang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
66
|
Kim H, Cao W, Oh G, Lee S, Shen A, Khadka D, Lee S, Sharma S, Kim SY, Choe S, Kwak TH, Kim J, Park R, So H. Augmentation of cellular NAD + by NQO1 enzymatic action improves age-related hearing impairment. Aging Cell 2019; 18:e13016. [PMID: 31353811 PMCID: PMC6718544 DOI: 10.1111/acel.13016] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 06/09/2019] [Accepted: 07/07/2019] [Indexed: 12/20/2022] Open
Abstract
Age-related hearing loss (ARHL) is a major neurodegenerative disorder and the leading cause of communication deficit in the elderly population, which remains largely untreated. The development of ARHL is a multifactorial event that includes both intrinsic and extrinsic factors. Recent studies suggest that NAD+ /NADH ratio may play a critical role in cellular senescence by regulating sirtuins, PARP-1, and PGC-1α. Nonetheless, the beneficial effect of direct modulation of cellular NAD+ levels on aging and age-related diseases has not been studied, and the underlying mechanisms remain obscure. Herein, we investigated the effect of β-lapachone (β-lap), a known plant-derived metabolite that modulates cellular NAD+ by conversion of NADH to NAD+ via the enzymatic action of NADH: quinone oxidoreductase 1 (NQO1) on ARHL in C57BL/6 mice. We elucidated that the reduction of cellular NAD+ during the aging process was an important contributor for ARHL; it facilitated oxidative stress and pro-inflammatory responses in the cochlear tissue through regulating sirtuins that alter various signaling pathways, such as NF-κB, p53, and IDH2. However, augmentation of NAD+ by β-lap effectively prevented ARHL and accompanying deleterious effects through reducing inflammation and oxidative stress, sustaining mitochondrial function, and promoting mitochondrial biogenesis in rodents. These results suggest that direct regulation of cellular NAD+ levels by pharmacological agents may be a tangible therapeutic option for treating various age-related diseases, including ARHL.
Collapse
Affiliation(s)
- Hyung‐Jin Kim
- Center for Metabolic Function Regulation (CMFR) and Department of Microbiology Wonkwang University School of Medicine Jeonbuk Korea
- NADIANBIO Ltd, Business Incubation Center Iksan Korea
| | - Wa Cao
- Center for Metabolic Function Regulation (CMFR) and Department of Microbiology Wonkwang University School of Medicine Jeonbuk Korea
| | - Gi‐Su Oh
- NADIANBIO Ltd, Business Incubation Center Iksan Korea
| | - SeungHoon Lee
- NADIANBIO Ltd, Business Incubation Center Iksan Korea
| | - AiHua Shen
- Center for Metabolic Function Regulation (CMFR) and Department of Microbiology Wonkwang University School of Medicine Jeonbuk Korea
| | - Dipendra Khadka
- Center for Metabolic Function Regulation (CMFR) and Department of Microbiology Wonkwang University School of Medicine Jeonbuk Korea
| | - Su‐Bin Lee
- Center for Metabolic Function Regulation (CMFR) and Department of Microbiology Wonkwang University School of Medicine Jeonbuk Korea
| | - Subham Sharma
- Center for Metabolic Function Regulation (CMFR) and Department of Microbiology Wonkwang University School of Medicine Jeonbuk Korea
| | - Seon Young Kim
- Center for Metabolic Function Regulation (CMFR) and Department of Microbiology Wonkwang University School of Medicine Jeonbuk Korea
| | - Seong‐Kyu Choe
- Center for Metabolic Function Regulation (CMFR) and Department of Microbiology Wonkwang University School of Medicine Jeonbuk Korea
| | - Tae Hwan Kwak
- NADIANBIO Ltd, Business Incubation Center Iksan Korea
| | - Jin‐Man Kim
- Department of Pathology and Infection Signaling Network Research Center Chungnam National University School of Medicine Daejeon Korea
| | - Raekil Park
- Department of Biomedical Science & Engineering, Institute of Integrated Technology Gwangju Institute of Science and Technology Gwangju Korea
| | - Hong‐Seob So
- Center for Metabolic Function Regulation (CMFR) and Department of Microbiology Wonkwang University School of Medicine Jeonbuk Korea
| |
Collapse
|
67
|
Barja G. Towards a unified mechanistic theory of aging. Exp Gerontol 2019; 124:110627. [DOI: 10.1016/j.exger.2019.05.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/08/2019] [Accepted: 05/30/2019] [Indexed: 12/18/2022]
|
68
|
Meal Timing, Aging, and Metabolic Health. Int J Mol Sci 2019; 20:ijms20081911. [PMID: 31003407 PMCID: PMC6514931 DOI: 10.3390/ijms20081911] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 12/14/2022] Open
Abstract
A growing body of evidence suggests that meal timing is an important factor for metabolic regulation and that the circadian clock tightly interacts with metabolic functions. The proper functioning of the circadian clock is critical for maintaining metabolic health. Therefore, chrononutrition, a novel discipline which investigates the relation between circadian rhythms, nutrition, and metabolism, has attracted increasing attention in recent years. Circadian rhythms are strongly affected by obesity, type 2 diabetes, and other dietary-induced metabolic diseases. With increasing age, the circadian system also undergoes significant changes which contribute to the dysregulation of metabolic rhythms. Metabolic diseases are a major health concern, particularly in light of a growing aging population, and effective approaches for their prevention and treatment are urgently needed. Recently, animal studies have impressively shown beneficial effects of several dietary patterns (e.g., caloric restriction or time-restricted feeding) on circadian rhythms and metabolic outcomes upon nutritional challenges. Whether these dietary patterns show the same beneficial effects in humans is, however, less well studied. As indicated by recent studies, dietary approaches might represent a promising, attractive, and easy-to-adapt strategy for the prevention and therapy of circadian and metabolic disturbances in humans of different age.
Collapse
|
69
|
Salvestrini V, Sell C, Lorenzini A. Obesity May Accelerate the Aging Process. Front Endocrinol (Lausanne) 2019; 10:266. [PMID: 31130916 PMCID: PMC6509231 DOI: 10.3389/fendo.2019.00266] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 04/10/2019] [Indexed: 12/27/2022] Open
Abstract
Lines of evidence from several studies have shown that increases in life expectancy are now accompanied by increased disability rate. The expanded lifespan of the aging population imposes a challenge on the continuous increase of chronic disease. The prevalence of overweight and obesity is increasing at an alarming rate in many parts of the world. Further to increasing the onset of metabolic imbalances, obesity leads to reduced life span and affects cellular and molecular processes in a fashion resembling aging. Nine key hallmarks of the aging process have been proposed. In this review, we will review these hallmarks and discuss pathophysiological changes that occur with obesity, that are similar to or contribute to those that occur during aging. We present and discuss the idea that obesity, in addition to having disease-specific effects, may accelerate the rate of aging affecting all aspects of physiology and thus shortening life span and health span.
Collapse
Affiliation(s)
- Valentina Salvestrini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Christian Sell
- Department of Pathology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Antonello Lorenzini
- Department of Biomedical and Neuromotor Sciences, Biochemistry Unit, University of Bologna, Bologna, Italy
- *Correspondence: Antonello Lorenzini
| |
Collapse
|