401
|
Abstract
The induction of brown-like adipocyte development in white adipose tissue (WAT) confers numerous metabolic benefits by decreasing adiposity and increasing energy expenditure. Therefore, WAT browning has gained considerable attention for its potential to reverse obesity and its associated co-morbidities. However, this perspective has been tainted by recent studies identifying the detrimental effects of inducing WAT browning. This review aims to highlight the adverse outcomes of both overactive and underactive browning activity, the harmful side effects of browning agents, as well as the molecular brake-switch system that has been proposed to regulate this process. Developing novel strategies that both sustain the metabolic improvements of WAT browning and attenuate the related adverse side effects is therefore essential for unlocking the therapeutic potential of browning agents in the treatment of metabolic diseases.
Collapse
|
402
|
Zhong H, Fang C, Fan Y, Lu Y, Wen B, Ren H, Hou G, Yang F, Xie H, Jie Z, Peng Y, Ye Z, Wu J, Zi J, Zhao G, Chen J, Bao X, Hu Y, Gao Y, Zhang J, Yang H, Wang J, Madsen L, Kristiansen K, Ni C, Li J, Liu S. Lipidomic profiling reveals distinct differences in plasma lipid composition in healthy, prediabetic, and type 2 diabetic individuals. Gigascience 2017; 6:1-12. [PMID: 28505362 PMCID: PMC5502363 DOI: 10.1093/gigascience/gix036] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 03/04/2017] [Accepted: 05/09/2017] [Indexed: 01/09/2023] Open
Abstract
The relationship between dyslipidemia and type 2 diabetes mellitus (T2D) has been extensively reported, but the global lipid profiles, especially in the East Asia population, associated with the development of T2D remain to be characterized. Liquid chromatography coupled to tandem mass spectrometry was applied to detect the global lipidome in the fasting plasma of 293 Chinese individuals, including 114 T2D patients, 81 prediabetic subjects, and 98 individuals with normal glucose tolerance (NGT). Both qualitative and quantitative analyses revealed a gradual change in plasma lipid features with T2D patients exhibiting characteristics close to those of prediabetic individuals, whereas they differed significantly from individuals with NGT. We constructed and validated a random forest classifier with 28 lipidomic features that effectively discriminated T2D from NGT or prediabetes. Most of the selected features significantly correlated with diabetic clinical indices. Hydroxybutyrylcarnitine was positively correlated with fasting plasma glucose, 2-hour postprandial glucose, glycated hemoglobin, and insulin resistance index (HOMA-IR). Lysophosphatidylcholines such as lysophosphatidylcholine (18:0), lysophosphatidylcholine (18:1), and lysophosphatidylcholine (18:2) were all negatively correlated with HOMA-IR. The altered plasma lipidome in Chinese T2D and prediabetic subjects suggests that lipid features may play a role in the pathogenesis of T2D and that such features may provide a basis for evaluating risk and monitoring disease development.
Collapse
Affiliation(s)
- Huanzi Zhong
- BGI-Shenzhen, Shenzhen 518083, China
- China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
- Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI-Shenzhen, Shenzhen 518083, China
| | - Chao Fang
- BGI-Shenzhen, Shenzhen 518083, China
- China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI-Shenzhen, Shenzhen 518083, China
| | | | - Yan Lu
- Suzhou Center for Disease Prevention and Control, Suzhou 215007, China
| | - Bo Wen
- BGI-Shenzhen, Shenzhen 518083, China
| | - Huahui Ren
- BGI-Shenzhen, Shenzhen 518083, China
- China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI-Shenzhen, Shenzhen 518083, China
| | - Guixue Hou
- Proteomics Division, BGI-Shenzhen, Shenzhen 518083, China
| | - Fangming Yang
- BGI-Shenzhen, Shenzhen 518083, China
- China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
- BGI Education Center, University of Chinese Academy of Sciences, Beijing, China
| | - Hailiang Xie
- BGI-Shenzhen, Shenzhen 518083, China
- China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
| | - Zhuye Jie
- BGI-Shenzhen, Shenzhen 518083, China
- China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
| | - Ye Peng
- BGI-Shenzhen, Shenzhen 518083, China
- China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Zhiqiang Ye
- BGI-Shenzhen, Shenzhen 518083, China
- China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
| | - Jiegen Wu
- BGI-Shenzhen, Shenzhen 518083, China
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Jin Zi
- Proteomics Division, BGI-Shenzhen, Shenzhen 518083, China
| | - Guoqing Zhao
- China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
| | - Jiayu Chen
- China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
| | - Xiao Bao
- China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
| | - Yihe Hu
- Suzhou Center for Disease Prevention and Control, Suzhou 215007, China
| | - Yan Gao
- Suzhou Center for Disease Prevention and Control, Suzhou 215007, China
| | - Jun Zhang
- Suzhou Center for Disease Prevention and Control, Suzhou 215007, China
| | - Huanming Yang
- BGI-Shenzhen, Shenzhen 518083, China
- China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
- James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | - Jian Wang
- BGI-Shenzhen, Shenzhen 518083, China
- China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
- James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | - Lise Madsen
- BGI-Shenzhen, Shenzhen 518083, China
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, 2100 Copenhagen Ø, Denmark
- National Institute of Nutrition and Seafood Research (NIFES), 5817 Bergen, Norway
| | - Karsten Kristiansen
- BGI-Shenzhen, Shenzhen 518083, China
- China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, 2100 Copenhagen Ø, Denmark
| | - Chuanming Ni
- Suzhou Center for Disease Prevention and Control, Suzhou 215007, China
| | - Junhua Li
- BGI-Shenzhen, Shenzhen 518083, China
- China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen 518083, China
- Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI-Shenzhen, Shenzhen 518083, China
| | - Siqi Liu
- BGI-Shenzhen, Shenzhen 518083, China
- Proteomics Division, BGI-Shenzhen, Shenzhen 518083, China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
403
|
Bilici M, Karakaya Arpaci D, Uygun Ilikhan S, Delikanli Corakci B, Bayraktaroglu T, Arasli M, Ozel Tekin I. Type 2 Diabetes Mellitus Associated with Premature Aging. IRANIAN RED CRESCENT MEDICAL JOURNAL 2017; 19. [DOI: 10.5812/ircmj.14024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
404
|
Caravia XM, Roiz-Valle D, Morán-Álvarez A, López-Otín C. Functional relevance of miRNAs in premature ageing. Mech Ageing Dev 2017; 168:10-19. [PMID: 28502819 DOI: 10.1016/j.mad.2017.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 03/30/2017] [Accepted: 05/09/2017] [Indexed: 02/06/2023]
Abstract
Ageing is a complex biological process characterized by the progressive loss of biological fitness due to the accumulation of macromolecular and cellular damage that affects most living organisms. Moreover, ageing is an important risk factor for many pathologies, including cardiovascular diseases, neurological disorders, and cancer. However, the ageing rate can be modulated by genetic, nutritional, and pharmacological factors, highlighting the concept of "ageing plasticity". Progeroid syndromes are a group of rare genetic diseases that resemble many characteristics of physiological ageing. Accordingly, studies on these diseases have been very useful for gaining mechanistic insights in ageing biology. In recent years, a great effort has been made in ageing research and several works have confirmed that geromiRs, the growing subgroup of miRNAs implicated in ageing, are able to modulate organismal lifespan. However, very little is still known about the impact of miRNA in premature ageing. In this review, we will address the functional relevance of this class of small non-coding RNAs in the regulation of the hallmarks of progeroid syndromes. In addition, we will discuss the potential strategies for managing progeria based on geromiR modulation.
Collapse
Affiliation(s)
- Xurde M Caravia
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain
| | - David Roiz-Valle
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain
| | - Alba Morán-Álvarez
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain; Centro de Investigación Biomédica en Red de Cáncer, Spain.
| |
Collapse
|
405
|
Chandrasekaran A, Idelchik MDPS, Melendez JA. Redox control of senescence and age-related disease. Redox Biol 2017; 11:91-102. [PMID: 27889642 PMCID: PMC5126126 DOI: 10.1016/j.redox.2016.11.005] [Citation(s) in RCA: 239] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 11/10/2016] [Indexed: 12/17/2022] Open
Abstract
The signaling networks that drive the aging process, associated functional deterioration, and pathologies has captured the scientific community's attention for decades. While many theories exist to explain the aging process, the production of reactive oxygen species (ROS) provides a signaling link between engagement of cellular senescence and several age-associated pathologies. Cellular senescence has evolved to restrict tumor progression but the accompanying senescence-associated secretory phenotype (SASP) promotes pathogenic pathways. Here, we review known biological theories of aging and how ROS mechanistically control senescence and the aging process. We also describe the redox-regulated signaling networks controlling the SASP and its important role in driving age-related diseases. Finally, we discuss progress in designing therapeutic strategies that manipulate the cellular redox environment to restrict age-associated pathology.
Collapse
Affiliation(s)
- Akshaya Chandrasekaran
- SUNY Polytechnic Institute, Colleges of Nanoscale Science and Engineering, 257 Fuller Road, Albany, NY 12203, USA
| | | | - J Andrés Melendez
- SUNY Polytechnic Institute, Colleges of Nanoscale Science and Engineering, 257 Fuller Road, Albany, NY 12203, USA.
| |
Collapse
|
406
|
Abe T, Hirasaka K, Nikawa T. Involvement of Cbl-b-mediated macrophage inactivation in insulin resistance. World J Diabetes 2017; 8:97-103. [PMID: 28344752 PMCID: PMC5348625 DOI: 10.4239/wjd.v8.i3.97] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 08/31/2016] [Accepted: 01/14/2017] [Indexed: 02/05/2023] Open
Abstract
Aging and overnutrition cause obesity in rodents and humans. It is well-known that obesity causes various diseases by producing insulin resistance (IR). Macrophages infiltrate the adipose tissue (AT) of obese individuals and cause chronic low-level inflammation associated with IR. Macrophage infiltration is regulated by the chemokines that are released from hypertrophied adipocytes and the immune cells in AT. Saturated fatty acids are recognized by toll-like receptor 4 (TLR4) and induce inflammatory responses in AT macrophages (ATMs). The inflammatory cytokines that are released from activated ATMs promote IR in peripheral organs, such as the liver, skeletal muscle and AT. Therefore, ATM activation is a therapeutic target for IR in obesity. The ubiquitin ligase Casitas b-lineage lymphoma-b (Cbl-b) appears to potently suppress macrophage migration and activation. Cbl-b is highly expressed in leukocytes and negatively regulates signals associated with migration and activation. Cbl-b deficiency enhances ATM accumulation and IR in aging- and diet-induced obese mice. Cbl-b inhibits migration-related signals and SFA-induced TLR4 signaling in ATMs. Thus, targeting Cbl-b may be a potential therapeutic strategy to reduce the IR induced by ATM activation. In this review, we summarize the regulatory functions of Cbl-b in ATMs.
Collapse
|
407
|
Espeland MA, Crimmins EM, Grossardt BR, Crandall JP, Gelfond JAL, Harris TB, Kritchevsky SB, Manson JE, Robinson JG, Rocca WA, Temprosa M, Thomas F, Wallace R, Barzilai N, Multimorbidity Clinical Trials Consortium. Clinical Trials Targeting Aging and Age-Related Multimorbidity. J Gerontol A Biol Sci Med Sci 2017; 72:355-361. [PMID: 28364543 PMCID: PMC5777384 DOI: 10.1093/gerona/glw220] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 10/12/2016] [Indexed: 01/17/2023] Open
Abstract
Background There is growing interest in identifying interventions that may increase health span by targeting biological processes underlying aging. The design of efficient and rigorous clinical trials to assess these interventions requires careful consideration of eligibility criteria, outcomes, sample size, and monitoring plans. Methods Experienced geriatrics researchers and clinical trialists collaborated to provide advice on clinical trial design. Results Outcomes based on the accumulation and incidence of age-related chronic diseases are attractive for clinical trials targeting aging. Accumulation and incidence rates of multimorbidity outcomes were developed by selecting at-risk subsets of individuals from three large cohort studies of older individuals. These provide representative benchmark data for decisions on eligibility, duration, and assessment protocols. Monitoring rules should be sensitive to targeting aging-related, rather than disease-specific, outcomes. Conclusions Clinical trials targeting aging are feasible, but require careful design consideration and monitoring rules.
Collapse
Affiliation(s)
- Mark A Espeland
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Eileen M Crimmins
- Davis School of Gerontology, University of Southern California, Los Angeles
| | - Brandon R Grossardt
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Jill P Crandall
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Jonathan A L Gelfond
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center, San Antonio
| | - Tamara B Harris
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, Bethesda, Maryland
| | - Stephen B Kritchevsky
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - JoAnn E Manson
- Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Walter A Rocca
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Marinella Temprosa
- The Biostatistics Center, The George Washington University, Rockville, Maryland
| | - Fridtjof Thomas
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis
| | | | - Nir Barzilai
- The Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York
| | | |
Collapse
|
408
|
Kuehne A, Hildebrand J, Soehle J, Wenck H, Terstegen L, Gallinat S, Knott A, Winnefeld M, Zamboni N. An integrative metabolomics and transcriptomics study to identify metabolic alterations in aged skin of humans in vivo. BMC Genomics 2017; 18:169. [PMID: 28201987 PMCID: PMC5312537 DOI: 10.1186/s12864-017-3547-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 02/02/2017] [Indexed: 11/12/2022] Open
Abstract
Background Aging human skin undergoes significant morphological and functional changes such as wrinkle formation, reduced wound healing capacity, and altered epidermal barrier function. Besides known age-related alterations like DNA-methylation changes, metabolic adaptations have been recently linked to impaired skin function in elder humans. Understanding of these metabolic adaptations in aged skin is of special interest to devise topical treatments that potentially reverse or alleviate age-dependent skin deterioration and the occurrence of skin disorders. Results We investigated the global metabolic adaptions in human skin during aging with a combined transcriptomic and metabolomic approach applied to epidermal tissue samples of young and old human volunteers. Our analysis confirmed known age-dependent metabolic alterations, e.g. reduction of coenzyme Q10 levels, and also revealed novel age effects that are seemingly important for skin maintenance. Integration of donor-matched transcriptome and metabolome data highlighted transcriptionally-driven alterations of metabolism during aging such as altered activity in upper glycolysis and glycerolipid biosynthesis or decreased protein and polyamine biosynthesis. Together, we identified several age-dependent metabolic alterations that might affect cellular signaling, epidermal barrier function, and skin structure and morphology. Conclusions Our study provides a global resource on the metabolic adaptations and its transcriptional regulation during aging of human skin. Thus, it represents a first step towards an understanding of the impact of metabolism on impaired skin function in aged humans and therefore will potentially lead to improved treatments of age related skin disorders. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3547-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andreas Kuehne
- Institute of Molecular Systems Biology, ETH Zurich, Auguste-Piccard-Hof 1, 8093, Zürich, Switzerland.,PhD Program Systems Biology, Life Science Zurich Graduate School, Zurich, Switzerland
| | - Janosch Hildebrand
- Coburg University of Applied Sciences and Arts, Friedrich-Streib-Straße 2, Coburg, 96450, Germany
| | - Joern Soehle
- Beiersdorf AG, R&D, Skin Research Center, Unnastrasse 48, Hamburg, 20253, Germany
| | - Horst Wenck
- Beiersdorf AG, R&D, Skin Research Center, Unnastrasse 48, Hamburg, 20253, Germany
| | - Lara Terstegen
- Beiersdorf AG, R&D, Skin Research Center, Unnastrasse 48, Hamburg, 20253, Germany
| | - Stefan Gallinat
- Beiersdorf AG, R&D, Skin Research Center, Unnastrasse 48, Hamburg, 20253, Germany
| | - Anja Knott
- Beiersdorf AG, R&D, Skin Research Center, Unnastrasse 48, Hamburg, 20253, Germany
| | - Marc Winnefeld
- Beiersdorf AG, R&D, Skin Research Center, Unnastrasse 48, Hamburg, 20253, Germany.
| | - Nicola Zamboni
- Institute of Molecular Systems Biology, ETH Zurich, Auguste-Piccard-Hof 1, 8093, Zürich, Switzerland.
| |
Collapse
|
409
|
The concept of ageing in evolutionary algorithms: Discussion and inspirations for human ageing. Mech Ageing Dev 2017; 163:8-14. [PMID: 28167122 DOI: 10.1016/j.mad.2017.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/30/2017] [Accepted: 02/02/2017] [Indexed: 10/20/2022]
Abstract
This paper discusses the concept of ageing as this applies to the operation of Evolutionary Algorithms, and examines its relationship to the concept of ageing as this is understood for human beings. Evolutionary Algorithms constitute a family of search algorithms which base their operation on an analogy from the evolution of species in nature. The paper initially provides the necessary knowledge on the operation of Evolutionary Algorithms, focusing on the use of ageing strategies during the implementation of the evolutionary process. Background knowledge on the concept of ageing, as this is defined scientifically for biological systems, is subsequently presented. Based on this information, the paper provides a comparison between the two ageing concepts, and discusses the philosophical inspirations which can be drawn for human ageing based on the operation of Evolutionary Algorithms.
Collapse
|
410
|
Braun F, Rinschen MM, Bartels V, Frommolt P, Habermann B, Hoeijmakers JHJ, Schumacher B, Dollé MET, Müller RU, Benzing T, Schermer B, Kurschat CE. Altered lipid metabolism in the aging kidney identified by three layered omic analysis. Aging (Albany NY) 2017; 8:441-57. [PMID: 26886165 PMCID: PMC4833139 DOI: 10.18632/aging.100900] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Aging-associated diseases and their comorbidities affect the life of a constantly growing proportion of the population in developed countries. At the center of these comorbidities are changes of kidney structure and function as age-related chronic kidney disease predisposes to the development of cardiovascular diseases such as stroke, myocardial infarction or heart failure. To detect molecular mechanisms involved in kidney aging, we analyzed gene expression profiles of kidneys from adult and aged wild-type mice by transcriptomic, proteomic and targeted lipidomic methodologies. Interestingly, transcriptome and proteome analyses revealed differential expression of genes primarily involved in lipid metabolism and immune response. Additional lipidomic analyses uncovered significant age-related differences in the total amount of phosphatidylethanolamines, phosphatidylcholines and sphingomyelins as well as in subspecies of phosphatidylserines and ceramides with age. By integration of these datasets we identified Aldh1a1, a key enzyme in vitamin A metabolism specifically expressed in the medullary ascending limb, as one of the most prominent upregulated proteins in old kidneys. Moreover, ceramidase Asah1 was highly expressed in aged kidneys, consistent with a decrease in ceramide C16. In summary, our data suggest that changes in lipid metabolism are involved in the process of kidney aging and in the development of chronic kidney disease.
Collapse
Affiliation(s)
- Fabian Braun
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Markus M Rinschen
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Valerie Bartels
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Department of Cardiology and Angiology, University of Münster, Münster, Germany
| | - Peter Frommolt
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | - Bianca Habermann
- Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany.,Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Jan H J Hoeijmakers
- Department of Cell Biology and Genetics, Medical Genetics Centre, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Björn Schumacher
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Institute for Genome Stability in Aging and Disease, Medical Faculty, University of Cologne, Cologne, Germany
| | - Martijn E T Dollé
- National Institute of Public Health and the Environment, Centre for Health Protection, Bilthoven, The Netherlands
| | - Roman-Ulrich Müller
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | - Christine E Kurschat
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
411
|
Fahrmann JF, Grapov D, DeFelice BC, Taylor S, Kim K, Kelly K, Wikoff WR, Pass H, Rom WN, Fiehn O, Miyamoto S. Serum phosphatidylethanolamine levels distinguish benign from malignant solitary pulmonary nodules and represent a potential diagnostic biomarker for lung cancer. Cancer Biomark 2017; 16:609-17. [PMID: 27002763 DOI: 10.3233/cbm-160602] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Recent computed tomography (CT) screening trials showed that it is effective for early detection of lung cancer, but were plagued by high false positive rates. Additional blood biomarker tests designed to complement CT screening and reduce false positive rates are highly desirable. OBJECTIVE Identify blood-based metabolite biomarkers for diagnosing lung cancer. MEHTODS Serum samples from subjects participating in a CT screening trial were analyzed using untargeted GC-TOFMS and HILIC-qTOFMS-based metabolomics. Samples were acquired prior to diagnosis (pre-diagnostic, n= 17), at-diagnosis (n= 25) and post-diagnosis (n= 19) of lung cancer and from subjects with benign nodules (n= 29). RESULTS Univariate analysis identified 40, 102 and 30 features which were significantly different between subjects with malignant (pre-, at- and post-diagnosis) solitary pulmonary nodules (SPNs) and benign SPNs, respectively. Ten metabolites were consistently different between subjects presenting malignant (pre- and at-diagnosis) or benign SPNs. Three of these 10 metabolites were phosphatidylethanolamines (PE) suggesting alterations in lipid metabolism. Accuracies of 77%, 83% and 78% in the pre-diagnosis group and 69%, 71% and 67% in the at-diagnosis group were determined for PE(34:2), PE(36:2) and PE(38:4), respectively. CONCLUSIONS This study demonstrates evidence of early metabolic alterations that can possibly distinguish malignant from benign SPNs. Further studies in larger pools of samples are warranted.
Collapse
Affiliation(s)
| | | | - Brian C DeFelice
- University of California, Davis Genome Center, Davis, California, CA, USA
| | - Sandra Taylor
- Division of Biostatistics, Department of Public Health Sciences, School of Medicine, University of California, Davis, California, CA, USA
| | - Kyoungmi Kim
- Division of Biostatistics, Department of Public Health Sciences, School of Medicine, University of California, Davis, California, CA, USA
| | - Karen Kelly
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California, Davis Medical Center, Sacramento, CA, USA
| | - William R Wikoff
- University of California, Davis Genome Center, Davis, California, CA, USA
| | - Harvey Pass
- Division of Thoracic Surgery, Department of Cardiothoracic Surgery, Langone Medical Center, New York University, New York, NY, USA
| | - William N Rom
- Division of Pulmonary, Critical Care, and Sleep, NYU School of Medicine, New York, NY, USA
| | - Oliver Fiehn
- University of California, Davis Genome Center, Davis, California, CA, USA.,Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi-Arabia
| | - Suzanne Miyamoto
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California, Davis Medical Center, Sacramento, CA, USA
| |
Collapse
|
412
|
Mathew R, Pal Bhadra M, Bhadra U. Insulin/insulin-like growth factor-1 signalling (IIS) based regulation of lifespan across species. Biogerontology 2017; 18:35-53. [DOI: 10.1007/s10522-016-9670-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 11/25/2016] [Indexed: 12/21/2022]
|
413
|
Gómez-Serrano M, Camafeita E, López JA, Rubio MA, Bretón I, García-Consuegra I, García-Santos E, Lago J, Sánchez-Pernaute A, Torres A, Vázquez J, Peral B. Differential proteomic and oxidative profiles unveil dysfunctional protein import to adipocyte mitochondria in obesity-associated aging and diabetes. Redox Biol 2016; 11:415-428. [PMID: 28064117 PMCID: PMC5220168 DOI: 10.1016/j.redox.2016.12.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 12/12/2016] [Accepted: 12/16/2016] [Indexed: 12/19/2022] Open
Abstract
Human age-related diseases, including obesity and type 2 diabetes (T2DM), have long been associated to mitochondrial dysfunction; however, the role for adipose tissue mitochondria in these conditions remains unknown. We have tackled the impact of aging and T2DM on adipocyte mitochondria from obese patients by quantitating not only the corresponding abundance changes of proteins, but also the redox alterations undergone by Cys residues thereof. For that, we have resorted to a high-throughput proteomic approach based on isobaric labeling, liquid chromatography and mass spectrometry. The alterations undergone by the mitochondrial proteome revealed aging- and T2DM-specific hallmarks. Thus, while a global decrease of oxidative phosphorylation (OXPHOS) subunits was found in aging, the diabetic patients exhibited a reduction of specific OXPHOS complexes as well as an up-regulation of the anti-oxidant response. Under both conditions, evidence is shown for the first time of a link between increased thiol protein oxidation and decreased protein abundance in adipose tissue mitochondria. This association was stronger in T2DM, where OXPHOS mitochondrial- vs. nuclear-encoded protein modules were found altered, suggesting impaired mitochondrial protein translocation and complex assembly. The marked down-regulation of OXPHOS oxidized proteins and the alteration of oxidized Cys residues related to protein import through the redox-active MIA (Mitochondrial Intermembrane space Assembly) pathway support that defects in protein translocation to the mitochondria may be an important underlying mechanism for mitochondrial dysfunction in T2DM and physiological aging. The present draft of redox targets together with the quantification of protein and oxidative changes may help to better understand the role of oxidative stress in both a physiological process like aging and a pathological condition like T2DM.
Collapse
Affiliation(s)
- María Gómez-Serrano
- Instituto de Investigaciones Biomédicas, Alberto Sols, (IIBM); Consejo Superior de Investigaciones Científicas & Universidad Autónoma de Madrid (CSIC-UAM), Madrid 28029, Spain
| | - Emilio Camafeita
- Laboratory of Cardiovascular Proteomics, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Juan A López
- Laboratory of Cardiovascular Proteomics, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Miguel A Rubio
- Department of Endocrinology, Hospital Clínico San Carlos (IDISSC), Facultad de Medicina, Universidad Complutense, Madrid 28040, Spain
| | - Irene Bretón
- Department of Endocrinology and Nutrition, Hospital General Universitario Gregorio Marañón (IISGM), Madrid 28007, Spain
| | - Inés García-Consuegra
- Instituto de Investigación, Hospital Universitario 12 de Octubre (i+12), Madrid 28041, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), U723, Madrid 28029, Spain
| | - Eva García-Santos
- Instituto de Investigaciones Biomédicas, Alberto Sols, (IIBM); Consejo Superior de Investigaciones Científicas & Universidad Autónoma de Madrid (CSIC-UAM), Madrid 28029, Spain
| | - Jesús Lago
- Department of Surgery, Hospital General Universitario Gregorio Marañón (IISGM), Madrid 28007, Spain
| | - Andrés Sánchez-Pernaute
- Department of Surgery, Hospital Clínico San Carlos (IDISSC), Facultad de Medicina, Universidad Complutense, Madrid 28040, Spain
| | - Antonio Torres
- Department of Surgery, Hospital Clínico San Carlos (IDISSC), Facultad de Medicina, Universidad Complutense, Madrid 28040, Spain
| | - Jesús Vázquez
- Laboratory of Cardiovascular Proteomics, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Belén Peral
- Instituto de Investigaciones Biomédicas, Alberto Sols, (IIBM); Consejo Superior de Investigaciones Científicas & Universidad Autónoma de Madrid (CSIC-UAM), Madrid 28029, Spain.
| |
Collapse
|
414
|
Kohanski RA, Deeks SG, Gravekamp C, Halter JB, High K, Hurria A, Fuldner R, Green P, Huebner R, Macchiarini F, Sierra F. Reverse geroscience: how does exposure to early diseases accelerate the age-related decline in health? Ann N Y Acad Sci 2016; 1386:30-44. [PMID: 27907230 DOI: 10.1111/nyas.13297] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 10/21/2016] [Indexed: 02/07/2023]
Abstract
Aging is the major risk factor for both the development of chronic diseases and loss of functional capacity. Geroscience provides links among the biology of aging, the biology of disease, and the physiology of frailty, three fields where enormous progress has been made in the last few decades. While, previously, the focus was on the role of aging in susceptibility to disease and disability, the other side of this relationship, which is the contribution of disease to aging, has been less explored at the molecular/cellular level. Indeed, the role of childhood or early adulthood exposure to chronic disease and/or treatment on accelerating aging phenotypes is well known in epidemiology, but the biological basis is poorly understood. A recent summit co-organized by the National Institutes of Health GeroScience Interest Group and the New York Academy of Sciences explored these relationships, using three chronic diseases as examples: cancer, HIV/AIDS, and diabetes. The epidemiological literature clearly indicates that early exposure to any of these diseases and/or their treatments results in an acceleration of the appearance of aging phenotypes, including loss of functional capacity and accelerated appearance of clinical symptoms of aging-related diseases not obviously related to the earlier event. The discussions at the summit focused on the molecular and cellular relationships between each of these diseases and the recently defined molecular and cellular pillars of aging. Two major conclusions from the meeting include the desire to refine an operational definition of aging and to concomitantly develop biomarkers of aging, in order to move from chronological to physiological age. The discussion also opened a dialogue on the possibility of improving late-life outcomes in patients affected by chronic disease by including age-delaying modalities along with the standard care for the disease in question.
Collapse
Affiliation(s)
- Ronald A Kohanski
- Division of Aging Biology, National Institute on Aging, NIH, Bethesda, Maryland
| | - Steven G Deeks
- Department of Medicine, University of California San Francisco, San Francisco, California
| | - Claudia Gravekamp
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Jeffrey B Halter
- Geriatrics Center and Institute of Gerontology, University of Michigan, Ann Arbor, Michigan
| | - Kevin High
- Department of Internal Medicine, Section on Infectious Diseases, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Arti Hurria
- City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, California
| | - Rebecca Fuldner
- Division of Aging Biology, National Institute on Aging, NIH, Bethesda, Maryland
| | - Paige Green
- Biobehavioral and Psychologic Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Robin Huebner
- Division of AIDS, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | | | - Felipe Sierra
- Division of Aging Biology, National Institute on Aging, NIH, Bethesda, Maryland
| |
Collapse
|
415
|
Cavadas C, Aveleira CA, Souza GFP, Velloso LA. The pathophysiology of defective proteostasis in the hypothalamus - from obesity to ageing. Nat Rev Endocrinol 2016; 12:723-733. [PMID: 27388987 DOI: 10.1038/nrendo.2016.107] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Hypothalamic dysfunction has emerged as an important mechanism involved in the development of obesity and its comorbidities, as well as in the process of ageing and age-related diseases, such as type 2 diabetes mellitus, hypertension and Alzheimer disease. In both obesity and ageing, inflammatory signalling is thought to coordinate many of the cellular events that lead to hypothalamic neuronal dysfunction. This process is triggered by the activation of signalling via the toll-like receptor 4 pathway and endoplasmic reticulum stress, which in turn results in intracellular inflammatory signalling. However, the process that connects inflammation with neuronal dysfunction is complex and includes several regulatory mechanisms that ultimately control the homeostasis of intracellular proteins and organelles (also known as 'proteostasis'). This Review discusses the evidence for the key role of proteostasis in the control of hypothalamic neurons and the involvement of this process in regulating whole-body energy homeostasis and lifespan.
Collapse
Affiliation(s)
- Cláudia Cavadas
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, 3004-504, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, 3004-504, Portugal
| | - Célia A Aveleira
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, 3004-504, Portugal
| | - Gabriela F P Souza
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Campinas, 1308-970, Brazil
| | - Lício A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Campinas, 1308-970, Brazil
| |
Collapse
|
416
|
Ghebre YT, Yakubov E, Wong WT, Krishnamurthy P, Sayed N, Sikora AG, Bonnen MD. Vascular Aging: Implications for Cardiovascular Disease and Therapy. TRANSLATIONAL MEDICINE (SUNNYVALE, CALIF.) 2016; 6:183. [PMID: 28932625 PMCID: PMC5602592 DOI: 10.4172/2161-1025.1000183] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The incidence and prevalence of cardiovascular disease is highest among the elderly, in part, due to deleterious effects of advancing age on the heart and blood vessels. Aging, a known cardiovascular risk factor, is progressively associated with structural and functional changes to the vasculature including hemodynamic disturbance due to increased oxidative stress, premature cellular senescence and impairments in synthesis and/or secretion of endothelium-derived vasoactive molecules. These molecular and physiological changes lead to vessel wall stiffening and thickening, as well as other vascular complications that culminate to loss of vascular tone regulation and endothelial function. Intriguingly, the vessel wall, a biochemically active structure composed of collagen, connective tissue, smooth muscle and endothelial cells, is adversely affected by processes involved in premature or normal aging. Notably, the inner most layer of the vessel wall, the endothelium, becomes senescent and dysfunctional with advancing age. As a result, its ability to release vasoactive molecules such as acetylcholine (ACh), prostacyclin (PGI2), endothelium-derived hyperpolarizing factor (EDHF), and nitric oxide (NO) is reduced and the cellular response to these molecules is also impaired. By contrast, the vascular endothelium increases its generation and release of reactive oxygen (ROS) and nitrogen (RNS) species, vasoconstrictors such as endothelin (ET) and angiotensin (AT), and endogenous inhibitors of NO synthases (NOSs) to block NO. This skews the balance of the endothelium in favor of the release of highly tissue reactive and harmful molecules that promote DNA damage, telomere erosion, senescence, as well as stiffened and hardened vessel wall that is prone to the development of hypertension, diabetes, atherosclerosis and other cardiovascular risk factors. This Review discusses the impact of advancing age on cardiovascular health, and highlights the cellular and molecular mechanisms that underlie age-associated vascular changes. In addition, the role of pharmacological interventions in preventing or delaying age-related cardiovascular disease is discussed.
Collapse
Affiliation(s)
- Yohannes T Ghebre
- Department of Radiation Oncology, Baylor College of Medicine, Houston, Texas, USA
| | - Eduard Yakubov
- phaRNA Comprehensive RNA Technologies, Houston, Texas, USA
| | - Wing Tak Wong
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nazish Sayed
- Department of Medicine, Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Andrew G Sikora
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - Mark D Bonnen
- Department of Radiation Oncology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
417
|
Sivarajasingam SP, Imami N, Johnson MR. Myometrial cytokines and their role in the onset of labour. J Endocrinol 2016; 231:R101-R119. [PMID: 27647860 DOI: 10.1530/joe-16-0157] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 09/19/2016] [Indexed: 12/12/2022]
Abstract
Human labour is an inflammatory event, physiologically driven by an interaction between hormonal and mechanical factors and pathologically associated with infection, bleeding and excessive uterine stretch. The initiation and communicators of inflammation is still not completely understood; however, a key role for cytokines has been implicated. We summarise the current understanding of the nature and role of cytokines, chemokines and hormones and their involvement in signalling within the myometrium particularly during labour.
Collapse
Affiliation(s)
- S P Sivarajasingam
- Department of Surgery and CancerImperial College London, Chelsea and Westminster Hospital, London, UK
| | - N Imami
- Department of MedicineImperial College London, London, UK
| | - M R Johnson
- Department of Surgery and CancerImperial College London, Chelsea and Westminster Hospital, London, UK
| |
Collapse
|
418
|
Palikaras K, Mari M, Petanidou B, Pasparaki A, Filippidis G, Tavernarakis N. Ectopic fat deposition contributes to age-associated pathology in Caenorhabditis elegans. J Lipid Res 2016; 58:72-80. [PMID: 27884963 DOI: 10.1194/jlr.m069385] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 11/06/2016] [Indexed: 01/08/2023] Open
Abstract
Age-dependent collapse of lipid homeostasis results in spillover of lipids and excessive fat deposition in nonadipose tissues. Ectopic fat contributes to lipotoxicity and has been implicated in the development of a metabolic syndrome that increases risk of age-associated diseases. However, the molecular mechanisms coupling ectopic fat accumulation with aging remain obscure. Here, we use nonlinear imaging modalities to visualize and quantify age-dependent ectopic lipid accumulation in Caenorhabditis elegans We find that aging is accompanied by pronounced deposition of lipids in nonadipose tissues, including the nervous system. Importantly, interventions that promote longevity such as low insulin signaling, germ-line loss, and dietary restriction, which effectively delay aging in evolutionary divergent organisms, diminish the rate of ectopic fat accumulation and the size of lipid droplets. Suppression of lipotoxic accumulation of fat in heterologous tissues is dependent on helix-loop-helix (HLH)-30/transcription factor EB (TFEB) and autophagy. Our findings in their totality highlight the pivotal role of HLH-30/TFEB and autophagic processes in the maintenance of lipid homeostasis during aging, in addition to establishing nonlinear imaging as a powerful tool for monitoring ectopic lipid droplet deposition in vivo.
Collapse
Affiliation(s)
- Konstantinos Palikaras
- Institute of Molecular Biology and Biotechnology Foundation for Research and Technology, Heraklion 71110, Crete, Greece
| | - Meropi Mari
- Institute of Molecular Biology and Biotechnology Foundation for Research and Technology, Heraklion 71110, Crete, Greece
| | - Barbara Petanidou
- Institute of Electronic Structure and Laser, Foundation for Research and Technology, Heraklion 71110, Crete, Greece.,Physics Department University of Crete, Heraklion 71003, Crete, Greece
| | - Angela Pasparaki
- Institute of Molecular Biology and Biotechnology Foundation for Research and Technology, Heraklion 71110, Crete, Greece
| | - George Filippidis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology, Heraklion 71110, Crete, Greece
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology Foundation for Research and Technology, Heraklion 71110, Crete, Greece .,Medical School, University of Crete, Heraklion 71003, Crete, Greece
| |
Collapse
|
419
|
Brewer RA, Gibbs VK, Smith DL. Targeting glucose metabolism for healthy aging. NUTRITION AND HEALTHY AGING 2016; 4:31-46. [PMID: 28035340 PMCID: PMC5166514 DOI: 10.3233/nha-160007] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Advancing age is the greatest single risk factor for numerous chronic diseases. Thus, the ability to target the aging process can facilitate improved healthspan and potentially lifespan. Lack of adequate glucoregulatory control remains a recurrent theme accompanying aging and chronic disease, while numerous longevity interventions result in maintenance of glucoregulatory control. In this review, we propose targeting glucose metabolism to enhance regulatory control as a means to ameliorate the aging process. We highlight that calorie restriction improves glucoregulatory control and extends both lifespan and healthspan in model organisms, but we also indicate more practical interventions (i.e., calorie restriction mimetics) are desirable for clinical application in humans. Of the calorie restriction mimetics being investigated, we focus on the type 2 diabetes drug acarbose, an α-glucosidase inhibitor that when taken with a meal, results in reduced enzymatic degradation and absorption of glucose from complex carbohydrates. We discuss alternatives to acarbose that yield similar physiologic effects and describe dietary sources (e.g., sweet potatoes, legumes, and berries) of bioactive compounds with α-glucosidase inhibitory activity. We indicate future research should include exploration of how non-caloric compounds like α-glucosidase inhibitors modify macronutrient metabolism prior to disease onset, which may guide nutritional/lifestyle interventions to support health and reduce age-related disease risk.
Collapse
Affiliation(s)
- Rachel A. Brewer
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Victoria K. Gibbs
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
- Nutrition Obesity Research Center, Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
- Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, AL, USA
- Nathan Shock Center of Excellence in the Biology of Aging, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Daniel L. Smith
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
- Nutrition Obesity Research Center, Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
- Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, AL, USA
- Nathan Shock Center of Excellence in the Biology of Aging, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
420
|
Stimulating S-adenosyl-l-methionine synthesis extends lifespan via activation of AMPK. Proc Natl Acad Sci U S A 2016; 113:11913-11918. [PMID: 27698120 DOI: 10.1073/pnas.1604047113] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Dietary restriction (DR), such as calorie restriction (CR) or methionine (Met) restriction, extends the lifespan of diverse model organisms. Although studies have identified several metabolites that contribute to the beneficial effects of DR, the molecular mechanism underlying the key metabolites responsible for DR regimens is not fully understood. Here we show that stimulating S-adenosyl-l-methionine (AdoMet) synthesis extended the lifespan of the budding yeast Saccharomyces cerevisiae The AdoMet synthesis-mediated beneficial metabolic effects, which resulted from consuming both Met and ATP, mimicked CR. Indeed, stimulating AdoMet synthesis activated the universal energy-sensing regulator Snf1, which is the S. cerevisiae ortholog of AMP-activated protein kinase (AMPK), resulting in lifespan extension. Furthermore, our findings revealed that S-adenosyl-l-homocysteine contributed to longevity with a higher accumulation of AdoMet only under the severe CR (0.05% glucose) conditions. Thus, our data uncovered molecular links between Met metabolites and lifespan, suggesting a unique function of AdoMet as a reservoir of Met and ATP for cell survival.
Collapse
|
421
|
Derkach KV, Ignatieva PA, Bogush IV, Balluzek MF, Shpakov AO. Changes in the hormonal status of cardiovascular and the thyroid systems in rats with 18-month type 2 diabetes mellitus. ADVANCES IN GERONTOLOGY 2016. [DOI: 10.1134/s2079057016040044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
422
|
Botelho M, Cavadas C. Neuropeptide Y: An Anti-Aging Player? Trends Neurosci 2016; 38:701-711. [PMID: 26549884 DOI: 10.1016/j.tins.2015.08.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 08/27/2015] [Accepted: 08/28/2015] [Indexed: 12/16/2022]
Abstract
Accumulating evidence suggests that neuropeptide Y (NPY) has a role in aging and lifespan determination. In this review, we critically discuss age-related changes in NPY levels in the brain, together with recent findings concerning the contribution of NPY to, and impact on, six hallmarks of aging, specifically: loss of proteostasis, stem cell exhaustion, altered intercellular communication, deregulated nutrient sensing, cellular senescence, and mitochondrial dysfunction. Understanding how NPY contributes to, and counteracts, these hallmarks of aging will open new avenues of research on limiting damage related to aging.
Collapse
Affiliation(s)
- Mariana Botelho
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Cláudia Cavadas
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
423
|
Vermeij WP, Dollé MET, Reiling E, Jaarsma D, Payan-Gomez C, Bombardieri CR, Wu H, Roks AJM, Botter SM, van der Eerden BC, Youssef SA, Kuiper RV, Nagarajah B, van Oostrom CT, Brandt RMC, Barnhoorn S, Imholz S, Pennings JLA, de Bruin A, Gyenis Á, Pothof J, Vijg J, van Steeg H, Hoeijmakers JHJ. Restricted diet delays accelerated ageing and genomic stress in DNA-repair-deficient mice. Nature 2016; 537:427-431. [PMID: 27556946 PMCID: PMC5161687 DOI: 10.1038/nature19329] [Citation(s) in RCA: 225] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/25/2016] [Indexed: 12/27/2022]
Abstract
Mice deficient in the DNA excision-repair gene Ercc1 (Ercc1∆/-) show numerous accelerated ageing features that limit their lifespan to 4-6 months. They also exhibit a 'survival response', which suppresses growth and enhances cellular maintenance. Such a response resembles the anti-ageing response induced by dietary restriction (also known as caloric restriction). Here we report that a dietary restriction of 30% tripled the median and maximal remaining lifespans of these progeroid mice, strongly retarding numerous aspects of accelerated ageing. Mice undergoing dietary restriction retained 50% more neurons and maintained full motor function far beyond the lifespan of mice fed ad libitum. Other DNA-repair-deficient, progeroid Xpg-/- (also known as Ercc5-/-) mice, a model of Cockayne syndrome, responded similarly. The dietary restriction response in Ercc1∆/- mice closely resembled the effects of dietary restriction in wild-type animals. Notably, liver tissue from Ercc1∆/- mice fed ad libitum showed preferential extinction of the expression of long genes, a phenomenon we also observed in several tissues ageing normally. This is consistent with the accumulation of stochastic, transcription-blocking lesions that affect long genes more than short ones. Dietary restriction largely prevented this declining transcriptional output and reduced the number of γH2AX DNA damage foci, indicating that dietary restriction preserves genome function by alleviating DNA damage. Our findings establish the Ercc1∆/- mouse as a powerful model organism for health-sustaining interventions, reveal potential for reducing endogenous DNA damage, facilitate a better understanding of the molecular mechanism of dietary restriction and suggest a role for counterintuitive dietary-restriction-like therapy for human progeroid genome instability syndromes and possibly neurodegeneration in general.
Collapse
Affiliation(s)
- W P Vermeij
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - M E T Dollé
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - E Reiling
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands.,Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - D Jaarsma
- Department of Neuroscience, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - C Payan-Gomez
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands.,Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Carrera 24, 63C-69 Bogotá, Colombia
| | - C R Bombardieri
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - H Wu
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - A J M Roks
- Department of Internal Medicine, Division of Vascular Medicine and Pharmacology, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - S M Botter
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands.,Laboratory for Orthopedic Research, Balgrist University Hospital, Forchstrasse 340, 8008, Zürich, Switzerland
| | - B C van der Eerden
- Department of Internal Medicine, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - S A Youssef
- Dutch Molecular Pathology Center, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, PO Box 80125, 3508 TC Utrecht, The Netherlands
| | - R V Kuiper
- Dutch Molecular Pathology Center, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, PO Box 80125, 3508 TC Utrecht, The Netherlands
| | - B Nagarajah
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - C T van Oostrom
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - R M C Brandt
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - S Barnhoorn
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - S Imholz
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - J L A Pennings
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands
| | - A de Bruin
- Dutch Molecular Pathology Center, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, PO Box 80125, 3508 TC Utrecht, The Netherlands.,Department of Pediatrics, Division Molecular Genetics, University Medical Center Groningen, PO Box 30001, 9700 RB Groningen, The Netherlands
| | - Á Gyenis
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - J Pothof
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - J Vijg
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, USA
| | - H van Steeg
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720 BA Bilthoven, The Netherlands.,Department of Human Genetics, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - J H J Hoeijmakers
- Department of Molecular Genetics, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands.,CECAD Forschungszentrum, Universität zu Köln, Joseph-Stelzmann-Straße 26, 50931 Köln, Germany
| |
Collapse
|
424
|
Huffman DM, Justice JN, Stout MB, Kirkland JL, Barzilai N, Austad SN. Evaluating Health Span in Preclinical Models of Aging and Disease: Guidelines, Challenges, and Opportunities for Geroscience. J Gerontol A Biol Sci Med Sci 2016; 71:1395-1406. [PMID: 27535967 PMCID: PMC5055649 DOI: 10.1093/gerona/glw106] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/22/2016] [Indexed: 12/14/2022] Open
Abstract
Life extension is no longer considered sufficient evidence of delayed aging in research animals. It must also be demonstrated that a broad swathe of health indicators have been extended. During a retreat of the Geroscience Network, a consortium of basic and clinical aging researchers, potential measures of mouse health were considered for their potential as easily standardized, highly informative metrics. Major health domains considered were neuromuscular, cognitive, cardiovascular, metabolic, and inflammatory functions as well as body composition and energetics and a multitude of assays interrogating these domains. A particularly sensitive metric of health is the ability to respond to, and recover, from stress. Therefore, the Network also considered stresses of human relevance that could be implemented in mouse models to assess frailty and resilience. Mouse models already exist for responses to forced immobility, cancer chemotherapy, infectious diseases, dietary challenges, and surgical stress, and it was felt that these could be employed to determine whether putative senescence-retarding interventions increased and extended organismal robustness. The Network discussed challenges in modeling age-related human chronic diseases and concluded that more attention needs to be paid to developing disease models with later age of onset, models of co- and multimorbidity, diversifying the strains and sexes commonly used in aging research, and considering additional species.
Collapse
Affiliation(s)
- Derek M Huffman
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York
| | - Jamie N Justice
- Department of Integrative Physiology, University of Colorado Boulder
| | - Michael B Stout
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Nir Barzilai
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York
| | | |
Collapse
|
425
|
Pan SY, de Groh M, Aziz A, Morrison H. Relation of insulin resistance with social-demographics, adiposity and behavioral factors in non-diabetic adult Canadians. J Diabetes Metab Disord 2016; 15:31. [PMID: 27525252 PMCID: PMC4982003 DOI: 10.1186/s40200-016-0253-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 08/04/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND Insulin resistance is a pathogenic factor for type II diabetes and has been associated with metabolic abnormalities and adverse clinical outcomes. The purpose of this study was to examine the relationship between insulin resistance and socio-demographics, adiposity and behavioral factors in the general, non-diabetic adult Canadian population. METHODS Data for 3515 non-diabetic adults aged 18 to 79 years from the Canadian Health Measures Survey (cycles 1 and 2, 2007-2011) were analyzed. Insulin resistance index was measured by the homeostasis model assessment of insulin resistance (HOMA-IR), and insulin resistance (IR) was defined as individuals in the highest quartile of the HOMA-IR index. Logistic regression models were used to examine the effect of demographics, lifestyle factors and adiposity measurements on HOMA-IR. RESULTS The risk of IR increased with age, particularly in men. Individuals had adjusted odds ratio (OR) (with corresponding 95 % confidence interval) of 5.97 (2.90-8.52) and 25.12 (15.20-41.51) associated with a body-mass-index (BMI) between 25.0 and < 30.0, or ≥30.0, of 9.23 (6.52-13.07) with abdominal obesity (waist circumstance ≥102 cm for men and ≥ 88 cm for women), of 8.72 (6.13-12.39) with a high waist-to-height ratio (>0.57), and of 6.30 (4.33-9.16) with a high waist-to-hip ratio (>0.90 for men and >0.85 for women). Physically inactive people and non-alcohol consumer also had a significantly higher odd of IR. CONCLUSIONS This study found that men and older, obese and physically inactive people were at increased risk for IR. Adiposity indices including BMI, waist circumstance, waist-to-height ratio and waist-to-hip ratio were highly associated with IR with similar magnitude of association.
Collapse
Affiliation(s)
- Sai Yi Pan
- Science Integration and Social Determinant Directorate, Public Health Agency of Canada, 785 Carling Avenue, AL 6809B, Ottawa, ON K1A 0K9 Canada
| | - Margaret de Groh
- Science Integration and Social Determinant Directorate, Public Health Agency of Canada, 785 Carling Avenue, AL 6809B, Ottawa, ON K1A 0K9 Canada
| | - Alfred Aziz
- Food Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON Canada
| | - Howard Morrison
- Science Integration and Social Determinant Directorate, Public Health Agency of Canada, 785 Carling Avenue, AL 6809B, Ottawa, ON K1A 0K9 Canada
| |
Collapse
|
426
|
da Costa JP, Vitorino R, Silva GM, Vogel C, Duarte AC, Rocha-Santos T. A synopsis on aging-Theories, mechanisms and future prospects. Ageing Res Rev 2016; 29:90-112. [PMID: 27353257 PMCID: PMC5991498 DOI: 10.1016/j.arr.2016.06.005] [Citation(s) in RCA: 239] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 06/23/2016] [Accepted: 06/23/2016] [Indexed: 12/31/2022]
Abstract
Answering the question as to why we age is tantamount to answering the question of what is life itself. There are countless theories as to why and how we age, but, until recently, the very definition of aging - senescence - was still uncertain. Here, we summarize the main views of the different models of senescence, with a special emphasis on the biochemical processes that accompany aging. Though inherently complex, aging is characterized by numerous changes that take place at different levels of the biological hierarchy. We therefore explore some of the most relevant changes that take place during aging and, finally, we overview the current status of emergent aging therapies and what the future holds for this field of research. From this multi-dimensional approach, it becomes clear that an integrative approach that couples aging research with systems biology, capable of providing novel insights into how and why we age, is necessary.
Collapse
Affiliation(s)
- João Pinto da Costa
- CESAM and Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal.
| | - Rui Vitorino
- Department of Medical Sciences, Institute for Biomedicine-iBiMED, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal; Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Gustavo M Silva
- Department of Biology, Center for Genomics and Systems Biology, NY, NY 10003, USA
| | - Christine Vogel
- Department of Biology, Center for Genomics and Systems Biology, NY, NY 10003, USA
| | - Armando C Duarte
- CESAM and Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| | - Teresa Rocha-Santos
- CESAM and Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| |
Collapse
|
427
|
Li T, Jiao W, Li W, Li H. Sex effect on insulin secretion and mitochondrial function in pancreatic beta cells of elderly Wistar rats. Endocr Res 2016; 41:167-79. [PMID: 26865180 DOI: 10.3109/07435800.2015.1124437] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AIMS/HYPOTHESIS Glucose tolerance progressively declines with age, and there is a high prevalence of type 2 diabetes in the elderly people. Previous studies have reported the sex differences in risk for type 2 diabetes, especially in the elderly people, whereas reasons for these sex differences remain poorly understood. This study aims to evaluate the effect of sex on glucose-stimulated insulin secretion and mitochondrial function in pancreatic beta cells of Wistar rats. METHODS 3-month-old and 18-month-old Wistar rats of both sexes were used. Insulin secretion of islets was analyzed by glucose-stimulated insulin secretion and islet perifusion assays; ATP content and oxygen consumption rate of islets were determined to evaluate the mitochondrial function. RESULTS Insulin secretion of islets under high glucose conditions declined significantly with age in both sexes. Glucose-stimulated insulin secretion of elderly female groups was markedly higher than that of male groups under high glucose conditions. Importantly, islets from elderly female groups showed higher mitochondrial function compared with male counterparts, evidenced by higher ATP content and oxygen consumption rate under high glucose conditions. It was also noted that mitochondrial biogenesis of islets from elderly female rats was significant higher compared with male rats. There were notable increases in expression of genes involved in mitochondrial biogenesis in islets from elderly female rats compared with male rats. CONCLUSION This study demonstrates a sex dimorphism in the age-associated impairment of pancreatic beta cell function in elderly rats, while the potential mechanism may be related to the sexual differences in mitochondrial biogenesis and function.
Collapse
Affiliation(s)
- Tianyi Li
- a Department of Elderly Endocrinology, First Affiliated Hospital , Zhengzhou University , Zhengzhou , China
| | - Wenjun Jiao
- a Department of Elderly Endocrinology, First Affiliated Hospital , Zhengzhou University , Zhengzhou , China
| | - Weifang Li
- a Department of Elderly Endocrinology, First Affiliated Hospital , Zhengzhou University , Zhengzhou , China
| | - Hua Li
- a Department of Elderly Endocrinology, First Affiliated Hospital , Zhengzhou University , Zhengzhou , China
| |
Collapse
|
428
|
Kim IH, Xu J, Liu X, Koyama Y, Ma HY, Diggle K, You YH, Schilling JM, Jeste D, Sharma K, Brenner DA, Kisseleva T. Aging increases the susceptibility of hepatic inflammation, liver fibrosis and aging in response to high-fat diet in mice. AGE (DORDRECHT, NETHERLANDS) 2016; 38:291-302. [PMID: 27578257 PMCID: PMC5061686 DOI: 10.1007/s11357-016-9938-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 07/14/2016] [Indexed: 04/17/2023]
Abstract
We aimed to investigate whether aging increases the susceptibility of hepatic and renal inflammation or fibrosis in response to high-fat diet (HFD) and explore the underlying genetic alterations. Middle (10 months old) and old (20 months old) aged, male C57BL/6N mice were fed either a low-fat diet (4 % fat) or HFD (60 % fat) for 4 months. Young (3 months old) aged mice were included as control group. HFD-induced liver and kidney injuries were analyzed by serum and urine assay, histologic staining, immunohistochemistry, and reverse-transcription real-time quantitative polymerase chain reaction. Total RNA sequencing with next-generation technology was done with RNA extracted from liver tissues. With HFD feeding, aged was associated with higher serum alanine aminotransferase levels, marked infiltration of hepatic macrophages, and increased expression of inflammatory cytokines (MCP1, TNF-α, IL-1β, IL-6, IL-12, IL-17A). Importantly, aged mice showed more advanced hepatic fibrosis and increased expression of fibrogenic markers (Col-I-α1, αSMA, TGF-β1, TGF-β2, TGFβRII, PDGF, PDGFRβII, TIMP1) in response to HFD. Aged mice fed on HFD also showed increased oxidative stress and TLR4 expression. In the total RNA seq and gene ontology analysis of liver, old-aged HFD group showed significant up-regulation of genes linked to innate immune response, immune response, defense response, inflammatory response compared to middle-aged HFD group. Meanwhile, aging and HFD feeding showed significant increase in glomerular size and mesangial area, higher urine albumin/creatinine ratio, and advanced renal inflammation or fibrosis. However, the difference of HFD-induced renal injury between old-aged group and middle-aged group was not significant. The susceptibility of hepatic fibrosis as well as hepatic inflammation in response to HFD was significantly increased with aging. In addition, aging was associated with glomerular alterations and increased renal inflammation or fibrosis, while the differential effect of aging on HFD-induced renal injury was not remarkable as shown in the liver.
Collapse
Affiliation(s)
- In Hee Kim
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive # 0702, La Jolla, San Diego, CA, 92093, USA
- Department of Internal Medicine, Chonbuk National University Medical School and Hospital, Jeonju, South Korea
| | - Jun Xu
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive # 0702, La Jolla, San Diego, CA, 92093, USA
| | - Xiao Liu
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive # 0702, La Jolla, San Diego, CA, 92093, USA
| | - Yukinori Koyama
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive # 0702, La Jolla, San Diego, CA, 92093, USA
| | - Hsiao-Yen Ma
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive # 0702, La Jolla, San Diego, CA, 92093, USA
| | - Karin Diggle
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive # 0702, La Jolla, San Diego, CA, 92093, USA
| | - Young-Hyun You
- Center for Renal Translational Medicine, Division of Nephrology-Hypertension, University of California, La Jolla, San Diego, CA, USA
| | - Jan M Schilling
- Department of Anesthesiology, University of California, La Jolla, San Diego, CA, USA
| | - Dilip Jeste
- Departments of Psychiatry and Neurosciences, and the Sam and Rose Stein Institute for Research on Aging, University of California, La Jolla, San Diego, CA, USA
| | - Kumar Sharma
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive # 0702, La Jolla, San Diego, CA, 92093, USA
| | - David A Brenner
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive # 0702, La Jolla, San Diego, CA, 92093, USA
| | - Tatiana Kisseleva
- Department of Medicine, School of Medicine, University of California, 9500 Gilman Drive # 0702, La Jolla, San Diego, CA, 92093, USA.
| |
Collapse
|
429
|
Bahrami SA, Bakhtiari N. Ursolic acid regulates aging process through enhancing of metabolic sensor proteins level. Biomed Pharmacother 2016; 82:8-14. [PMID: 27470332 DOI: 10.1016/j.biopha.2016.04.047] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 04/23/2016] [Accepted: 04/25/2016] [Indexed: 02/06/2023] Open
Abstract
We previously reported that Ursolic Acid (UA) ameliorates skeletal muscle performance through satellite cells proliferation and cellular energy status. In studying the potential role of the hypothalamus in aging, we developed a strategy to pursue UA effects on the hypothalamus anti-aging proteins such as; SIRT1, SIRT6, PGC-1β and α-Klotho. In this study, we used a model of aging animals (C57BL/6). UA dissolved in Corn oil (20mg/ml) and then administrated (200mg/Kg i.p injection) to mice, twice daily for 7days. After treatment times, the mice perfused and the hypothalamus isolated for preparing of tissue to Immunofluorescence microscopy. The data illustrated that UA significantly increased SIRT1 (∼3.5±0.3 folds) and SIRT-6 (∼1.5±0.2 folds) proteins overexpression (P<0.001). In addition, our results showed that UA enhanced α-Klotho (∼3.3±0.3) and PGC-1β (∼2.6±0.2 folds) proteins levels (P<0. 01). In this study, data were analyzed using SPSS 16 (ANOVA test). To the best of our knowledge, it seems that UA through enhancing of anti-aging biomarkers (SIRT1 and SIRT6) and PGC-1β in hypothalamus regulates aging-process and attenuates mitochondrial-related diseases. In regard to the key role of α-Klotho in aging, our data indicate that UA may be on the horizon to forestall diseases of aging.
Collapse
Affiliation(s)
- Soroush Alaghehband Bahrami
- Department of Biochemistry, Faculty of Basic Sciences, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
| | - Nuredin Bakhtiari
- Department of Biochemistry, Faculty of Basic Sciences, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran.
| |
Collapse
|
430
|
Fuku N, Díaz-Peña R, Arai Y, Abe Y, Pareja-Galeano H, Sanchis-Gomar F, Santos-Lozano A, Zempo H, Naito H, Murakami H, Miyachi M, Venturini L, Ricevuti G, Nobuyoshi H, Emanuele E, Lucia A. rs2802292 polymorphism in the FOXO3A gene and exceptional longevity in two ethnically distinct cohorts. Maturitas 2016; 92:110-114. [PMID: 27621247 DOI: 10.1016/j.maturitas.2016.07.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 07/24/2016] [Accepted: 07/26/2016] [Indexed: 01/14/2023]
Abstract
OBJECTIVES Previous studies have indicated that the rs2802292 polymorphism in the human forkhead box O3A (FOXO3A) gene might be associated with exceptional longevity (EL, i.e., living 100+ years), although the results are conflicting. STUDY DESIGN AND MAIN OUTCOME MEASURES Using a case-control design, we investigated the distribution of the rs2802292 polymorphism in two ethnically distinct cohorts of centenarians (cases) and younger adults (controls). The first cohort included Japanese individuals (733 centenarians and 820 controls) and the second was from Northern Italy (79 disease-free centenarians and 316 controls). RESULTS No statistically significant association was found between the rs2802292 polymorphism and EL in either cohort (either examined in their entirety or in a sex-based analysis). CONCLUSIONS In light of our negative findings, further research and resequencing efforts are needed to shed more light on the potential association between EL and FOXO3A polymorphisms.
Collapse
Affiliation(s)
- Noriyuki Fuku
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Roberto Díaz-Peña
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile; Hospital Universitari Institut Pere Mata, IISPV, URV. CIBERSAM, Reus, Spain.
| | - Yasumichi Arai
- Center for Supercentenarian Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Yukiko Abe
- Center for Supercentenarian Medical Research, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | - Hirofumi Zempo
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Hisashi Naito
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Haruka Murakami
- Department of Health Promotion and Exercise, National Institute of Health and Nutrition, NIBIOHN, Tokyo, Japan
| | - Motohiko Miyachi
- Department of Health Promotion and Exercise, National Institute of Health and Nutrition, NIBIOHN, Tokyo, Japan
| | - Letizia Venturini
- Department of Internal Medicine and Therapeutics, Cellular Pathophysiology and Clinical Immunology Laboratory, University of Pavia, Pavia, Italy
| | - Giovanni Ricevuti
- Department of Internal Medicine and Therapeutics, Cellular Pathophysiology and Clinical Immunology Laboratory, University of Pavia, Pavia, Italy
| | - Hirose Nobuyoshi
- Center for Supercentenarian Medical Research, Keio University School of Medicine, Tokyo, Japan
| | | | - Alejandro Lucia
- European University and Research Institute i + 12, Madrid, Spain
| |
Collapse
|
431
|
Rückgang der Nierenfunktion im Alter. Z Gerontol Geriatr 2016; 49:469-76. [DOI: 10.1007/s00391-016-1109-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 06/14/2016] [Accepted: 06/23/2016] [Indexed: 10/21/2022]
|
432
|
Abstract
Aging is a universal phenomenon in metazoans, characterized by a general decline of the organism physiology associated with an increased risk of mortality and morbidity. Aging of an organism correlates with a decline in function of its cells, as shown for muscle, immune, and neuronal cells. As the DNA content of most cells within an organism remains largely identical throughout the life span, age-associated transcriptional changes must be achieved by epigenetic mechanisms. However, how aging may impact on the epigenetic state of cells is only beginning to be understood. In light of a growing number of studies demonstrating that noncoding RNAs can provide molecular signals that regulate expression of protein-coding genes and define epigenetic states of cells, we hypothesize that noncoding RNAs could play a direct role in inducing age-associated profiles of gene expression. In this context, the role of long noncoding RNAs (lncRNAs) as regulators of gene expression might be important for the overall transcriptional landscape observed in aged human cells. The possible functions of lncRNAs and other noncoding RNAs, and their roles in the regulation of aging-related cellular pathways will be analyzed.
Collapse
|
433
|
Zhang L, Chopp M, Zhang Y, Xiong Y, Li C, Sadry N, Rhaleb I, Lu M, Zhang ZG. Diabetes Mellitus Impairs Cognitive Function in Middle-Aged Rats and Neurological Recovery in Middle-Aged Rats After Stroke. Stroke 2016; 47:2112-8. [PMID: 27387991 DOI: 10.1161/strokeaha.115.012578] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 06/10/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND PURPOSE Diabetes mellitus (DM) is a common metabolic disease among the middle-aged and older population, which leads to an increase of stroke incidence and poor stroke recovery. The present study was designed to investigate the impact of DM on brain damage and on ischemic brain repair after stroke in aging animals. METHODS DM was induced in middle-aged rats (13 months) by administration of nicotinamide and streptozotocin. Rats with confirmed hyperglycemia status 30 days after nicotinamide-streptozotocin injection and age-matched non-DM rats were subjected to embolic middle cerebral artery occlusion. RESULTS Middle-aged rats subjected to nicotinamide-streptozotocin injection became hyperglycemic and developed cognitive deficits 2 months after induction of DM. Histopathologic analysis revealed that there was sporadic vascular disruption, including cerebral microvascular thrombosis, blood-brain barrier leakage, and loss of paravascular aquaporin-4 in the hippocampi. Importantly, middle-aged DM rats subjected to stroke had exacerbated sensorimotor and cognitive deficits compared with age-matched non-DM ischemic rats during stroke recovery. Compared with age-matched non-DM ischemic rats, DM ischemic rats exhibited aggravated neurovascular disruption in the bilateral hippocampi and white matter, suppressed stroke-induced neurogenesis and oligodendrogenesis, and impaired dendritic/spine plasticity. However, DM did not enlarge infarct volume. CONCLUSIONS Our data suggest that DM exacerbates neurovascular damage and hinders brain repair processes, which likely contribute to the impairment of stroke recovery.
Collapse
Affiliation(s)
- Li Zhang
- From the Department of Neurology (L.Z., M.C., C.L., N.S., I.R., Z.G.Z.), Department of Neurosurgery (Y.Z., Y.X.), and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.).
| | - Michael Chopp
- From the Department of Neurology (L.Z., M.C., C.L., N.S., I.R., Z.G.Z.), Department of Neurosurgery (Y.Z., Y.X.), and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Yanlu Zhang
- From the Department of Neurology (L.Z., M.C., C.L., N.S., I.R., Z.G.Z.), Department of Neurosurgery (Y.Z., Y.X.), and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Ye Xiong
- From the Department of Neurology (L.Z., M.C., C.L., N.S., I.R., Z.G.Z.), Department of Neurosurgery (Y.Z., Y.X.), and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Chao Li
- From the Department of Neurology (L.Z., M.C., C.L., N.S., I.R., Z.G.Z.), Department of Neurosurgery (Y.Z., Y.X.), and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Neema Sadry
- From the Department of Neurology (L.Z., M.C., C.L., N.S., I.R., Z.G.Z.), Department of Neurosurgery (Y.Z., Y.X.), and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Imane Rhaleb
- From the Department of Neurology (L.Z., M.C., C.L., N.S., I.R., Z.G.Z.), Department of Neurosurgery (Y.Z., Y.X.), and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Mei Lu
- From the Department of Neurology (L.Z., M.C., C.L., N.S., I.R., Z.G.Z.), Department of Neurosurgery (Y.Z., Y.X.), and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Zheng Gang Zhang
- From the Department of Neurology (L.Z., M.C., C.L., N.S., I.R., Z.G.Z.), Department of Neurosurgery (Y.Z., Y.X.), and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| |
Collapse
|
434
|
Suchiang K, Sharma R. Age-dependent modulation of fasting and long-term dietary restriction on acetylcholinesterase in non-neuronal tissues of mice. Mol Cell Biochem 2016; 419:135-45. [PMID: 27379505 DOI: 10.1007/s11010-016-2757-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 06/21/2016] [Indexed: 12/18/2022]
Abstract
Dietary restriction (DR) without malnutrition is a robust intervention that extends lifespan and slows the onset of nervous system deficit and age-related diseases in diverse organisms. Acetylcholinesterase (AChE), a thoroughly studied enzyme better known for hydrolyzing acetylcholine (ACh) in neuronal tissues, has recently been linked with multiple unrelated biological functions in different non-neuronal tissues. In the present study, the activity and protein expression level of AChE in liver, heart, and kidney of young (1 month), adult (6 month), and aged (18 month) mice were investigated. We also studied age- and tissue-specific changes in AChE activity and protein expression level after the mice were subjected to 24-h fasting and long-term DR. Our results showed that AChE activity and protein expression in kidney and heart of aged mice decreased significantly in comparison with young mice. On the contrary, long-term DR decreases the AChE activity and the protein expression level in all tissues irrespective of ages studied. We summarized that changes in AChE with age in different tissues studied reflects its different roles at different phases of an organism's life. Conversely, the cumulative modulation manifested in the form of lowering AChE by long-term DR may prevent the futile synthesis and accumulation of unwanted AChE besides the added compensatory benefit of enhanced ACh availability needed during the period of starvation. This, in turn, may help in preventing the declining homeostatic roles of this important neurotransmitter in different tissues.
Collapse
Affiliation(s)
- Kitlangki Suchiang
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, 605 014, India.
| | - Ramesh Sharma
- Department of Biochemistry, North-Eastern Hill University, Shillong, Meghalaya, 793 022, India
| |
Collapse
|
435
|
Seo C, Hwang YH, Kim Y, Joo BS, Yee ST, Kim CM, Paik MJ. Metabolomic study of aging in mouse plasma by gas chromatography–mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1025:1-6. [DOI: 10.1016/j.jchromb.2016.04.052] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 04/19/2016] [Accepted: 04/30/2016] [Indexed: 11/16/2022]
|
436
|
Barzilai N, Crandall JP, Kritchevsky SB, Espeland MA. Metformin as a Tool to Target Aging. Cell Metab 2016; 23:1060-1065. [PMID: 27304507 PMCID: PMC5943638 DOI: 10.1016/j.cmet.2016.05.011] [Citation(s) in RCA: 719] [Impact Index Per Article: 79.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 05/16/2016] [Accepted: 05/25/2016] [Indexed: 02/07/2023]
Abstract
Aging has been targeted by genetic and dietary manipulation and by drugs in order to increase lifespan and health span in numerous models. Metformin, which has demonstrated protective effects against several age-related diseases in humans, will be tested in the TAME (Targeting Aging with Metformin) trial, as the initial step in the development of increasingly effective next-generation drugs.
Collapse
Affiliation(s)
- Nir Barzilai
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Jill P Crandall
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Stephen B Kritchevsky
- Wake Forest Older Americans Independence Center and the Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Mark A Espeland
- Wake Forest Older Americans Independence Center and the Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
437
|
Irianto J, Pfeifer CR, Ivanovska IL, Swift J, Discher DE. Nuclear lamins in cancer. Cell Mol Bioeng 2016; 9:258-267. [PMID: 27570565 PMCID: PMC4999255 DOI: 10.1007/s12195-016-0437-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 04/12/2016] [Indexed: 01/25/2023] Open
Abstract
Dysmorphic nuclei are commonly seen in cancers and provide strong motivation for studying the main structural proteins of nuclei, the lamins, in cancer. Past studies have also demonstrated the significance of microenvironment mechanics to cancer progression, which is extremely interesting because the lamina was recently shown to be mechanosensitive. Here, we review current knowledge relating cancer progression to lamina biophysics. Lamin levels can constrain cancer cell migration in 3D and thereby impede tumor growth, and lamins can also protect a cancer cell's genome. In addition, lamins can influence transcriptional regulators (RAR, SRF, YAP/TAZ) and chromosome conformation in lamina associated domains. Further investigation of the roles for lamins in cancer and even DNA damage may lead to new therapies or at least to a clearer understanding of lamins as bio-markers in cancer progression.
Collapse
Affiliation(s)
- Jerome Irianto
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Charlotte R. Pfeifer
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Irena L. Ivanovska
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joe Swift
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dennis E. Discher
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
438
|
Appelman-Dijkstra NM, Rijndorp M, Biermasz NR, Dekkers OM, Pereira AM. Effects of discontinuation of growth hormone replacement in adult GH-deficient patients: a cohort study and a systematic review of the literature. Eur J Endocrinol 2016; 174:705-16. [PMID: 26944562 DOI: 10.1530/eje-15-1086] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 03/04/2016] [Indexed: 11/08/2022]
Abstract
BACKGROUND Recombinant human growth hormone (rhGH) replacement is advocated in adult growth hormone-deficient (GHD) patients to increase bone mass and improve lipid profile, body composition, and quality of life. The long-term effects of discontinuation of rhGh replacement are unknown. METHODS This cohort study and systematic review aim to evaluate the long-term metabolic effects of discontinuation of rhGh replacement in adult GHD patients, with a subgroup analyses according to age (< or > 60 years). Data on anthropometry, lipids, glucose, and bone mass density (BMD) were assessed for 3 years after discontinuation. RESULTS Cohort study included 64 patients who had discontinued rhGh replacement for >12 months. Fat percentage increased from 31.5±9.5% to 33.8±9.0% (mean difference 2.3, P=0.003). BMI decreased only in subjects <60 years (P=0.014). Glucose, total cholesterol, and LDL-cholesterol levels did not change; however, the percentage of patients on statins increased slightly from 39% to 44%. HDL-C concentration increased only in patients <60 years (mean difference 0.2, P=0.043). Lumbar spine BMD did not change; however, femoral neck BMD and bone turnover markers decreased in subjects <60 years (P=0.001). Systematic review included eight studies (n=166 patients) with a follow-up duration of 6-18 months. Of the Please check the edit of the sentence 'Of the eight studies "'.eight studies, three qualified as low risk of bias and five as having an intermediate risk of bias. None of the studies reported handling of statins, bisphosphonates, and glucose-lowering medication or excluded patients using these medications. CONCLUSIONS In this study, discontinuation of rhGh replacement resulted in metabolic changes only in patients <60 years after 3 years. Further research warrants to determine the optimal strategies for (dis)continuation of rhGh replacement in adult patients with GHD.
Collapse
Affiliation(s)
- Natasha M Appelman-Dijkstra
- Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The Netherlands
| | - Marnick Rijndorp
- Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The Netherlands
| | - Nienke R Biermasz
- Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The Netherlands
| | - Olaf M Dekkers
- Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The Netherlands Department of Clinical EpidemiologyLeiden University Medical Center, Leiden, The Netherlands Department of Clinical EpidemiologyAarhus University, Aarhus, Denmark
| | - Alberto M Pereira
- Department of MedicineDivision of Endocrinology and Center for Endocrine Tumors, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
439
|
Reis FCG, Branquinho JLO, Brandão BB, Guerra BA, Silva ID, Frontini A, Thomou T, Sartini L, Cinti S, Kahn CR, Festuccia WT, Kowaltowski AJ, Mori MA. Fat-specific Dicer deficiency accelerates aging and mitigates several effects of dietary restriction in mice. Aging (Albany NY) 2016; 8:1201-1222. [PMID: 27241713 PMCID: PMC4931827 DOI: 10.18632/aging.100970] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 05/15/2016] [Indexed: 01/08/2023]
Abstract
Aging increases the risk of type 2 diabetes, and this can be prevented by dietary restriction (DR). We have previously shown that DR inhibits the downregulation of miRNAs and their processing enzymes - mainly Dicer - that occurs with aging in mouse white adipose tissue (WAT). Here we used fat-specific Dicer knockout mice (AdicerKO) to understand the contributions of adipose tissue Dicer to the metabolic effects of aging and DR. Metabolomic data uncovered a clear distinction between the serum metabolite profiles of Lox control and AdicerKO mice, with a notable elevation of branched-chain amino acids (BCAA) in AdicerKO. These profiles were associated with reduced oxidative metabolism and increased lactate in WAT of AdicerKO mice and were accompanied by structural and functional changes in mitochondria, particularly under DR. AdicerKO mice displayed increased mTORC1 activation in WAT and skeletal muscle, where Dicer expression is not affected. This was accompanied by accelerated age-associated insulin resistance and premature mortality. Moreover, DR-induced insulin sensitivity was abrogated in AdicerKO mice. This was reverted by rapamycin injection, demonstrating that insulin resistance in AdicerKO mice is caused by mTORC1 hyperactivation. Our study evidences a DR-modulated role for WAT Dicer in controlling metabolism and insulin resistance.
Collapse
Affiliation(s)
- Felipe C. G. Reis
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Jéssica L. O. Branquinho
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Bruna B. Brandão
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Beatriz A. Guerra
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ismael D. Silva
- Department of Gynecology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Andrea Frontini
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Thomas Thomou
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Loris Sartini
- Department of Clinical and Experimental Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Saverio Cinti
- Department of Clinical and Experimental Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - C. Ronald Kahn
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - William T. Festuccia
- Departament of Physiology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Alicia J. Kowaltowski
- Department of Biochemistry, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Marcelo A. Mori
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
- Department of Biochemistry and Tissue Biology, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, Brazil
| |
Collapse
|
440
|
Huffman DM, Schafer MJ, LeBrasseur NK. Energetic interventions for healthspan and resiliency with aging. Exp Gerontol 2016; 86:73-83. [PMID: 27260561 DOI: 10.1016/j.exger.2016.05.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 05/03/2016] [Accepted: 05/24/2016] [Indexed: 12/16/2022]
Abstract
Several behavioral and pharmacological strategies improve longevity, which is indicative of delayed organismal aging, with the most effective interventions extending both life- and healthspan. In free living creatures, maintaining health and function into old age requires resilience against a multitude of stressors. Conversely, in experimental settings, conventional housing of rodents limits exposure to such challenges, thereby obscuring an accurate assessment of resilience. Caloric restriction (CR) and exercise, as well as pharmacologic strategies (resveratrol, rapamycin, metformin, senolytics), are well established to improve indices of health and aging, but some paradoxical effects have been observed on resilience. For instance, CR potently retards the onset of age-related diseases, and improves lifespan to a greater extent than exercise in a variety of models. However, exercise has proven more consistently beneficial to organismal resilience against a broad array of stressors, including infections, surgery, wound healing and frailty. CR can improve cellular stress defenses and protect from frailty, but also impairs the response to infections, bed rest and healing. How an intervention will impact not only longevity, health and function, but also resiliency, is critical to better understanding translational implications. Thus, organismal robustness represents a critical, albeit understudied aspect of aging, which needs more careful attention in order to better inform on how putative age-delaying strategies will impact preservation of health and function in response to stressors with aging in humans.
Collapse
Affiliation(s)
- Derek M Huffman
- Department of Molecular Pharmacology and Medicine, Albert Einstein College of Medicine, Bronx, NY, USA; Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Marissa J Schafer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
441
|
Aunan JR, Watson MM, Hagland HR, Søreide K. Molecular and biological hallmarks of ageing. Br J Surg 2016; 103:e29-46. [PMID: 26771470 DOI: 10.1002/bjs.10053] [Citation(s) in RCA: 191] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 10/16/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND Ageing is the inevitable time-dependent decline in physiological organ function that eventually leads to death. Age is a major risk factor for many of the most common medical conditions, such as cardiovascular disease, cancer, diabetes and Alzheimer's disease. This study reviews currently known hallmarks of ageing and their clinical implications. METHODS A literature search of PubMed/MEDLINE was conducted covering the last decade. RESULTS Average life expectancy has increased dramatically over the past century and is estimated to increase even further. Maximum longevity, however, appears unchanged, suggesting a universal limitation to the human organism. Understanding the underlying molecular processes of ageing and health decline may suggest interventions that, if used at an early age, can prevent, delay, alleviate or even reverse age-related diseases. Hallmarks of ageing can be grouped into three main categories. The primary hallmarks cause damage to cellular functions: genomic instability, telomere attrition, epigenetic alterations and loss of proteostasis. These are followed by antagonistic responses to such damage: deregulated nutrient sensing, altered mitochondrial function and cellular senescence. Finally, integrative hallmarks are possible culprits of the clinical phenotype (stem cell exhaustion and altered intercellular communication), which ultimately contribute to the clinical effects of ageing as seen in physiological loss of reserve, organ decline and reduced function. CONCLUSION The sum of these molecular hallmarks produces the clinical picture of the elderly surgical patient: frailty, sarcopenia, anaemia, poor nutrition and a blunted immune response system. Improved understanding of the ageing processes may give rise to new biomarkers of risk or prognosis, novel treatment targets and translational approaches across disciplines that may improve outcomes.
Collapse
Affiliation(s)
- J R Aunan
- Gastrointestinal Translational Research Unit, Molecular Laboratory, Stavanger University Hospital, Stavanger, Norway.,Department of Gastrointestinal Surgery, Stavanger University Hospital, Stavanger, Norway
| | - M M Watson
- Gastrointestinal Translational Research Unit, Molecular Laboratory, Stavanger University Hospital, Stavanger, Norway.,Department of Gastrointestinal Surgery, Stavanger University Hospital, Stavanger, Norway
| | - H R Hagland
- Gastrointestinal Translational Research Unit, Molecular Laboratory, Stavanger University Hospital, Stavanger, Norway.,Centre for Organelle Research (CORE), Faculty of Science and Technology, University of Stavanger, Stavanger, Norway
| | - K Søreide
- Gastrointestinal Translational Research Unit, Molecular Laboratory, Stavanger University Hospital, Stavanger, Norway.,Department of Gastrointestinal Surgery, Stavanger University Hospital, Stavanger, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
442
|
Pérez LM, Pareja-Galeano H, Sanchis-Gomar F, Emanuele E, Lucia A, Gálvez BG. 'Adipaging': ageing and obesity share biological hallmarks related to a dysfunctional adipose tissue. J Physiol 2016; 594:3187-207. [PMID: 26926488 DOI: 10.1113/jp271691] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 01/21/2016] [Indexed: 12/15/2022] Open
Abstract
The increasing ageing of our societies is accompanied by a pandemic of obesity and related cardiometabolic disorders. Progressive dysfunction of the white adipose tissue is increasingly recognized as an important hallmark of the ageing process, which in turn contributes to metabolic alterations, multi-organ damage and a systemic pro-inflammatory state ('inflammageing'). On the other hand, obesity, the paradigm of adipose tissue dysfunction, shares numerous biological similarities with the normal ageing process such as chronic inflammation and multi-system alterations. Accordingly, understanding the interplay between accelerated ageing related to obesity and adipose tissue dysfunction is critical to gain insight into the ageing process in general as well as into the pathophysiology of obesity and other related conditions. Here we postulate the concept of 'adipaging' to illustrate the common links between ageing and obesity and the fact that, to a great extent, obese adults are prematurely aged individuals.
Collapse
Affiliation(s)
- Laura M Pérez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Helios Pareja-Galeano
- Universidad Europea de Madrid, Spain.,Research Institute Hospital 12 de Octubre ('i+12'), Madrid, Spain
| | | | | | - Alejandro Lucia
- Universidad Europea de Madrid, Spain.,Research Institute Hospital 12 de Octubre ('i+12'), Madrid, Spain
| | - Beatriz G Gálvez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Universidad Europea de Madrid, Spain
| |
Collapse
|
443
|
Gómez-Serrano M, Camafeita E, García-Santos E, López JA, Rubio MA, Sánchez-Pernaute A, Torres A, Vázquez J, Peral B. Proteome-wide alterations on adipose tissue from obese patients as age-, diabetes- and gender-specific hallmarks. Sci Rep 2016; 6:25756. [PMID: 27160966 PMCID: PMC4861930 DOI: 10.1038/srep25756] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 04/21/2016] [Indexed: 12/12/2022] Open
Abstract
Obesity is a main global health issue and an outstanding cause of morbidity and mortality predisposing to type 2 diabetes (T2DM) and cardiovascular diseases. Huge research efforts focused on gene expression, cellular signalling and metabolism in obesity have improved our understanding of these disorders; nevertheless, to bridge the gap between the regulation of gene expression and changes in signalling/metabolism, protein levels must be assessed. We have extensively analysed visceral adipose tissue from age-, T2DM- and gender-matched obese patients using high-throughput proteomics and systems biology methods to identify new biomarkers for the onset of T2DM in obesity, as well as to gain insight into the influence of aging and gender in these disorders. About 250 proteins showed significant abundance differences in the age, T2DM and gender comparisons. In diabetic patients, remarkable gender-specific hallmarks were discovered regarding redox status, immune response and adipose tissue accumulation. Both aging and T2DM processes were associated with mitochondrial remodelling, albeit through well-differentiated proteome changes. Systems biology analysis highlighted mitochondrial proteins that could play a key role in the age-dependent pathophysiology of T2DM. Our findings could serve as a framework for future research in Translational Medicine directed at improving the quality of life of obese patients.
Collapse
Affiliation(s)
- María Gómez-Serrano
- Instituto de Investigaciones Biomédicas, Alberto Sols, (IIBM); Consejo Superior de Investigaciones Científicas &Universidad Autónoma de Madrid (CSIC-UAM), Madrid, 28029, Spain
| | - Emilio Camafeita
- Laboratory of Cardiovascular Proteomics, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, 28029, Spain
| | - Eva García-Santos
- Instituto de Investigaciones Biomédicas, Alberto Sols, (IIBM); Consejo Superior de Investigaciones Científicas &Universidad Autónoma de Madrid (CSIC-UAM), Madrid, 28029, Spain
| | - Juan A López
- Laboratory of Cardiovascular Proteomics, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, 28029, Spain
| | - Miguel A Rubio
- Department of Endocrinology, Hospital Clínico San Carlos (IDISSC), Facultad de Medicina, Universidad Complutense, Madrid, 28040, Spain
| | - Andrés Sánchez-Pernaute
- Department of Surgery, Hospital Clínico San Carlos (IDISSC), Facultad de Medicina, Universidad Complutense, Madrid, 28040, Spain
| | - Antonio Torres
- Department of Surgery, Hospital Clínico San Carlos (IDISSC), Facultad de Medicina, Universidad Complutense, Madrid, 28040, Spain
| | - Jesús Vázquez
- Laboratory of Cardiovascular Proteomics, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, 28029, Spain
| | - Belén Peral
- Instituto de Investigaciones Biomédicas, Alberto Sols, (IIBM); Consejo Superior de Investigaciones Científicas &Universidad Autónoma de Madrid (CSIC-UAM), Madrid, 28029, Spain.,CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Spain
| |
Collapse
|
444
|
Morgan A, Mooney K, Wilkinson S, Pickles N, Mc Auley M. Cholesterol metabolism: A review of how ageing disrupts the biological mechanisms responsible for its regulation. Ageing Res Rev 2016; 27:108-124. [PMID: 27045039 DOI: 10.1016/j.arr.2016.03.008] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/22/2016] [Accepted: 03/30/2016] [Indexed: 02/06/2023]
Abstract
Cholesterol plays a vital role in the human body as a precursor of steroid hormones and bile acids, in addition to providing structure to cell membranes. Whole body cholesterol metabolism is maintained by a highly coordinated balancing act between cholesterol ingestion, synthesis, absorption, and excretion. The aim of this review is to discuss how ageing interacts with these processes. Firstly, we will present an overview of cholesterol metabolism. Following this, we discuss how the biological mechanisms which underpin cholesterol metabolism are effected by ageing. Included in this discussion are lipoprotein dynamics, cholesterol absorption/synthesis and the enterohepatic circulation/synthesis of bile acids. Moreover, we discuss the role of oxidative stress in the pathological progression of atherosclerosis and also discuss how cholesterol biosynthesis is effected by both the mammalian target of rapamycin and sirtuin pathways. Next, we examine how diet and alterations to the gut microbiome can be used to mitigate the impact ageing has on cholesterol metabolism. We conclude by discussing how mathematical models of cholesterol metabolism can be used to identify therapeutic interventions.
Collapse
|
445
|
Castillo-Quan JI, Li L, Kinghorn KJ, Ivanov DK, Tain LS, Slack C, Kerr F, Nespital T, Thornton J, Hardy J, Bjedov I, Partridge L. Lithium Promotes Longevity through GSK3/NRF2-Dependent Hormesis. Cell Rep 2016; 15:638-650. [PMID: 27068460 PMCID: PMC4850359 DOI: 10.1016/j.celrep.2016.03.041] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 01/31/2016] [Accepted: 03/10/2016] [Indexed: 01/06/2023] Open
Abstract
The quest to extend healthspan via pharmacological means is becoming increasingly urgent, both from a health and economic perspective. Here we show that lithium, a drug approved for human use, promotes longevity and healthspan. We demonstrate that lithium extends lifespan in female and male Drosophila, when administered throughout adulthood or only later in life. The life-extending mechanism involves the inhibition of glycogen synthase kinase-3 (GSK-3) and activation of the transcription factor nuclear factor erythroid 2-related factor (NRF-2). Combining genetic loss of the NRF-2 repressor Kelch-like ECH-associated protein 1 (Keap1) with lithium treatment revealed that high levels of NRF-2 activation conferred stress resistance, while low levels additionally promoted longevity. The discovery of GSK-3 as a therapeutic target for aging will likely lead to more effective treatments that can modulate mammalian aging and further improve health in later life.
Collapse
Affiliation(s)
- Jorge Iván Castillo-Quan
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London WC1E 6BT, UK; Max Planck Institute for Biology of Ageing, Joseph-Stelzmann Strasse 9-b, 50931 Köln, Germany; Department of Molecular Neuroscience, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Li Li
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London WC1E 6BT, UK; Department of Molecular Neuroscience, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Kerri J Kinghorn
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London WC1E 6BT, UK; Department of Molecular Neuroscience, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Dobril K Ivanov
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Luke S Tain
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann Strasse 9-b, 50931 Köln, Germany
| | - Cathy Slack
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London WC1E 6BT, UK; Max Planck Institute for Biology of Ageing, Joseph-Stelzmann Strasse 9-b, 50931 Köln, Germany
| | - Fiona Kerr
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London WC1E 6BT, UK
| | - Tobias Nespital
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann Strasse 9-b, 50931 Köln, Germany
| | - Janet Thornton
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - John Hardy
- Department of Molecular Neuroscience, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - Ivana Bjedov
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London WC1E 6BT, UK; UCL Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | - Linda Partridge
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London WC1E 6BT, UK; Max Planck Institute for Biology of Ageing, Joseph-Stelzmann Strasse 9-b, 50931 Köln, Germany.
| |
Collapse
|
446
|
Brouwers B, Dalmasso B, Hatse S, Laenen A, Kenis C, Swerts E, Neven P, Smeets A, Schöffski P, Wildiers H. Biological ageing and frailty markers in breast cancer patients. Aging (Albany NY) 2016; 7:319-33. [PMID: 25989735 PMCID: PMC4468313 DOI: 10.18632/aging.100745] [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] [Indexed: 11/25/2022]
Abstract
Older cancer patients are a highly heterogeneous population in terms of global health and physiological reserves, and it is often difficult to determine the best treatment. Moreover, clinical tools currently used to assess global health require dedicated time and lack a standardized end score. Circulating markers of biological age and/or fitness could complement or partially substitute the existing screening tools. In this study we explored the relationship of potential ageing/frailty biomarkers with age and clinical frailty. On a population of 82 young and 162 older non-metastatic breast cancer patients, we measured mean leukocyte telomere length and plasma levels of interleukin-6 (IL-6), regulated upon activation, normal T cell expressed and secreted (RANTES), monocyte chemotactic protein 1 (MCP-1), insulin-like growth factor 1 (IGF-1). We also developed a new tool to summarize clinical frailty, designated Leuven Oncogeriatric Frailty Score (LOFS), by integrating GA results in a single, semi-continuous score. LOFS' median score was 8, on a scale from 0=frail to 10=fit. IL-6 levels were associated with chronological age in both groups and with clinical frailty in older breast cancer patients, whereas telomere length, IGF-1 and MCP-1 only correlated with age. Plasma IL-6 should be further explored as frailty biomarker in cancer patients.
Collapse
Affiliation(s)
- Barbara Brouwers
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Bruna Dalmasso
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium.,Department of Internal Medicine, Istituto di Ricerca a Carattere Clinico e Scientifico (IRCCS), Azienda Ospedaliera Universitaria (AOU) San Martino Istituto Nazionale Tumori (IST), Genoa, Italy
| | - Sigrid Hatse
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Annouschka Laenen
- Interuniversity Centre for Biostatistics and Statistical Bioinformatics, Leuven, Belgium
| | - Cindy Kenis
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Evalien Swerts
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Patrick Neven
- Leuven Multidisciplinary Breast Center, University Hospitals Leuven, Belgium
| | - Ann Smeets
- Leuven Multidisciplinary Breast Center, University Hospitals Leuven, Belgium
| | - Patrick Schöffski
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Hans Wildiers
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven, and Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium.,Leuven Multidisciplinary Breast Center, University Hospitals Leuven, Belgium
| |
Collapse
|
447
|
Martins R, Lithgow GJ, Link W. Long live FOXO: unraveling the role of FOXO proteins in aging and longevity. Aging Cell 2016; 15:196-207. [PMID: 26643314 PMCID: PMC4783344 DOI: 10.1111/acel.12427] [Citation(s) in RCA: 515] [Impact Index Per Article: 57.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2015] [Indexed: 12/19/2022] Open
Abstract
Aging constitutes the key risk factor for age‐related diseases such as cancer and cardiovascular and neurodegenerative disorders. Human longevity and healthy aging are complex phenotypes influenced by both environmental and genetic factors. The fact that genetic contribution to lifespan strongly increases with greater age provides basis for research on which “protective genes” are carried by long‐lived individuals. Studies have consistently revealed FOXO (Forkhead box O) transcription factors as important determinants in aging and longevity. FOXO proteins represent a subfamily of transcription factors conserved from Caenorhabditis elegans to mammals that act as key regulators of longevity downstream of insulin and insulin‐like growth factor signaling. Invertebrate genomes have one FOXO gene, while mammals have four FOXO genes: FOXO1, FOXO3, FOXO4, and FOXO6. In mammals, this subfamily is involved in a wide range of crucial cellular processes regulating stress resistance, metabolism, cell cycle arrest, and apoptosis. Their role in longevity determination is complex and remains to be fully elucidated. Throughout this review, the mechanisms by which FOXO factors contribute to longevity will be discussed in diverse animal models, from Hydra to mammals. Moreover, compelling evidence of FOXOs as contributors for extreme longevity and health span in humans will be addressed.
Collapse
Affiliation(s)
- Rute Martins
- Regenerative Medicine Program Department of Biomedical Sciences and Medicine University of Algarve Campus de Gambelas 8005‐139 Faro Portugal
| | | | - Wolfgang Link
- Regenerative Medicine Program Department of Biomedical Sciences and Medicine University of Algarve Campus de Gambelas 8005‐139 Faro Portugal
- Centre for Biomedical Research (CBMR) University of Algarve Campus de Gambelas 8005‐139 Faro Portugal
| |
Collapse
|
448
|
Sierra F. The Emergence of Geroscience as an Interdisciplinary Approach to the Enhancement of Health Span and Life Span. Cold Spring Harb Perspect Med 2016; 6:a025163. [PMID: 26931460 PMCID: PMC4817738 DOI: 10.1101/cshperspect.a025163] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Research on the biology of aging has accelerated rapidly in the last two decades. It is now at the point where translation of the findings into useful approaches to improve the health of the elderly population seems possible. In trying to fill that gap, a new field termed geroscience will be articulated here that attempts to identify the biological underpinnings for the age-dependency of most chronic diseases. Herein, I will review the major conceptual issues leading to the formulation of geroscience as a field, as well as give examples of current areas of inquiry in which basic aging biology research could lead to therapeutic approaches to address age-related chronic diseases, not one at a time, but most of them in unison.
Collapse
Affiliation(s)
- Felipe Sierra
- Division of Aging Biology, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, 20892
| |
Collapse
|
449
|
Krishna KB, Stefanovic-Racic M, Dedousis N, Sipula I, O'Doherty RM. Similar degrees of obesity induced by diet or aging cause strikingly different immunologic and metabolic outcomes. Physiol Rep 2016; 4:4/6/e12708. [PMID: 27033445 PMCID: PMC4814885 DOI: 10.14814/phy2.12708] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 01/20/2016] [Indexed: 01/07/2023] Open
Abstract
In obesity, adipose tissue (AT) and liver are infiltrated with Th‐1 polarized immune cells, which are proposed to play an important role in the pathogenesis of the metabolic abnormalities of obesity. Aging is also associated with increased adiposity, but the effects of this increase on inflammation and associated metabolic dysfunction are poorly understood. To address this issue, we assessed insulin resistance (IR) and AT and liver immunophenotype in aged, lean (AL) and aged, obese (AO) mice, all of whom were maintained on a standard chow diet (11% fat diet) throughout their lives. For comparison, these variables were also assessed in young, lean (YL) and young diet‐induced obese mice (41% fat diet, YO). Despite similar body weight and fat accumulation, YO mice were substantially more IR and had greater liver steatosis compared to AO mice. YO also had elevated infiltration of macrophages/dendritic cells in AT and liver, but these increases were absent in AO. Furthermore, liver immune cells of YO were more Th‐1 polarized then AO. Notably, aging was associated with accumulation of T cells, but this occurred independent of obesity. Together, the data suggest that reduced inflammation in AO underlies the improved insulin sensitivity and lowered steatosis compared to YO.
Collapse
Affiliation(s)
- Kanthi B Krishna
- Division of Pediatric Endocrinology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Maja Stefanovic-Racic
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nikolaos Dedousis
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ian Sipula
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert M O'Doherty
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
450
|
Song J, Kim J. Role of Sirtuins in Linking Metabolic Syndrome with Depression. Front Cell Neurosci 2016; 10:86. [PMID: 27065808 PMCID: PMC4814520 DOI: 10.3389/fncel.2016.00086] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/21/2016] [Indexed: 12/20/2022] Open
Abstract
Depression is now widely regarded as a common disabling disorder that affects negatively the social functioning all over the world. Depression is associated with diverse phenomenon in brain such as neuroinflammation, synaptic dysfunction, and cognitive deficit. Recent studies reported that depression occurs by various metabolic changes, leading to metabolic syndrome. Sirtuins (SIRTs) are NAD+-dependent class III histone deacetylases, known to regulate diverse biological mechanism such as longevity, genomic stability, and inflammation. The modulation of sirtuin activity has been highlighted as a promising approach to reduce neurodegenerative processes. In this review, we summarize the recent discoveries regarding the potential relationship between SIRTs and depression caused by metabolic disorders (Mets). Ultimately, we suggest the possibility that SIRTs will be novel targets to alleviate neuropathogenesis induced by depression.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Biomedical Engineering, Dongguk University Seoul, South Korea
| | - Jongpil Kim
- Department of Biomedical Engineering, Dongguk University Seoul, South Korea
| |
Collapse
|