301
|
Mikkola TM, Salonen MK, Kajantie E, Kautiainen H, Eriksson JG. Associations of Fat and Lean Body Mass with Circulating Amino Acids in Older Men and Women. J Gerontol A Biol Sci Med Sci 2021; 75:885-891. [PMID: 31095700 DOI: 10.1093/gerona/glz126] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Indexed: 12/16/2022] Open
Abstract
Circulating amino acids are potential markers of body composition. Previous studies are mainly limited to middle age and focus on either fat or lean mass, thereby ignoring overall body composition. We investigated the associations of fat and lean body mass with circulating amino acids in older men and women. We studied 594 women and 476 men from the Helsinki Birth Cohort Study (age 62-74 years). Bioelectrical impedance analysis was used to indicate two main body compartments by fat (fat mass/height2) and lean mass indices (lean mass/height2), dichotomized based on sex-specific medians. Eight serum amino acids were quantified using nuclear magnetic resonance spectroscopy. General linear models were adjusted for age, smoking, and fasting glucose. Higher lean mass index (LMI) was associated with higher concentrations of branched-chain amino acids in both sexes (p ≤ .001). In men, LMI was also positively associated with tyrosine (p = .006) and inversely with glycine (p < .001). Higher fat mass index was associated with higher concentrations of all branched-chain amino acids, aromatic amino acids (phenylalanine and tyrosine), and alanine in both sexes (p ≤ .008). Associations between body composition and amino acids are largely similar in older men and women. The associations are largely similar to those previously observed in younger adults.
Collapse
Affiliation(s)
| | - Minna K Salonen
- Folkhälsan Research Center, Helsinki, Finland.,Public Health Promotion Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Eero Kajantie
- Public Health Promotion Unit, National Institute for Health and Welfare, Helsinki, Finland.,PEDEGO Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, Finland.,Department of Clinical and Molecular Medicine, Norwegian University for Science and Technology, Trondheim, Norway.,Children's Hospital, Helsinki University Hospital and University of Helsinki, Finland
| | - Hannu Kautiainen
- Folkhälsan Research Center, Helsinki, Finland.,Primary Health Care Unit, Kuopio University Hospital, Finland
| | - Johan G Eriksson
- Folkhälsan Research Center, Helsinki, Finland.,Public Health Promotion Unit, National Institute for Health and Welfare, Helsinki, Finland.,Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Finland.,Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research.,Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore
| |
Collapse
|
302
|
Bcl-xL as a Modulator of Senescence and Aging. Int J Mol Sci 2021; 22:ijms22041527. [PMID: 33546395 PMCID: PMC7913597 DOI: 10.3390/ijms22041527] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 02/06/2023] Open
Abstract
Many features of aging result from the incapacity of cells to adapt to stress conditions. When cells are overwhelmed by stress, they can undergo senescence to avoid unrestricted growth of damaged cells. Recent findings have proven that cellular senescence is more than that. A specific grade of senescence promotes embryo development, tissue remodeling and wound healing. However, constant stresses and a weakening immune system can lead to senescence chronicity with aging. The accumulation of senescent cells is directly related to tissue dysfunction and age-related pathologies. Centenarians, the most aged individuals, should accumulate senescent cells and suffer from their deleterious effects, however, they enjoy a compression of morbidity. We have shown that they overexpress B-cell lymphoma-extra large (Bcl-xL). Bcl-xL could avoid an excessive burden of senescent cells through the regulation of intrinsic apoptosis, mitochondrial bioenergetics and oxidative stress. On the other hand, Bcl-xL maintains a fully functional immune system that ensures an efficient clearance of senescent cells. Moreover, there is a paradox, as inhibitors of Bcl-xL have been employed as senolytic agents, which have been shown to protect from aging in animal models. In this review, we aim to discuss how Bcl-xL could modulate senescence-associated harmful effects in centenarians, protecting them from the burden of accumulation of senescent cells.
Collapse
|
303
|
Effects of Long-Term DHA Supplementation and Physical Exercise on Non-Alcoholic Fatty Liver Development in Obese Aged Female Mice. Nutrients 2021; 13:nu13020501. [PMID: 33546405 PMCID: PMC7913512 DOI: 10.3390/nu13020501] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
Obesity and aging are associated to non-alcoholic fatty liver disease (NAFLD) development. Here, we investigate whether long-term feeding with a docosahexaenoic acid (DHA)-enriched diet and aerobic exercise, alone or in combination, are effective in ameliorating NAFLD in aged obese mice. Two-month-old female C57BL/6J mice received control or high fat diet (HFD) for 4 months. Then, the diet-induced obese (DIO) mice were distributed into four groups: DIO, DIO + DHA (15% dietary lipids replaced by a DHA-rich concentrate), DIO + EX (treadmill running), and DIO + DHA + EX up to 18 months. The DHA-rich diet reduced liver steatosis in DIO mice, decreasing lipogenic genes (Dgat2, Scd1, Srebp1c), and upregulated lipid catabolism genes (Hsl/Acox) expression. A similar pattern was observed in the DIO + EX group. The combination of DHA + exercise potentiated an increase in Cpt1a and Ppara genes, and AMPK activation, key regulators of fatty acid oxidation. Exercise, alone or in combination with DHA, significantly reversed the induction of proinflammatory genes (Mcp1, Il6, Tnfα, Tlr4) in DIO mice. DHA supplementation was effective in preventing the alterations induced by the HFD in endoplasmic reticulum stress-related genes (Ern1/Xbp1) and autophagy markers (LC3II/I ratio, p62, Atg7). In summary, long-term DHA supplementation and/or exercise could be helpful to delay NAFLD progression during aging in obesity.
Collapse
|
304
|
Zhang B, Gems D. Gross ways to live long: Parasitic worms as an anti-inflammaging therapy? eLife 2021; 10:65180. [PMID: 33526169 PMCID: PMC7853715 DOI: 10.7554/elife.65180] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 01/05/2021] [Indexed: 12/11/2022] Open
Abstract
Evolutionary medicine argues that disease can arise because modern conditions do not match those in which we evolved. For example, a decline in exposure to commensal microbes and gastrointestinal helminths in developed countries has been linked to increased prevalence of allergic and autoimmune inflammatory disorders (the hygiene hypothesis). Accordingly, probiotic therapies that restore ‘old friend’ microbes and helminths have been explored as Darwinian treatments for these disorders. A further possibility is that loss of old friend commensals also increases the sterile, aging-associated inflammation known as inflammaging, which contributes to a range of age-related diseases, including cardiovascular disease, dementia, and cancer. Interestingly, Crowe et al., 2020 recently reported that treatment with a secreted glycoprotein from a parasitic nematode can protect against murine aging by induction of anti-inflammatory mechanisms. Here, we explore the hypothesis that restorative helminth therapy would have anti-inflammaging effects. Could worm infections provide broad-spectrum protection against age-related disease?
Collapse
Affiliation(s)
- Bruce Zhang
- Institute of Healthy Ageing, and Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| | - David Gems
- Institute of Healthy Ageing, and Research Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| |
Collapse
|
305
|
Wilhelmsen A, Tsintzas K, Jones SW. Recent advances and future avenues in understanding the role of adipose tissue cross talk in mediating skeletal muscle mass and function with ageing. GeroScience 2021; 43:85-110. [PMID: 33528828 PMCID: PMC8050140 DOI: 10.1007/s11357-021-00322-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/01/2021] [Indexed: 12/15/2022] Open
Abstract
Sarcopenia, broadly defined as the age-related decline in skeletal muscle mass, quality, and function, is associated with chronic low-grade inflammation and an increased likelihood of adverse health outcomes. The regulation of skeletal muscle mass with ageing is complex and necessitates a delicate balance between muscle protein synthesis and degradation. The secretion and transfer of cytokines, long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), both discretely and within extracellular vesicles, have emerged as important communication channels between tissues. Some of these factors have been implicated in regulating skeletal muscle mass, function, and pathologies and may be perturbed by excessive adiposity. Indeed, adipose tissue participates in a broad spectrum of inter-organ communication and obesity promotes the accumulation of macrophages, cellular senescence, and the production and secretion of pro-inflammatory factors. Pertinently, age-related sarcopenia has been reported to be more prevalent in obesity; however, such effects are confounded by comorbidities and physical activity level. In this review, we provide evidence that adiposity may exacerbate age-related sarcopenia and outline some emerging concepts of adipose-skeletal muscle communication including the secretion and processing of novel myokines and adipokines and the role of extracellular vesicles in mediating inter-tissue cross talk via lncRNAs and miRNAs in the context of sarcopenia, ageing, and obesity. Further research using advances in proteomics, transcriptomics, and techniques to investigate extracellular vesicles, with an emphasis on translational, longitudinal human studies, is required to better understand the physiological significance of these factors, the impact of obesity upon them, and their potential as therapeutic targets in combating muscle wasting.
Collapse
Affiliation(s)
- Andrew Wilhelmsen
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Kostas Tsintzas
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK.
| | - Simon W Jones
- Institute of Inflammation and Ageing, MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, Queen Elizabeth Hospital, The University of Birmingham, Birmingham, UK
| |
Collapse
|
306
|
Martyniak K, Wei F, Ballesteros A, Meckmongkol T, Calder A, Gilbertson T, Orlovskaya N, Coathup MJ. Do polyunsaturated fatty acids protect against bone loss in our aging and osteoporotic population? Bone 2021; 143:115736. [PMID: 33171312 DOI: 10.1016/j.bone.2020.115736] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 11/02/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023]
Abstract
Age-related bone loss is inevitable in both men and women and there will soon be more people of extreme old age than ever before. Osteoporosis is a common chronic disease and as the proportion of older people, rate of obesity and the length of life increases, a rise in age-related degenerating bone diseases, disability, and prolonged dependency is projected. Fragility fractures are one of the most severe complications associated with both primary and secondary osteoporosis and current treatment strategies target weight-bearing exercise and pharmacological intervention, both with limited long-term success. Obesity and osteoporosis are intimately interrelated, and diet is a variable that plays a significant role in bone regeneration and repair. The Western Diet is characterized by its unhealthy components, specifically excess amounts of saturated fat intake. This review examines the impact of saturated and polyunsaturated fatty acid consumption on chronic inflammation, osteogenesis, bone architecture, and strength and explores the hypothesis that dietary polyunsaturated fats have a beneficial effect on osteogenesis, reducing bone loss by decreasing chronic inflammation, and activating bone resorption through key cellular and molecular mechanisms in our aging population. We conclude that aging, obesity and a diet high in saturated fatty acids significantly impairs bone regeneration and repair and that consumption of ω-3 polyunsaturated fatty acids is associated with significantly increased bone regeneration, improved microarchitecture and structural strength. However, ω-6 polyunsaturated fatty acids were typically pro-inflammatory and have been associated with an increased fracture risk. This review suggests a potential role for ω-3 fatty acids as a non-pharmacological dietary method of reducing bone loss in our aging population. We also conclude that contemporary amendments to the formal nutritional recommendations made by the Food and Nutrition Board may be necessary such that our aging population is directly considered.
Collapse
Affiliation(s)
- Kari Martyniak
- Biionix Cluster, University of Central Florida, Orlando, FL, United States; Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Fei Wei
- Biionix Cluster, University of Central Florida, Orlando, FL, United States; Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Amelia Ballesteros
- Biionix Cluster, University of Central Florida, Orlando, FL, United States; Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Teerin Meckmongkol
- Biionix Cluster, University of Central Florida, Orlando, FL, United States; Department of General Surgery, Nemours Children's Hospital, Orlando, FL, United States
| | - Ashley Calder
- Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Timothy Gilbertson
- Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Nina Orlovskaya
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, United States
| | - Melanie J Coathup
- Biionix Cluster, University of Central Florida, Orlando, FL, United States; Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, United States.
| |
Collapse
|
307
|
Abstract
Significance: Cell senescence was originally defined by an acute loss of replicative capacity and thus believed to be restricted to proliferation-competent cells. More recently, senescence has been recognized as a cellular stress and damage response encompassing multiple pathways or senescence domains, namely DNA damage response, cell cycle arrest, senescence-associated secretory phenotype, senescence-associated mitochondrial dysfunction, autophagy/mitophagy dysfunction, nutrient and stress signaling, and epigenetic reprogramming. Each of these domains is activated during senescence, and all appear to interact with each other. Cell senescence has been identified as an important driver of mammalian aging. Recent Advances: Activation of all these senescence domains has now also been observed in a wide range of post-mitotic cells, suggesting that senescence as a stress response can occur in nondividing cells temporally uncoupled from cell cycle arrest. Here, we review recent evidence for post-mitotic cell senescence and speculate about its possible relevance for mammalian aging. Critical Issues: Although a majority of senescence domains has been found to be activated in a range of post-mitotic cells during aging, independent confirmation of these results is still lacking for most of them. Future Directions: To define whether post-mitotic senescence plays a significant role as a driver of aging phenotypes in tissues such as brain, muscle, heart, and others. Antioxid. Redox Signal. 34, 308-323.
Collapse
Affiliation(s)
- Thomas von Zglinicki
- Ageing Research Laboratories, Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom.,Molecular Biology and Genetics, Arts and Sciences Faculty, Near East University, Nicosia, Turkey
| | - Tengfei Wan
- Ageing Research Laboratories, Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Satomi Miwa
- Ageing Research Laboratories, Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
308
|
Bermudez V, Salazar J, Martínez MS, Olivar LC, Nava M, Rojas M, Ortega Á, Añez R, Toledo A, Rojas J, Chacín M, Rodríguez JE, D'Marco L, Cano C. Age-specific waist circumference cutoff-points for abdominal obesity diagnosis: a personalized strategy for a large Venezuelan population. J Diabetes Metab Disord 2021; 20:217-227. [PMID: 34178833 DOI: 10.1007/s40200-021-00735-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 01/05/2021] [Indexed: 10/22/2022]
Abstract
Background Evidence shows that the ageing process is a determining factor in fat distribution, composition, and functionality. The goal of this research was to determine cut-off points for waist circumference according to age in the adult population from Maracaibo city, Venezuela. Methodology The Metabolic Syndrome Prevalence Study is a descriptive, cross-sectional study with multi-stage randomized sampling. In this post-hoc analysis 1902 individuals ≥18 years and from both sexes were evaluated. Waist circumference ROC curves were built for each age group and sex, using metabolic phenotypes for classification. Results 52.2% (n = 992) were women, and the mean age was 38.7 ± 2. Cut-off points obtained for the <30 years age group were: 91 cm for women (Sensitivity: 96,8%, Specificity: 97,7%) and 94 cm for men (Sensitivity:100%, Specificity: 99,2%); for 30-49 years: women 94 cm (Sensitivity: 93.7%, Specificity: 97.1%) and men 95 cm (Sensitivity: 97.3%, Specificity: 100%); for ≥50 years: women 94 cm (Sensitivity: 91.8%, Specificity: 86.7%) and men 101 cm (Sensitivity: 100%, Specificity: 100%). Conclusion The use of specific cut-off points according to age groups is proposed to determine abdominal obesity in Maracaibo city due to the underestimation seen in young people and the overestimation observed in older people when using a unique cut-off point.
Collapse
Affiliation(s)
- Valmore Bermudez
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Juan Salazar
- Endocrine and Metabolic Disease Research Center. School of Medicine, University of Zulia, Maracaibo, 4004 Venezuela
| | - María Sofía Martínez
- Endocrine and Metabolic Disease Research Center. School of Medicine, University of Zulia, Maracaibo, 4004 Venezuela
| | - Luis Carlos Olivar
- Endocrine and Metabolic Disease Research Center. School of Medicine, University of Zulia, Maracaibo, 4004 Venezuela
| | - Manuel Nava
- Endocrine and Metabolic Disease Research Center. School of Medicine, University of Zulia, Maracaibo, 4004 Venezuela
| | - Milagros Rojas
- Endocrine and Metabolic Disease Research Center. School of Medicine, University of Zulia, Maracaibo, 4004 Venezuela
| | - Ángel Ortega
- Endocrine and Metabolic Disease Research Center. School of Medicine, University of Zulia, Maracaibo, 4004 Venezuela
| | - Roberto Añez
- Departamento de Endocrinología y Nutrición, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Alexandra Toledo
- Endocrine and Metabolic Disease Research Center. School of Medicine, University of Zulia, Maracaibo, 4004 Venezuela
| | - Joselyn Rojas
- Pulmonary and Critical Care Medicine Department, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Maricarmen Chacín
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Johel E Rodríguez
- Facultad de Ingenierías, Universidad Simón Bolívar, Cúcuta, Colombia
| | - Luis D'Marco
- Hospital Clínico de Valencia, INCLIVA, Servicio de Nefrología, Valencia, Spain
| | - Clímaco Cano
- Endocrine and Metabolic Disease Research Center. School of Medicine, University of Zulia, Maracaibo, 4004 Venezuela
| |
Collapse
|
309
|
Ting KK, Coleman P, Zhao Y, Vadas MA, Gamble JR. The aging endothelium. VASCULAR BIOLOGY 2021; 3:R35-R47. [PMID: 33880430 PMCID: PMC8052565 DOI: 10.1530/vb-20-0013] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 01/12/2021] [Indexed: 01/10/2023]
Abstract
Cellular senescence is now recognized as one of the hallmarks of aging. Herein, we examine current findings on senescence of the vascular endothelium and its impacts on age-related vascular diseases. Endothelial senescence can result in systemic metabolic changes, implicating senescence in chronic diseases such as diabetes, obesity and atherosclerosis. Senolytics, drugs that eliminate senescent cells, afford new therapeutic strategies for control of these chronic diseases.
Collapse
Affiliation(s)
- Ka Ka Ting
- Centre for the Endothelium Vascular Biology Program Centenary Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Paul Coleman
- Centre for the Endothelium Vascular Biology Program Centenary Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Yang Zhao
- Centre for the Endothelium Vascular Biology Program Centenary Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Mathew A Vadas
- Centre for the Endothelium Vascular Biology Program Centenary Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Jennifer R Gamble
- Centre for the Endothelium Vascular Biology Program Centenary Institute, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
310
|
Cao J, Li J, Yang X, Li P, Yao Z, Han D, Ying L, Wang L, Tian J. Transcriptomics analysis for the identification of potential age-related genes and cells associated with three major urogenital cancers. Sci Rep 2021; 11:641. [PMID: 33436826 PMCID: PMC7803945 DOI: 10.1038/s41598-020-80065-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/16/2020] [Indexed: 02/08/2023] Open
Abstract
Age is one of the most important risk factors of the occurrence for tumor patients. The majority of patients with urogenital cancers are the elderly, whose clinical characteristics are greatly affected by age and ageing. Our study aimed to explore age-related genes, cells, and biological changes in three common urogenital cancers via integrative bioinformatics analysis. First, mRNA (count format) and clinical data for bladder cancer, prostate cancer and renal cell carcinoma were downloaded from the Cancer Genome Atlas (TCGA). Through the comparison of clinicopathological characteristics, genes expression and cells infiltration between the old group and the young group, it was found that the clinical characteristics, genes and cells in the tumor microenvironment of different ages were quite different. And 4 key cells, 14 hub genes and some potential pathways were identified and considered as important factors. More importantly, we analyzed the differential landscape of the genes and cells from different perspectives, and confirmed its importance. In conclusion, we identified genes and cell types associated with age-related changes in the tumour microenvironment in urogenital cancer patients. These genes and cell types may play a critical role in the age-associated differences in clinicopathological characteristics among urogenital cancers, thus providing a link between ageing and cancer occurrence. The findings of this study may pave the way for the development of age-tailored approaches to treat cancer and other age-related diseases.
Collapse
Affiliation(s)
- Jinlong Cao
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
- Key Laboratory of Urological Diseases of Gansu Provincial, Lanzhou, 730000, People's Republic of China
| | - Jianpeng Li
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
- Key Laboratory of Urological Diseases of Gansu Provincial, Lanzhou, 730000, People's Republic of China
| | - Xin Yang
- Reproductive Medicine Center, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Pan Li
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
- Key Laboratory of Urological Diseases of Gansu Provincial, Lanzhou, 730000, People's Republic of China
| | - Zhiqiang Yao
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
- Key Laboratory of Urological Diseases of Gansu Provincial, Lanzhou, 730000, People's Republic of China
| | - Dali Han
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
- Key Laboratory of Urological Diseases of Gansu Provincial, Lanzhou, 730000, People's Republic of China
| | - Lijun Ying
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
- Key Laboratory of Urological Diseases of Gansu Provincial, Lanzhou, 730000, People's Republic of China
| | - Lijie Wang
- Department of Gynecology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Junqiang Tian
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China.
- Key Laboratory of Urological Diseases of Gansu Provincial, Lanzhou, 730000, People's Republic of China.
| |
Collapse
|
311
|
Sazonova MA, Sinyov VV, Ryzhkova AI, Sazonova MD, Kirichenko TV, Khotina VA, Khasanova ZB, Doroschuk NA, Karagodin VP, Orekhov AN, Sobenin IA. Some Molecular and Cellular Stress Mechanisms Associated with Neurodegenerative Diseases and Atherosclerosis. Int J Mol Sci 2021; 22:E699. [PMID: 33445687 PMCID: PMC7828120 DOI: 10.3390/ijms22020699] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic stress is a combination of nonspecific adaptive reactions of the body to the influence of various adverse stress factors which disrupt its homeostasis, and it is also a corresponding state of the organism's nervous system (or the body in general). We hypothesized that chronic stress may be one of the causes occurence of several molecular and cellular types of stress. We analyzed literary sources and considered most of these types of stress in our review article. We examined genes and mutations of nuclear and mitochondrial genomes and also molecular variants which lead to various types of stress. The end result of chronic stress can be metabolic disturbance in humans and animals, leading to accumulation of reactive oxygen species (ROS), oxidative stress, energy deficiency in cells (due to a decrease in ATP synthesis) and mitochondrial dysfunction. These changes can last for the lifetime and lead to severe pathologies, including neurodegenerative diseases and atherosclerosis. The analysis of literature allowed us to conclude that under the influence of chronic stress, metabolism in the human body can be disrupted, mutations of the mitochondrial and nuclear genome and dysfunction of cells and their compartments can occur. As a result of these processes, oxidative, genotoxic, and cellular stress can occur. Therefore, chronic stress can be one of the causes forthe occurrence and development of neurodegenerative diseases and atherosclerosis. In particular, chronic stress can play a large role in the occurrence and development of oxidative, genotoxic, and cellular types of stress.
Collapse
Affiliation(s)
- Margarita A. Sazonova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia; (V.V.S.); (A.I.R.); (M.D.S.); (T.V.K.); (V.A.K.); (V.P.K.); (A.N.O.); (I.A.S.)
- Laboratory of Medical Genetics, National Medical Research Center of Cardiology, 121552 Moscow, Russia; (Z.B.K.); (N.A.D.)
| | - Vasily V. Sinyov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia; (V.V.S.); (A.I.R.); (M.D.S.); (T.V.K.); (V.A.K.); (V.P.K.); (A.N.O.); (I.A.S.)
- Laboratory of Medical Genetics, National Medical Research Center of Cardiology, 121552 Moscow, Russia; (Z.B.K.); (N.A.D.)
| | - Anastasia I. Ryzhkova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia; (V.V.S.); (A.I.R.); (M.D.S.); (T.V.K.); (V.A.K.); (V.P.K.); (A.N.O.); (I.A.S.)
| | - Marina D. Sazonova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia; (V.V.S.); (A.I.R.); (M.D.S.); (T.V.K.); (V.A.K.); (V.P.K.); (A.N.O.); (I.A.S.)
| | - Tatiana V. Kirichenko
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia; (V.V.S.); (A.I.R.); (M.D.S.); (T.V.K.); (V.A.K.); (V.P.K.); (A.N.O.); (I.A.S.)
- Laboratory of Medical Genetics, National Medical Research Center of Cardiology, 121552 Moscow, Russia; (Z.B.K.); (N.A.D.)
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Research Institute of Human Morphology, 117418 Moscow, Russia
| | - Victoria A. Khotina
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia; (V.V.S.); (A.I.R.); (M.D.S.); (T.V.K.); (V.A.K.); (V.P.K.); (A.N.O.); (I.A.S.)
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Research Institute of Human Morphology, 117418 Moscow, Russia
| | - Zukhra B. Khasanova
- Laboratory of Medical Genetics, National Medical Research Center of Cardiology, 121552 Moscow, Russia; (Z.B.K.); (N.A.D.)
| | - Natalya A. Doroschuk
- Laboratory of Medical Genetics, National Medical Research Center of Cardiology, 121552 Moscow, Russia; (Z.B.K.); (N.A.D.)
| | - Vasily P. Karagodin
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia; (V.V.S.); (A.I.R.); (M.D.S.); (T.V.K.); (V.A.K.); (V.P.K.); (A.N.O.); (I.A.S.)
- Department of Commodity Science and Expertise, Plekhanov Russian University of Economics, 125993 Moscow, Russia
| | - Alexander N. Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia; (V.V.S.); (A.I.R.); (M.D.S.); (T.V.K.); (V.A.K.); (V.P.K.); (A.N.O.); (I.A.S.)
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Research Institute of Human Morphology, 117418 Moscow, Russia
- Institute for Atherosclerosis Research, Skolkovo Innovative Centre, 143024 Moscow, Russia
| | - Igor A. Sobenin
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia; (V.V.S.); (A.I.R.); (M.D.S.); (T.V.K.); (V.A.K.); (V.P.K.); (A.N.O.); (I.A.S.)
- Laboratory of Medical Genetics, National Medical Research Center of Cardiology, 121552 Moscow, Russia; (Z.B.K.); (N.A.D.)
| |
Collapse
|
312
|
Stojakovic A, Trushin S, Sheu A, Khalili L, Chang SY, Li X, Christensen T, Salisbury JL, Geroux RE, Gateno B, Flannery PJ, Dehankar M, Funk CC, Wilkins J, Stepanova A, O'Hagan T, Galkin A, Nesbitt J, Zhu X, Tripathi U, Macura S, Tchkonia T, Pirtskhalava T, Kirkland JL, Kudgus RA, Schoon RA, Reid JM, Yamazaki Y, Kanekiyo T, Zhang S, Nemutlu E, Dzeja P, Jaspersen A, Kwon YIC, Lee MK, Trushina E. Partial inhibition of mitochondrial complex I ameliorates Alzheimer's disease pathology and cognition in APP/PS1 female mice. Commun Biol 2021; 4:61. [PMID: 33420340 PMCID: PMC7794523 DOI: 10.1038/s42003-020-01584-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 12/08/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's Disease (AD) is a devastating neurodegenerative disorder without a cure. Here we show that mitochondrial respiratory chain complex I is an important small molecule druggable target in AD. Partial inhibition of complex I triggers the AMP-activated protein kinase-dependent signaling network leading to neuroprotection in symptomatic APP/PS1 female mice, a translational model of AD. Treatment of symptomatic APP/PS1 mice with complex I inhibitor improved energy homeostasis, synaptic activity, long-term potentiation, dendritic spine maturation, cognitive function and proteostasis, and reduced oxidative stress and inflammation in brain and periphery, ultimately blocking the ongoing neurodegeneration. Therapeutic efficacy in vivo was monitored using translational biomarkers FDG-PET, 31P NMR, and metabolomics. Cross-validation of the mouse and the human transcriptomic data from the NIH Accelerating Medicines Partnership-AD database demonstrated that pathways improved by the treatment in APP/PS1 mice, including the immune system response and neurotransmission, represent mechanisms essential for therapeutic efficacy in AD patients.
Collapse
Affiliation(s)
- Andrea Stojakovic
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Sergey Trushin
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Anthony Sheu
- Institute for Translational Neuroscience, University of Minnesota Twin Cities, 2101 6th Street SE, Minneapolis, MN, 55455, USA
| | - Layla Khalili
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Su-Youne Chang
- Department of Neurologic Surgery, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Xing Li
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Trace Christensen
- Microscopy and Cell Analysis Core, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Jeffrey L Salisbury
- Microscopy and Cell Analysis Core, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Rachel E Geroux
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Benjamin Gateno
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Padraig J Flannery
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Mrunal Dehankar
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Cory C Funk
- Institute for Systems Biology, Seattle, WA, 98109-5263, USA
| | - Jordan Wilkins
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Anna Stepanova
- Division of Neonatology, Department of Pediatrics, Columbia University, 116th St & Broadway, New York, NY, 10027, USA
| | - Tara O'Hagan
- Division of Neonatology, Department of Pediatrics, Columbia University, 116th St & Broadway, New York, NY, 10027, USA
| | - Alexander Galkin
- Division of Neonatology, Department of Pediatrics, Columbia University, 116th St & Broadway, New York, NY, 10027, USA
| | - Jarred Nesbitt
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Xiujuan Zhu
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Utkarsh Tripathi
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Slobodan Macura
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Tamar Pirtskhalava
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Rachel A Kudgus
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Renee A Schoon
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Joel M Reid
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Yu Yamazaki
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Song Zhang
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Emirhan Nemutlu
- Faculty of Pharmacy, Department of Analytical Chemistry, Hacettepe University, Sihhiye, Ankara, 06100, Turkey
| | - Petras Dzeja
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Adam Jaspersen
- Microscopy and Cell Analysis Core, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Ye In Christopher Kwon
- Institute for Translational Neuroscience, University of Minnesota Twin Cities, 2101 6th Street SE, Minneapolis, MN, 55455, USA
| | - Michael K Lee
- Institute for Translational Neuroscience, University of Minnesota Twin Cities, 2101 6th Street SE, Minneapolis, MN, 55455, USA
| | - Eugenia Trushina
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA.
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA.
| |
Collapse
|
313
|
Wu Q, Li B, Li J, Sun S, Yuan J, Sun S. Cancer-associated adipocytes as immunomodulators in cancer. Biomark Res 2021; 9:2. [PMID: 33413697 PMCID: PMC7792018 DOI: 10.1186/s40364-020-00257-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 12/21/2020] [Indexed: 02/08/2023] Open
Abstract
Cancer-associated adipocytes (CAAs), as a main component of the tumor-adipose microenvironment (TAME), have various functions, including remodeling the extracellular matrix and interacting with tumor cells or infiltrated leukocytes through a variety of mutual signals. Here, we summarize the primary interplay among CAAs, the immune response and cancer with a focus on the mechanistic aspects of these relationships. Finally, unifying our understanding of CAAs with the immune cell function may be an effective method to enhance the efficacy of immunotherapeutic and conventional treatments.
Collapse
Affiliation(s)
- Qi Wu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, 238 Ziyang Road, Wuhan, 430060, Hubei Province, P. R. China
| | - Bei Li
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Ziyang Road, Wuhan, 430060, Hubei Province, P. R. China
| | - Juanjuan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, 238 Ziyang Road, Wuhan, 430060, Hubei Province, P. R. China
| | - Si Sun
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Jingping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, 238 Ziyang Road, Wuhan, 430060, Hubei Province, P. R. China.
| | - Shengrong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, 238 Ziyang Road, Wuhan, 430060, Hubei Province, P. R. China.
| |
Collapse
|
314
|
Bellard AM, Cornelissen PL, Mian E, Cazzato V. The ageing body: contributing attitudinal factors towards perceptual body size estimates in younger and middle-aged women. Arch Womens Ment Health 2021; 24:93-105. [PMID: 32562005 PMCID: PMC7929965 DOI: 10.1007/s00737-020-01046-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 05/21/2020] [Indexed: 02/06/2023]
Abstract
Over-estimation of body size, a core feature of eating disorders (EDs), has been well-documented both in young healthy and ED individuals. Yet, evidence that altered body perception might also affect older women is limited. Here, we examined whether attitudinal components of body image (i.e. the feelings an individual has about their body size and shape) might affect perceived actual and ideal body shape self-estimates in midlife, similarly to younger women. Thirty-two younger (mean age, 24.22 years) and 33 middle-aged (mean age, 53.79 years) women took part to a computerized body perception assessment of perceived, actual and ideal aspects of body image. Body mass index (BMI), societal and interpersonal aspects of appearance ideals, measured by means of Sociocultural Attitudes towards Appearance Questionnaire (SATAQ-4), and assessment of body uneasiness and concerns for specific body parts, measured by Body Uneasiness Test (BUT-A/B) scales, were also investigated. Younger and middle-aged women with larger BMI showed greater discrepancy in perceptual distortions from their perceived actual body size. However, middle-aged women with greater body part concerns overestimated their perceived body size, as opposed to younger women who were almost accurate. Unlike middle-aged women, younger women with higher body part concerns desired slimmer ideal body image than their perceived actual. Results suggest that distortions in the perceived actual and ideal body size self-estimates of younger and middle-aged women are best explained by a combination of BMI, body part concerns and the particular age group to which a participant belonged. In the future, a personalized approach for the assessment of women's perceptions and concerns of specific body areas during lifespan should be adopted.
Collapse
Affiliation(s)
- Ashleigh M. Bellard
- grid.4425.70000 0004 0368 0654School of Psychology, Faculty of Health, Liverpool John Moores University, Liverpool, UK
| | - Piers L. Cornelissen
- grid.42629.3b0000000121965555Department of Psychology, Northumbria University, Newcastle, UK
| | - Emanuel Mian
- Emotifood Body Image & Eating Disorders Unit, 20900 Monza, Italy
| | - Valentina Cazzato
- School of Psychology, Faculty of Health, Liverpool John Moores University, Liverpool, UK.
| |
Collapse
|
315
|
Sadie-Van Gijsen H. Is Adipose Tissue the Fountain of Youth? The Impact of Adipose Stem Cell Aging on Metabolic Homeostasis, Longevity, and Cell-Based Therapies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1286:225-250. [PMID: 33725357 DOI: 10.1007/978-3-030-55035-6_16] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Aging is driven by four interlinked processes: (1) low-grade sterile inflammation; (2) macromolecular and organelle dysfunction, including DNA damage, telomere erosion, and mitochondrial dysfunction; (3) stem cell dysfunction; and (4) an accumulation of senescent cells in tissues. Adipose tissue is not immune to the effects of time, and all four of these processes contribute to a decline of adipose tissue function with advanced age. This decline is associated with an increase in metabolic disorders. Conversely, optimally functioning adipose tissue generates signals that promote longevity. As tissue-resident progenitor cells that actively participate in adipose tissue homeostasis and dysregulation, adipose stem cells (ASCs) have emerged as a key feature in the relationship between age and adipose tissue function. This review will give a mechanistic overview of the myriad ways in which age affects ASC function and, conversely, how ASC function contribute to healthspan and lifespan. A central mediator in this relationship is the degree of resilience of ASCs to maintain stemness into advanced age and the consequent preservation of adipose tissue function, in particular subcutaneous fat. The last sections of this review will discuss therapeutic options that target senescent ASCs to extend healthspan and lifespan, as well as ASC-based therapies that can be used to treat age-related pathologies, and collectively, these therapeutic applications may transform the way we age.
Collapse
Affiliation(s)
- Hanél Sadie-Van Gijsen
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg Campus, Parow, South Africa.
| |
Collapse
|
316
|
Tao W, Yu Z, Han JDJ. A digitized catalog of COVID-19 epidemiology data. QUANTITATIVE BIOLOGY 2021. [DOI: 10.15302/j-qb-020-0230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
317
|
Li Z, Xu K, Zhao S, Guo Y, Chen H, Ni J, Liu Q, Wang Z. SPATA4 improves aging-induced metabolic dysfunction through promotion of preadipocyte differentiation and adipose tissue expansion. Aging Cell 2021; 20:e13282. [PMID: 33314576 PMCID: PMC7811838 DOI: 10.1111/acel.13282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/25/2020] [Accepted: 10/28/2020] [Indexed: 12/13/2022] Open
Abstract
Spermatogenesis‐associated protein 4 (SPATA4) is conserved across multiple species. However, the function of this gene remains largely unknown. In this study, we generated Spata4 transgenic mice to explore tissue‐specific function of SPATA4. Spata4 overexpression mice displayed increased subcutaneous fat tissue compared with wild‐type littermates at an old age, while this difference was not observed in younger mice. Aging‐induced ectopic fat distribution, inflammation, and insulin resistance were also significantly attenuated by SPATA4. In vitro, SPATA4 promoted preadipocyte differentiation through activation of the ERK1/2 and C/EBPβ pathway and increased the expression of adipokines. These data suggest SPATA4 can regulate lipid accumulation in a tissue‐specific manner and improve aging‐induced dysmetabolic syndromes. Clarifying the mechanism of SPATA4 functioning in lipid metabolism might provide novel therapeutic targets for disease interventions.
Collapse
Affiliation(s)
- Zhongchi Li
- Protein Science Key Laboratory of the Ministry of Education School of Pharmaceutical Sciences Tsinghua University Beijing China
| | - Kang Xu
- Protein Science Key Laboratory of the Ministry of Education School of Pharmaceutical Sciences Tsinghua University Beijing China
| | - Sen Zhao
- Key Laboratory of Big Data for Spinal Deformities Peking Union Medical College Hospital Beijing China
| | - Yannan Guo
- Protein Science Key Laboratory of the Ministry of Education School of Pharmaceutical Sciences Tsinghua University Beijing China
| | - Huiling Chen
- Protein Science Key Laboratory of the Ministry of Education School of Pharmaceutical Sciences Tsinghua University Beijing China
| | - Jianquan Ni
- School of Medicine Tsinghua University Beijing China
| | - Qingfei Liu
- Protein Science Key Laboratory of the Ministry of Education School of Pharmaceutical Sciences Tsinghua University Beijing China
| | - Zhao Wang
- Protein Science Key Laboratory of the Ministry of Education School of Pharmaceutical Sciences Tsinghua University Beijing China
| |
Collapse
|
318
|
Wu Q, He S, Zhu Y, Pu S, Zhou Z. Antiobesity Effects of Adipose-Derived Stromal/Stem Cells in a Naturally Aged Mouse Model. Obesity (Silver Spring) 2021; 29:133-142. [PMID: 33185001 DOI: 10.1002/oby.23036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/14/2020] [Accepted: 08/22/2020] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Adipose-derived stromal/stem cells (ASCs) have multilineage differentiation potential and functional properties, as well as applications for cell-based therapies in tissue repair and regeneration. However, there is a lack of evidence regarding the efficacy of ASCs as an antiobesity agent in aged organisms. This study aimed to clarify the effectiveness of ASCs at treating obesity using a naturally aged mouse model. METHODS Old (22 months) C57BL/6J mice with transplanted young-mice (2 months) donor ASCs were measured for weight change, biochemistry, cytokines, hormone secretion, cell senescence, lipid metabolism, and functional changes of ASCs. RESULTS The results indicated that old mice treated with ASCs showed antiaging and antiobesity effects such as significant loss of body and organ weight, improved stem cell plasticity, increased antioxidant capacity (superoxide dismutase and catalase), improved liver and kidney function, improved lipid metabolism, and increased hormone secretion (sex hormone-binding globulin, thyrotropin, and leptin). Treatment with ASCs decreased cell senescence and suppressed secretion of inflammatory agents (interleukin-6 and tumor necrosis factor alpha). CONCLUSIONS Traditional drugs used in the treatment of obesity have limitations and are unsuitable for the elderly. Based on the results, the future use of ASCs as primary antiaging and antiobesity agents is suggested because of their positive effects on aged animals.
Collapse
Affiliation(s)
- Qiong Wu
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Shuangli He
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Yu Zhu
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Shiming Pu
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Zuping Zhou
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| |
Collapse
|
319
|
Chen X, Yi Z, Wong GT, Hasan KMM, Kwan JS, Ma AC, Chang RC. Is exercise a senolytic medicine? A systematic review. Aging Cell 2021; 20:e13294. [PMID: 33378138 PMCID: PMC7811843 DOI: 10.1111/acel.13294] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/15/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022] Open
Abstract
Cellular senescence, a state of irreversible growth arrest triggered by various stressors, engages in a category of pathological processes, whereby senescent cells accumulate in mitotic tissues. Senolytics as novel medicine against aging and various diseases through the elimination of senescent cells has emerged rapidly in recent years. Exercise is a potent anti‐aging and anti‐chronic disease medicine, which has shown the capacity to lower the markers of cellular senescence over the past decade. However, whether exercise is a senolytic medicine for aging and various diseases remains unclear. Here, we have conducted a systematic review of the published literature studying the senolytic effects of exercise or physical activity on senescent cells under various states in both human and animal models. Exercise can reduce the markers of senescent cells in healthy humans, while it lowered the markers of senescent cells in obese but not healthy animals. The discrepancy between human and animal studies may be due to the relatively small volume of research and the variations in markers of senescent cells, types of cells/tissues, and health conditions. These findings suggest that exercise has senolytic properties under certain conditions, which warrant further investigations.
Collapse
Affiliation(s)
- Xiang‐Ke Chen
- Laboratory of Neurodegenerative Diseases School of Biomedical Sciences LKS Faculty of Medicine The University of Hong Kong Hong Kong China
| | - Zhen‐Ni Yi
- Department of Health Technology and Informatics Hong Kong Polytechnic University Hong Kong China
| | - Gordon Tin‐Chun Wong
- Department of Anaesthesiology LKS Faculty of Medicine The University of Hong Kong Hong Kong China
| | - Kazi Md. Mahmudul Hasan
- Department of Health Technology and Informatics Hong Kong Polytechnic University Hong Kong China
| | | | - Alvin Chun‐Hang Ma
- Department of Health Technology and Informatics Hong Kong Polytechnic University Hong Kong China
| | - Raymond Chuen‐Chung Chang
- Laboratory of Neurodegenerative Diseases School of Biomedical Sciences LKS Faculty of Medicine The University of Hong Kong Hong Kong China
- State Key Laboratory of Brain and Cognitive Sciences The University of Hong Kong Hong Kong China
| |
Collapse
|
320
|
Rouault C, Marcelin G, Adriouch S, Rose C, Genser L, Ambrosini M, Bichet JC, Zhang Y, Marquet F, Aron-Wisnewsky J, Poitou C, André S, Dérumeaux G, Guerre-Millo M, Clément K. Senescence-associated β-galactosidase in subcutaneous adipose tissue associates with altered glycaemic status and truncal fat in severe obesity. Diabetologia 2021; 64:240-254. [PMID: 33125520 DOI: 10.1007/s00125-020-05307-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 09/01/2020] [Indexed: 01/02/2023]
Abstract
AIM/HYPOTHESIS Altered adipose tissue secretory profile contributes to insulin resistance and type 2 diabetes in obesity. Preclinical studies have identified senescent cells as a cellular source of proinflammatory factors in adipose tissue of obese mice. In humans, potential links with obesity comorbidities are poorly defined. Here, we investigated adipose tissue senescent status and relationships with metabolic complications in human obesity. METHODS The study includes a prospective cohort of 227 individuals with severe obesity. A photometric method was used to quantify senescence-associated β-galactosidase (SA-β-gal) activity in paired subcutaneous and omental adipose tissue biopsies obtained during gastric surgery. Gene and secretory profiling was performed in adipose tissue biopsies and in human primary pre-adipocytes in the presence or absence of senolytic drugs targeting senescent cells. Participants were phenotyped for anthropometric and bioclinical variables, metabolic complications and gastric surgery-induced improvement to address relationships with adipose tissue SA-β-gal. RESULTS SA-β-gal activity was sevenfold higher in subcutaneous than in omental adipose tissue and not associated with BMI or chronological age. Several factors, including insulin-like growth factor binding protein 3 (IGFBP3), plasminogen activator inhibitor 1 (PAI1), C-C motif chemokine ligand 2 (CCL2) and IL-6, were upregulated in subcutaneous adipose tissue in relation with SA-β-gal (p for linear trend across tertiles <0.05) and in pre-adipocytes cultured with inflammatory macrophage conditioned media. Senolytic treatment reduced SA-β-gal staining and normalised these alterations. In the whole population, subcutaneous adipose tissue SA-β-gal activity was positively associated with serum leptin, markers of insulin resistance and increased trunk fat mass. Metabolic complications, including type 2 diabetes and dyslipidaemia, were more prevalent in patients with high levels of SA-β-gal, but improved with bariatric surgery whatever the initial adipose tissue senescent status. CONCLUSIONS/INTERPRETATION This study highlights a phenotype of senescence in adipose tissue of severely obese individuals, which characterises prominently subcutaneous fat depots. Subcutaneous adipose tissue senescence is significantly linked to altered glucose metabolism and body fat distribution. Elimination of senescent cells through senolytic treatment could alleviate metabolic complications in severely obese people. Graphical abstract.
Collapse
Affiliation(s)
- Christine Rouault
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches (Nutriomics), Paris, France
| | - Geneviève Marcelin
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches (Nutriomics), Paris, France
| | - Solia Adriouch
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches (Nutriomics), Paris, France
| | - Cindy Rose
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches (Nutriomics), Paris, France
| | - Laurent Genser
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches (Nutriomics), Paris, France
- Assistance Publique Hôpitaux de Paris, Visceral Surgery Department, Pitié-Salpêtrière Hospital, Paris, France
| | - Marc Ambrosini
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches (Nutriomics), Paris, France
| | - Jean-Christophe Bichet
- Assistance Publique Hôpitaux de Paris, Service de Chirurgie et Cancérologie Gynécologique et Mammaire, Pitié-Salpêtrière Hospital, Paris, France
| | - Yanyan Zhang
- Inserm U955, Université Paris-Est Créteil (UPEC), Créteil, France
| | - Florian Marquet
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches (Nutriomics), Paris, France
| | - Judith Aron-Wisnewsky
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches (Nutriomics), Paris, France
- Assistance Publique Hôpitaux de Paris, Nutrition Department, CRNH Ile de France, Pitié-Salpêtrière Hospital, Paris, France
| | - Christine Poitou
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches (Nutriomics), Paris, France
- Assistance Publique Hôpitaux de Paris, Nutrition Department, CRNH Ile de France, Pitié-Salpêtrière Hospital, Paris, France
| | - Sébastien André
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches (Nutriomics), Paris, France
| | - Geneviève Dérumeaux
- Inserm U955, Université Paris-Est Créteil (UPEC), Créteil, France
- Assistance Publique Hôpitaux de Paris, Department of Cardiology, Henri Mondor Hospital, DHU-ATVB, Créteil, France
| | - Michèle Guerre-Millo
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches (Nutriomics), Paris, France
| | - Karine Clément
- Sorbonne Université, Inserm, Nutrition and Obesities: Systemic Approaches (Nutriomics), Paris, France.
- Assistance Publique Hôpitaux de Paris, Nutrition Department, CRNH Ile de France, Pitié-Salpêtrière Hospital, Paris, France.
| |
Collapse
|
321
|
Grandys M, Majerczak J, Zapart-Bukowska J, Duda K, Kulpa JK, Zoladz JA. Lowered Serum Testosterone Concentration Is Associated With Enhanced Inflammation and Worsened Lipid Profile in Men. Front Endocrinol (Lausanne) 2021; 12:735638. [PMID: 34566895 PMCID: PMC8459752 DOI: 10.3389/fendo.2021.735638] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 08/16/2021] [Indexed: 11/13/2022] Open
Abstract
The negative relationship between testosterone and inflammatory cytokines has been reported for decades, although the exact mechanisms of their interactions are still not clear. At the same time, little is known about the relation between androgens and acute phase proteins. Therefore, in this investigation, we aimed to study the relationship between androgen status and inflammatory acute phase reactants in a group of men using multi-linear regression analysis. Venous blood samples were taken from 149 men ranging in age from 18 to 77 years. Gonadal androgens [testosterone (T) and free testosterone (fT)], acute phase reactants [C-reactive protein (CRP), ferritin (FER), alpha-1-acid glycoprotein (AAG), and interleukin-6 (IL-6)], cortisol (C), and lipid profile concentrations were determined. It was demonstrated that the markers of T and fT were negatively correlated with all acute phase proteins (CRP, FER, and AAG; p < 0.02) and the blood lipid profile [total cholesterol (TC), low-density lipoprotein (LDL), and triglycerides (TG); p < 0.03]. Multivariate analysis showed that T, fT, and the fT/C ratio were inversely correlated with the CRP, AAG, and FER concentrations independently of age and blood lipids. When adjustment for BMI was made, T, fT, and the fT/C ratio were negatively correlated with the AAG concentrations only. In addition, it was demonstrated that gonadal androgens were positively correlated with physical activity level (p < 0.01). We have concluded that a lowered serum T concentration may promote inflammatory processes independently of adipose tissue and age through a reduced inhibition of inflammatory cytokine synthesis, which leads to enhanced acute phase protein production. Therefore, a low serum T concentration appears to be an independent risk factor in the development of atherosclerosis and cardiovascular diseases. Moreover, the positive correlation between testosterone and physical activity level suggests that exercise training attenuates the age-related decrease in gonadal androgens and, in this way, may reduce the enhancement of systemic low-grade inflammation in aging men.
Collapse
Affiliation(s)
- Marcin Grandys
- Department of Muscle Physiology, Institute of Basic Sciences, Faculty of Rehabilitation, University School of Physical Education, Krakow, Poland
- Faculty of Health Sciences, Jagiellonian University Medical College, Krakow, Poland
- *Correspondence: Marcin Grandys, ; Jerzy A. Zoladz,
| | - Joanna Majerczak
- Faculty of Health Sciences, Jagiellonian University Medical College, Krakow, Poland
- Department of Neurobiology, Poznan University of Physical Education, Poznań, Poland
| | - Justyna Zapart-Bukowska
- Department of Muscle Physiology, Institute of Basic Sciences, Faculty of Rehabilitation, University School of Physical Education, Krakow, Poland
| | - Krzysztof Duda
- Department of Nursing, Institute of Health Protection, The State Higher School of Vocational Education, Tarnow, Poland
| | - Jan K. Kulpa
- Department of Clinical Biochemistry, Centre of Oncology, Maria Sklodowska-Curie Memorial Institute, Cracow Branch, Krakow, Poland
| | - Jerzy A. Zoladz
- Department of Muscle Physiology, Institute of Basic Sciences, Faculty of Rehabilitation, University School of Physical Education, Krakow, Poland
- Faculty of Health Sciences, Jagiellonian University Medical College, Krakow, Poland
- *Correspondence: Marcin Grandys, ; Jerzy A. Zoladz,
| |
Collapse
|
322
|
Siemienowicz KJ, Coukan F, Franks S, Rae MT, Duncan WC. Aberrant subcutaneous adipogenesis precedes adult metabolic dysfunction in an ovine model of polycystic ovary syndrome (PCOS). Mol Cell Endocrinol 2021; 519:111042. [PMID: 33010309 DOI: 10.1016/j.mce.2020.111042] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/25/2020] [Accepted: 09/27/2020] [Indexed: 12/13/2022]
Abstract
Polycystic ovary syndrome (PCOS) affects over 10% of women. Insulin resistance, elevated free fatty acids (FFAs) and increased adiposity are key factors contributing to metabolic dysfunction in PCOS. We hypothesised that aberrant adipogenesis during adolescence, and downstream metabolic perturbations, contributes to the metabolic phenotype of adult PCOS. We used prenatally androgenised (PA) sheep as a clinically realistic model of PCOS. During adolescence, but not during fetal or early life of PA sheep, adipogenesis was decreased in subcutaneous adipose tissue (SAT) accompanied by decreased leptin, adiponectin, and increased FFAs. In adulthood, PA sheep developed adipocyte hypertrophy in SAT paralleled by increased expression of inflammatory markers, elevated FFAs and increased expression of genes linked to fat accumulation in visceral adipose tissue. This study provides better understanding into the pathophysiology of PCOS from puberty to adulthood and identifies opportunity for early clinical intervention to normalise adipogenesis and ameliorate the metabolic phenotype.
Collapse
Affiliation(s)
- Katarzyna J Siemienowicz
- MRC Centre for Reproductive Health, The University of Edinburgh, Edinburgh, EH16 4TJ, UK; School of Applied Sciences, Edinburgh Napier University, Edinburgh, EH11 4BN, UK.
| | - Flavien Coukan
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, EH11 4BN, UK
| | - Stephen Franks
- Institute of Reproductive and Developmental Biology, Imperial College, London, UK
| | - Mick T Rae
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, EH11 4BN, UK
| | - W Colin Duncan
- MRC Centre for Reproductive Health, The University of Edinburgh, Edinburgh, EH16 4TJ, UK
| |
Collapse
|
323
|
Chen X, Yi Z, Wong GT, Hasan KMM, Kwan JS, Ma AC, Chang RC. Is exercise a senolytic medicine? A systematic review. Aging Cell 2020. [DOI: https://doi.org/10.1111/acel.13294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Xiang‐Ke Chen
- Laboratory of Neurodegenerative Diseases School of Biomedical Sciences LKS Faculty of Medicine The University of Hong Kong Hong Kong China
| | - Zhen‐Ni Yi
- Department of Health Technology and Informatics Hong Kong Polytechnic University Hong Kong China
| | - Gordon Tin‐Chun Wong
- Department of Anaesthesiology LKS Faculty of Medicine The University of Hong Kong Hong Kong China
| | - Kazi Md. Mahmudul Hasan
- Department of Health Technology and Informatics Hong Kong Polytechnic University Hong Kong China
| | | | - Alvin Chun‐Hang Ma
- Department of Health Technology and Informatics Hong Kong Polytechnic University Hong Kong China
| | - Raymond Chuen‐Chung Chang
- Laboratory of Neurodegenerative Diseases School of Biomedical Sciences LKS Faculty of Medicine The University of Hong Kong Hong Kong China
- State Key Laboratory of Brain and Cognitive Sciences The University of Hong Kong Hong Kong China
| |
Collapse
|
324
|
The Effects of Continuous and Withdrawal Voluntary Wheel Running Exercise on the Expression of Senescence-Related Genes in the Visceral Adipose Tissue of Young Mice. Int J Mol Sci 2020; 22:ijms22010264. [PMID: 33383848 PMCID: PMC7794976 DOI: 10.3390/ijms22010264] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 11/17/2022] Open
Abstract
Obesity has become a global medical problem. The upregulation of senescence-related markers in adipose tissue may cause impairment of adipose tissue and disorders of systemic metabolism. Weight control through diet has been found to ameliorate senescence in the adipose tissue. Exercise is also important in maintaining a healthy lifestyle, however, very few researchers have examined the relationship between senescence-related markers in adipose tissue. Dietary restriction is also reported to have a legacy effect, wherein the effects are maintained for some periods after the termination of the intervention. However, very few researchers have examined the relationship between exercise and senescence-related markers in adipose tissue. Besides, there is no study on the long-term effects of exercise. Hence, we investigated whether the exercise could change the expression of senescence-related genes in the visceral adipose tissue of young mice and whether there was a legacy effect of exercise for 10 weeks after the termination of exercise. Four-week-old male ICR mice were assigned to one of the three groups: 20 weeks of sedentary condition, 20 weeks of voluntary wheel running exercise, or 10 weeks of exercise followed by 10 weeks of sedentary condition. The mice showed decreased expression in genes related to senescence and senescence-associated secretory phenotype, such as p53, p16, and IL-6, in the visceral adipose tissue in response to exercise. These effects were maintained for 10 weeks after the mice stopped exercising. Our study is the first report that exercise reduces the expression of senescence-related genes in the visceral adipose tissue of young mice, and that exercise causes the legacy effect.
Collapse
|
325
|
De Tollenaere M, Chapuis E, Lapierre L, Bracq M, Hubert J, Lambert C, Sandré J, Auriol D, Scandolera A, Reynaud R. Overall renewal of skin lipids with Vetiver extract for a complete anti-ageing strategy. Int J Cosmet Sci 2020; 43:165-180. [PMID: 33253416 PMCID: PMC8246832 DOI: 10.1111/ics.12678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/20/2020] [Accepted: 11/26/2020] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Skin lipids are essential in every compartment of the skin where they play a key role in various biological functions. Interestingly, their role is central in the maintenance of hydration which is related to skin barrier function and in the skin structure through adipose tissue. It is well described today that skin lipids are affected by ageing giving skin sagging, wrinkles and dryness. Thereby, developing cosmetic actives able to reactivate skin lipids would be an efficient ant-ageing strategy. Due to the strong commitment of our scientists to innovate responsibly and create value, they designed a high value active ingredient named here as Vetiver extract, using a ground-breaking upcycling approach. We evidenced that this unique extract was able to reactivate globally the skin lipids production, bringing skin hydration and plumping effect for mature skin. METHOD In order to demonstrate the global renewal of lipids, we evaluated the lipids synthesis on cutaneous cells that produce lipids such as keratinocytes, sebocytes and adipocytes then on Reconstructed Human Epidermis and skin explants. We evaluated the expression of proteins involved in ceramides transport and barrier cornification. We then evaluated hydration and sebaceous parameters on a panel of mature volunteers. RESULTS We firstly demonstrated that Vetiver extract induced sebum production from human sebocytes cells lines but also improved its quality as observed by the production of specific antimicrobial lipids. Secondly, we demonstrated that Vetiver extract was able to restore skin barrier with the increase of skin lipids neosynthesis on Reconstructed Human Epidermis and skin explants. We also evidenced that Vetiver extract stimulated the lipids transport and epidermal cornification. Finally, Vetiver extract showed a significant effect on adipogenesis and maturation of adipocytes at in vitro and ex vivo models. We confirmed all these activities by showing that Vetiver extract improved sebum production and brought hydration through an increase of lipids content and their conformation. Vetiver extract induced an improvement of skin fatigue and a plumping effect by acting deeply on adipose tissue. CONCLUSION In conclusion, we developed an active ingredient able to bring anti-ageing effect for mature skin by a global increase of skin lipids.
Collapse
Affiliation(s)
| | - Emilie Chapuis
- Givaudan France SAS, Research and Development, Argenteuil, France
| | - Laura Lapierre
- Givaudan France SAS, Research and Development, Argenteuil, France
| | - Marine Bracq
- Givaudan France SAS, Research and Development, Argenteuil, France
| | | | - Carole Lambert
- Givaudan France SAS, Research and Development, Argenteuil, France
| | - Jérome Sandré
- Chirurgien plasticien et esthétique, Polyclinique de Courlancy, Reims, France
| | - Daniel Auriol
- Givaudan France SAS, Research and Development, Argenteuil, France
| | | | - Romain Reynaud
- Givaudan France SAS, Research and Development, Argenteuil, France
| |
Collapse
|
326
|
Das S, Chattopadhyay D, Chatterjee SK, Mondal SA, Majumdar SS, Mukhopadhyay S, Saha N, Velayutham R, Bhattacharya S, Mukherjee S. Increase in PPARγ inhibitory phosphorylation by Fetuin-A through the activation of Ras-MEK-ERK pathway causes insulin resistance. Biochim Biophys Acta Mol Basis Dis 2020; 1867:166050. [PMID: 33359696 DOI: 10.1016/j.bbadis.2020.166050] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 11/13/2020] [Accepted: 12/14/2020] [Indexed: 12/20/2022]
Abstract
Obesity induced insulin resistance is primarily regulated by the inhibitory phosphorylation of peroxisome proliferator-activated receptor γ at serine 273 (PPARγS273) which has been shown to be regulated by MEK and ERK. An upstream regulatory molecule of this pathway could be a therapeutic option. Here we analyzed the involvement of Fetuin-A (FetA), a key hepato-adipokine implicated in insulin resistance, as an upstream regulator molecule for the regulation of PPARγ inhibitory phosphorylation. Mice fed with standard diet (SD), high fat diet (HFD) and HFD with FetA knockdown (HFD-FetAKD) were used to examine the role of FetA on PPARγS273 phosphorylation in adipocytes. The mechanism of regulation and its effect on skeletal muscle were studied using primary adipocytes, 3T3-L1 (preadipocyte) and C2C12 (myotube) cell lines. Increased FetA in HFD mice strongly correlated with augmentation of PPARγS273 phosphorylation in inflamed adipocytes while knockdown of FetA suppressed it. This effect of FetA was mediated through the activation of Ras which in turn activated MEK and ERK. On addressing how FetA could stimulate activation of Ras, we found that FetA triggered TNFα in inflamed adipocytes which induced Ras activation. The ensuing sharp fall in adiponectin level attenuated AMPK activation in skeletal muscle cells affecting mitochondrial ATP production. Our data reveal the essential role of FetA induced activation of Ras in regulating PPARγ inhibitory phosphorylation through Ras-MEK-ERK pathway which downregulates adiponectin disrupting skeletal muscle mitochondrial bioenergetics. Thus, FetA mediated PPARγ inactivation has adverse consequences upon adipocyte-myocyte crosstalk leading to disruption of energy homeostasis and loss of insulin sensitivity.
Collapse
Affiliation(s)
- Snehasis Das
- Endocrinology and Metabolism Laboratory, Department of Zoology, Siksha Bhavana (Institute of Science), Visva-Bharati (A Central University), Santiniketan - 731235, India
| | - Dipanjan Chattopadhyay
- Endocrinology and Metabolism Laboratory, Department of Zoology, Siksha Bhavana (Institute of Science), Visva-Bharati (A Central University), Santiniketan - 731235, India
| | - Subhendu K Chatterjee
- Endocrinology and Metabolism Laboratory, Department of Zoology, Siksha Bhavana (Institute of Science), Visva-Bharati (A Central University), Santiniketan - 731235, India
| | - Samim Ali Mondal
- Department of Endocrinology & Metabolism, Institute of Post-Graduate Medical Education & Research-Seth Sukhlal Karnani Memorial (IPGME&R-SSKM) Hospital, Kolkata 700025, India
| | | | - Satinath Mukhopadhyay
- Department of Endocrinology & Metabolism, Institute of Post-Graduate Medical Education & Research-Seth Sukhlal Karnani Memorial (IPGME&R-SSKM) Hospital, Kolkata 700025, India
| | - Nirmalendu Saha
- Department of Zoology, North-Eastern Hill University, Shillong 793022, India
| | | | - Samir Bhattacharya
- Endocrinology and Metabolism Laboratory, Department of Zoology, Siksha Bhavana (Institute of Science), Visva-Bharati (A Central University), Santiniketan - 731235, India
| | - Sutapa Mukherjee
- Endocrinology and Metabolism Laboratory, Department of Zoology, Siksha Bhavana (Institute of Science), Visva-Bharati (A Central University), Santiniketan - 731235, India.
| |
Collapse
|
327
|
Gaspar LS, Sousa C, Álvaro AR, Cavadas C, Mendes AF. Common risk factors and therapeutic targets in obstructive sleep apnea and osteoarthritis: An unexpectable link? Pharmacol Res 2020; 164:105369. [PMID: 33352231 DOI: 10.1016/j.phrs.2020.105369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/11/2020] [Accepted: 12/09/2020] [Indexed: 10/22/2022]
Abstract
Osteoarthritis (OA) and Obstructive Sleep Apnea (OSA) are two highly prevalent chronic diseases for which effective therapies are urgently needed. Recent epidemiologic studies, although scarce, suggest that the concomitant occurrence of OA and OSA is associated with more severe manifestations of both diseases. Moreover, OA and OSA share risk factors, such as aging and metabolic disturbances, and co-morbidities, including cardiovascular and metabolic diseases, sleep deprivation and depression. Whether this coincidental occurrence is fortuitous or involves cause-effect relationships is unknown. This review aims at collating and integrating present knowledge on both diseases by providing a brief overview of their epidemiology and pathophysiology, analyzing current evidences relating OA and OSA and discussing potential common mechanisms by which they can aggravate each other. Such mechanisms constitute potential therapeutic targets whose pharmacological modulation may provide more efficient ways of reducing the consequences of OA and OSA and, thus, lessen the huge individual and social burden that they impose.
Collapse
Affiliation(s)
- Laetitia S Gaspar
- Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal; PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Cátia Sousa
- Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Ana Rita Álvaro
- Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Cláudia Cavadas
- Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
| | - Alexandrina Ferreira Mendes
- Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
328
|
Adamczyk-Grochala J, Lewinska A. Nano-Based Theranostic Tools for the Detection and Elimination of Senescent Cells. Cells 2020; 9:E2659. [PMID: 33322013 PMCID: PMC7764355 DOI: 10.3390/cells9122659] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/05/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
The progressive accumulation of apoptosis-resistant and secretory active senescent cells (SCs) in animal and human aged tissues may limit lifespan and healthspan and lead to age-related diseases such as cancer, neurodegenerative disorders, and metabolic syndrome. Thus, SCs are suggested targets in anti-aging therapy. In the last two decades, a number of nanomaterials have gained much attention as innovative tools in theranostic applications due to their unique properties improving target visualization, drug and gene delivery, controlled drug release, effective diagnosis, and successful therapy. Although the healthcare industry has focused on a plethora of applications of nanomaterials, it remains elusive how nanomaterials may modulate cellular senescence, a hallmark of aging. In this review paper, we consider novel nanotechnology-based strategies for healthspan promotion and the prevention of age-related dysfunctions that are based on the delivery of therapeutic compounds capable to preferentially killing SCs (nano-senolytics) and/or modulating a proinflammatory secretome (nano-senomorphics/nano-senostatics). Recent examples of SC-targeted nanomaterials and the mechanisms underlying different aspects of the nanomaterial-mediated senolysis are presented and discussed.
Collapse
Affiliation(s)
- Jagoda Adamczyk-Grochala
- Department of Biotechnology, Institute of Biology and Biotechnology, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| | - Anna Lewinska
- Department of Biotechnology, Institute of Biology and Biotechnology, University of Rzeszow, Pigonia 1, 35-310 Rzeszow, Poland
| |
Collapse
|
329
|
Xiang QY, Tian F, Du X, Xu J, Zhu LY, Guo LL, Wen T, Liu YS, Liu L. Postprandial triglyceride-rich lipoproteins-induced premature senescence of adipose-derived mesenchymal stem cells via the SIRT1/p53/Ac-p53/p21 axis through oxidative mechanism. Aging (Albany NY) 2020; 12:26080-26094. [PMID: 33316776 PMCID: PMC7803527 DOI: 10.18632/aging.202298] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 11/06/2020] [Indexed: 12/26/2022]
Abstract
The accumulation of senescent adipose-derived mesenchymal stem cells (AMSCs) in subcutaneous white adipose tissue (WAT) is the main cause for the deterioration of WAT and the subsequent age-related disorders in obesity. The number of AMSCs staining positively for senescence-associated-β-galactosidase (SA-β-Gal) increased significantly after incubation with postprandial triglyceride-rich lipoproteins (TRL), accompanied by an impaired cell proliferation capacity and increased expression of inflammatory factors. Besides, the expression of anti-aging protein, silent mating-type information regulation 2 homolog 1 (SIRT1), was downregulated significantly, while those of acetylated p53 (Ac-p53), total p53, and p21 proteins were upregulated significantly during postprandial TRL-induced premature senescence of AMSCs. Furthermore, the production of intracellular reactive oxygen species (ROS) in the TRL group increased significantly, while pretreatment with the ROS scavenger N-acetyl-L-cysteine effectively attenuated the premature senescence of AMSCs by decreasing ROS production and upregulating SIRT1 level. Thus, postprandial TRL induced premature senescence of AMSCs through the SIRT1/p53/Ac-p53/p21 axis, partly through increased oxidative stress.
Collapse
Affiliation(s)
- Qun-Yan Xiang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, PR China.,Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha 410011, Hunan, PR China.,Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha 410011, Hunan, PR China.,Cardiovascular Disease Research Center of Hunan Province, Changsha 410011, Hunan, PR China.,Department of Geriatrics, Institute of Aging and Geriatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, PR China
| | - Feng Tian
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, PR China.,Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha 410011, Hunan, PR China.,Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha 410011, Hunan, PR China.,Cardiovascular Disease Research Center of Hunan Province, Changsha 410011, Hunan, PR China.,Department of Geriatric Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, Henan, PR China
| | - Xiao Du
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, PR China.,Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha 410011, Hunan, PR China.,Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha 410011, Hunan, PR China.,Cardiovascular Disease Research Center of Hunan Province, Changsha 410011, Hunan, PR China
| | - Jin Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, PR China.,Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha 410011, Hunan, PR China.,Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha 410011, Hunan, PR China.,Cardiovascular Disease Research Center of Hunan Province, Changsha 410011, Hunan, PR China
| | - Li-Yuan Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, PR China.,Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha 410011, Hunan, PR China.,Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha 410011, Hunan, PR China.,Cardiovascular Disease Research Center of Hunan Province, Changsha 410011, Hunan, PR China
| | - Li-Ling Guo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, PR China.,Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha 410011, Hunan, PR China.,Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha 410011, Hunan, PR China.,Cardiovascular Disease Research Center of Hunan Province, Changsha 410011, Hunan, PR China
| | - Tie Wen
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, PR China.,Emergency Medicine and Difficult Disease Institute, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, PR China
| | - You-Shuo Liu
- Department of Geriatrics, Institute of Aging and Geriatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, PR China
| | - Ling Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, PR China.,Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha 410011, Hunan, PR China.,Modern Cardiovascular Disease Clinical Technology Research Center of Hunan Province, Changsha 410011, Hunan, PR China.,Cardiovascular Disease Research Center of Hunan Province, Changsha 410011, Hunan, PR China
| |
Collapse
|
330
|
Mann SN, Hadad N, Nelson Holte M, Rothman AR, Sathiaseelan R, Ali Mondal S, Agbaga MP, Unnikrishnan A, Subramaniam M, Hawse J, Huffman DM, Freeman WM, Stout MB. Health benefits attributed to 17α-estradiol, a lifespan-extending compound, are mediated through estrogen receptor α. eLife 2020; 9:59616. [PMID: 33289482 PMCID: PMC7744101 DOI: 10.7554/elife.59616] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
Metabolic dysfunction underlies several chronic diseases, many of which are exacerbated by obesity. Dietary interventions can reverse metabolic declines and slow aging, although compliance issues remain paramount. 17α-estradiol treatment improves metabolic parameters and slows aging in male mice. The mechanisms by which 17α-estradiol elicits these benefits remain unresolved. Herein, we show that 17α-estradiol elicits similar genomic binding and transcriptional activation through estrogen receptor α (ERα) to that of 17β-estradiol. In addition, we show that the ablation of ERα completely attenuates the beneficial metabolic effects of 17α-E2 in male mice. Our findings suggest that 17α-E2 may act through the liver and hypothalamus to improve metabolic parameters in male mice. Lastly, we also determined that 17α-E2 improves metabolic parameters in male rats, thereby proving that the beneficial effects of 17α-E2 are not limited to mice. Collectively, these studies suggest ERα may be a drug target for mitigating chronic diseases in male mammals.
Collapse
Affiliation(s)
- Shivani N Mann
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Niran Hadad
- The Jackson Laboratory, Bar Harbor, United States
| | - Molly Nelson Holte
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Alicia R Rothman
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Roshini Sathiaseelan
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Samim Ali Mondal
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Martin-Paul Agbaga
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Archana Unnikrishnan
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | | | - John Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Derek M Huffman
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, United States
| | - Willard M Freeman
- Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, United States.,Oklahoma City Veterans Affairs Medical Center, Oklahoma City, United States
| | - Michael B Stout
- Department of Nutritional Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, United States.,Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| |
Collapse
|
331
|
Abstract
Over the last decade, our understanding of the physiological role of senescent cells has drastically evolved, from merely indicators of cellular stress and ageing to having a central role in regeneration and repair. Increasingly, studies have identified senescent cells and the senescence-associated secretory phenotype (SASP) as being critical in the regenerative process following injury; however, the timing and context at which the senescence programme is activated can lead to distinct outcomes. For example, a transient induction of senescent cells followed by rapid clearance at the early stages following injury promotes repair, while the long-term accumulation of senescent cells impairs tissue function and can lead to organ failure. A key role of the SASP is the recruitment of immune cells to the site of injury and the subsequent elimination of senescent cells. Among these cell types are macrophages, which have well-documented regulatory roles in all stages of regeneration and repair. However, while the role of senescent cells and macrophages in this process is starting to be explored, the specific interactions between these cell types and how these are important in the different stages of injury/reparative response still require further investigation. In this review, we consider the current literature regarding the interaction of these cell types, how their cooperation is important for regeneration and repair, and what questions remain to be answered to advance the field.
Collapse
|
332
|
Kaur J, Farr JN. Cellular senescence in age-related disorders. Transl Res 2020; 226:96-104. [PMID: 32569840 PMCID: PMC7572662 DOI: 10.1016/j.trsl.2020.06.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 02/06/2023]
Abstract
Much of the population is now faced with an enormous burden of age-associated chronic diseases. Recent discoveries in geroscience indicate that healthspan in model organisms such as mice can be manipulated by targeting cellular senescence, a hallmark mechanism of aging, defined as an irreversible proliferative arrest that occurs when cells experience oncogenic or other diverse forms of damage. Senescent cells and their proinflammatory secretome have emerged as contributors to age-related tissue dysfunction and morbidity. Cellular senescence has causal roles in mediating osteoporosis, frailty, cardiovascular diseases, osteoarthritis, pulmonary fibrosis, renal diseases, neurodegenerative diseases, hepatic steatosis, and metabolic dysfunction. Therapeutically targeting senescent cells in mice can prevent, delay, or alleviate each of these conditions. Therefore, senotherapeutic approaches, including senolytics and senomorphics, that either selectively eliminate senescent cells or interfere with their ability to promote tissue dysfunction, are gaining momentum as potential realistic strategies to abrogate human senescence to thereby compress morbidity and extend healthspan.
Collapse
Affiliation(s)
- Japneet Kaur
- Division of Endocrinology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester Minnesota; Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Mayo Clinic, Rochester Minnesota
| | - Joshua N Farr
- Division of Endocrinology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester Minnesota; Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Mayo Clinic, Rochester Minnesota; Division of Physiology and Biomedical Engineering; Mayo Clinic College of Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
333
|
Xu L, Lu Y, Li N, Zhao Q, Li K, Zhang Y, Liu Y, Li C, Cheng X. Cross-sectional associations of adipokines and abdominal fat distribution with aging in men. Aging Male 2020; 23:1576-1582. [PMID: 33499713 DOI: 10.1080/13685538.2021.1876020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE To investigate the relationship of adipokines and abdominal fat distribution with aging in men. METHODS In a cross-sectional study, a total of 218 participants aged 40-79 years were recruited as a subset of the Prospective Urban Rural Epidemiology (PURE) China Action on Spine and Hip status (CASH) study population. Analysis of variance (ANOVA) and multivariable regression were used to estimate the associations of interest. RESULTS With the increasing of age, waist circumference, waist-to-hip ratio, waist-to-height ratio, total adipose tissue (TAT), visceral adipose tissue (VAT), VAT/subcutaneous adipose tissue (SAT), leptin, adiponectin-to-leptin ratio, and human monocyte chemo-attractant protein-1 (MCP-1) increased significantly (p < 0.05), while adiponectin decreased significantly (p < 0.05). Adiponectin, adiponectin/leptin, and adiponectin/resistin varied inversely with the VAT quartiles (p < 0.05). There was a significant negative correlation among adiponectin, adiponectin-to-leptin ratio, adiponectin-to-resistin ratio, and all the body fat distribution parameters. VAT was inversely and significantly associated with adiponectin, adiponectin-to-leptin ratio, and adiponectin-to-resistin ratio (p < 0.05). CONCLUSIONS It showed that aging, abdominal fat distribution, and adipokines were related with each other, which support the hypothesis that regulation of VAT and adipokines is closely linked to aging.
Collapse
Affiliation(s)
- Li Xu
- Department of Radiology, Beijing Jishuitan Hospital, Beijing, China
| | - Yanhui Lu
- Department of Endocrinology, the Second Medical Centre, Chinese PLA General Hospital, National Clinical Research Centre for Geriatric Disease, Beijing, China
| | - Nan Li
- Department of Endocrinology, the Second Medical Centre, Chinese PLA General Hospital, National Clinical Research Centre for Geriatric Disease, Beijing, China
| | - Qian Zhao
- International Medical Center, Sichuan University West China Hospital, Chengdu, China
| | - Kai Li
- Department of Radiology, Beijing Jishuitan Hospital, Beijing, China
| | - Yong Zhang
- Department of Radiology, Beijing Jishuitan Hospital, Beijing, China
| | - Yandong Liu
- Department of Radiology, Beijing Jishuitan Hospital, Beijing, China
| | - Chunlin Li
- Department of Endocrinology, the Second Medical Centre, Chinese PLA General Hospital, National Clinical Research Centre for Geriatric Disease, Beijing, China
| | - Xiaoguang Cheng
- Department of Radiology, Beijing Jishuitan Hospital, Beijing, China
| |
Collapse
|
334
|
Wueest S, Lucchini FC, Haim Y, Rudich A, Konrad D. Depletion of ASK1 blunts stress-induced senescence in adipocytes. Adipocyte 2020; 9:535-541. [PMID: 32930631 PMCID: PMC7714422 DOI: 10.1080/21623945.2020.1815977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Increasing energy expenditure via induction of browning in white adipose tissue has emerged as a potential strategy to treat obesity and associated metabolic complications. We previously reported that ASK1 inhibition in adipocytes protected from high-fat diet (HFD) or lipopolysaccharide (LPS)-mediated downregulation of UCP1 both in vitro and in vivo. Conversely, adipocyte-specific ASK1 overexpression attenuated cold-induction of UCP-1 in inguinal fat. Herein, we provide evidence that both TNFα-mediated and HFD-induced activation of p38 MAPK in white adipocytes are ASK1-dependent. Moreover, expression of senescence markers was reduced in HFD-fed adipocyte-specific ASK1 knockout mice. Similarly, LPS-induced upregulation of senescence markers was blunted in ASK1-depleted adipocytes. Thus, our study identifies a previously unknown role for ASK1 in the induction of stress-induced senescence.
Collapse
Affiliation(s)
- Stephan Wueest
- Division of Pediatric Endocrinology and Diabetology, University Children’s Hospital, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital, Zurich, Switzerland
| | - Fabrizio C. Lucchini
- Division of Pediatric Endocrinology and Diabetology, University Children’s Hospital, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Yulia Haim
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Assaf Rudich
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology, University Children’s Hospital, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
335
|
Mechanisms of adipose tissue extracellular matrix alterations in an in vitro model of adipocytes hypoxia and aging. Mech Ageing Dev 2020; 192:111374. [DOI: 10.1016/j.mad.2020.111374] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/12/2020] [Accepted: 09/25/2020] [Indexed: 12/17/2022]
|
336
|
Zamboni M, Nori N, Brunelli A, Zoico E. How does adipose tissue contribute to inflammageing? Exp Gerontol 2020; 143:111162. [PMID: 33253807 DOI: 10.1016/j.exger.2020.111162] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/09/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022]
Abstract
Across aging, white adipose tissue (WAT) undergoes significant changes in quantity and distribution, with an increase in visceral adipose tissue, ectopic fat deposition and a decline in gluteofemoral subcutaneous depot. In particular, WAT becomes dysfunctional with an increase in production of inflammatory peptides and a decline of those with anti-inflammatory activity and infiltration of inflammatory cells. Moreover, dysfunction of WAT is characterized by preadipocyte differentiation decline, increased oxidative stress and mitochondrial dysfunction, reduction in vascularization and hypoxia, increased fibrosis and senescent cell accumulation. WAT changes represent an important hallmark of the aging process and may be responsible for the systemic pro-inflammatory state ("inflammageing") typical of aging itself, leading to age-related metabolic alterations. This review focuses on mechanisms linking age-related WAT changes to inflammageing.
Collapse
Affiliation(s)
- Mauro Zamboni
- Division of Geriatric Medicine, Department of Surgery, Dentistry, Pediatric and Gynecology, University of Verona, Verona, Italy.
| | - Nicole Nori
- Division of Geriatric Medicine, Department of Medicine, University of Verona, Verona, Italy
| | - Anna Brunelli
- Division of Geriatric Medicine, Department of Medicine, University of Verona, Verona, Italy
| | - Elena Zoico
- Division of Geriatric Medicine, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
337
|
Frasca D, Blomberg BB. Aging induces B cell defects and decreased antibody responses to influenza infection and vaccination. IMMUNITY & AGEING 2020; 17:37. [PMID: 33292323 PMCID: PMC7674578 DOI: 10.1186/s12979-020-00210-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/16/2020] [Indexed: 12/15/2022]
Abstract
Background Aging is characterized by a progressive decline in the capacity of the immune system to fight influenza virus infection and to respond to vaccination. Among the several factors involved, in addition to increased frailty and high-risk conditions, the age-associated decrease in cellular and humoral immune responses plays a relevant role. This is in large part due to inflammaging, the chronic low-grade inflammatory status of the elderly, associated with intrinsic inflammation of the immune cells and decreased immune function. Results Aging is usually associated with reduced influenza virus-specific and influenza vaccine-specific antibody responses but some elderly individuals with higher pre-exposure antibody titers, due to a previous infection or vaccination, have less probability to get infected. Examples of this exception are the elderly individuals infected during the 2009 pandemic season who made antibodies with broader epitope recognition and higher avidity than those made by younger individuals. Several studies have allowed the identification of B cell intrinsic defects accounting for sub-optimal antibody responses of elderly individuals. These defects include 1) reduced class switch recombination, responsible for the generation of a secondary response of class switched antibodies, 2) reduced de novo somatic hypermutation of the antibody variable region, 3) reduced binding and neutralization capacity, as well as binding specificity, of the secreted antibodies, 4) increased epigenetic modifications that are associated with lower antibody responses, 5) increased frequencies of inflammatory B cell subsets, and 6) shorter telomeres. Conclusions Although influenza vaccination represents the most effective way to prevent influenza infection, vaccines with greater immunogenicity are needed to improve the response of elderly individuals. Recent advances in technology have made possible a broad approach to better understand the age-associated changes in immune cells, needed to design tailored vaccines and effective therapeutic strategies that will be able to improve the immune response of vulnerable individuals.
Collapse
Affiliation(s)
- Daniela Frasca
- Department of Microbiology and Immunology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, RMSB 3146A, 1600 NW 10th Ave, Miami, FL, 33136, USA.
| | - Bonnie B Blomberg
- Department of Microbiology and Immunology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, RMSB 3146A, 1600 NW 10th Ave, Miami, FL, 33136, USA
| |
Collapse
|
338
|
Bartke A, Brannan S, Hascup E, Hascup K, Darcy J. Energy Metabolism and Aging. World J Mens Health 2020; 39:222-232. [PMID: 33151044 PMCID: PMC7994661 DOI: 10.5534/wjmh.200112] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/04/2020] [Accepted: 08/28/2020] [Indexed: 12/21/2022] Open
Abstract
Aging is strongly related to energy metabolism, but the underlying processes and mechanisms are complex and incompletely understood. Restricting energy intake and reducing metabolic rate can slow the rate of aging and extend longevity, implying a reciprocal relationship between energy metabolism and life expectancy. However, increased energy expenditure has also been associated with improved health and longer life. In both experimental animals and humans, reduced body temperature has been related to extended longevity. However, recent findings on the function of thermogenic (brown or beige) adipose tissue produced intense interest in increasing the amount of energy expended for thermogenesis to prevent and/or treat obesity, improve metabolic health, and extend life. Evidence available to-date indicates that increasing adipose tissue thermogenesis by pharmacologic, environmental, or genetic interventions can indeed produce significant metabolic benefits, which are associated with improved chances for healthy aging and long life.
Collapse
Affiliation(s)
- Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, USA.
| | - Savannah Brannan
- Department of Biology, University of Illinois Springfield, Springfield, IL, USA
| | - Erin Hascup
- Department of Neurology and Center for Alzheimer's Disease and Related Disorders, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kevin Hascup
- Department of Neurology and Center for Alzheimer's Disease and Related Disorders, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Justin Darcy
- Joslin Diabetes Center, Section on Integrative Physiology and Metabolism, Harvard Medical School One Joslin Place, Boston, MA, USA
| |
Collapse
|
339
|
Nadra K, André M, Marchaud E, Kestemont P, Braccini F, Cartier H, Kéophiphath M, Fanian F. A hyaluronic acid-based filler reduces lipolysis in human mature adipocytes and maintains adherence and lipid accumulation of long-term differentiated human preadipocytes. J Cosmet Dermatol 2020; 20:1474-1482. [PMID: 33150734 PMCID: PMC8246837 DOI: 10.1111/jocd.13794] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 07/22/2020] [Accepted: 09/22/2020] [Indexed: 12/14/2022]
Abstract
The beneficial role of subcutaneous adipose tissue in skin rejuvenation derived from its capacity to fill the under‐layer volumes but also from its ability to regulate the extracellular matrix production by dermis fibroblasts. Hyaluronic acid (HA), a major component of the extracellular matrix, is a commonly used injectable dermal filler showing excellent efficiencies to maintain tissue augmentation even after its biodegradation. To improve their stability, the HA molecules can also be “cross‐linked” to each other. The effects of cross‐linked HA‐based fillers on the dermal structure are well known. For safety reasons, most of the physicians prefer to use the blunt cannula for injections. However, evidences showed that the cannula could not be located in the dermis, but it passes through immediate hypodermis and the long‐lasting effect of cross‐linked HA‐based fillers may be related to its effects on adipose tissue. To test whether cross‐linked HA has a direct effect on human adipocytes, we treated isolated adipocytes and precursors cells from human skin donors with cross‐linked HA. Biochemical and cellular analysis demonstrated that treatment by cross‐linked HA showed beneficial effects on differentiated cell adherence and survival as well as reduced basal and induced lipolysis in fully mature adipocytes. Taken together, these data showed that cross‐linked HA promoted cell adherence and preserved the adipogenic capacity of preadipocytes during prolonged cell culture, bringing additional evidences of the beneficial role of cross‐linked HA‐based fillers in maintenance of the subcutaneous fat mass. This first study could defend a preventive approach to facial volume loss during natural aging.
Collapse
|
340
|
Generation of a p16 Reporter Mouse and Its Use to Characterize and Target p16 high Cells In Vivo. Cell Metab 2020; 32:814-828.e6. [PMID: 32949498 DOI: 10.1016/j.cmet.2020.09.006] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 12/25/2022]
Abstract
Cell senescence plays a key role in age-associated organ dysfunction, but the in vivo pathogenesis is largely unclear. Here, we generated a p16-CreERT2-tdTomato mouse model to analyze the in vivo characteristics of p16high cells at a single-cell level. We found tdTomato-positive p16high cells detectable in all organs, which were enriched with age. We also found that these cells failed to proliferate and had half-lives ranging from 2.6 to 4.2 months, depending on the tissue examined. Single-cell transcriptomics in the liver and kidneys revealed that p16high cells were present in various cell types, though most dominant in hepatic endothelium and in renal proximal and distal tubule epithelia, and that these cells exhibited heterogeneous senescence-associated phenotypes. Further, elimination of p16high cells ameliorated nonalcoholic steatohepatitis-related hepatic lipidosis and immune cell infiltration. Our new mouse model and single-cell analysis provide a powerful resource to enable the discovery of previously unidentified senescence functions in vivo.
Collapse
|
341
|
Affiliation(s)
- Diane I. Duncan
- Department of Plastic Surgery Plastic Surgical Associates of Fort Collins, P. C. Fort Collins Colorado USA
| |
Collapse
|
342
|
Kirkland JL, Tchkonia T. Senolytic drugs: from discovery to translation. J Intern Med 2020; 288:518-536. [PMID: 32686219 PMCID: PMC7405395 DOI: 10.1111/joim.13141] [Citation(s) in RCA: 634] [Impact Index Per Article: 126.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/31/2020] [Accepted: 06/09/2020] [Indexed: 12/14/2022]
Abstract
Senolytics are a class of drugs that selectively clear senescent cells (SC). The first senolytic drugs Dasatinib, Quercetin, Fisetin and Navitoclax were discovered using a hypothesis-driven approach. SC accumulate with ageing and at causal sites of multiple chronic disorders, including diseases accounting for the bulk of morbidity, mortality and health expenditures. The most deleterious SC are resistant to apoptosis and have up-regulation of anti-apoptotic pathways which defend SC against their own inflammatory senescence-associated secretory phenotype (SASP), allowing them to survive, despite killing neighbouring cells. Senolytics transiently disable these SCAPs, causing apoptosis of those SC with a tissue-destructive SASP. Because SC take weeks to reaccumulate, senolytics can be administered intermittently - a 'hit-and-run' approach. In preclinical models, senolytics delay, prevent or alleviate frailty, cancers and cardiovascular, neuropsychiatric, liver, kidney, musculoskeletal, lung, eye, haematological, metabolic and skin disorders as well as complications of organ transplantation, radiation and cancer treatment. As anticipated for agents targeting the fundamental ageing mechanisms that are 'root cause' contributors to multiple disorders, potential uses of senolytics are protean, potentially alleviating over 40 conditions in preclinical studies, opening a new route for treating age-related dysfunction and diseases. Early pilot trials of senolytics suggest they decrease senescent cells, reduce inflammation and alleviate frailty in humans. Clinical trials for diabetes, idiopathic pulmonary fibrosis, Alzheimer's disease, COVID-19, osteoarthritis, osteoporosis, eye diseases and bone marrow transplant and childhood cancer survivors are underway or beginning. Until such studies are done, it is too early for senolytics to be used outside of clinical trials.
Collapse
Affiliation(s)
- J L Kirkland
- From the, Mayo Clinic Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| | - T Tchkonia
- From the, Mayo Clinic Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| |
Collapse
|
343
|
Covarrubias AJ, Kale A, Perrone R, Lopez-Dominguez JA, Pisco AO, Kasler HG, Schmidt MS, Heckenbach I, Kwok R, Wiley CD, Wong HS, Gibbs E, Iyer SS, Basisty N, Wu Q, Kim IJ, Silva E, Vitangcol K, Shin KO, Lee YM, Riley R, Ben-Sahra I, Ott M, Schilling B, Scheibye-Knudsen M, Ishihara K, Quake SR, Newman J, Brenner C, Campisi J, Verdin E. Senescent cells promote tissue NAD + decline during ageing via the activation of CD38 + macrophages. Nat Metab 2020; 2:1265-1283. [PMID: 33199924 PMCID: PMC7908681 DOI: 10.1038/s42255-020-00305-3] [Citation(s) in RCA: 256] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 09/25/2020] [Indexed: 11/08/2022]
Abstract
Declining tissue nicotinamide adenine dinucleotide (NAD) levels are linked to ageing and its associated diseases. However, the mechanism for this decline is unclear. Here, we show that pro-inflammatory M1-like macrophages, but not naive or M2 macrophages, accumulate in metabolic tissues, including visceral white adipose tissue and liver, during ageing and acute responses to inflammation. These M1-like macrophages express high levels of the NAD-consuming enzyme CD38 and have enhanced CD38-dependent NADase activity, thereby reducing tissue NAD levels. We also find that senescent cells progressively accumulate in visceral white adipose tissue and liver during ageing and that inflammatory cytokines secreted by senescent cells (the senescence-associated secretory phenotype, SASP) induce macrophages to proliferate and express CD38. These results uncover a new causal link among resident tissue macrophages, cellular senescence and tissue NAD decline during ageing and offer novel therapeutic opportunities to maintain NAD levels during ageing.
Collapse
Affiliation(s)
- Anthony J Covarrubias
- Buck Institute for Research on Aging, Novato, CA, USA
- UCSF Department of Medicine, San Francisco, CA, USA
| | - Abhijit Kale
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | | | | | | | - Mark S Schmidt
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Indra Heckenbach
- Buck Institute for Research on Aging, Novato, CA, USA
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ryan Kwok
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Hoi-Shan Wong
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Eddy Gibbs
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Shankar S Iyer
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Qiuxia Wu
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Ik-Jung Kim
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Elena Silva
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Kyong-Oh Shin
- College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Republic of Korea
| | - Yong-Moon Lee
- College of Pharmacy, Chungbuk National University, Cheongju, Republic of Korea
| | | | - Issam Ben-Sahra
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL, USA
| | - Melanie Ott
- Gladstone Institutes, Virology and Immunology, San Francisco, CA, USA
| | | | - Morten Scheibye-Knudsen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Katsuhiko Ishihara
- Immunology and Molecular Genetics, Kawasaki Medical School, Kurashiki, Japan
| | - Stephen R Quake
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - John Newman
- Buck Institute for Research on Aging, Novato, CA, USA
- UCSF Department of Medicine, San Francisco, CA, USA
| | - Charles Brenner
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Diabetes & Cancer Metabolism, City of Hope National Medical Center, Duarte, CA, USA
| | | | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA.
- UCSF Department of Medicine, San Francisco, CA, USA.
| |
Collapse
|
344
|
Lilja S, Oldenburg J, Pointner A, Dewald L, Lerch M, Hippe B, Switzeny O, Haslberger A. Epigallocatechin Gallate Effectively Affects Senescence and Anti-SASP via SIRT3 in 3T3-L1 Preadipocytes in Comparison with Other Bioactive Substances. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4793125. [PMID: 33149809 PMCID: PMC7603628 DOI: 10.1155/2020/4793125] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/28/2020] [Accepted: 10/09/2020] [Indexed: 01/10/2023]
Abstract
AIM We investigated different bioactive compounds including epigallocatechin gallate (EGCG), anthocyanidin, resveratrol, phloretin, spermidine, butyrate, and β-hydroxybutyrate with regard to their effect on SIRT3 via NRF2 and modulation of the proinflammatory senescence-associated secretory phenotype (SASP) in senescence induced 3T3-L1 preadipocytes. METHODS For induction of senescence, 3T3-L1 preadipocytes were incubated with bromodeoxyuridine (BrdU) for 8 days. Cell cycle inhibition was observed, and β-galactosidase activity was measured. After BrdU treatment, cells were treated with different bioactive compounds in various concentrations for 96 h. ELISA was used for determining proinflammatory cytokine IL6 in SASP cells. RESULTS CDKN1a increased significantly after BrdU incubation compared to untreated control (p < 0.01). All secondary plant ingredients used for treatment, but not anthocyanidin 50 μM, decrease CDKN1a expression (p < 0.05), whereas most endogenous substances did not attenuate CDKN1a. IL6 secretion positively correlated with CDKN1a (p < 0.01), whereas EGCG could diminish both, IL6 and CDKN1a with the strongest effect (p < 0.01). Although NRF2 positively correlated with SIRT3 activation (p < 0.05), only resveratrol (p < 0.01) and anthocyanidin (p < 0.05) could activate NRF2 significantly. Solely anthocyanidin 50 μM (p < 0.05) and 100 μM (p < 0.01) and EGCG 50 μM (p < 0.01) could increase SIRT3 expression. Activation of SIRT3 with EGCG correlated with lowered IL6 secretion significantly (p < 0.05) but not with anthocyanidin. CONCLUSION Accumulation of senescent cells in adipose tissue plays an important role in obesity and age-related diseases. SIRT3, located in the mitochondria, can regulate ROS via different pathways. Thus, targeting SIRT3 activating compounds such as EGCG may delay senescence of cells and senescence induced inflammatory processes.
Collapse
Affiliation(s)
- Stephanie Lilja
- Department of Nutritional Sciences, University of Vienna, 1090 Vienna, Austria
| | - Julia Oldenburg
- Department of Nutritional Sciences, University of Vienna, 1090 Vienna, Austria
| | - Angelika Pointner
- Department of Nutritional Sciences, University of Vienna, 1090 Vienna, Austria
| | - Laura Dewald
- Department of Nutritional Sciences, University of Vienna, 1090 Vienna, Austria
| | - Mariam Lerch
- Department of Nutritional Sciences, University of Vienna, 1090 Vienna, Austria
| | - Berit Hippe
- HealthBioCare GmbH Nußdorferstraße 67, 1090 Wien, Austria
| | | | | |
Collapse
|
345
|
Activation of LKB1 rescues 3T3-L1 adipocytes from senescence induced by Sirt1 knock-down: a pivotal role of LKB1 in cellular aging. Aging (Albany NY) 2020; 12:18942-18956. [PMID: 33040052 PMCID: PMC7732306 DOI: 10.18632/aging.104052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 08/24/2020] [Indexed: 01/24/2023]
Abstract
Previous reports have shown that excess calorie intake promotes p53 dependent senescence in mouse adipose tissues. The objective of the current study was to address the mechanism underlying this observation, i.e. adipocyte aging. Using cultured 3T3-L1 cells, we investigated the involvement of energy regulators Sirt1, AMPK, and LKB1 in senescence. Fifteen days post differentiation, Sirt1 knock-down increased senescence-associated beta-galactosidase (SA-β-Gal) staining by 20-40% (p<0.05, n=12) and both cyclin kinase inhibitor p21Cip and chemokine receptor IL8Rb expression by 2-4 fold. ATP and expression of mitochondria Complex 1 were also reduced by 30% and 50%, respectively (p<0.05, n=4). Such energy depletion may have caused the observed increase in AMPK activity, despite LKB1 activity downregulation. This association between Sirt1 and LKB1 activity was confirmed in vivo in mouse adipose tissue. Upregulation of LKB1 activity by expression of the Sirt1-insensitive LKB1-K48R mutant in 3T3-L1 cells completely prevented the senescence-associated changes of Sirt1 knock-down. In addition, cellular senescence, which also occurs in cultured primary human aortic endothelial cells, was largely prevented by ectopic expression of LKB1. These results suggest that LKB1 plays a pivotal role in cellular senescence occurring in adipocytes and other cell types.
Collapse
|
346
|
Espeland MA, Gaussoin SA, Bahnson J, Vaughan EM, Knowler WC, Simpson FR, Hazuda HP, Johnson KC, Munshi MN, Coday M, Pi-Sunyer X. Impact of an 8-Year Intensive Lifestyle Intervention on an Index of Multimorbidity. J Am Geriatr Soc 2020; 68:2249-2256. [PMID: 33267558 PMCID: PMC8299520 DOI: 10.1111/jgs.16672] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 05/06/2020] [Accepted: 05/10/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND/OBJECTIVES Type 2 diabetes mellitus and obesity are sometimes described as conditions that accelerate aging. Multidomain lifestyle interventions have shown promise to slow the accumulation of age-related diseases, a hallmark of aging. However, they have not been assessed among at-risk individuals with these two conditions. We examined the relative impact of 8 years of a multidomain lifestyle intervention on an index of multimorbidity. DESIGN Randomized controlled clinical trial comparing an intensive lifestyle intervention (ILI) that targeted weight loss through caloric restriction and increased physical activity with a control condition of diabetes support and education (DSE). SETTING Sixteen U.S. academic centers. PARTICIPANTS A total of 5,145 volunteers, aged 45 to 76, with established type 2 diabetes mellitus and overweight or obesity who met eligibility criteria for a randomized controlled clinical trial. MEASUREMENTS A multimorbidity index that included nine age-related chronic diseases and death was tracked over 8 years of intervention delivery. RESULTS Among individuals assigned to DSE, the multimorbidity index scores increased by an average of .98 (95% confidence interval [CI] = .94-1.02) over 8 years, compared with .89 (95% CI = .85-.93) among those in the multidomain ILI, which was a 9% difference (P = .003). Relative intervention effects were similar among individuals grouped by baseline body mass index, age, and sex, and they were greater for those with lower levels of multimorbidity index scores at baseline. CONCLUSIONS Increases in multimorbidity over time among adults with overweight or obesity and type 2 diabetes mellitus may be slowed by multidomain ILI. J Am Geriatr Soc 68:2249-2256, 2020.
Collapse
Affiliation(s)
- Mark A. Espeland
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC
| | - Sarah A. Gaussoin
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC
| | - Judy Bahnson
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC
| | | | - William C. Knowler
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ
| | - Felicia R. Simpson
- Department of Mathematics, Winston-Salem State University, Winston-Salem, NC 27110
| | - Helen P. Hazuda
- Department of Clinical Epidemiology, University of Texas Health Science Center, San Antonio, TX
| | - Karen C. Johnson
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Medha N. Munshi
- Joslin Geriatric Diabetes Program, Joslin Diabetes Center, Boston, MA
| | - Mace Coday
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Xavier Pi-Sunyer
- Department of Medicine, Columbia University School of Medicine, New York, NY
| |
Collapse
|
347
|
Spinelli R, Parrillo L, Longo M, Florese P, Desiderio A, Zatterale F, Miele C, Raciti GA, Beguinot F. Molecular basis of ageing in chronic metabolic diseases. J Endocrinol Invest 2020; 43:1373-1389. [PMID: 32358737 PMCID: PMC7481162 DOI: 10.1007/s40618-020-01255-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/10/2020] [Indexed: 02/06/2023]
Abstract
AIM Over the last decades, the shift in age distribution towards older ages and the progressive ageing which has occurred in most populations have been paralleled by a global epidemic of obesity and its related metabolic disorders, primarily, type 2 diabetes (T2D). Dysfunction of the adipose tissue (AT) is widely recognized as a significant hallmark of the ageing process that, in turn, results in systemic metabolic alterations. These include insulin resistance, accumulation of ectopic lipids and chronic inflammation, which are responsible for an elevated risk of obesity and T2D onset associated to ageing. On the other hand, obesity and T2D, the paradigms of AT dysfunction, share many physiological characteristics with the ageing process, such as an increased burden of senescent cells and epigenetic alterations. Thus, these chronic metabolic disorders may represent a state of accelerated ageing. MATERIALS AND METHODS A more precise explanation of the fundamental ageing mechanisms that occur in AT and a deeper understanding of their role in the interplay between accelerated ageing and AT dysfunction can be a fundamental leap towards novel therapies that address the causes, not just the symptoms, of obesity and T2D, utilizing strategies that target either senescent cells or DNA methylation. RESULTS In this review, we summarize the current knowledge of the pathways that lead to AT dysfunction in the chronological ageing process as well as the pathophysiology of obesity and T2D, emphasizing the critical role of cellular senescence and DNA methylation. CONCLUSION Finally, we highlight the need for further research focused on targeting these mechanisms.
Collapse
Affiliation(s)
- R Spinelli
- Department of Translation Medicine, Federico II University of Naples, 80131, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - L Parrillo
- Department of Translation Medicine, Federico II University of Naples, 80131, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - M Longo
- Department of Translation Medicine, Federico II University of Naples, 80131, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - P Florese
- Department of Translation Medicine, Federico II University of Naples, 80131, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - A Desiderio
- Department of Translation Medicine, Federico II University of Naples, 80131, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - F Zatterale
- Department of Translation Medicine, Federico II University of Naples, 80131, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - C Miele
- Department of Translation Medicine, Federico II University of Naples, 80131, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - G Alexander Raciti
- Department of Translation Medicine, Federico II University of Naples, 80131, Naples, Italy
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy
| | - F Beguinot
- Department of Translation Medicine, Federico II University of Naples, 80131, Naples, Italy.
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131, Naples, Italy.
| |
Collapse
|
348
|
Hall BM, Gleiberman AS, Strom E, Krasnov PA, Frescas D, Vujcic S, Leontieva OV, Antoch MP, Kogan V, Koman IE, Zhu Y, Tchkonia T, Kirkland JL, Chernova OB, Gudkov AV. Immune checkpoint protein VSIG4 as a biomarker of aging in murine adipose tissue. Aging Cell 2020; 19:e13219. [PMID: 32856419 PMCID: PMC7576241 DOI: 10.1111/acel.13219] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 05/26/2020] [Accepted: 07/03/2020] [Indexed: 12/20/2022] Open
Abstract
Adipose tissue is recognized as a major source of systemic inflammation with age, driving age-related tissue dysfunction and pathogenesis. Macrophages (Mφ) are central to these changes yet adipose tissue Mφ (ATMs) from aged mice remain poorly characterized. To identify biomarkers underlying changes in aged adipose tissue, we performed an unbiased RNA-seq analysis of ATMs from young (8-week-old) and healthy aged (80-week-old) mice. One of the genes identified, V-set immunoglobulin-domain-containing 4 (VSIG4/CRIg), encodes a Mφ-associated complement receptor and B7 family-related immune checkpoint protein. Here, we demonstrate that Vsig4 expression is highly upregulated with age in perigonadal white adipose tissue (gWAT) in two mouse strains (inbred C57BL/6J and outbred NIH Swiss) independent of gender. The accumulation of VSIG4 was mainly attributed to a fourfold increase in the proportion of VSIG4+ ATMs (13%-52%). In a longitudinal study, VSIG4 expression in gWAT showed a strong correlation with age within a cohort of male and female mice and correlated strongly with physiological frailty index (PFI, a multi-parameter assessment of health) in male mice. Our results indicate that VSIG4 is a novel biomarker of aged murine ATMs. VSIG4 expression was also found to be elevated in other aging tissues (e.g., thymus) and was strongly induced in tumor-adjacent stroma in cases of spontaneous and xenograft lung cancer models. VSIG4 expression was recently associated with cancer and several inflammatory diseases with diagnostic and prognostic potential in both mice and humans. Further investigation is required to determine whether VSIG4-positive Mφ contribute to immunosenescence and/or systemic age-related deficits.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Olga V. Leontieva
- Department of Pharmacology and TherapeuticsRoswell Park Comprehensive Cancer CenterBuffaloNYUSA
| | - Marina P. Antoch
- Department of Pharmacology and TherapeuticsRoswell Park Comprehensive Cancer CenterBuffaloNYUSA
| | - Valeria Kogan
- Institute for Translational ResearchAriel UniversityArielIsrael
| | - Igor E. Koman
- Institute for Translational ResearchAriel UniversityArielIsrael
| | - Yi Zhu
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMNUSA
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMNUSA
| | | | | | - Andrei V. Gudkov
- Everon Biosciences IncBuffaloNYUSA
- Department of Cell Stress BiologyRoswell Park Comprehensive Cancer CenterBuffaloNYUSA
- Genome Protection IncBuffaloNYUSA
| |
Collapse
|
349
|
Al Suraih MS, Trussoni CE, Splinter PL, LaRusso NF, O’Hara SP. Senescent cholangiocytes release extracellular vesicles that alter target cell phenotype via the epidermal growth factor receptor. Liver Int 2020; 40:2455-2468. [PMID: 32558183 PMCID: PMC7669612 DOI: 10.1111/liv.14569] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/18/2020] [Accepted: 06/05/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Primary sclerosing cholangitis (PSC) is a chronic liver disease characterized by peribiliary inflammation and fibrosis. Cholangiocyte senescence is a prominent feature of PSC. Here, we hypothesize that extracellular vesicles (EVs) from senescent cholangiocytes influence the phenotype of target cells. METHODS EVs were isolated from normal human cholangiocytes (NHCs), cholangiocytes from PSC patients and NHCs experimentally induced to senescence. NHCs, malignant human cholangiocytes (MHCs) and monocytes were exposed to 108 EVs from each donor cell population and assessed for proliferation, MAPK activation and migration. Additionally, we isolated EVs from plasma of wild-type and Mdr2-/- mice (a murine model of PSC), and assessed mouse monocyte activation. RESULTS EVs exhibited the size and protein markers of exosomes. The number of EVs released from senescent human cholangiocytes was increased; similarly, the EVs in plasma from Mdr2-/- mice were increased. Additionally, EVs from senescent cholangiocytes were enriched in multiple growth factors, including EGF. NHCs exposed to EVs from senescent cholangiocytes showed increased NRAS and ERK1/2 activation. Moreover, EVs from senescent cholangiocytes promoted proliferation of NHCs and MHCs, findings that were blocked by erlotinib, an EGF receptor inhibitor. Furthermore, EVs from senescent cholangiocytes induced EGF-dependent Interleukin 1-beta and Tumour necrosis factor expression and migration of human monocytes; similarly, Mdr2-/- mouse plasma EVs induced activation of mouse monocytes. CONCLUSIONS The data continue to support the importance of cholangiocyte senescence in PSC pathogenesis, directly implicate EVs in cholangiocyte proliferation, malignant progression and immune cell activation and migration, and identify novel therapeutic approaches for PSC.
Collapse
Affiliation(s)
- Mohammed S. Al Suraih
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota. 55905.,Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota. 55905
| | - Christy E. Trussoni
- Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota. 55905
| | - Patrick L. Splinter
- Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota. 55905
| | - Nicholas F. LaRusso
- Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota. 55905
| | - Steven P. O’Hara
- Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota. 55905
| |
Collapse
|
350
|
Parvizi M, Ryan ZC, Ebtehaj S, Arendt BK, Lanza IR. The secretome of senescent preadipocytes influences the phenotype and function of cells of the vascular wall. Biochim Biophys Acta Mol Basis Dis 2020; 1867:165983. [PMID: 33002577 DOI: 10.1016/j.bbadis.2020.165983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/16/2020] [Accepted: 09/24/2020] [Indexed: 01/10/2023]
Abstract
Senescent cells accumulate in numerous tissues in several chronic conditions such as aging, obesity, and diabetes. These cells are in a state of irreversible cell-cycle arrest and secrete inflammatory cytokines, chemokines and other immune modulators that have paracrine effects on nearby tissues. Adipose tissue, in particular, harbors senescent cells, which have been linked with numerous chronic conditions and age-related comorbidities. Here we performed a series of in vitro experiments to determine the influence of senescent preadipocytes on key cell types found in vessel walls, including vascular smooth muscle cells (VSMCs), endothelial cells (ECs), macrophages (MQs), and adipose-derived stromal/stem cells (ASCs). Primary human preadipocytes were irradiated to trigger a senescence-like phenotype. VSMCs, ECs, MQs, and ASCs were exposed to conditioned media collected from irradiated preadipocytes or control preadipocytes. Additional experiments were performed where VSMCs, ECs, MQs, and ASCs were co-cultured with irradiated or control preadipocytes. The secretome of irradiated cells induced an inflammatory phenotype, decreased cell viability, disrupted proliferation and migration, and impaired metabolic function of these cell types in vitro. These maladaptive changes in response to senescent cell exposure provide early evidence in support of a hypothesis that senescent preadipocytes trigger phenotypic and functional changes in key cellular components of blood vessels that may contribute to vascular disease.
Collapse
Affiliation(s)
- Mojtaba Parvizi
- Division of Endocrinology and Metabolism, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Zachary C Ryan
- Division of Endocrinology and Metabolism, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Sanam Ebtehaj
- Department of Cardiovascular Diseases, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Bonnie K Arendt
- Division of Endocrinology and Metabolism, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Ian R Lanza
- Division of Endocrinology and Metabolism, Mayo Clinic College of Medicine, Rochester, MN, USA.
| |
Collapse
|