551
|
Troike KM, Henry BE, Jensen EA, Young JA, List EO, Kopchick JJ, Berryman DE. Impact of Growth Hormone on Regulation of Adipose Tissue. Compr Physiol 2017. [PMID: 28640444 DOI: 10.1002/cphy.c160027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Increasing prevalence of obesity and obesity-related conditions worldwide has necessitated a more thorough understanding of adipose tissue (AT) and expanded the scope of research in this field. AT is now understood to be far more complex and dynamic than previously thought, which has also fueled research to reevaluate how hormones, such as growth hormone (GH), alter the tissue. In this review, we will introduce properties of AT important for understanding how GH alters the tissue, such as anatomical location of depots and adipokine output. We will provide an overview of GH structure and function and define several human conditions and cognate mouse lines with extremes in GH action that have helped shape our understanding of GH and AT. A detailed discussion of the GH/AT relationship will be included that addresses adipokine production, immune cell populations, lipid metabolism, senescence, differentiation, and fibrosis, as well as brown AT and beiging of white AT. A brief overview of how GH levels are altered in an obese state, and the efficacy of GH as a therapeutic option to manage obesity will be given. As we will reveal, the effects of GH on AT are numerous, dynamic and depot-dependent. © 2017 American Physiological Society. Compr Physiol 7:819-840, 2017.
Collapse
Affiliation(s)
- Katie M Troike
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,School of Applied Health Sciences and Wellness, College of Health Sciences and Professions, Ohio University, Athens, Ohio, USA
| | - Brooke E Henry
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,School of Applied Health Sciences and Wellness, College of Health Sciences and Professions, Ohio University, Athens, Ohio, USA
| | - Elizabeth A Jensen
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA.,Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, Ohio, USA
| | - Jonathan A Young
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, Ohio, USA.,Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, Ohio, USA
| | - Edward O List
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, Ohio, USA
| | - John J Kopchick
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA.,Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, Ohio, USA
| | - Darlene E Berryman
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| |
Collapse
|
552
|
Gaborit B, Sengenes C, Ancel P, Jacquier A, Dutour A. Role of Epicardial Adipose Tissue in Health and Disease: A Matter of Fat? Compr Physiol 2017. [PMID: 28640452 DOI: 10.1002/cphy.c160034] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Epicardial adipose tissue (EAT) is a small but very biologically active ectopic fat depot that surrounds the heart. Given its rapid metabolism, thermogenic capacity, unique transcriptome, secretory profile, and simply measurability, epicardial fat has drawn increasing attention among researchers attempting to elucidate its putative role in health and cardiovascular diseases. The cellular crosstalk between epicardial adipocytes and cells of the vascular wall or myocytes is high and suggests a local role for this tissue. The balance between protective and proinflammatory/profibrotic cytokines, chemokines, and adipokines released by EAT seem to be a key element in atherogenesis and could represent a future therapeutic target. EAT amount has been found to predict clinical coronary outcomes. EAT can also modulate cardiac structure and function. Its amount has been associated with atrial fibrillation, coronary artery disease, and sleep apnea syndrome. Conversely, a beiging fat profile of EAT has been identified. In this review, we describe the current state of knowledge regarding the anatomy, physiology and pathophysiological role of EAT, and the factors more globally leading to ectopic fat development. We will also highlight the most recent findings on the origin of this ectopic tissue, and its association with cardiac diseases. © 2017 American Physiological Society. Compr Physiol 7:1051-1082, 2017.
Collapse
Affiliation(s)
- Bénédicte Gaborit
- NORT, Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France.,Endocrinology Metabolic Diseases, and Nutrition Department, Pole ENDO, APHM, Aix-Marseille Univ, Marseille, France
| | - Coralie Sengenes
- STROMALab, Université de Toulouse, EFS, ENVT, Inserm U1031, ERL CNRS 5311, CHU Rangueil, Toulouse, France
| | - Patricia Ancel
- NORT, Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France
| | - Alexis Jacquier
- CNRS UMR 7339, Centre de Résonance Magnétique Biologique et Médicale (CRMBM), Marseille, France.,Radiology department, CHU La Timone, Marseille, France
| | - Anne Dutour
- NORT, Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France.,Endocrinology Metabolic Diseases, and Nutrition Department, Pole ENDO, APHM, Aix-Marseille Univ, Marseille, France
| |
Collapse
|
553
|
Ogrodnik M, Miwa S, Tchkonia T, Tiniakos D, Wilson CL, Lahat A, Day CP, Burt A, Palmer A, Anstee QM, Grellscheid SN, Hoeijmakers JHJ, Barnhoorn S, Mann DA, Bird TG, Vermeij WP, Kirkland JL, Passos JF, von Zglinicki T, Jurk D. Cellular senescence drives age-dependent hepatic steatosis. Nat Commun 2017; 8:15691. [PMID: 28608850 PMCID: PMC5474745 DOI: 10.1038/ncomms15691] [Citation(s) in RCA: 726] [Impact Index Per Article: 90.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 04/20/2017] [Indexed: 02/06/2023] Open
Abstract
The incidence of non-alcoholic fatty liver disease (NAFLD) increases with age. Cellular senescence refers to a state of irreversible cell-cycle arrest combined with the secretion of proinflammatory cytokines and mitochondrial dysfunction. Senescent cells contribute to age-related tissue degeneration. Here we show that the accumulation of senescent cells promotes hepatic fat accumulation and steatosis. We report a close correlation between hepatic fat accumulation and markers of hepatocyte senescence. The elimination of senescent cells by suicide gene-meditated ablation of p16Ink4a-expressing senescent cells in INK-ATTAC mice or by treatment with a combination of the senolytic drugs dasatinib and quercetin (D+Q) reduces overall hepatic steatosis. Conversely, inducing hepatocyte senescence promotes fat accumulation in vitro and in vivo. Mechanistically, we show that mitochondria in senescent cells lose the ability to metabolize fatty acids efficiently. Our study demonstrates that cellular senescence drives hepatic steatosis and elimination of senescent cells may be a novel therapeutic strategy to reduce steatosis.
Collapse
Affiliation(s)
- Mikolaj Ogrodnik
- Newcastle University Institute for Ageing, Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Satomi Miwa
- Newcastle University Institute for Ageing, Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, USA
| | - Dina Tiniakos
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Department of Pathology, Aretaieio Hospital, Medical School, National & Kapodistrian University of Athens, Athens 11528, Greece
| | - Caroline L. Wilson
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Albert Lahat
- Department of Biosciences, Durham University, Durham DH1 3LE, UK
| | - Christoper P. Day
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Liver Unit, Newcastle upon Tyne Hospitals NHS Trust, Freeman Hospital, Newcastle upon Tyne NE7 7DN, UK
| | - Alastair Burt
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- The University of Adelaide, Faculty of Health Science, North Terrace, Adelaide, South Australia 5005, Australia
| | - Allyson Palmer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, USA
| | - Quentin M. Anstee
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | | | - Jan H J. Hoeijmakers
- Department of Molecular Genetics, Erasmus University Medical Center, PO Box 2040, Rotterdam 3000 CA, The Netherlands
- CECAD Forschungszentrum, Universität zu Köln, Joseph-Stelzmann-Straße 26, Köln 50931, Germany
| | - Sander Barnhoorn
- Department of Molecular Genetics, Erasmus University Medical Center, PO Box 2040, Rotterdam 3000 CA, The Netherlands
| | - Derek A. Mann
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Thomas G. Bird
- MRC Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| | - Wilbert P. Vermeij
- Department of Molecular Genetics, Erasmus University Medical Center, PO Box 2040, Rotterdam 3000 CA, The Netherlands
| | - James L. Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, USA
| | - João F. Passos
- Newcastle University Institute for Ageing, Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Thomas von Zglinicki
- Newcastle University Institute for Ageing, Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Diana Jurk
- Newcastle University Institute for Ageing, Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| |
Collapse
|
554
|
Short-Term Dietary Restriction Rescues Mice From Lethal Abdominal Sepsis and Endotoxemia and Reduces the Inflammatory/Coagulant Potential of Adipose Tissue. Crit Care Med 2017; 44:e509-19. [PMID: 26646465 DOI: 10.1097/ccm.0000000000001475] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVES Visceral adipose tissue is a major site for expression of proinflammatory and procoagulant genes during acute systemic inflammation. In this study, we tested whether the loss of fat mass by dietary restriction would remove the major source of these factors resulting in improved tolerance to sepsis and endotoxemia. DESIGN Prospective, laboratory controlled experiments. SETTING Aging and critical care research laboratory in a university hospital. SUBJECTS Middle-aged (12-month old) male C57BL/6 mice. INTERVENTIONS Mice were subjected to 40% dietary restriction for 3 weeks followed by induction of abdominal sepsis or endotoxemia by intraperitoneal injection with cecal slurry or lipopolysaccharide, respectively. MEASUREMENTS AND MAIN RESULTS Compared with freely fed mice, dietary restricted mice exhibited dramatically improved survival (80% vs 0% after sepsis; p < 0.001 and 86% vs 12% after endotoxemia; p = 0.013) and significantly reduced visceral fat-derived messenger RNA expression of interleukin-6, thrombospondin-1, plasminogen activator inhibitor-1, and tissue factor, which positively correlated with fat mass. Plasma levels of interleukin-6 were significantly reduced by dietary restriction and correlated with adipose interleukin-6 messenger RNA levels and fat mass (p < 0.001; R = 0.64 and 0.89). In vitro culture of visceral fat explants from naive dietary restricted mice showed significantly reduced interleukin-6 secretion compared with that from freely fed mice in response to lipopolysaccharide. Analysis of major adipose immune cell populations by flow cytometry demonstrated that macrophages were the only cell population reduced by dietary restriction and that CD11c/CD206 (M2-type) and CD11c/CD206 (double negative) macrophages, in addition to T cells, are the major immune cell populations that produce interleukin-6 in middle-aged mice during systemic inflammation. CONCLUSIONS Short-term dietary restriction drastically improved the survival outcome of middle-aged mice during both polymicrobial sepsis and sterile endotoxemia. Improved survival was accompanied by a significantly attenuated inflammatory response in adipose tissue, which is likely due to alterations of both fat mass quantity and qualitative changes, including a reduction in macrophage populations.
Collapse
|
555
|
Jin B, Chen X, Xing L, Xu W, Fu X, Zhu J, Mou X, Wang Z, Shu J. Tissue-specific effects of estrogen on glycerol channel aquaporin 7 expression in an ovariectomized mouse model of menopause. Climacteric 2017; 20:385-390. [PMID: 28489425 DOI: 10.1080/13697137.2017.1319920] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Elevated fat mass and redistribution of body fat are commonly observed in postmenopausal women. Aquaporin 7 (AQP7), a unique glycerol permeable integral membrane protein, has been associated with the onset of obesity. We hypothesized that estrogen supplementation could counteract this fat accumulation and redistribution through tissue-specific modulation of AQP7. METHODS We measured fat depot weight, adipocyte size, and the expression of AQP7 and glycerol kinase (GK) in visceral and subcutaneous fat tissues of ovariectomized mice supplemented with or without 17β-estradiol. RESULTS Removal of the ovaries resulted in a significant decrease in AQP7 expression and an increase in GK expression in visceral adipocyte tissue; expression of AQP7 and GK in subcutaneous adipose tissue remained unaltered. Supplementation with estrogen significantly restored the visceral, but not subcutaneous, fat depot mass and adipocyte size to those of sham-operated mice. A marked increase in the expression of AQP7 and a reduction of GK were observed selectively in the visceral fat depots in estrogen-treated mice. CONCLUSIONS Our results suggest that estrogen has tissue-specific effects on AQP7 expression, and modulation of AQP7 by estrogen alters the balance of adipocyte metabolism between adipose tissue depots.
Collapse
Affiliation(s)
- B Jin
- a Department of Reproductive Endocrinology , Zhejiang Provincial People's Hospital , Hangzhou , Zhejiang , PR China
| | - X Chen
- a Department of Reproductive Endocrinology , Zhejiang Provincial People's Hospital , Hangzhou , Zhejiang , PR China
| | - L Xing
- a Department of Reproductive Endocrinology , Zhejiang Provincial People's Hospital , Hangzhou , Zhejiang , PR China
| | - W Xu
- a Department of Reproductive Endocrinology , Zhejiang Provincial People's Hospital , Hangzhou , Zhejiang , PR China
| | - X Fu
- a Department of Reproductive Endocrinology , Zhejiang Provincial People's Hospital , Hangzhou , Zhejiang , PR China
| | - J Zhu
- a Department of Reproductive Endocrinology , Zhejiang Provincial People's Hospital , Hangzhou , Zhejiang , PR China
| | - X Mou
- b Clinical Research Institute , Zhejiang Provincial People's Hospital , Hangzhou , Zhejiang , PR China
| | - Z Wang
- b Clinical Research Institute , Zhejiang Provincial People's Hospital , Hangzhou , Zhejiang , PR China
| | - J Shu
- a Department of Reproductive Endocrinology , Zhejiang Provincial People's Hospital , Hangzhou , Zhejiang , PR China
| |
Collapse
|
556
|
Roichman A, Kanfi Y, Glazz R, Naiman S, Amit U, Landa N, Tinman S, Stein I, Pikarsky E, Leor J, Cohen HY. SIRT6 Overexpression Improves Various Aspects of Mouse Healthspan. J Gerontol A Biol Sci Med Sci 2017; 72:603-615. [PMID: 27519885 DOI: 10.1093/gerona/glw152] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 07/18/2016] [Indexed: 01/08/2023] Open
Abstract
The extension in human lifespan in the last century results in a significant increase in incidence of age related diseases. It is therefore crucial to identify key factors that control elderly healthspan. Similar to dietary restriction, mice overexpressing the NAD+ dependent protein deacylase SIRT6 (MOSES) live longer and have reduced IGF-1 levels. However, it is as yet unknown whether SIRT6 also affects various healthspan parameters. Here, a range of age related phenotypes was evaluated in MOSES mice. In comparison to their wild-type (WT) littermates, old MOSES mice showed amelioration of a variety of age-related disorders, including: improved glucose tolerance, younger hormonal profile, reduced age-related adipose inflammation and increased physical activity. The increased activity was accompanied with increased muscle AMP-activated protein kinase (AMPK) activity. Altogether, these results indicate that overexpression of SIRT6 in mice retards important aspects of the aging process and suggest SIRT6 to be a potential therapeutic target for the treatment of a set of age-related disorders.
Collapse
Affiliation(s)
- Asael Roichman
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Yariv Kanfi
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Renana Glazz
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Shoshana Naiman
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Uri Amit
- Tamman and Neufeld Cardiovascular Research Institute, Sheba Center of Regenerative Medicine, Sheba Medical Center, Tel Aviv University, Israel
| | - Natalie Landa
- Tamman and Neufeld Cardiovascular Research Institute, Sheba Center of Regenerative Medicine, Sheba Medical Center, Tel Aviv University, Israel
| | - Simon Tinman
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Ilan Stein
- The Lautenberg Center for Immunology and Cancer Research, IMRIC, Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Eli Pikarsky
- The Lautenberg Center for Immunology and Cancer Research, IMRIC, Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Jonathan Leor
- Tamman and Neufeld Cardiovascular Research Institute, Sheba Center of Regenerative Medicine, Sheba Medical Center, Tel Aviv University, Israel
| | - Haim Y Cohen
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| |
Collapse
|
557
|
Sarcopenic obesity or obese sarcopenia: A cross talk between age-associated adipose tissue and skeletal muscle inflammation as a main mechanism of the pathogenesis. Ageing Res Rev 2017; 35:200-221. [PMID: 27702700 DOI: 10.1016/j.arr.2016.09.008] [Citation(s) in RCA: 519] [Impact Index Per Article: 64.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/23/2016] [Accepted: 09/26/2016] [Indexed: 02/08/2023]
Abstract
Sarcopenia, an age-associated decline in skeletal muscle mass coupled with functional deterioration, may be exacerbated by obesity leading to higher disability, frailty, morbidity and mortality rates. In the combination of sarcopenia and obesity, the state called sarcopenic obesity (SOB), some key age- and obesity-mediated factors and pathways may aggravate sarcopenia. This review will analyze the mechanisms underlying the pathogenesis of SOB. In obese adipose tissue (AT), adipocytes undergo hypertrophy, hyperplasia and activation resulted in accumulation of pro-inflammatory macrophages and other immune cells as well as dysregulated production of various adipokines that together with senescent cells and the immune cell-released cytokines and chemokines create a local pro-inflammatory status. In addition, obese AT is characterized by excessive production and disturbed capacity to store lipids, which accumulate ectopically in skeletal muscle. These intramuscular lipids and their derivatives induce mitochondrial dysfunction characterized by impaired β-oxidation capacity and increased reactive oxygen species formation providing lipotoxic environment and insulin resistance as well as enhanced secretion of some pro-inflammatory myokines capable of inducing muscle dysfunction by auto/paracrine manner. In turn, by endocrine manner, these myokines may exacerbate AT inflammation and also support chronic low grade systemic inflammation (inflammaging), overall establishing a detrimental vicious circle maintaining AT and skeletal muscle inflammation, thus triggering and supporting SOB development. Under these circumstances, we believe that AT inflammation dominates over skeletal muscle inflammation. Thus, in essence, it redirects the vector of processes from "sarcopenia→obesity" to "obesity→sarcopenia". We therefore propose that this condition be defined as "obese sarcopenia", to reflect the direction of the pathological pathway.
Collapse
|
558
|
Niedernhofer LJ, Kirkland JL, Ladiges W. Molecular pathology endpoints useful for aging studies. Ageing Res Rev 2017; 35:241-249. [PMID: 27721062 DOI: 10.1016/j.arr.2016.09.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 09/15/2016] [Accepted: 09/30/2016] [Indexed: 12/12/2022]
Abstract
The first clinical trial aimed at targeting fundamental processes of aging will soon be launched (TAME: Targeting Aging with Metformin). In its wake is a robust pipeline of therapeutic interventions that have been demonstrated to extend lifespan or healthspan of preclinical models, including rapalogs, antioxidants, anti-inflammatory agents, and senolytics. This ensures that if the TAME trial is successful, numerous additional clinical trials are apt to follow. But a significant impediment to these trials remains the question of what endpoints should be measured? The design of the TAME trial very cleverly skirts around this based on the fact that there are decades of data on metformin in humans, providing unequaled clarity of what endpoints are most likely to yield a positive outcome. But for a new chemical entity, knowing what endpoints to measure remains a formidable challenge. For economy's sake, and to achieve results in a reasonable time frame, surrogate markers of lifespan and healthy aging are desperately needed. This review provides a comprehensive analysis of molecular endpoints that are currently being used as indices of age-related phenomena (e.g., morbidity, frailty, mortality) and proposes an approach for validating and prioritizing these endpoints.
Collapse
Affiliation(s)
- L J Niedernhofer
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL 33458, United States.
| | - J L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, United States
| | - W Ladiges
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, WA 98195, United States
| |
Collapse
|
559
|
Hanh NTH, Tuyet LT, Dao DTA, Tao Y, Chu DT. Childhood Obesity Is a High-risk Factor for Hypertriglyceridemia: A Case-control Study in Vietnam. Osong Public Health Res Perspect 2017; 8:138-146. [PMID: 28540158 PMCID: PMC5441441 DOI: 10.24171/j.phrp.2017.8.2.06] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 03/29/2017] [Accepted: 04/06/2017] [Indexed: 12/25/2022] Open
Abstract
Objectives To investigate the relationship between dyslipidemia and obesity status among Viet-namese adolescents. Methods In this case-control study, 282 adolescents (6–11 years), including 88 obese cases and 194 normal-weight controls, were recruited from a population-based cross-sectional study from two provinces in Vietnam. The anthropometric, blood lipid, and other laboratory test results of the study subjects were analyzed. Results Obese children tended to have more visceral fat (Pearson’s r = 0.795, p < 0.0001) than subcutaneous fat (Pearson’s r = 0.754, p < 0.0001), and this difference was associated with an increase in blood triglyceride level (Pearson’s r = 0.232, p < 0.05) and a strikingly high rate of hypertriglyceridemia (38.6%). We also found that birth weight and parental body mass index were related to the status of obesity among the study subjects. However, only birth weight was significantly higher in the obese group than in the normal weight group. These findings indicate the effect of prenatal nutrition on childhood obesity. Furthermore, high-birth weight children had a surprisingly high rate of obesity. Conclusion Together, our data suggest that obesity increased the risk for hypertriglyceridemia, which was, at least partially, due to prenatal nutrition.
Collapse
Affiliation(s)
| | - Le Thi Tuyet
- Hanoi National University of Education, Hanoi, Vietnam
| | | | - Yang Tao
- College of Food Science and Technology, Nanjing Agricultural University, China
| | - Dinh-Toi Chu
- Hanoi National University of Education, Hanoi, Vietnam.,Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway
| |
Collapse
|
560
|
Cellular Senescence: A Translational Perspective. EBioMedicine 2017; 21:21-28. [PMID: 28416161 PMCID: PMC5514381 DOI: 10.1016/j.ebiom.2017.04.013] [Citation(s) in RCA: 692] [Impact Index Per Article: 86.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/06/2017] [Accepted: 04/06/2017] [Indexed: 12/17/2022] Open
Abstract
Cellular senescence entails essentially irreversible replicative arrest, apoptosis resistance, and frequently acquisition of a pro-inflammatory, tissue-destructive senescence-associated secretory phenotype (SASP). Senescent cells accumulate in various tissues with aging and at sites of pathogenesis in many chronic diseases and conditions. The SASP can contribute to senescence-related inflammation, metabolic dysregulation, stem cell dysfunction, aging phenotypes, chronic diseases, geriatric syndromes, and loss of resilience. Delaying senescent cell accumulation or reducing senescent cell burden is associated with delay, prevention, or alleviation of multiple senescence-associated conditions. We used a hypothesis-driven approach to discover pro-survival Senescent Cell Anti-apoptotic Pathways (SCAPs) and, based on these SCAPs, the first senolytic agents, drugs that cause senescent cells to become susceptible to their own pro-apoptotic microenvironment. Several senolytic agents, which appear to alleviate multiple senescence-related phenotypes in pre-clinical models, are beginning the process of being translated into clinical interventions that could be transformative. Cellular senescence is among the aging processes underlying chronic diseases, loss of resilience, and geriatric syndromes. Senolytics selectively induce senescent cell apoptosis. They delay or alleviate multiple disorders in preclinical studies. If senolytics are demonstrated to be effective and safe in clinical trials, they could be transformative.
Collapse
|
561
|
Rui Y, Cheng J, Qin L, Shan C, Chang J, Wang G, Wan Z. Effects of vitamin D and resveratrol on metabolic associated markers in liver and adipose tissue from SAMP8 mice. Exp Gerontol 2017; 93:16-28. [PMID: 28411010 DOI: 10.1016/j.exger.2017.03.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 03/17/2017] [Accepted: 03/19/2017] [Indexed: 12/31/2022]
Abstract
SAMP8 mice exhibit multiple metabolic characteristics associated with age, and it is a suitable candidate for researching aging associated metabolic dysfunction. OBJECTIVES We aimed to 1) explore how key metabolic markers will be altered in both liver and adipose tissue with aging in SAMP8 mice; and 2) how the combination of vitamin D (VD) with resveratrol (RSV) will affect aging associated metabolic impairment in liver and adipose tissue from SAMP8 mice. METHODS SAMP8 mice and their control SAMR1 mice were divided into 5 groups, i.e. SAMR1, SAMP8, SAMP8 mice supplemented with VD, RSV and VD combined with RSV group, respectively. At the end of the intervention, glucose and insulin tolerance, p-AMP-activated protein kinase (AMPK) and amyloid precursor protein (APP), and endoplasmic reticulum (ER) stress markers in liver and adipose tissue, adiponectin secretion, p-NF-κBp65 and TNF-α protein expression in adipose tissue were determined. RESULTS Compared to SAMR1 control, SAMP8 mice demonstrate impaired glucose tolerance and reduction in circulating adiponectin level; in the liver, SAMP8 mice have reduction in p-Aktser473, elevation in PTP1B and APP, p-eIF2α, GRP78 and p-JNK protein expression. In epididymal (EPI) fat, SAMP8 mice also have elevated p-Aktser473 and PTP1B compared to SAMR1 mice. In both epididymal (EPI) and subcutaneous (SC) fat, there were elevated ER stress markers, reduced p-AMPK and elevated APP, as well as elevated p-NF-κBp65 and TNF-α protein expression from SAMP8 compared to SAMR1 mice. In liver, the combined intervention significantly restored p-Aktser473, p-eIF2α and p-JNK protein expression. In both EPI and SC fat, the combined intervention is effective for reducing p-NF-κB p65 and TNF-α in both fat depot, while only partially reduced ER stress markers in SC fat. As for adiponectin, their combination is unable to reverse reduction in adiponectin level. Adiponectin secretion in SC fat from VD, RSV and VDRSV group were also significantly reduced compared to SAMR1. CONCLUSION The combined intervention might exert greater beneficial effects for reversing aging associated metabolic dysfunction in liver and adipose tissue from SAMP8 mice.
Collapse
Affiliation(s)
- Yehua Rui
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, PR China
| | - Jinbo Cheng
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, PR China
| | - Liqiang Qin
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, PR China
| | - Cheng Shan
- University of Waterloo, Waterloo, Ontario, Canada
| | - Jie Chang
- Department of Occupational and Environmental Health, School of Public Health, Soochow University, 199 Ren'ai Road, Suzhou 215123, PR China
| | - Guiping Wang
- Laboratory Animal Center, Soochow University, 199 Ren'ai Road, Suzhou 215123, PR China
| | - Zhongxiao Wan
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, PR China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Disease, Soochow University, 199 Ren'ai Road, Suzhou 215123, PR China.
| |
Collapse
|
562
|
Darcy J, McFadden S, Bartke A. Altered structure and function of adipose tissue in long-lived mice with growth hormone-related mutations. Adipocyte 2017; 6:69-75. [PMID: 28425851 DOI: 10.1080/21623945.2017.1308990] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
A major focus of biogerontology is elucidating the role(s) of the endocrine system in aging and the accumulation of age-related diseases. Endocrine control of mammalian longevity was first reported in Ames dwarf (Prop1df) mice, which are long-lived due to a recessive Prop1 loss-of-function mutation resulting in deficiency of growth hormone (GH), thyroid-stimulating hormone, and prolactin. Following this report, several other GH-related mutants with altered longevity have been described including long-lived Snell dwarf and growth hormone receptor knockout mice, and short-lived GH overexpressing transgenic mice. One of the emerging areas of interest in these mutant mice is the role of adipose tissue in their altered healthspan and lifespan. Here, we provide an overview of the alterations in body composition of GH-related mutants, as well as the altered thermogenic potential of their brown adipose tissue and the altered cellular senescence and adipokine production of their white adipose tissue.
Collapse
Affiliation(s)
- Justin Darcy
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, Illinois, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - Samuel McFadden
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| |
Collapse
|
563
|
McCulloch K, Litherland GJ, Rai TS. Cellular senescence in osteoarthritis pathology. Aging Cell 2017; 16:210-218. [PMID: 28124466 PMCID: PMC5334539 DOI: 10.1111/acel.12562] [Citation(s) in RCA: 263] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2016] [Indexed: 12/19/2022] Open
Abstract
Cellular senescence is a state of stable proliferation arrest of cells. The senescence pathway has many beneficial effects and is seen to be activated in damaged/stressed cells, as well as during embryonic development and wound healing. However, the persistence and accumulation of senescent cells in various tissues can also impair function and have been implicated in the pathogenesis of many age‐related diseases. Osteoarthritis (OA), a severely debilitating chronic condition characterized by progressive tissue remodeling and loss of joint function, is the most prevalent disease of the synovial joints, and increasing age is the primary OA risk factor. The profile of inflammatory and catabolic mediators present during the pathogenesis of OA is strikingly similar to the secretory profile observed in ‘classical’ senescent cells. During OA, chondrocytes (the sole cell type present within articular cartilage) exhibit increased levels of various senescence markers, such as senescence‐associated beta‐galactosidase (SAβGal) activity, telomere attrition, and accumulation of p16ink4a. This suggests the hypothesis that senescence of cells within joint tissues may play a pathological role in the causation of OA. In this review, we discuss the mechanisms by which senescent cells may predispose synovial joints to the development and/or progression of OA, as well as touching upon various epigenetic alterations associated with both OA and senescence.
Collapse
Affiliation(s)
- Kendal McCulloch
- Institute of Biomedical and Environmental Health Research; University of the West of Scotland; Paisley PA1 2BE UK
| | - Gary J. Litherland
- Institute of Biomedical and Environmental Health Research; University of the West of Scotland; Paisley PA1 2BE UK
| | - Taranjit Singh Rai
- Institute of Biomedical and Environmental Health Research; University of the West of Scotland; Paisley PA1 2BE UK
| |
Collapse
|
564
|
Abstract
Older adults make up an ever-increasing number of patients presenting for surgery, and a significant percentage of these patients will be frail. Frailty is a geriatric syndrome that has been conceptualized as decreased reserve when confronted with stressors, although the precise definition of frailty has not been easy to standardize. The 2 most popular approaches to define frailty are the phenotypic approach and the deficit accumulation approach, although at least 20 tools have been developed, which has made comparison across studies difficult. In epidemiologic studies, baseline frailty has been associated with poor outcomes in both community cohorts and hospitalized patients. Specifically in cardiac surgery (including transcatheter aortic valve implantation procedures), frailty has been strongly associated with postoperative mortality and morbidity, and thus frailty likely improves the identification of high-risk patients beyond known risk scores. For perioperative physicians then, the question arises of how to incorporate this information into perioperative care. To date, 2 thrusts of research and clinical practice have emerged: (1) preoperative identification of high-risk patients to guide both patient expectations and surgical decision-making; and (2) perioperative optimization strategies for frail patients. However, despite the strong association of frailty and poor outcomes, there is a lack of well-designed trials that have examined perioperative interventions with a specific focus on frail patients undergoing cardiac surgery. Thus, in many cases, principles of geriatric care may need to be applied. Further research is needed to standardize and implement the feasible definitions of frailty and examine perioperative interventions for frail patients undergoing cardiac surgery.
Collapse
Affiliation(s)
- Antonio Graham
- From the *Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland; and †Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | | |
Collapse
|
565
|
Song X, Narzt MS, Nagelreiter IM, Hohensinner P, Terlecki-Zaniewicz L, Tschachler E, Grillari J, Gruber F. Autophagy deficient keratinocytes display increased DNA damage, senescence and aberrant lipid composition after oxidative stress in vitro and in vivo. Redox Biol 2017; 11:219-230. [PMID: 28012437 PMCID: PMC5192251 DOI: 10.1016/j.redox.2016.12.015] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 12/15/2016] [Accepted: 12/16/2016] [Indexed: 01/05/2023] Open
Abstract
Autophagy allows cells fundamental adaptations to metabolic needs and to stress. Using autophagic bulk degradation cells can clear crosslinked macromolecules and damaged organelles that arise under redox stress. Accumulation of such debris results in cellular dysfunction and is observed in aged tissue and senescent cells. Conversely, promising anti-aging strategies aim at inhibiting the mTOR pathway and thereby activating autophagy, to counteract aging associated damage. We have inactivated autophagy related 7 (Atg7), an essential autophagy gene, in murine keratinocytes (KC) and have found in an earlier study that this resulted in increased baseline oxidative stress and reduced capacity to degrade crosslinked proteins after oxidative ultraviolet stress. To investigate whether autophagy deficiency would promote cellular aging, we studied how Atg7 deficient (KO) and Atg7 bearing cells (WT) would respond to stress induced by paraquat (PQ), an oxidant drug commonly used to induce cellular senescence. Atg7 deficient KC displayed increased prostanoid signaling and a pro- mitotic gene expression signature as compared to the WT. After exposure to PQ, both WT and KO cells showed an inflammatory and stress-related transcriptomic response. However, the Atg7 deficient cells additionally showed drastic DNA damage- and cell cycle arrest signaling. Indeed, DNA fragmentation and -oxidation were strongly increased in the stressed Atg7 deficient cells upon PQ stress but also after oxidizing ultraviolet A irradiation. Damage associated phosphorylated histone H2AX (γH2AX) foci were increased in the nuclei, whereas expression of the nuclear lamina protein lamin B1 was strongly decreased. Similarly, in both, PQ treated mouse tail skin explants and in UVA irradiated mouse tail skin, we found a strong increase in γH2AX positive nuclei within the basal layer of Atg7 deficient epidermis. Atg7 deficiency significantly affected expression of lipid metabolic genes. Therefore we performed lipid profiling of keratinocytes which demonstrated a major dysregulation of cellular lipid metabolism. We found accumulation of autophagy agonisitic free fatty acids, whereas triglyceride levels were strongly decreased. Together, our data show that in absence of Atg7/autophagy the resistance of keratinocytes to intrinsic and environmental oxidative stress was severely impaired and resulted in DNA damage, cell cycle arrest and a disturbed lipid phenotype, all typical for premature cell aging.
Collapse
Affiliation(s)
- Xiuzu Song
- Department of Dermatology, Medical University of Vienna, Währinger Gürtel 18-20, Leitstelle 7J, A-1090 Vienna, Austria; Department of Dermatology, The Third Hospital of Hangzhou, 38 Xihu Road, Hangzhou, Zhejiang, 310009, PR China
| | - Marie Sophie Narzt
- Department of Dermatology, Medical University of Vienna, Währinger Gürtel 18-20, Leitstelle 7J, A-1090 Vienna, Austria
| | - Ionela Mariana Nagelreiter
- Department of Dermatology, Medical University of Vienna, Währinger Gürtel 18-20, Leitstelle 7J, A-1090 Vienna, Austria
| | - Philipp Hohensinner
- Department of Internal Medicine II - Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria
| | - Lucia Terlecki-Zaniewicz
- Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, Muthgasse 18, 1190 Vienna, Austria; Christian Doppler Laboratory for Biotechnology of Skin Aging, Austria
| | - Erwin Tschachler
- Department of Dermatology, Medical University of Vienna, Währinger Gürtel 18-20, Leitstelle 7J, A-1090 Vienna, Austria
| | - Johannes Grillari
- Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, Muthgasse 18, 1190 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Muthgasse 18, 1190 Vienna, Austria; Christian Doppler Laboratory for Biotechnology of Skin Aging, Austria
| | - Florian Gruber
- Department of Dermatology, Medical University of Vienna, Währinger Gürtel 18-20, Leitstelle 7J, A-1090 Vienna, Austria; Christian Doppler Laboratory for Biotechnology of Skin Aging, Austria.
| |
Collapse
|
566
|
Molecular connections of obesity and aging: a focus on adipose protein 53 and retinoblastoma protein. Biogerontology 2017; 18:321-332. [PMID: 28357524 DOI: 10.1007/s10522-017-9698-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 03/27/2017] [Indexed: 12/13/2022]
Abstract
Obesity is an induced health problem that human beings have been facing with non-optimal treatment so far. Humans are on average getting fatter with age, and obesity and aging interact each other to shorten lifetime and decrease life quality. Obesity also causes several aging related-disorders such as cancer, strokes, cardiovascular disease, high blood pressure and type 2 diabetes. So, the molecular connections between aging and obesity are promising targets for bio-medical researches and innovative therapies of many health problems. In this review, we discuss the findings of adipose p53 and Rb-two central molecular linkages between aging and obesity-on lipid metabolism and obesity.
Collapse
|
567
|
O'Hara SP, Splinter PL, Trussoni CE, Pisarello MJL, Loarca L, Splinter NS, Schutte BF, LaRusso NF. ETS Proto-oncogene 1 Transcriptionally Up-regulates the Cholangiocyte Senescence-associated Protein Cyclin-dependent Kinase Inhibitor 2A. J Biol Chem 2017; 292:4833-4846. [PMID: 28184004 PMCID: PMC5377799 DOI: 10.1074/jbc.m117.777409] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 02/06/2017] [Indexed: 12/13/2022] Open
Abstract
Primary sclerosing cholangitis (PSC) is a chronic, fibroinflammatory cholangiopathy (disease of the bile ducts) of unknown pathogenesis. We reported that cholangiocyte senescence features prominently in PSC and that neuroblastoma RAS viral oncogene homolog (NRAS) is activated in PSC cholangiocytes. Additionally, persistent microbial insult (e.g. LPSs) induces cyclin-dependent kinase inhibitor 2A (CDKN2A/p16INK4a) expression and senescence in cultured cholangiocytes in an NRAS-dependent manner. However, the molecular mechanisms involved in LPS-induced cholangiocyte senescence and NRAS-dependent regulation of CDKN2A remain unclear. Using our in vitro senescence model, we found that LPS-induced CDKN2A expression coincided with a 4.5-fold increase in ETS1 (ETS proto-oncogene 1) mRNA, suggesting that ETS1 is involved in regulating CDKN2A This idea was confirmed by RNAi-mediated suppression or genetic deletion of ETS1, which blocked CDKN2A expression and reduced cholangiocyte senescence. Furthermore, site-directed mutagenesis of a predicted ETS-binding site within the CDKN2A promoter abolished luciferase reporter activity. Pharmacological inhibition of RAS/MAPK reduced ETS1 and CDKN2A protein expression and CDKN2A promoter-driven luciferase activity by ∼50%. In contrast, constitutively active NRAS expression induced ETS1 and CDKN2A protein expression, whereas ETS1 RNAi blocked this increase. Chromatin immunoprecipitation-PCR detected increased ETS1 and histone 3 lysine 4 trimethylation (H3K4Me3) at the CDKN2A promoter following LPS-induced senescence. Additionally, phospho-ETS1 expression was increased in cholangiocytes of human PSC livers and in the Abcb4 (Mdr2)-/- mouse model of PSC. These data pinpoint ETS1 and H3K4Me3 as key transcriptional regulators in NRAS-induced expression of CDKN2A, and this regulatory axis may therefore represent a potential therapeutic target for PSC treatment.
Collapse
Affiliation(s)
- Steven P O'Hara
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Patrick L Splinter
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Christy E Trussoni
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Maria J Lorenzo Pisarello
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Lorena Loarca
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Noah S Splinter
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Bryce F Schutte
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| | - Nicholas F LaRusso
- From the Division of Gastroenterology and Hepatology, and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Minnesota 55905
| |
Collapse
|
568
|
Elrayess MA, Almuraikhy S, Kafienah W, Al-Menhali A, Al-Khelaifi F, Bashah M, Zarkovic K, Zarkovic N, Waeg G, Alsayrafi M, Jaganjac M. 4-hydroxynonenal causes impairment of human subcutaneous adipogenesis and induction of adipocyte insulin resistance. Free Radic Biol Med 2017; 104:129-137. [PMID: 28088621 DOI: 10.1016/j.freeradbiomed.2017.01.015] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/26/2016] [Accepted: 01/09/2017] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Increased adipose production of 4-hydroxynonenal (4-HNE), a bioreactive aldehyde, directly correlates with obesity and insulin resistance. The aim of this study was to elucidate the impact of 4-HNE in mediating adipocyte differentiation and function in two metabolically distinct obese groups; the insulin sensitive (IS) and the insulin resistant (IR). METHODS Subcutaneous (SC) adipose tissues were obtained from eighteen clinically well characterized obese premenopausal women undergoing weight reduction surgery. Cellular distribution of 4-HNE in the form of protein adducts was determined by immunohistochemistry in addition to its effect on oxidative stress, cell growth, adipogenic capacity and insulin signaling in preadipocytes derived from the IS and IR participants. RESULTS 4-HNE was detected in the SC adipose tissue in different cell types with the highest level detected in adipocytes and blood vessels. Short and long-term in vitro treatment of SC preadipocytes with 4-HNE caused inhibition of their growth and increased production of reactive oxygen species (ROS) and antioxidant enzymes. Repeated 4-HNE treatment led to a greater reduction in the adipogenic capacity of preadipocytes from IS subjects compared to IR and caused dephosphorylation of IRS-1 and p70S6K while activating GSK3α/β and BAD, triggering an IR phenotype. CONCLUSION These data suggest that 4-HNE-induced oxidative stress plays a role in the regulation of preadipocyte growth, differentiation and insulin signaling and may therefore contribute to adipose tissue metabolic dysfunction associated with insulin resistance.
Collapse
Affiliation(s)
| | - Shamma Almuraikhy
- Anti Doping Lab Qatar, Sports City, Doha, Qatar; School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Wael Kafienah
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | | | | | - Moataz Bashah
- Bariatric and Metabolic Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Kamelija Zarkovic
- Department of Pathology, Medical Faculty University of Zagreb, Clinical Hospital Centre Zagreb, Zagreb, Croatia
| | - Neven Zarkovic
- Laboratory for Oxidative Stress, Department of Molecular Medicine, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Georg Waeg
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | | | | |
Collapse
|
569
|
DNA methylation map in circulating leukocytes mirrors subcutaneous adipose tissue methylation pattern: a genome-wide analysis from non-obese and obese patients. Sci Rep 2017; 7:41903. [PMID: 28211912 PMCID: PMC5314866 DOI: 10.1038/srep41903] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 12/09/2016] [Indexed: 02/07/2023] Open
Abstract
The characterization of the epigenetic changes within the obesity-related adipose tissue will provide new insights to understand this metabolic disorder, but adipose tissue is not easy to sample in population-based studies. We aimed to evaluate the capacity of circulating leukocytes to reflect the adipose tissue-specific DNA methylation status of obesity susceptibility. DNA samples isolated from subcutaneous adipose tissue and circulating leukocytes were hybridized in the Infinium HumanMethylation 450 BeadChip. Data were compared between samples from obese (n = 45) and non-obese (n = 8–10) patients by Wilcoxon-rank test, unadjusted for cell type distributions. A global hypomethylation of the differentially methylated CpG sites (DMCpGs) was observed in the obese subcutaneous adipose tissue and leukocytes. The overlap analysis yielded a number of genes mapped by the common DMCpGs that were identified to reflect the obesity state in the leukocytes. Specifically, the methylation levels of FGFRL1, NCAPH2, PNKD and SMAD3 exhibited excellent and statistically significant efficiencies in the discrimination of obesity from non-obesity status (AUC > 0.80; p < 0.05) and a great correlation between both tissues. Therefore, the current study provided new and valuable DNA methylation biomarkers of obesity-related adipose tissue pathogenesis through peripheral blood analysis, an easily accessible and minimally invasive biological material instead of adipose tissue.
Collapse
|
570
|
Correa-de-Araujo R, Harris-Love MO, Miljkovic I, Fragala MS, Anthony BW, Manini TM. The Need for Standardized Assessment of Muscle Quality in Skeletal Muscle Function Deficit and Other Aging-Related Muscle Dysfunctions: A Symposium Report. Front Physiol 2017; 8:87. [PMID: 28261109 PMCID: PMC5310167 DOI: 10.3389/fphys.2017.00087] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/31/2017] [Indexed: 12/12/2022] Open
Abstract
A growing body of scientific literature suggests that not only changes in skeletal muscle mass, but also other factors underpinning muscle quality, play a role in the decline in skeletal muscle function and impaired mobility associated with aging. A symposium on muscle quality and the need for standardized assessment was held on April 28, 2016 at the International Conference on Frailty and Sarcopenia Research in Philadelphia, Pennsylvania. The purpose of this symposium was to provide a venue for basic science and clinical researchers and expert clinicians to discuss muscle quality in the context of skeletal muscle function deficit and other aging-related muscle dysfunctions. The present article provides an expanded introduction concerning the emerging definitions of muscle quality and a potential framework for scientific inquiry within the field. Changes in muscle tissue composition, based on excessive levels of inter- and intra-muscular adipose tissue and intramyocellular lipids, have been found to adversely impact metabolism and peak force generation. However, methods to easily and rapidly assess muscle tissue composition in multiple clinical settings and with minimal patient burden are needed. Diagnostic ultrasound and other assessment methods continue to be developed for characterizing muscle pathology, and enhanced sonography using sensors to provide user feedback and improve reliability is currently the subject of ongoing investigation and development. In addition, measures of relative muscle force such as specific force or grip strength adjusted for body size have been proposed as methods to assess changes in muscle quality. Furthermore, performance-based assessments of muscle power via timed tests of function and body size estimates, are associated with lower extremity muscle strength may be responsive to age-related changes in muscle quality. Future aims include reaching consensus on the definition and standardized assessments of muscle quality, and providing recommendations to address critical clinical and technology research gaps within the field.
Collapse
Affiliation(s)
- Rosaly Correa-de-Araujo
- Division of Geriatrics and Clinical Gerontology, National Institute on Aging, National Institutes of Health, U.S. Department of Health and Human Services Bethesda, MD, USA
| | - Michael O Harris-Love
- Muscle Morphology, Mechanics and Performance Laboratory, Clinical Research Center - Human Performance Research Unit, Veterans Affairs Medical CenterWashington, DC, USA; Geriatrics and Extended Care Service/Research Service, Veterans Affairs Medical CenterWashington, DC, USA; Department of Exercise and Nutritional Sciences, Milken Institute School of Public Health, The George Washington UniversityWashington, DC, USA
| | - Iva Miljkovic
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh Pittsburgh, PA, USA
| | | | - Brian W Anthony
- Laboratory for Manufacturing and Productivity, Massachusetts Institute of TechnologyCambridge, MA, USA; Medical Electronic Device Realization Center, Massachusetts Institute of TechnologyCambridge, MA, USA
| | - Todd M Manini
- Department of Aging & Geriatric Research, Institute on Aging, University of Florida College of Medicine Gainesville, FL, USA
| |
Collapse
|
571
|
Growing Older and Staying Positive: Associations Between Diverse Aging Women’s Perceptions of Age and Body Satisfaction. JOURNAL OF ADULT DEVELOPMENT 2017. [DOI: 10.1007/s10804-016-9256-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
572
|
Frasca D, Diaz A, Romero M, Vazquez T, Blomberg BB. Obesity induces pro-inflammatory B cells and impairs B cell function in old mice. Mech Ageing Dev 2017; 162:91-99. [PMID: 28111127 DOI: 10.1016/j.mad.2017.01.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 01/03/2017] [Accepted: 01/07/2017] [Indexed: 12/14/2022]
Abstract
We analyzed B cells infiltrating the Visceral Adipose Tissue (VAT) of young and old mice to identify contributors to the phenotypic and functional changes observed in aged B cells. We found higher percentages of pro-inflammatory (age-associated B cells, ABC) and less Follicular (FO) B cells in VAT as compared to spleen. VAT B cells express higher pro-adipogenic and inflammatory markers, including NF-kB, as compared to splenic B cells, and also secrete IgG2c antibodies, some of which have been shown to be autoimmune in other studies. Moreover, we found that in the presence of an adipocyte-conditioned medium FO B cells differentiated into ABC. Additional results showed production of pro-inflammatory chemokines by the adipocytes, suggesting a mechanism for B cell attraction by the VAT. These results are the first to show a direct effect of adipocytes on the generation of pro-inflammatory B cells.
Collapse
Affiliation(s)
- Daniela Frasca
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33101, USA.
| | - Alain Diaz
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33101, USA
| | - Maria Romero
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33101, USA
| | - Thomas Vazquez
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33101, USA
| | - Bonnie B Blomberg
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33101, USA
| |
Collapse
|
573
|
Soto-Gamez A, Demaria M. Therapeutic interventions for aging: the case of cellular senescence. Drug Discov Today 2017; 22:786-795. [PMID: 28111332 DOI: 10.1016/j.drudis.2017.01.004] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/30/2016] [Accepted: 01/10/2017] [Indexed: 12/19/2022]
Abstract
Organismal aging is a multifactorial process characterized by the onset of degenerative conditions and cancer. One of the key drivers of aging is cellular senescence, a state of irreversible growth arrest induced by many pro-tumorigenic stresses. Senescent cells accumulate late in life and at sites of age-related pathologies, where they contribute to disease onset and progression through complex cell and non-cell autonomous effects. Here, we summarize the mechanisms by which cellular senescence can promote aging, and we offer an extensive description of current potential pharmacological interventions for senescent cells, highlighting limitations and suggesting alternatives.
Collapse
Affiliation(s)
- Abel Soto-Gamez
- University of Groningen, European Institute for the Biology of Aging (ERIBA), University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Marco Demaria
- University of Groningen, European Institute for the Biology of Aging (ERIBA), University Medical Center Groningen (UMCG), Groningen, The Netherlands.
| |
Collapse
|
574
|
Letra L, Santana I. The Influence of Adipose Tissue on Brain Development, Cognition, and Risk of Neurodegenerative Disorders. ADVANCES IN NEUROBIOLOGY 2017; 19:151-161. [PMID: 28933064 DOI: 10.1007/978-3-319-63260-5_6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The brain is a highly metabolic organ and thus especially vulnerable to changes in peripheral metabolism, including those induced by obesity-associated adipose tissue dysfunction. In this context, it is likely that the development and maturation of neurocognitive circuits may also be affected and modulated by metabolic environmental factors, beginning in utero. It is currently recognized that maternal obesity, either pre-gestational or gestational, negatively influences fetal brain development and elevates the risk of cognitive impairment and neuropsychiatric disorders in the offspring. During infancy and adolescence, obesity remains a limiting factor for healthy neurodevelopment, especially affecting executive functions but also attention, visuospatial ability, and motor skills. In middle age, obesity seems to induce an accelerated brain aging and thus may increase the risk of age-related neurodegenerative diseases such as Alzheimer's disease. In this chapter we review and discuss experimental and clinical evidence focusing on the influence of adipose tissue dysfunction on neurodevelopment and cognition across lifespan, as well as some possible mechanistic links, namely the role of the most well studied adipokines.
Collapse
Affiliation(s)
- Liliana Letra
- Institute of Physiology, Institute for Biomedical Imaging and Life Sciences-IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal. .,Neurology Department, Centro Hospitalar do Baixo Vouga, Aveiro, Portugal.
| | - Isabel Santana
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
575
|
Cellular Mechanisms Driving Sex Differences in Adipose Tissue Biology and Body Shape in Humans and Mouse Models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1043:29-51. [PMID: 29224089 DOI: 10.1007/978-3-319-70178-3_3] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Sex differences in adipose tissue distribution and the metabolic, endocrine, and immune functions of different anatomical fat depots have been described, but they are incompletely documented in the literature. It is becoming increasingly clear that adipose depots serve distinct functions in males and females and have specific physiological roles. However, the mechanisms that regulate the size and function of specific adipose tissues in men and women remain poorly understood. New insights from mouse models have advanced our understanding of depot differences in adipose growth and remodeling via the proliferation and differentiation of adipose progenitors that can expand adipocyte number in the tissue or simply replace dysfunctional older and larger adipocytes. A limited ability of a depot to expand or remodel can lead to excessive adipocyte hypertrophy, which is often correlated with metabolic dysfunction. However, the relationship of adipocyte size and function varies by depot and sex. For example, femoral adipose tissues of premenopausal women appear to have a greater capacity for adipose expansion via hyperplasia and hypertrophy; although larger, these gluteal-femoral adipocytes remain insulin sensitive. The microenvironment of specific depots, including the composition of the extracellular matrix and cellular composition, as well as cell-autonomous genetic differences, influences sex- and depot-dependent metabolic and growth properties. Although there are some species differences, studies of the molecular and physiological determinants of sex differences in adipocyte growth and function in humans and rodents are both needed for understanding sex differences in health and disease.
Collapse
|
576
|
SNEV hPrp19/hPso4 Regulates Adipogenesis of Human Adipose Stromal Cells. Stem Cell Reports 2016; 8:21-29. [PMID: 28041875 PMCID: PMC5233435 DOI: 10.1016/j.stemcr.2016.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 12/01/2016] [Accepted: 12/01/2016] [Indexed: 11/26/2022] Open
Abstract
Aging is accompanied by loss of subcutaneous adipose tissue. This may be due to reduced differentiation capacity or deficiency in DNA damage repair (DDR) factors. Here we investigated the role of SNEVhPrp19/hPso4, which was implicated in DDR and senescence evasion, in adipogenic differentiation of human adipose stromal cells (hASCs). We showed that SNEV is induced during adipogenesis and localized both in the nucleus and in the cytoplasm. Knockdown of SNEV perturbed adipogenic differentiation and led to accumulation of DNA damage in hASCs upon oxidative stress. In addition, we demonstrated that SNEV is required for fat deposition in Caenorhabditis elegans. Consequently, we tested other DDR factors and found that WRN is also required for adipogenesis in both models. These results demonstrate that SNEV regulates adipogenesis in hASCs and indicate that DDR capacity in general might be a pre-requisite for this process. SNEV is required for adipogenic differentiation of human adipose stromal cells SNEV modulates pro- and anti-adipogenic signaling pathways SNEV regulates DNA repair capacity of human adipose stromal cells SNEV modulates organismal fat deposition in Caenorhabditis elegans
Collapse
|
577
|
Kocher C, Christiansen M, Martin S, Adams C, Wehner P, Gress T, Santanam N. Sexual dimorphism in obesity-related genes in the epicardial fat during aging. J Physiol Biochem 2016; 73:215-224. [PMID: 28012156 DOI: 10.1007/s13105-016-0542-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 12/06/2016] [Indexed: 01/07/2023]
Abstract
Aging increases the risk of cardiovascular disease and metabolic syndrome. Alterations in epicardial fat play an important pathophysiological role in coronary artery disease and hypertension. We investigated the impact of normal aging on obesity-related genes in epicardial fat. Sex-specific changes in obesity-related genes with aging in epicardial fat (EF) were determined in young (6 months) and old (30/36 months) female and male, Fischer 344 × Brown Norway hybrid (FBN) rats, using a rat obesity RT2 PCR Array. Circulating sex hormone levels, body and heart weights were determined. Statistical significance was determined using two-tailed Student's t test and Pearson's correlation. Our results revealed sex-specific differences in obesity-related genes with aging. Dramatic changes in the expression profile of obesity-related genes in EF with aging in female, but not in male, FBN rats were observed. The older (30 months) female rats had more significant variations in the abundance of obesity-related genes in the EF compared to that seen in younger female rats or both age groups in male rats. A correlation of changes in obesity-related genes in EF to heart weights was observed in female rats, but not in male rats with aging. No correlation was observed to circulating sex hormone levels. Our findings indicate a dysfunctional EF in female rats with aging compared to male rats. These findings, with further functional validation, might help explain the sex differences in cardiovascular risk and mortality associated with aging observed in humans.
Collapse
Affiliation(s)
- Caitlin Kocher
- Department of Pharmacology, Physiology, & Toxicology, Joan C Edwards School of Medicine, Marshall University, One John Marshall Dr, Huntington, WV, 25755, USA
| | - Matthew Christiansen
- Department of Pharmacology, Physiology, & Toxicology, Joan C Edwards School of Medicine, Marshall University, One John Marshall Dr, Huntington, WV, 25755, USA
| | - Sarah Martin
- Department of Pharmacology, Physiology, & Toxicology, Joan C Edwards School of Medicine, Marshall University, One John Marshall Dr, Huntington, WV, 25755, USA
| | - Christopher Adams
- Department of Medicine and Cardiology, Joan C Edwards School of Medicine, Marshall University, One John Marshall Dr, Huntington, WV, 25755, USA
| | - Paulette Wehner
- Department of Medicine and Cardiology, Joan C Edwards School of Medicine, Marshall University, One John Marshall Dr, Huntington, WV, 25755, USA
| | - Todd Gress
- Department of Medicine and Cardiology, Joan C Edwards School of Medicine, Marshall University, One John Marshall Dr, Huntington, WV, 25755, USA
| | - Nalini Santanam
- Department of Pharmacology, Physiology, & Toxicology, Joan C Edwards School of Medicine, Marshall University, One John Marshall Dr, Huntington, WV, 25755, USA.
- Department of Medicine and Cardiology, Joan C Edwards School of Medicine, Marshall University, One John Marshall Dr, Huntington, WV, 25755, USA.
| |
Collapse
|
578
|
Chauvet V, Jouaville S, Garbez N, Martins I. [Removal of senescent cells: towards a better future?]. Med Sci (Paris) 2016; 32:1030-1033. [PMID: 28008848 DOI: 10.1051/medsci/20163211023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Affiliation(s)
| | | | - Nicolas Garbez
- M1 Biologie Santé, Université Paris-Saclay, 91405 Orsay, France
| | - Isabelle Martins
- Mort cellulaire et sénescence, Inserm U1030, Institut Gustave Roussy, 114, rue Edouard Vaillant, 94800 Villejuif, France
| |
Collapse
|
579
|
Identification of Potential Key lncRNAs and Genes Associated with Aging Based on Microarray Data of Adipocytes from Mice. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9181702. [PMID: 28097151 PMCID: PMC5209599 DOI: 10.1155/2016/9181702] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 10/19/2016] [Accepted: 11/01/2016] [Indexed: 11/18/2022]
Abstract
Objective. This study aimed to screen potential crucial lncRNAs and genes involved in aging. Methods. The data of 9 peripheral white adipocytes, respectively, taken from male C57BL/6J mice (6 months, 14 months, and 18 months of age) in GSE25905 were used in this study. Differentially time series expressed lncRNA genes (DE-lncRNAs) and mRNA genes (DEGs) were identified. After cluster analysis of lncRNAs expression pattern, target genes of DE-lncRNAs were predicted from the DEGs, and functional analysis for target genes was conducted. Results. A total of 8301 time series-related DEGs and 43 time series-related DE-lncRNAs were identified. Among them, 41 DE-lncRNAs targeted 1880 DEGs. The DEGs positively regulated by DE-lncRNAs were mainly related to the development of blood vessel and the pathways of cholesterol biosynthesis and elastic fibre formation. Furthermore, the DEGs negatively regulated by DE-lncRNAs were correlated with protein metabolism. Conclusion. These DE-lncRNAs and DEGs are potentially involved in the process of aging.
Collapse
|
580
|
Martyniak K, Masternak MM. Changes in adipose tissue cellular composition during obesity and aging as a cause of metabolic dysregulation. Exp Gerontol 2016; 94:59-63. [PMID: 27939445 DOI: 10.1016/j.exger.2016.12.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 12/02/2016] [Accepted: 12/07/2016] [Indexed: 12/17/2022]
Abstract
Adipose tissue represents complex endocrine organ containing several different cellular populations including adipocytes, pre-adipocytes, mesenchymal stem cells, macrophages and lymphocytes. It is well establishing that these populations are not static but alter during obesity and aging. Changes in cellular populations alter inflammatory status and other common metabolic complications arise, therefore adipose tissue cellular composition helps dictate its endocrine and regulatory function. During excessive weight gain in obese individuals and as we age there is shift towards increase populations of inflammatory macrophages with a decrease of regulatory T cell. This altered cellular composition promote chronic low grade inflammation negatively affecting mesenchymal stem cell progenitor self-renewal, which result in deterioration of adipogenesis and increased cellular stress in adipocytes. All these changes promote metabolic disorders including age- or obese-related insulin resistance leading to type 2 diabetes.
Collapse
Affiliation(s)
- Kari Martyniak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32827, United States
| | - Michal M Masternak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32827, United States; Department of Head and Neck Surgery, The Greater Poland Cancer Centre, Poznan, Poland.
| |
Collapse
|
581
|
Transient remission of nonalcoholic fatty liver disease decreases the risk of incident type 2 diabetes mellitus in Japanese men. Eur J Gastroenterol Hepatol 2016; 28:1443-1449. [PMID: 27603300 DOI: 10.1097/meg.0000000000000736] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION It is unclear how the transient remission of nonalcoholic fatty liver disease (NAFLD) affects incident type 2 diabetes mellitus (T2DM). Here, we sought to determine the effect of the transient remission of NAFLD on incident T2DM in Japanese men. MATERIALS AND METHODS We used a population-based health check-up program. The primary outcome was set as incident T2DM. We divided the participants who showed NAFLD at the time of enrollment into three groups according to their clinical course of NAFLD: the Regression group, in which the participants showed a regression of NAFLD and no relapse during the follow-up period; the Transient Remission group, in which participants achieved a transient remission of NAFLD, but had a relapse of NAFLD; and the Persistent group, in which participants showed NAFLD throughout the follow-up. The Never group of participants who did not show NAFLD throughout the follow-up served as a reference. RESULTS The incidence rates of T2DM in the Never group, the Regression group, the Transient Remission group, and the Persistent group were 4.7% (62/1306), 9.2% (14/153), 18.0% (25/139), and 35.1% (120/342), respectively. In a multivariate Cox regression analysis with covariates, the adjusted hazard ratios for incident T2DM compared with the Never group were as follows: Regression group: 1.08 [95% confidence interval (CI) 0.53-2.04, P=0.81], Transient Remission group: 2.12 (95% CI 1.22-3.57, P<0.01), and Persistent group: 3.44 (95% CI 2.29-5.21, P<0.001). The adjusted hazard ratio of the Transient Remission group was significantly lower than that of the Persistent group (P<0.05). CONCLUSION Transient remission of NAFLD significantly decreased the risk of developing T2DM.
Collapse
|
582
|
Palikaras K, Mari M, Petanidou B, Pasparaki A, Filippidis G, Tavernarakis N. Ectopic fat deposition contributes to age-associated pathology in Caenorhabditis elegans. J Lipid Res 2016; 58:72-80. [PMID: 27884963 DOI: 10.1194/jlr.m069385] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 11/06/2016] [Indexed: 01/08/2023] Open
Abstract
Age-dependent collapse of lipid homeostasis results in spillover of lipids and excessive fat deposition in nonadipose tissues. Ectopic fat contributes to lipotoxicity and has been implicated in the development of a metabolic syndrome that increases risk of age-associated diseases. However, the molecular mechanisms coupling ectopic fat accumulation with aging remain obscure. Here, we use nonlinear imaging modalities to visualize and quantify age-dependent ectopic lipid accumulation in Caenorhabditis elegans We find that aging is accompanied by pronounced deposition of lipids in nonadipose tissues, including the nervous system. Importantly, interventions that promote longevity such as low insulin signaling, germ-line loss, and dietary restriction, which effectively delay aging in evolutionary divergent organisms, diminish the rate of ectopic fat accumulation and the size of lipid droplets. Suppression of lipotoxic accumulation of fat in heterologous tissues is dependent on helix-loop-helix (HLH)-30/transcription factor EB (TFEB) and autophagy. Our findings in their totality highlight the pivotal role of HLH-30/TFEB and autophagic processes in the maintenance of lipid homeostasis during aging, in addition to establishing nonlinear imaging as a powerful tool for monitoring ectopic lipid droplet deposition in vivo.
Collapse
Affiliation(s)
- Konstantinos Palikaras
- Institute of Molecular Biology and Biotechnology Foundation for Research and Technology, Heraklion 71110, Crete, Greece
| | - Meropi Mari
- Institute of Molecular Biology and Biotechnology Foundation for Research and Technology, Heraklion 71110, Crete, Greece
| | - Barbara Petanidou
- Institute of Electronic Structure and Laser, Foundation for Research and Technology, Heraklion 71110, Crete, Greece.,Physics Department University of Crete, Heraklion 71003, Crete, Greece
| | - Angela Pasparaki
- Institute of Molecular Biology and Biotechnology Foundation for Research and Technology, Heraklion 71110, Crete, Greece
| | - George Filippidis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology, Heraklion 71110, Crete, Greece
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology Foundation for Research and Technology, Heraklion 71110, Crete, Greece .,Medical School, University of Crete, Heraklion 71003, Crete, Greece
| |
Collapse
|
583
|
Luo T, Liu H, Feng W, Liu D, Du J, Sun J, Wang W, Han X, Guo J, Amizuka N, Li X, Li M. Adipocytes enhance expression of osteoclast adhesion-related molecules through the CXCL12/CXCR4 signalling pathway. Cell Prolif 2016; 50. [PMID: 27868262 DOI: 10.1111/cpr.12317] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 10/18/2016] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES The purpose of this study was to investigate effects of adipocytes on osteoclast adhesion-related molecules. MATERIALS AND METHODS ST2 cells, a cloned stromal cell line from mouse bone marrow, able to differentiate into adipocytes, were cultured in serum-free α-MEM which was then collected to be used as adipocyte-conditioned medium (ADIPO CM). RAW264.7 cells were cultured in ADIPO CM in the presence of RANKL, and bone marrow-derived macrophages were cultured in ADIPO CM in the presence of RANKL and macrophage-colony stimulating factor to induce osteoclast differentiation. TRAP staining, resorption pit assay, qRT-PCR and western blotting assays were performed. RESULTS ELISAs revealed that CXCL12 was abundant in ADIPO CM and CCK-8 assay revealed no proliferation of RAW264.7 cells after exogenous CXCL12 treatment. ADIPO CM enhanced osteoclast formation and resorption, both by RAW264.7 cells and BMMs. In addition, exogenous CXCL12 efficiently potentiated formation of TRAP-positive osteoclast and resorption by RAW264.7 cells. Western blotting and qRT-PCR suggested that ADIPO CM or combined treatment with exogenous CXCL12 caused significant increase in expression of NFAT2, src and osteoclast adhesion-related molecules, including β3 integrin, CD44 and osteopontin. However, these promotional effects were largely abrogated on treatment of AMD3100, a CXCR4 antagonist. CONCLUSIONS Adipocytes promoted osteoclast differentiation, function and expression of adhesion-related molecules through the CXCL12/CXCR4 signalling pathway.
Collapse
Affiliation(s)
- Tingting Luo
- School of Stomatology, Shanxi Medical University, Taiyuan, China.,Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Bone Metabolism, School of Stomatology Shandong University, Jinan, China
| | - Hongrui Liu
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Bone Metabolism, School of Stomatology Shandong University, Jinan, China
| | - Wei Feng
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Bone Metabolism, School of Stomatology Shandong University, Jinan, China
| | - Di Liu
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Bone Metabolism, School of Stomatology Shandong University, Jinan, China
| | - Juan Du
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Bone Metabolism, School of Stomatology Shandong University, Jinan, China
| | - Jing Sun
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Bone Metabolism, School of Stomatology Shandong University, Jinan, China
| | - Wei Wang
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Bone Metabolism, School of Stomatology Shandong University, Jinan, China
| | - Xiuchun Han
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Bone Metabolism, School of Stomatology Shandong University, Jinan, China
| | - Jie Guo
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Bone Metabolism, School of Stomatology Shandong University, Jinan, China
| | - Norio Amizuka
- Department of Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Xianqi Li
- School of Stomatology, Shanxi Medical University, Taiyuan, China.,Department of Hard Tissue Research, Graduate School of Oral Medicine, Matsumoto Dental University, Shiojiri, Japan
| | - Minqi Li
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Bone Metabolism, School of Stomatology Shandong University, Jinan, China
| |
Collapse
|
584
|
Aldiss P, Davies G, Woods R, Budge H, Sacks HS, Symonds ME. 'Browning' the cardiac and peri-vascular adipose tissues to modulate cardiovascular risk. Int J Cardiol 2016; 228:265-274. [PMID: 27865196 PMCID: PMC5236060 DOI: 10.1016/j.ijcard.2016.11.074] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 11/05/2016] [Indexed: 01/02/2023]
Abstract
Excess visceral adiposity, in particular that located adjacent to the heart and coronary arteries is associated with increased cardiovascular risk. In the pathophysiological state, dysfunctional adipose tissue secretes an array of factors modulating vascular function and driving atherogenesis. Conversely, brown and beige adipose tissues utilise glucose and lipids to generate heat and are associated with improved cardiometabolic health. The cardiac and thoracic perivascular adipose tissues are now understood to be composed of brown adipose tissue in the healthy state and undergo a brown-to-white transition i.e. during obesity which may be a driving factor of cardiovascular disease. In this review we discuss the risks of excess cardiac and vascular adiposity and potential mechanisms by which restoring the brown phenotype i.e. “re-browning” could potentially be achieved in clinically relevant populations. Epicardial, paracardial and thoracic perivascular adipose tissues resemble BAT at birth. Despite ‘whitening’ in early life these depots remain metabolically active and potentially thermogenic into adulthood. Obesity induces further ‘whitening’ and inflammation in these depots likely driving the atherogenesis. Maintaining or inducing the brown phenotype in these depots could prevent atherosclerotic disease.
Collapse
Affiliation(s)
- Peter Aldiss
- The Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, School of Medicine, University Hospital, University of Nottingham, Nottingham, UK, NG7 2UH
| | - Graeme Davies
- The Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, School of Medicine, University Hospital, University of Nottingham, Nottingham, UK, NG7 2UH
| | - Rachel Woods
- The Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, School of Medicine, University Hospital, University of Nottingham, Nottingham, UK, NG7 2UH
| | - Helen Budge
- The Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, School of Medicine, University Hospital, University of Nottingham, Nottingham, UK, NG7 2UH
| | - Harold S Sacks
- VA Greater Los Angeles Healthcare System, Endocrinology and Diabetes Division, and Department of Medicine David Geffen School of Medicine, Los Angeles, CA 90073, USA
| | - Michael E Symonds
- The Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, School of Medicine, University Hospital, University of Nottingham, Nottingham, UK, NG7 2UH.
| |
Collapse
|
585
|
Leite JSM, Cruzat VF, Krause M, Homem de Bittencourt PI. Physiological regulation of the heat shock response by glutamine: implications for chronic low-grade inflammatory diseases in age-related conditions. ACTA ACUST UNITED AC 2016. [DOI: 10.1186/s41110-016-0021-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
586
|
Chen JR, Lazarenko OP, Blackburn ML, Rose S, Frye RE, Badger TM, Andres A, Shankar K. Maternal Obesity Programs Senescence Signaling and Glucose Metabolism in Osteo-Progenitors From Rat and Human. Endocrinology 2016; 157:4172-4183. [PMID: 27653035 DOI: 10.1210/en.2016-1408] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Nutritional status during intrauterine and early postnatal life impacts the risk of chronic diseases, presumably via epigenetic mechanisms. However, evidence on the impact of gestational events on regulation of embryonic bone cell fate is sparse. We investigated the effects of maternal obesity on fetal osteoblast development in both rodents and humans. Female rats were fed control or an obesogenic high-fat diet (HFD) for 12 weeks and mated with male rats fed control diets, and respective maternal diets were continued during pregnancy. Embryonic rat osteogenic calvarial cells (EOCCs) were taken from gestational day 18.5 fetuses from control and HFD dams. EOCCs from HFD obese dams showed increases in p53/p21-mediated cell senescence signaling but decreased glucose metabolism. Decreased aerobic glycolysis in HFD-EOCCs was associated with decreased osteoblastic cell differentiation and proliferation. Umbilical cord human mesenchymal stem cells (MSCs) from 24 pregnant women (12 obese and 12 lean) along with placentas were collected upon delivery. The umbilical cord MSCs of obese mothers displayed less potential toward osteoblastogenesis and more towards adipogenesis. Human MSCs and placenta from obese mothers also exhibited increased cell senescence signaling, whereas MSCs showed decreased glucose metabolism and insulin resistance. Finally, we showed that overexpression of p53 linked increased cell senescence signaling and decreased glucose metabolism in fetal osteo-progenitors from obese rats and humans. These findings suggest programming of fetal preosteoblastic cell senescence signaling and glucose metabolism by maternal obesity.
Collapse
Affiliation(s)
- Jin-Ran Chen
- Arkansas Children's Nutrition Center (J.-R.C., O.P.L., M.L.B., T.M.B., A.A., K.S.), Department of Pediatrics (J.-R.C., O.P.L., M.L.B., R.E.F., T.M.B., A.A., K.S.), University of Arkansas for Medical Sciences, and Arkansas Children's Hospital Research Institute (S.R., R.E.F.), Little Rock, Arkansas 72202
| | - Oxana P Lazarenko
- Arkansas Children's Nutrition Center (J.-R.C., O.P.L., M.L.B., T.M.B., A.A., K.S.), Department of Pediatrics (J.-R.C., O.P.L., M.L.B., R.E.F., T.M.B., A.A., K.S.), University of Arkansas for Medical Sciences, and Arkansas Children's Hospital Research Institute (S.R., R.E.F.), Little Rock, Arkansas 72202
| | - Michael L Blackburn
- Arkansas Children's Nutrition Center (J.-R.C., O.P.L., M.L.B., T.M.B., A.A., K.S.), Department of Pediatrics (J.-R.C., O.P.L., M.L.B., R.E.F., T.M.B., A.A., K.S.), University of Arkansas for Medical Sciences, and Arkansas Children's Hospital Research Institute (S.R., R.E.F.), Little Rock, Arkansas 72202
| | - Shannon Rose
- Arkansas Children's Nutrition Center (J.-R.C., O.P.L., M.L.B., T.M.B., A.A., K.S.), Department of Pediatrics (J.-R.C., O.P.L., M.L.B., R.E.F., T.M.B., A.A., K.S.), University of Arkansas for Medical Sciences, and Arkansas Children's Hospital Research Institute (S.R., R.E.F.), Little Rock, Arkansas 72202
| | - Richard E Frye
- Arkansas Children's Nutrition Center (J.-R.C., O.P.L., M.L.B., T.M.B., A.A., K.S.), Department of Pediatrics (J.-R.C., O.P.L., M.L.B., R.E.F., T.M.B., A.A., K.S.), University of Arkansas for Medical Sciences, and Arkansas Children's Hospital Research Institute (S.R., R.E.F.), Little Rock, Arkansas 72202
| | - Thomas M Badger
- Arkansas Children's Nutrition Center (J.-R.C., O.P.L., M.L.B., T.M.B., A.A., K.S.), Department of Pediatrics (J.-R.C., O.P.L., M.L.B., R.E.F., T.M.B., A.A., K.S.), University of Arkansas for Medical Sciences, and Arkansas Children's Hospital Research Institute (S.R., R.E.F.), Little Rock, Arkansas 72202
| | - Aline Andres
- Arkansas Children's Nutrition Center (J.-R.C., O.P.L., M.L.B., T.M.B., A.A., K.S.), Department of Pediatrics (J.-R.C., O.P.L., M.L.B., R.E.F., T.M.B., A.A., K.S.), University of Arkansas for Medical Sciences, and Arkansas Children's Hospital Research Institute (S.R., R.E.F.), Little Rock, Arkansas 72202
| | - Kartik Shankar
- Arkansas Children's Nutrition Center (J.-R.C., O.P.L., M.L.B., T.M.B., A.A., K.S.), Department of Pediatrics (J.-R.C., O.P.L., M.L.B., R.E.F., T.M.B., A.A., K.S.), University of Arkansas for Medical Sciences, and Arkansas Children's Hospital Research Institute (S.R., R.E.F.), Little Rock, Arkansas 72202
| |
Collapse
|
587
|
Almuraikhy S, Kafienah W, Bashah M, Diboun I, Jaganjac M, Al-Khelaifi F, Abdesselem H, Mazloum NA, Alsayrafi M, Mohamed-Ali V, Elrayess MA. Interleukin-6 induces impairment in human subcutaneous adipogenesis in obesity-associated insulin resistance. Diabetologia 2016; 59:2406-2416. [PMID: 27342408 PMCID: PMC5506102 DOI: 10.1007/s00125-016-4031-3] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 06/02/2016] [Indexed: 12/22/2022]
Abstract
AIMS/HYPOTHESIS A subset of obese individuals remains insulin sensitive by mechanisms as yet unclear. The hypothesis that maintenance of normal subcutaneous (SC) adipogenesis accounts, at least partially, for this protective phenotype and whether it can be abrogated by chronic exposure to IL-6 was investigated. METHODS Adipose tissue biopsies were collected from insulin-sensitive (IS) and insulin-resistant (IR) individuals undergoing weight-reduction surgery. Adipocyte size, pre-adipocyte proportion of stromal vascular fraction (SVF)-derived cells, adipogenic capacity and gene expression profiles of isolated pre-adipocytes were determined, along with local in vitro IL-6 secretion. Adipogenic capacity was further assessed in response to exogenous IL-6 application. RESULTS Despite being equally obese, IR individuals had significantly lower plasma leptin and adiponectin levels and higher IL-6 levels compared with age-matched IS counterparts. Elevated systemic IL-6 in IR individuals was associated with hyperplasia of adipose tissue-derived SVF cells, despite higher frequency of hypertrophied adipocytes. SC pre-adipocytes from these tissues exhibited lower adipogenic capacity accompanied by downregulation of PPARγ (also known as PPARG) and CEBPα (also known as CEBPA) and upregulation of GATA3 expression. Impaired adipogenesis in IR individuals was further associated with increased adipose secretion of IL-6. Treatment of IS-derived SC pre-adipocytes with IL-6 reduced their adipogenic capacity to levels of the IR group. CONCLUSIONS/INTERPRETATION Obesity-associated insulin resistance is marked by impaired SC adipogenesis, mediated, at least in a subset of individuals, by elevated local levels of IL-6. Understanding the molecular mechanisms underlying reduced adipogenic capacity in IR individuals could help target appropriate therapeutic strategies aimed at those at greatest risk of insulin resistance and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Shamma Almuraikhy
- Anti-Doping Lab Qatar, Sports City Road, P.O. Box 27775, Doha, Qatar
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Wael Kafienah
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Moataz Bashah
- Bariatric and Metabolic Surgery, Hamad Medical Corporation, Doha, Qatar
| | - Ilhame Diboun
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Doha, Qatar
| | - Morana Jaganjac
- Anti-Doping Lab Qatar, Sports City Road, P.O. Box 27775, Doha, Qatar
| | | | - Houari Abdesselem
- Microbiology and Immunology, Weill Cornell Medicine Qatar, Doha, Qatar
| | - Nayef A Mazloum
- Microbiology and Immunology, Weill Cornell Medicine Qatar, Doha, Qatar
| | | | - Vidya Mohamed-Ali
- Anti-Doping Lab Qatar, Sports City Road, P.O. Box 27775, Doha, Qatar
| | | |
Collapse
|
588
|
Ioannidou A, Goulielmaki E, Garinis GA. DNA Damage: From Chronic Inflammation to Age-Related Deterioration. Front Genet 2016; 7:187. [PMID: 27826317 PMCID: PMC5078321 DOI: 10.3389/fgene.2016.00187] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/07/2016] [Indexed: 12/15/2022] Open
Abstract
To lessen the "wear and tear" of existence, cells have evolved mechanisms that continuously sense DNA lesions, repair DNA damage and restore the compromised genome back to its native form. Besides genome maintenance pathways, multicellular organisms may also employ adaptive and innate immune mechanisms to guard themselves against bacteria or viruses. Recent evidence points to reciprocal interactions between DNA repair, DNA damage responses and aspects of immunity; both self-maintenance and defense responses share a battery of common players and signaling pathways aimed at safeguarding our bodily functions over time. In the short-term, this functional interplay would allow injured cells to restore damaged DNA templates or communicate their compromised state to the microenvironment. In the long-term, however, it may result in the (premature) onset of age-related degeneration, including cancer. Here, we discuss the beneficial and unrewarding outcomes of DNA damage-driven inflammation in the context of tissue-specific pathology and disease progression.
Collapse
Affiliation(s)
- Anna Ioannidou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-HellasHeraklion, Greece; Department of Biology, University of CreteHeraklion, Greece
| | - Evi Goulielmaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas Heraklion, Greece
| | - George A Garinis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-HellasHeraklion, Greece; Department of Biology, University of CreteHeraklion, Greece
| |
Collapse
|
589
|
Pazienza V, Panebianco C, Rappa F, Memoli D, Borghesan M, Cannito S, Oji A, Mazza G, Tamburrino D, Fusai G, Barone R, Bolasco G, Villarroya F, Villarroya J, Hatsuzawa K, Cappello F, Tarallo R, Nakanishi T, Vinciguerra M. Histone macroH2A1.2 promotes metabolic health and leanness by inhibiting adipogenesis. Epigenetics Chromatin 2016; 9:45. [PMID: 27800025 PMCID: PMC5078890 DOI: 10.1186/s13072-016-0098-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 10/17/2016] [Indexed: 12/13/2022] Open
Abstract
Background Obesity has tremendous impact on the health systems. Its epigenetic bases are unclear. MacroH2A1 is a variant of histone H2A, present in two alternatively exon-spliced isoforms macroH2A1.1 and macroH2A1.2, regulating cell plasticity and proliferation, during pluripotency and tumorigenesis. Their role in adipose tissue plasticity is unknown. Results Here, we show evidence that macroH2A1.1 protein levels in the visceral adipose tissue of obese humans positively correlate with BMI, while macroH2A1.2 is nearly absent. We thus introduced a constitutive GFP-tagged transgene for macroH2A1.2 in mice, and we characterized their metabolic health upon being fed a standard chow diet or a high fat diet. Despite unchanged food intake, these mice exhibit lower adipose mass and improved glucose metabolism both under a chow and an obesogenic diet. In the latter regimen, transgenic mice display smaller pancreatic islets and significantly less inflammation. MacroH2A1.2 overexpression in the mouse adipose tissue induced dramatic changes in the transcript levels of key adipogenic genes; genomic analyses comparing pre-adipocytes to mature adipocytes uncovered only minor changes in macroH2A1.2 genomic distribution upon adipogenic differentiation and suggested differential cooperation with transcription factors. MacroH2A1.2 overexpression markedly inhibited adipogenesis, while overexpression of macroH2A1.1 had opposite effects. Conclusions MacroH2A1.2 is an unprecedented chromatin component powerfully promoting metabolic health by modulating anti-adipogenic transcriptional networks in the differentiating adipose tissue. Strategies aiming at enhancing macroH2A1.2 expression might counteract excessive adiposity in humans. Electronic supplementary material The online version of this article (doi:10.1186/s13072-016-0098-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Valerio Pazienza
- Gastroenterology Unit, IRCCS "Casa Sollievo della Sofferenza" Hospital, 71013 San Giovanni Rotondo, Italy
| | - Concetta Panebianco
- Gastroenterology Unit, IRCCS "Casa Sollievo della Sofferenza" Hospital, 71013 San Giovanni Rotondo, Italy
| | - Francesca Rappa
- Department of Experimental Biomedicine and Clinical Neurosciences, Section of Human Anatomy, University of Palermo, 90127 Palermo, Italy ; Department of Legal, Society and Sport Sciences, University of Palermo, 90133 Palermo, Italy ; Euro-Mediterranean Institute of Science and Technology (IEMEST), 90146 Palermo, Italy
| | - Domenico Memoli
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry 'Schola Medica Salernitana', University of Salerno, 84081 Baronissi, SA Italy
| | - Michela Borghesan
- Gastroenterology Unit, IRCCS "Casa Sollievo della Sofferenza" Hospital, 71013 San Giovanni Rotondo, Italy ; Institute for Liver and Digestive Health, University College London (UCL), Royal Free Hospital, London, NW3 2PF UK
| | - Sara Cannito
- Gastroenterology Unit, IRCCS "Casa Sollievo della Sofferenza" Hospital, 71013 San Giovanni Rotondo, Italy
| | - Asami Oji
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 5650871 Japan
| | - Giuseppe Mazza
- Institute for Liver and Digestive Health, University College London (UCL), Royal Free Hospital, London, NW3 2PF UK
| | - Domenico Tamburrino
- Centre for HPB Surgery and Liver Transplantation, Royal Free Hospital, London, NW3 2QG UK
| | - Giuseppe Fusai
- Centre for HPB Surgery and Liver Transplantation, Royal Free Hospital, London, NW3 2QG UK
| | - Rosario Barone
- Department of Experimental Biomedicine and Clinical Neurosciences, Section of Human Anatomy, University of Palermo, 90127 Palermo, Italy ; Euro-Mediterranean Institute of Science and Technology (IEMEST), 90146 Palermo, Italy
| | - Giulia Bolasco
- Mouse Biology Unit, European Molecular Biology Laboratory (EMBL), 00015 Monterotondo, Italy
| | - Francesc Villarroya
- Departament de Bioquimica i Biologia Molecular, Institut de Biomedicina de la Universitat de Barcelona (IBUB), and CIBER Fisiopatologia de la Obesidad y Nutricion, University of Barcelona, Barcelona, 08007 Spain ; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBEROBN) ISCIII, Madrid, Spain
| | - Joan Villarroya
- Departament de Bioquimica i Biologia Molecular, Institut de Biomedicina de la Universitat de Barcelona (IBUB), and CIBER Fisiopatologia de la Obesidad y Nutricion, University of Barcelona, Barcelona, 08007 Spain ; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBEROBN) ISCIII, Madrid, Spain
| | | | - Francesco Cappello
- Department of Experimental Biomedicine and Clinical Neurosciences, Section of Human Anatomy, University of Palermo, 90127 Palermo, Italy ; Euro-Mediterranean Institute of Science and Technology (IEMEST), 90146 Palermo, Italy
| | - Roberta Tarallo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry 'Schola Medica Salernitana', University of Salerno, 84081 Baronissi, SA Italy
| | - Tomoko Nakanishi
- Faculty of Medicine, Tottori University, Yonago, 683-8503 Japan ; The Institute of Medical Sciences, University of Tokyo, Tokyo, 108-8639 Japan
| | - Manlio Vinciguerra
- Gastroenterology Unit, IRCCS "Casa Sollievo della Sofferenza" Hospital, 71013 San Giovanni Rotondo, Italy ; Euro-Mediterranean Institute of Science and Technology (IEMEST), 90146 Palermo, Italy ; Institute for Liver and Digestive Health, University College London (UCL), Royal Free Hospital, London, NW3 2PF UK ; Center for Translational Medicine (CTM), International Clinical Research Center (ICRC), St. Anne's University Hospital, Brno, 656 91 Czech Republic
| |
Collapse
|
590
|
Rivera-Gonzalez GC, Shook BA, Andrae J, Holtrup B, Bollag K, Betsholtz C, Rodeheffer MS, Horsley V. Skin Adipocyte Stem Cell Self-Renewal Is Regulated by a PDGFA/AKT-Signaling Axis. Cell Stem Cell 2016; 19:738-751. [PMID: 27746098 DOI: 10.1016/j.stem.2016.09.002] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 07/14/2016] [Accepted: 09/11/2016] [Indexed: 12/15/2022]
Abstract
Tissue growth and maintenance requires stem cell populations that self-renew, proliferate, and differentiate. Maintenance of white adipose tissue (WAT) requires the proliferation and differentiation of adipocyte stem cells (ASCs) to form postmitotic, lipid-filled mature adipocytes. Here we use the dynamic adipogenic program that occurs during hair growth to uncover an unrecognized regulator of ASC self-renewal and proliferation, PDGFA, which activates AKT signaling to drive and maintain the adipogenic program in the skin. Pdgfa expression is reduced in aged ASCs and is required for ASC proliferation and maintenance in the dermis, but not in other WATs. Our molecular and genetic studies uncover PI3K/AKT2 as a direct PDGFA target that is activated in ASCs during WAT hyperplasia and is functionally required for dermal ASC proliferation. Our data therefore reveal active mechanisms that regulate ASC self-renewal in the skin and show that distinct regulatory mechanisms operate in different WAT depots.
Collapse
Affiliation(s)
| | - Brett A Shook
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Johanna Andrae
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Brandon Holtrup
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Katherine Bollag
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Matthew S Rodeheffer
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA; Section of Comparative Medicine, Yale University, New Haven, CT 06520, USA
| | - Valerie Horsley
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA; Department of Dermatology, Yale School of Medicine, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
591
|
Shook B, Rivera Gonzalez G, Ebmeier S, Grisotti G, Zwick R, Horsley V. The Role of Adipocytes in Tissue Regeneration and Stem Cell Niches. Annu Rev Cell Dev Biol 2016; 32:609-631. [PMID: 27146311 PMCID: PMC5157158 DOI: 10.1146/annurev-cellbio-111315-125426] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Classically, white adipose tissue (WAT) was considered an inert component of connective tissue but is now appreciated as a major regulator of metabolic physiology and endocrine homeostasis. Recent work defining how WAT develops and expands in vivo emphasizes the importance of specific locations of WAT or depots in metabolic regulation. Interestingly, mature white adipocytes are integrated into several tissues. A new perspective regarding the in vivo regulation and function of WAT in these tissues has highlighted an essential role of adipocytes in tissue homeostasis and regeneration. Finally, there has been significant progress in understanding how mature adipocytes regulate the pathology of several diseases. In this review, we discuss these novel roles of WAT in the homeostasis and regeneration of epithelial, muscle, and immune tissues and how they contribute to the pathology of several disorders.
Collapse
Affiliation(s)
- Brett Shook
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06520;
| | - Guillermo Rivera Gonzalez
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06520;
| | - Sarah Ebmeier
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06520;
| | | | - Rachel Zwick
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06520;
| | - Valerie Horsley
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06520;
- Department of Dermatology, Yale University, New Haven, Connecticut 06520
| |
Collapse
|
592
|
Pinti M, Appay V, Campisi J, Frasca D, Fülöp T, Sauce D, Larbi A, Weinberger B, Cossarizza A. Aging of the immune system: Focus on inflammation and vaccination. Eur J Immunol 2016; 46:2286-2301. [PMID: 27595500 PMCID: PMC5156481 DOI: 10.1002/eji.201546178] [Citation(s) in RCA: 318] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 08/20/2016] [Accepted: 08/30/2016] [Indexed: 12/21/2022]
Abstract
Major advances in preventing, delaying, or curing individual pathologies are responsible for an increasingly long life span in the developed parts of our planet, and indeed reaching eight to nine decades of life is nowadays extremely frequent. However, medical and sanitary advances have not prevented or delayed the underlying cause of the disparate pathologies occurring in the elderly: aging itself. The identification of the basis of the aging processes that drives the multiple pathologies and loss of function typical of older individuals is a major challenge in current aging research. Among the possible causes, an impairment of the immune system plays a major role, and indeed numerous studies have described immunological changes which occur with age. Far from the intention of being exhaustive, this review will focus on recent advances and views on the role that modifications of cell signalling and remodelling of the immune response play during human aging and longevity, paying particular attention to phenomena which are linked to the so called inflammaging process, such as dysregulation of innate immunity, altered T-cell or B-cell maturation and differentiation, as well as to the implications of immune aging for vaccination strategies in the elderly.
Collapse
Affiliation(s)
- Marcello Pinti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Victor Appay
- Sorbonne Universités, UPMC Univ. Paris 06, DHU FAST, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Judith Campisi
- USA and Lawrence Berkeley National Laboratory, Buck Institute for Research on Aging, Berkeley, CA, USA
| | - Daniela Frasca
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Tamas Fülöp
- Division of Geriatrics, Department of Medicine, Research Center on Aging, University of Sherbrooke, Canada
| | - Delphine Sauce
- Sorbonne Universités, UPMC Univ. Paris 06, DHU FAST, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Aging and Immunity Program, A*STAR, Singapore
| | - Birgit Weinberger
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Andrea Cossarizza
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia School of Medicine, Modena, Italy.
| |
Collapse
|
593
|
Abstract
Maintaining an active and engaged social life is a critical component of aging well, and women are generally more socially active than men. However, as women age their self-perceptions of their bodies may reduce social behaviors and consequently, increase depressive symptoms. Because little is known about how body image is associated with social engagement and depressive symptoms among aging women, four aspects of body image: satisfaction with cosmetic features, body function, physical appearance, and weight were assessed among women aged 65 and older (n = 123). Regression analyses indicated that cosmetic appearance, body function, and physical appearance were associated with depressive symptoms, whereas satisfaction with weight was unrelated. Further, both greater satisfaction with cosmetic features and body function were associated with higher levels of social engagement, and social engagement mediated the association between these aspects of body satisfaction and depressive symptoms. The findings indicate that specific age-relevant aspects of body satisfaction are linked to social behavior and depression among aging women, and reduced body satisfaction may lead to lower social engagement, and consequently aging women's health and well-being may be diminished.
Collapse
Affiliation(s)
- Natalie J Sabik
- a Health Studies Program , University of Rhode Island , Kingston , Rhode Island , USA
| |
Collapse
|
594
|
Lettieri Barbato D, Tatulli G, Aquilano K, Ciriolo MR. Mitochondrial Hormesis links nutrient restriction to improved metabolism in fat cell. Aging (Albany NY) 2016; 7:869-81. [PMID: 26540513 PMCID: PMC4637211 DOI: 10.18632/aging.100832] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Fasting promotes longevity by reprogramming metabolic and stress resistance pathways. However, although the impact on adipose tissue physiology through hormonal inputs is well established, the direct role of fasting on adipose cells is poorly understood. Herein we show that white and beige adipocytes, as well as mouse epididymal and subcutaneous adipose depots, respond to nutrient scarcity by acquiring a brown-like phenotype. Indeed, they improve oxidative metabolism through modulating the expression of mitochondrial-and nuclear-encoded oxidative phosphorylation genes as well as mitochondrial stress defensive proteins (UCP1, SOD2). Such adaptation is placed in a canonical mitohormetic response that proceeds via mitochondrial reactive oxygen species (mtROS) production and redistribution of FoxO1 transcription factor into nucleus. Nuclear FoxO1 (nFoxO1) mediates retrograde communication by inducing the expression of mitochondrial oxidative and stress defensive genes. Collectively, our findings describe an unusual white/beige fat cell response to nutrient availability highlighting another health-promoting mechanism of fasting.
Collapse
Affiliation(s)
| | | | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy.,IRCCS San Raffaele Roma, 00163 Rome, Italy
| | - Maria R Ciriolo
- Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| |
Collapse
|
595
|
Levine JA, Kaihara KA, Layden BT, Wicksteed B. Long-term activation of PKA in β-cells provides sustained improvement to glucose control, insulin sensitivity and body weight. Islets 2016; 8:125-34. [PMID: 27340937 PMCID: PMC5029204 DOI: 10.1080/19382014.2016.1198457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Type 2 diabetes is associated with obesity, insulin resistance and β-cell failure. Therapeutic aims are to reduce adiposity, improve insulin sensitivity and enhance β-cell function. However, it has been proposed that chronically increasing insulin release leads to β-cell exhaustion and failure. We previously developed mice to have increased activity of the cAMP-dependent protein kinase (PKA), specifically in β-cells (β-caPKA mice). β-caPKA mice have enhanced acute phase insulin release, which is the primary determinant of the efficacy of glucose clearance. Here these mice were used to determine the sustainability of enhanced insulin secretion, and to characterize peripheral effects of enhanced β-cell function. Increased PKA activity was induced by tamoxifen administration at 10 weeks of age. Male mice were aged to 12 months of age and female mice to 16 months. Glucose control in both male and female β-caPKA mice was significantly improved relative to littermate controls with ad libitum feeding, upon refeeding after fasting, and in glucose tolerance tests. In female mice insulin release was both greater and more rapid than in controls. Female mice were more insulin sensitive than controls. Male and female β-caPKA mice had lower body weights than controls. DEXA analysis of male mice revealed that this was due to reduced adiposity and not due to changes in lean body mass. This study indicates that targeting β-cells to enhance insulin release is sustainable, maintains insulin sensitivity and reduces body weight. These data identify β-cell PKA activity as a novel target for obesity therapies.
Collapse
Affiliation(s)
- Joshua A. Levine
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Kelly A. Kaihara
- Committee for Molecular Metabolism and Nutrition, The University of Chicago, Chicago, IL, USA
- Bio-Rad Laboratories, Hercules, CA, USA
| | - Brian T. Layden
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| | - Barton Wicksteed
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Committee for Molecular Metabolism and Nutrition, The University of Chicago, Chicago, IL, USA
- CONTACT Barton Wicksteed Division of Endocrinology, Metabolism and Molecular Medicine, Tarry Building 15-735 300 East Superior St., Chicago, IL 60611-3008, USA
| |
Collapse
|
596
|
Stout MB, Swindell WR, Zhi X, Rohde K, List EO, Berryman DE, Kopchick JJ, Gesing A, Fang Y, Masternak MM. Transcriptome profiling reveals divergent expression shifts in brown and white adipose tissue from long-lived GHRKO mice. Oncotarget 2016; 6:26702-15. [PMID: 26436954 PMCID: PMC4694946 DOI: 10.18632/oncotarget.5760] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 08/29/2015] [Indexed: 12/24/2022] Open
Abstract
Mice lacking the growth hormone receptor (GHRKO) exhibit improved lifespan and healthspan due to loss of growth hormone signaling. Both the distribution and activity of brown and white adipose tissue (BAT and WAT) are altered in GHRKO mice, but the contribution of each tissue to age-related phenotypes has remained unclear. We therefore used whole-genome microarrays to evaluate transcriptional differences in BAT and WAT depots between GHRKO and normal littermates at six months of age. Our findings reveal a unique BAT transcriptome as well as distinctive responses of BAT to Ghr ablation. BAT from GHRKO mice exhibited elevated expression of genes associated with mitochondria and metabolism, along with reduced expression of genes expressed by monocyte-derived cells (dendritic cells [DC] and macrophages). Largely the opposite was observed in WAT, with increased expression of DC-expressed genes and reduced expression of genes associated with metabolism, cellular respiration and the mitochondrial inner envelope. These findings demonstrate divergent response patterns of BAT and WAT to loss of GH signaling in GHRKO mice. These patterns suggest both BAT and WAT contribute in different ways to phenotypes in GHRKO mice, with Ghr ablation blunting inflammation in BAT as well as cellular metabolism and mitochondrial biogenesis in WAT.
Collapse
Affiliation(s)
- Michael B Stout
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | | | - Xu Zhi
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - Kyle Rohde
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - Edward O List
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Darlene E Berryman
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - John J Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Adam Gesing
- Department of Oncological Endocrinology, Medical University of Lodz, Lodz, Poland
| | - Yimin Fang
- Geriatrics Research Laboratory, Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Michal M Masternak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA.,Department of Head and Neck Surgery, The Greater Poland Cancer Centre, Poznan, Poland
| |
Collapse
|
597
|
Huffman DM, Justice JN, Stout MB, Kirkland JL, Barzilai N, Austad SN. Evaluating Health Span in Preclinical Models of Aging and Disease: Guidelines, Challenges, and Opportunities for Geroscience. J Gerontol A Biol Sci Med Sci 2016; 71:1395-1406. [PMID: 27535967 PMCID: PMC5055649 DOI: 10.1093/gerona/glw106] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/22/2016] [Indexed: 12/14/2022] Open
Abstract
Life extension is no longer considered sufficient evidence of delayed aging in research animals. It must also be demonstrated that a broad swathe of health indicators have been extended. During a retreat of the Geroscience Network, a consortium of basic and clinical aging researchers, potential measures of mouse health were considered for their potential as easily standardized, highly informative metrics. Major health domains considered were neuromuscular, cognitive, cardiovascular, metabolic, and inflammatory functions as well as body composition and energetics and a multitude of assays interrogating these domains. A particularly sensitive metric of health is the ability to respond to, and recover, from stress. Therefore, the Network also considered stresses of human relevance that could be implemented in mouse models to assess frailty and resilience. Mouse models already exist for responses to forced immobility, cancer chemotherapy, infectious diseases, dietary challenges, and surgical stress, and it was felt that these could be employed to determine whether putative senescence-retarding interventions increased and extended organismal robustness. The Network discussed challenges in modeling age-related human chronic diseases and concluded that more attention needs to be paid to developing disease models with later age of onset, models of co- and multimorbidity, diversifying the strains and sexes commonly used in aging research, and considering additional species.
Collapse
Affiliation(s)
- Derek M Huffman
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York
| | - Jamie N Justice
- Department of Integrative Physiology, University of Colorado Boulder
| | - Michael B Stout
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Nir Barzilai
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York
| | | |
Collapse
|
598
|
Xu X, Byles J, Shi Z, McElduff P, Hall J. Dietary pattern transitions, and the associations with BMI, waist circumference, weight and hypertension in a 7-year follow-up among the older Chinese population: a longitudinal study. BMC Public Health 2016; 16:743. [PMID: 27502827 PMCID: PMC4977626 DOI: 10.1186/s12889-016-3425-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 07/30/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Few studies explored the effects of nutritional changes on body mass index (BMI), weight (Wt), waist circumference (WC) and hypertension, especially for the older Chinese population. METHODS By using China Health and Nutrition Survey 2004-2011 waves, a total of 6348 observations aged ≥ 60 were involved in the study. The number of participants dropped from 2197 in 2004, to 1763 in 2006, 1303 in 2009, and 1085 in 2011. Dietary information was obtained from participants using 24 hour-recall over three consecutive days. Height, Wt, WC, systolic and diastolic blood pressure were also measured in each survey year. The dietary pattern was derived by exploratory factor analysis using principal component analysis methods. Linear Mixed Models were used to investigate associations of dietary patterns with BMI, Wt and WC. Generalized Estimating Equation models were used to assess the associations between dietary patterns and hypertension. RESULTS Over time, older people's diets were shifting towards a modern dietary pattern (high intake of dairy, fruit, cakes and fast food). Traditional and modern dietary patterns had distinct associations with BMI, Wt and WC. Participants with a diet in the highest quartile for traditional composition had a β (difference in mean) of -0.23 (95 % CI: -0.44; -0.02) for BMI decrease, β of -0.90 (95 % CI: -1.42; -0.37) for Wt decrease; and β of -1.57 (95 % CI: -2.32; -0.83) for WC decrease. However, participants with a diet in the highest quartile for modern diet had a β of 0.29 (95 % CI: 0.12; 0.47) for BMI increase; β of 1.02 (95 % CI: 0.58; 1.46) for Wt increase; and β of 1.44 (95 % CI: 0.78; 2.10) for Wt increase. No significant associations were found between dietary patterns and hypertension. CONCLUSIONS We elucidate the associations between dietary pattern and change in BMI, Wt, WC and hypertension in a 7-year follow-up study. The strong association between favourable body composition and traditional diet, compared with an increase in BMI, WC and Wt with modern diet suggests that there is an urgent need to develop age-specific dietary guideline for older Chinese people.
Collapse
Affiliation(s)
- Xiaoyue Xu
- Priority Research Centre for Gender, Health and Ageing, School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
- Centre for Clinical Epidemiology and Biostatistics, School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Julie Byles
- Priority Research Centre for Gender, Health and Ageing, School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Zumin Shi
- School of Medicine, University of Adelaide, Adelaide, Australia
| | - Patrick McElduff
- Centre for Clinical Epidemiology and Biostatistics, School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - John Hall
- Centre for Clinical Epidemiology and Biostatistics, School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| |
Collapse
|
599
|
Schulz TJ, Graja A, Huang TL, Xue R, An D, Poehle-Kronawitter S, Lynes MD, Tolkachov A, O'Sullivan LE, Hirshman MF, Schupp M, Goodyear LJ, Mishina Y, Tseng YH. Loss of BMP receptor type 1A in murine adipose tissue attenuates age-related onset of insulin resistance. Diabetologia 2016; 59:1769-77. [PMID: 27209464 PMCID: PMC4930470 DOI: 10.1007/s00125-016-3990-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 04/26/2016] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS Adipose tissue dysfunction is a prime risk factor for the development of metabolic disease. Bone morphogenetic proteins (BMPs) have previously been implicated in adipocyte formation. Here, we investigate the role of BMP signalling in adipose tissue health and systemic glucose homeostasis. METHODS We employed the Cre/loxP system to generate mouse models with conditional ablation of BMP receptor 1A in differentiating and mature adipocytes, as well as tissue-resident myeloid cells. Metabolic variables were assessed by glucose and insulin tolerance testing, insulin-stimulated glucose uptake and gene expression analysis. RESULTS Conditional deletion of Bmpr1a using the aP2 (also known as Fabp4)-Cre strain resulted in a complex phenotype. Knockout mice were clearly resistant to age-related impairment of insulin sensitivity during normal and high-fat-diet feeding and showed significantly improved insulin-stimulated glucose uptake in brown adipose tissue and skeletal muscle. Moreover, knockouts displayed significant reduction of variables of adipose tissue inflammation. Deletion of Bmpr1a in myeloid cells had no impact on insulin sensitivity, while ablation of Bmpr1a in mature adipocytes partially recapitulated the initial phenotype from aP2-Cre driven deletion. Co-cultivation of macrophages with pre-adipocytes lacking Bmpr1a markedly reduced expression of proinflammatory genes. CONCLUSIONS/INTERPRETATION Our findings show that altered BMP signalling in adipose tissue affects the tissue's metabolic properties and systemic insulin resistance by altering the pattern of immune cell infiltration. The phenotype is due to ablation of Bmpr1a specifically in pre-adipocytes and maturing adipocytes rather than an immune cell-autonomous effect. Mechanistically, we provide evidence for a BMP-mediated direct crosstalk between pre-adipocytes and macrophages.
Collapse
Affiliation(s)
- Tim J Schulz
- German Institute of Human Nutrition (DIfE), Department of Adipocyte Development and Nutrition, 114-116, Arthur-Scheunert Allee, 14558, Potsdam-Nuthetal, Germany.
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, One Joslin Place, Boston, MA, 02215, USA.
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.
| | - Antonia Graja
- German Institute of Human Nutrition (DIfE), Department of Adipocyte Development and Nutrition, 114-116, Arthur-Scheunert Allee, 14558, Potsdam-Nuthetal, Germany
| | - Tian Lian Huang
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, One Joslin Place, Boston, MA, 02215, USA
| | - Ruidan Xue
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, One Joslin Place, Boston, MA, 02215, USA
| | - Ding An
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, One Joslin Place, Boston, MA, 02215, USA
| | - Sophie Poehle-Kronawitter
- German Institute of Human Nutrition (DIfE), Department of Adipocyte Development and Nutrition, 114-116, Arthur-Scheunert Allee, 14558, Potsdam-Nuthetal, Germany
| | - Matthew D Lynes
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, One Joslin Place, Boston, MA, 02215, USA
| | - Alexander Tolkachov
- Charité University School of Medicine, Institute of Pharmacology, Center for Cardiovascular Research, Berlin, Germany
| | - Lindsay E O'Sullivan
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, One Joslin Place, Boston, MA, 02215, USA
| | - Michael F Hirshman
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, One Joslin Place, Boston, MA, 02215, USA
| | - Michael Schupp
- Charité University School of Medicine, Institute of Pharmacology, Center for Cardiovascular Research, Berlin, Germany
| | - Laurie J Goodyear
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, One Joslin Place, Boston, MA, 02215, USA
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, One Joslin Place, Boston, MA, 02215, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
600
|
Castro JP, Grune T, Speckmann B. The two faces of reactive oxygen species (ROS) in adipocyte function and dysfunction. Biol Chem 2016; 397:709-24. [DOI: 10.1515/hsz-2015-0305] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/08/2016] [Indexed: 12/11/2022]
Abstract
Abstract
White adipose tissue (WAT) is actively involved in the regulation of whole-body energy homeostasis via storage/release of lipids and adipokine secretion. Current research links WAT dysfunction to the development of metabolic syndrome (MetS) and type 2 diabetes (T2D). The expansion of WAT during oversupply of nutrients prevents ectopic fat accumulation and requires proper preadipocyte-to-adipocyte differentiation. An assumed link between excess levels of reactive oxygen species (ROS), WAT dysfunction and T2D has been discussed controversially. While oxidative stress conditions have conclusively been detected in WAT of T2D patients and related animal models, clinical trials with antioxidants failed to prevent T2D or to improve glucose homeostasis. Furthermore, animal studies yielded inconsistent results regarding the role of oxidative stress in the development of diabetes. Here, we discuss the contribution of ROS to the (patho)physiology of adipocyte function and differentiation, with particular emphasis on sources and nutritional modulators of adipocyte ROS and their functions in signaling mechanisms controlling adipogenesis and functions of mature fat cells. We propose a concept of ROS balance that is required for normal functioning of WAT. We explain how both excessive and diminished levels of ROS, e.g. resulting from over supplementation with antioxidants, contribute to WAT dysfunction and subsequently insulin resistance.
Collapse
|