601
|
Berry DC, Jiang Y, Graff JM. Emerging Roles of Adipose Progenitor Cells in Tissue Development, Homeostasis, Expansion and Thermogenesis. Trends Endocrinol Metab 2016; 27:574-585. [PMID: 27262681 PMCID: PMC10947416 DOI: 10.1016/j.tem.2016.05.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/26/2016] [Accepted: 05/02/2016] [Indexed: 01/10/2023]
Abstract
Stem or progenitor cells are an essential component for the development, homeostasis, expansion, and regeneration of many tissues. Within white adipose tissue (WAT) reside vascular-resident adipose progenitor cells (APCs) that can proliferate and differentiate into either white or beige/brite adipocytes, which may control adiposity. Recent studies have begun to show that APCs can be manipulated to control adiposity and counteract 'diabesity'. However, much remains unknown about the identity of APCs and how they may control adiposity in response to homeostatic and external cues. Here, we discuss recent advances in our understanding of adipose progenitors and cover a range of topics, including the stem cell/progenitor lineage, their niche, their developmental and adult roles, and their role in cold-induced beige/brite adipocyte formation.
Collapse
Affiliation(s)
- Daniel C Berry
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center 5323, Harry Hines Blvd, Dallas, TX 75235, USA
| | - Yuwei Jiang
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center 5323, Harry Hines Blvd, Dallas, TX 75235, USA
| | - Jonathan M Graff
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center 5323, Harry Hines Blvd, Dallas, TX 75235, USA; Department of Molecular Biology, University of Texas Southwestern Medical Center 5323, Harry Hines Blvd, Dallas, TX 75235, USA.
| |
Collapse
|
602
|
Metabolic Control of Longevity. Cell 2016; 166:802-821. [DOI: 10.1016/j.cell.2016.07.031] [Citation(s) in RCA: 524] [Impact Index Per Article: 58.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/15/2016] [Accepted: 07/20/2016] [Indexed: 12/19/2022]
|
603
|
Oishi Y, Manabe I. Macrophages in age-related chronic inflammatory diseases. NPJ Aging Mech Dis 2016; 2:16018. [PMID: 28721272 PMCID: PMC5515003 DOI: 10.1038/npjamd.2016.18] [Citation(s) in RCA: 194] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/21/2016] [Accepted: 05/25/2016] [Indexed: 12/12/2022] Open
Abstract
Chronic inflammation is the common pathological basis for such age-associated diseases as cardiovascular disease, diabetes, cancer and Alzheimer’s disease. A multitude of bodily changes occur with aging that contribute to the initiation and development of inflammation. In particular, the immune system of elderly individuals often exhibits diminished efficiency and fidelity, termed immunosenescence. But, although immune responses to new pathogens and vaccines are impaired, immunosenescence is also characterized by a basal systemic inflammatory state. This alteration in immune system function likely promotes chronic inflammation. Changes in the tissue microenvironment, such as the accumulation of cell debris, and systemic changes in metabolic and hormonal signals, also likely contribute to the development of chronic inflammation. Monocyte/macrophage lineage cells are crucial to these age-associated changes, which culminate in the development of chronic inflammatory diseases. In this review, we will summarize the diverse physiological and pathological roles of macrophages in the chronic inflammation underlying age-associated diseases.
Collapse
Affiliation(s)
- Yumiko Oishi
- Department of Cellular and Molecular Medicine, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ichiro Manabe
- Department of Aging Research, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
604
|
Zhang ZQ, Lu QG, Huang J, Jiao CY, Huang SM, Mao LM. Maternal and cord blood adiponectin levels in relation to post-natal body size in infants in the first year of life: a prospective study. BMC Pregnancy Childbirth 2016; 16:189. [PMID: 27459998 PMCID: PMC4962456 DOI: 10.1186/s12884-016-0978-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 07/18/2016] [Indexed: 11/18/2022] Open
Abstract
Background Adiponectin is an adipocyte hormone involved in energy homeostasis and metabolism. However, its role in early infancy is poorly understood. Methods We recruited a total of 443 pregnant women and their children in this prospective study. Cord blood samples were successfully obtained from 331 neonates. Maternal and umbilical blood serum adiponectin were measured. The weight-, height- and BMI-for-age Z scores of infants at birth and at 3, 6 and 12 months of age were assessed. Results Multiple linear regression analysis indicated that cord blood but not maternal serum adiponectin was positively associated with all of the anthropometric measures at birth (P < 0.01). Using Generalized Estimating Equation model after adjustment for sex, time, maternal age, gestational age, prepregnancy BMI, weight gain during pregnancy, maternal education, parity, history of miscarriage and mode of delivery, for every 1-μg/ml increment of maternal serum adiponectin, the height-for-age Z score during the first year of life increased by 0.026 (P =0.013) on average, and the height-for-age Z score of infants in the highest quartile of maternal serum adiponectin was 0.270 (95 % CI: 0.013–0.527) higher than those in the lowest quartile. The changes in weight-for-age Z score from birth decreased by 0.67 × 10−2 on average with every 1-μg/ml additional increase of cord blood adiponectin (P = 0.047). The infants in the highest quartile of cord blood adiponectin showed a −0.368 (95 % CI, −0.701–−0.035) decrease in weight-for-age Z score change from birth compared with those in the lowest quartile. Conclusions Cord blood adiponectin concentration is a determinant of infant birth size and weight gain in the first year of life. Circulating maternal adiponectin during pregnancy may predict postnatal height growth. Electronic supplementary material The online version of this article (doi:10.1186/s12884-016-0978-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhe-Qing Zhang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Qing-Gui Lu
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Jie Huang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Chang-Ya Jiao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Shao-Ming Huang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Li-Mei Mao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
605
|
Lettieri Barbato D, Aquilano K. Feast and famine: Adipose tissue adaptations for healthy aging. Ageing Res Rev 2016; 28:85-93. [PMID: 27223996 DOI: 10.1016/j.arr.2016.05.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/16/2016] [Accepted: 05/16/2016] [Indexed: 12/18/2022]
Abstract
Proper adipose tissue function controls energy balance with favourable effects on metabolic health and longevity. The molecular and metabolic asset of adipose tissue quickly and dynamically readapts in response to nutrient fluctuations. Once delivered into cells, nutrients are managed by mitochondria that represent a key bioenergetics node. A persistent nutrient overload generates mitochondrial exhaustion and uncontrolled reactive oxygen species ((mt)ROS) production. In adipocytes, metabolic/molecular reorganization is triggered culminating in the acquirement of a hypertrophic and hypersecretory phenotype that accelerates aging. Conversely, dietary regimens such as caloric restriction or time-controlled fasting endorse mitochondrial functionality and (mt)ROS-mediated signalling, thus promoting geroprotection. In this perspective view, we argued some important molecular and metabolic aspects related to adipocyte response to nutrient stress. Finally we delineated hypothetical routes by which molecularly and metabolically readapted adipose tissue promotes healthy aging.
Collapse
|
606
|
Kruglikov IL, Scherer PE. Skin aging: are adipocytes the next target? Aging (Albany NY) 2016; 8:1457-69. [PMID: 27434510 PMCID: PMC4993342 DOI: 10.18632/aging.100999] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 07/07/2016] [Indexed: 01/09/2023]
Abstract
Dermal white adipose tissue (dWAT) is increasingly appreciated as a special fat depot. The adipocytes in this depot exert a variety of unique effects on their surrounding cells and can undergo massive phenotypic changes. Significant modulation of dWAT content can be observed both in intrinsically and extrinsically aged skin. Specifically, skin that has been chronically photo-damaged displays a reduction of the dWAT volume, caused by the replacement of adipocytes by fibrotic structures. This is likely to be caused by the recently uncovered process described as "adipocyte-myofibroblast transition" (AMT). In addition, contributions of dermal adipocytes to the skin aging processes are also indirectly supported by spatial correlations between the prevalence of hypertrophic scarring and the appearance of signs of skin aging in different ethnic groups. These observations could elevate dermal adipocytes to prime targets in strategies aimed at counteracting skin aging.
Collapse
Affiliation(s)
| | - Philipp E. Scherer
- Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
607
|
Effects of Anthocyanin and Flavanol Compounds on Lipid Metabolism and Adipose Tissue Associated Systemic Inflammation in Diet-Induced Obesity. Mediators Inflamm 2016; 2016:2042107. [PMID: 27365896 PMCID: PMC4913062 DOI: 10.1155/2016/2042107] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/05/2016] [Indexed: 11/21/2022] Open
Abstract
Background. Naturally occurring substances from the flavanol and anthocyanin family of polyphenols have been proposed to exert beneficial effects in the course of obesity. We hypothesized that their effects on attenuating obesity-induced dyslipidemia as well as the associated inflammatory sequelae especially have health-promoting potential. Methods. Male C57BL/6J mice (n = 52) received a control low-fat diet (LFD; 10 kcal% fat) for 6 weeks followed by 24 weeks of either LFD (n = 13) or high-fat diet (HFD; 45 kcal% fat; n = 13) or HFD supplemented with 0.1% w/w of the flavanol compound epicatechin (HFD+E; n = 13) or an anthocyanin-rich bilberry extract (HFD+B; n = 13). Energy substrate utilization was determined by indirect calorimetry in a subset of mice following the dietary switch and at the end of the experiment. Blood samples were collected at baseline and at 3 days and 4, 12, and 20 weeks after dietary switch and analyzed for systemic lipids and proinflammatory cytokines. Adipose tissue (AT) histopathology and inflammatory gene expression as well as hepatic lipid content were analyzed after sacrifice. Results. The switch from a LFD to a HFD lowered the respiratory exchange ratio and increased plasma cholesterol and hepatic lipid content. These changes were not attenuated by HFD+E or HFD+B. Furthermore, the polyphenol compounds could not prevent HFD-induced systemic rise of TNF-α levels. Interestingly, a significant reduction in Tnf gene expression in HFD+B mice was observed in the AT. Furthermore, HFD+B, but not HFD+E, significantly prevented the early upregulation of circulating neutrophil chemoattractant mKC. However, no differences in AT histopathology were observed between the HFD types. Conclusion. Supplementation of HFD with an anthocyanin-rich bilberry extract but not with the flavanol epicatechin may exert beneficial effects on the systemic early inflammatory response associated with diet-induced obesity. These systemic effects were transient and not observed after prolongation of HFD-feeding (24 weeks). On the tissue level, long-term treatment with bilberry attenuated TNF-α expression in adipose tissue.
Collapse
|
608
|
Schafer MJ, White TA, Evans G, Tonne JM, Verzosa GC, Stout MB, Mazula DL, Palmer AK, Baker DJ, Jensen MD, Torbenson MS, Miller JD, Ikeda Y, Tchkonia T, van Deursen JM, Kirkland JL, LeBrasseur NK. Exercise Prevents Diet-Induced Cellular Senescence in Adipose Tissue. Diabetes 2016; 65:1606-15. [PMID: 26983960 PMCID: PMC4878429 DOI: 10.2337/db15-0291] [Citation(s) in RCA: 193] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 02/29/2016] [Indexed: 12/12/2022]
Abstract
Considerable evidence implicates cellular senescence in the biology of aging and chronic disease. Diet and exercise are determinants of healthy aging; however, the extent to which they affect the behavior and accretion of senescent cells within distinct tissues is not clear. Here we tested the hypothesis that exercise prevents premature senescent cell accumulation and systemic metabolic dysfunction induced by a fast-food diet (FFD). Using transgenic mice that express EGFP in response to activation of the senescence-associated p16(INK4a) promoter, we demonstrate that FFD consumption causes deleterious changes in body weight and composition as well as in measures of physical, cardiac, and metabolic health. The harmful effects of the FFD were associated with dramatic increases in several markers of senescence, including p16, EGFP, senescence-associated β-galactosidase, and the senescence-associated secretory phenotype (SASP) specifically in visceral adipose tissue. We show that exercise prevents the accumulation of senescent cells and the expression of the SASP while nullifying the damaging effects of the FFD on parameters of health. We also demonstrate that exercise initiated after long-term FFD feeding reduces senescent phenotype markers in visceral adipose tissue while attenuating physical impairments, suggesting that exercise may provide restorative benefit by mitigating accrued senescent burden. These findings highlight a novel mechanism by which exercise mediates its beneficial effects and reinforces the effect of modifiable lifestyle choices on health span.
Collapse
Affiliation(s)
- Marissa J Schafer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN
| | - Thomas A White
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN
| | - Glenda Evans
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN
| | - Jason M Tonne
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN
| | | | - Michael B Stout
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Daniel L Mazula
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN
| | - Allyson K Palmer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN
| | - Darren J Baker
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN
| | - Michael D Jensen
- Division of Endocrinology, Department of Medicine, Mayo Clinic, Rochester, MN
| | | | - Jordan D Miller
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN Department of Surgery, Mayo Clinic, Rochester, MN
| | - Yasuhiro Ikeda
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN
| | - Jan M van Deursen
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Nathan K LeBrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN
| |
Collapse
|
609
|
Martín V, Castilla J, Godoy P, Delgado-Rodríguez M, Soldevila N, Fernández-Villa T, Molina AJ, Astray J, Castro A, González-Candelas F, Mayoral JM, Quintana JM, Domínguez Á. High Body Mass Index as a Risk Factor for Hospitalization Due to Influenza: A Case–Control Study. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.arbr.2015.11.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
610
|
Zhu Y, Tchkonia T, Fuhrmann‐Stroissnigg H, Dai HM, Ling YY, Stout MB, Pirtskhalava T, Giorgadze N, Johnson KO, Giles CB, Wren JD, Niedernhofer LJ, Robbins PD, Kirkland JL. Identification of a novel senolytic agent, navitoclax, targeting the Bcl-2 family of anti-apoptotic factors. Aging Cell 2016; 15:428-35. [PMID: 26711051 PMCID: PMC4854923 DOI: 10.1111/acel.12445] [Citation(s) in RCA: 762] [Impact Index Per Article: 84.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2015] [Indexed: 12/29/2022] Open
Abstract
Clearing senescent cells extends healthspan in mice. Using a hypothesis‐driven bioinformatics‐based approach, we recently identified pro‐survival pathways in human senescent cells that contribute to their resistance to apoptosis. This led to identification of dasatinib (D) and quercetin (Q) as senolytics, agents that target some of these pathways and induce apoptosis preferentially in senescent cells. Among other pro‐survival regulators identified was Bcl‐xl. Here, we tested whether the Bcl‐2 family inhibitors, navitoclax (N) and TW‐37 (T), are senolytic. Like D and Q, N is senolytic in some, but not all types of senescent cells: N reduced viability of senescent human umbilical vein epithelial cells (HUVECs), IMR90 human lung fibroblasts, and murine embryonic fibroblasts (MEFs), but not human primary preadipocytes, consistent with our previous finding that Bcl‐xl siRNA is senolytic in HUVECs, but not preadipocytes. In contrast, T had little senolytic activity. N targets Bcl‐2, Bcl‐xl, and Bcl‐w, while T targets Bcl‐2, Bcl‐xl, and Mcl‐1. The combination of Bcl‐2, Bcl‐xl, and Bcl‐w siRNAs was senolytic in HUVECs and IMR90 cells, while combination of Bcl‐2, Bcl‐xl, and Mcl‐1 siRNAs was not. Susceptibility to N correlated with patterns of Bcl‐2 family member proteins in different types of human senescent cells, as has been found in predicting response of cancers to N. Thus, N is senolytic and acts in a potentially predictable cell type‐restricted manner. The hypothesis‐driven, bioinformatics‐based approach we used to discover that dasatinib (D) and quercetin (Q) are senolytic can be extended to increase the repertoire of senolytic drugs, including additional cell type‐specific senolytic agents.
Collapse
Affiliation(s)
- Yi Zhu
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester MN USA
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester MN USA
| | | | - Haiming M. Dai
- Center of Medical Physics and Technology Hefei Institutes of Physical Sciences Hefei China
| | - Yuanyuan Y. Ling
- Department of Metabolism and Aging The Scripps Research Institute Jupiter FL USA
| | - Michael B. Stout
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester MN USA
| | | | - Nino Giorgadze
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester MN USA
| | - Kurt O. Johnson
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester MN USA
| | - Cory B. Giles
- Arthritis and Clinical Immunology Research Program Oklahoma Medical Research Foundation Oklahoma City OK USA
| | - Jonathan D. Wren
- Arthritis and Clinical Immunology Research Program Oklahoma Medical Research Foundation Oklahoma City OK USA
| | | | - Paul D. Robbins
- Department of Metabolism and Aging The Scripps Research Institute Jupiter FL USA
| | - James L. Kirkland
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester MN USA
| |
Collapse
|
611
|
"Inflammaging" as a Druggable Target: A Senescence-Associated Secretory Phenotype-Centered View of Type 2 Diabetes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1810327. [PMID: 27340505 PMCID: PMC4908264 DOI: 10.1155/2016/1810327] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/09/2016] [Indexed: 12/22/2022]
Abstract
Aging is a complex phenomenon driven by a variety of molecular alterations. A relevant feature of aging is chronic low-grade inflammation, termed “inflammaging.” In type 2 diabetes mellitus (T2DM), many elements of aging appear earlier or are overrepresented, including consistent inflammaging. T2DM patients have an increased death rate, associated with an incremented inflammatory score. The source of this inflammation is debated. Recently, the senescence-associated secretory phenotype (SASP) has been proposed as the main origin of inflammaging in both aging and T2DM. Different pathogenic mechanisms linked to T2DM progression and complications development have been linked to senescence and SASP, that is, oxidative stress and endoplasmic reticulum (ER) stress. Here we review the latest data connecting oxidative and ER stress with the SASP in the context of aging and T2DM, with emphasis on endothelial cells (ECs) and endothelial dysfunction. Moreover, since current medical practice is insufficient to completely suppress the increased death rate of diabetic patients, we propose a SASP-centered view of T2DM as a futuristic therapeutic option, possibly opening new prospects by moving the attention from one-organ studies of diabetes complications to a wider targeting of the aging process.
Collapse
|
612
|
Midgley AC, Morris G, Phillips AO, Steadman R. 17β-estradiol ameliorates age-associated loss of fibroblast function by attenuating IFN-γ/STAT1-dependent miR-7 upregulation. Aging Cell 2016; 15:531-41. [PMID: 26931423 PMCID: PMC4854905 DOI: 10.1111/acel.12462] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2016] [Indexed: 12/20/2022] Open
Abstract
Age‐related defects in fibroblast differentiation and functionality were previously shown to be associated with impaired hyaluronan (HA) synthase 2 (HAS2) and epidermal growth factor receptor (EGFR) function, as a result of upregulated microRNA‐7 (miR‐7) expression. In aging fibroblasts, inhibiting miR‐7 prevented the dysregulation of the HA‐mediated CD44/EGFR signaling pathway. Here, we investigated transcriptional upregulation of miR‐7 and implicated the age‐associated over‐activation of JAK/STAT1 as a primary candidate. STAT1 binding sites were identified on the putative miR‐7 promoter and stimulation of fibroblasts with the inflammatory cytokine, interferon‐γ (IFN‐γ), significantly increased miR‐7 transcriptional activity and resulted in upregulated miR‐7 and loss of EGFR. Additionally, we demonstrated a role for the anti‐inflammatory steroid, 17β‐estradiol (E2), in the attenuation of miR‐7 expression. E2 stimulation promoted estrogen receptor (ER) interactions with the miR‐7 putative promoter and suppressed miR‐7 expression. E2 also attenuated STAT1 expression and activity. Furthermore, treatments with E2 restored fibroblast functionality, including proliferation, migration and differentiation, key events in effective wound healing. In light of our findings, we propose that the regulation of miR‐7 by pro‐ and anti‐inflammatory mediators plays a wider role than previously thought. The modulation of fibroblast functions and ultimately wound healing by miR‐7 activators or inhibitors could provide realistic targets for the restoration of chronic wound healing capabilities in the elderly.
Collapse
Affiliation(s)
- Adam C. Midgley
- Institute of Nephrology, Wales Kidney Research Unit; Division of Infection and Immunity; Cardiff University; College of Biomedical and Life Sciences; University Hospital of Wales; Heath Park Wales CF14 4XN UK
| | - Glyn Morris
- Institute of Nephrology, Wales Kidney Research Unit; Division of Infection and Immunity; Cardiff University; College of Biomedical and Life Sciences; University Hospital of Wales; Heath Park Wales CF14 4XN UK
| | - Aled O. Phillips
- Institute of Nephrology, Wales Kidney Research Unit; Division of Infection and Immunity; Cardiff University; College of Biomedical and Life Sciences; University Hospital of Wales; Heath Park Wales CF14 4XN UK
| | - Robert Steadman
- Institute of Nephrology, Wales Kidney Research Unit; Division of Infection and Immunity; Cardiff University; College of Biomedical and Life Sciences; University Hospital of Wales; Heath Park Wales CF14 4XN UK
| |
Collapse
|
613
|
Liang X, Yang Q, Fu X, Rogers CJ, Wang B, Pan H, Zhu MJ, Nathanielsz PW, Du M. Maternal obesity epigenetically alters visceral fat progenitor cell properties in male offspring mice. J Physiol 2016; 594:4453-66. [PMID: 27060371 DOI: 10.1113/jp272123] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 03/29/2016] [Indexed: 02/01/2023] Open
Abstract
KEY POINTS Maternal obesity reduces adipogenic progenitor density in offspring adipose tissue. The ability of adipose tissue expansion in the offspring of obese mothers is limited and is associated with metabolic dysfunction of adipose tissue when challenged with a high-fat diet. Maternal obesity induces DNA demethylation in the promoter of zinc finger protein 423, which renders progenitor cells with a high adipogenic capacity. Maternal obesity demonstrates long-term effects on the adipogenic capacity of progenitor cells in offspring adipose tissue, demonstrating a developmental programming effect. ABSTRACT Maternal obesity (MO) programs offspring obesity and metabolic disorders, although the underlying mechanisms remain poorly defined. Progenitor cells are the source of new adipocytes. The present study aimed to test whether MO epigenetically predisposes adipocyte progenitors in the fat of offspring to adipogenic differentiation and subsequent depletion, which leads to a failure of adipose tissue plasticity under positive energy balance, contributing to adipose tissue metabolic dysfunction. C57BL/6 female mice were fed either a control diet (10% energy from fat) or a high-fat diet (45% energy from fat) for 8 weeks before mating. Male offspring of control (Con) and obese (OB) dams were weaned onto a regular (Reg) or obesogenic (Obe) diet until 3 months of age. At weaning, male OB offspring had a higher expression of Zinc finger protein 423 (zfp423), a key transcription factor in adipogenesis, as well as lower DNA methylation of its promoter in progenitors of epididymal fat compared to Con offspring, which was correlated with enhanced adipogenic differentiation. At 3 months of age, progenitor density was 30.9 ± 9.7% lower in OB/Obe compared to Con/Obe mice, accompanied by a limited expansion of the adipocyte number when challenged with a high-energy diet. This difference was associated with lower DNA methylation in the zfp423 promoter in the epididymal fat of OB/Obe offspring, which was correlated with greater macrophage chemotactic protein-1 and hypoxia-inducible factor 1α expression. In summary, MO epigenetically limits the expansion capacity of offspring adipose tissue, providing an explanation for the adipose metabolic dysfunction of male offspring in the setting of MO.
Collapse
Affiliation(s)
- Xingwei Liang
- Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Qiyuan Yang
- Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Xing Fu
- Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Carl J Rogers
- Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Bo Wang
- Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Hong Pan
- Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Mei-Jun Zhu
- School of Food Sciences, Washington State University, Pullman, WA, USA
| | - Peter W Nathanielsz
- Wyoming Pregnancy and Life Course Health Centre, Department of Animal Science, University of Wyoming, Laramie, Wyoming, USA
| | - Min Du
- Washington Centre for Muscle Biology and Department of Animal Sciences, Washington State University, Pullman, WA, USA.,Beijing Advanced Innovation Centre for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
614
|
Xu M, Tchkonia T, Kirkland JL. Perspective: Targeting the JAK/STAT pathway to fight age-related dysfunction. Pharmacol Res 2016; 111:152-154. [PMID: 27241018 DOI: 10.1016/j.phrs.2016.05.015] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 05/09/2016] [Accepted: 05/10/2016] [Indexed: 01/05/2023]
Abstract
Senescent cells accumulate in a variety of tissues with aging. They can develop a senescence-associated secretory phenotype (SASP) that entails secretion of inflammatory cytokines, chemokines, proteases, and growth factors. These SASP components can alter the microenvironment within tissues and affect the function of neighboring cells, which can eventually lead to local and systemic dysfunction. The JAK pathway is more highly activate in senescent than non-senescent cells. Inhibition of the JAK pathway suppresses the SASP in senescent cells and alleviates age-related tissue dysfunction. Targeting senescent cells could be a promising way to improve healthspan in aged population.
Collapse
Affiliation(s)
- Ming Xu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, United States.
| |
Collapse
|
615
|
Pérez LM, Pareja-Galeano H, Sanchis-Gomar F, Emanuele E, Lucia A, Gálvez BG. 'Adipaging': ageing and obesity share biological hallmarks related to a dysfunctional adipose tissue. J Physiol 2016; 594:3187-207. [PMID: 26926488 DOI: 10.1113/jp271691] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 01/21/2016] [Indexed: 12/15/2022] Open
Abstract
The increasing ageing of our societies is accompanied by a pandemic of obesity and related cardiometabolic disorders. Progressive dysfunction of the white adipose tissue is increasingly recognized as an important hallmark of the ageing process, which in turn contributes to metabolic alterations, multi-organ damage and a systemic pro-inflammatory state ('inflammageing'). On the other hand, obesity, the paradigm of adipose tissue dysfunction, shares numerous biological similarities with the normal ageing process such as chronic inflammation and multi-system alterations. Accordingly, understanding the interplay between accelerated ageing related to obesity and adipose tissue dysfunction is critical to gain insight into the ageing process in general as well as into the pathophysiology of obesity and other related conditions. Here we postulate the concept of 'adipaging' to illustrate the common links between ageing and obesity and the fact that, to a great extent, obese adults are prematurely aged individuals.
Collapse
Affiliation(s)
- Laura M Pérez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Helios Pareja-Galeano
- Universidad Europea de Madrid, Spain.,Research Institute Hospital 12 de Octubre ('i+12'), Madrid, Spain
| | | | | | - Alejandro Lucia
- Universidad Europea de Madrid, Spain.,Research Institute Hospital 12 de Octubre ('i+12'), Madrid, Spain
| | - Beatriz G Gálvez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Universidad Europea de Madrid, Spain
| |
Collapse
|
616
|
Kim EJ, Kim YK, Kim MK, Kim S, Kim JY, Lee DH, Chung JH. UV-induced inhibition of adipokine production in subcutaneous fat aggravates dermal matrix degradation in human skin. Sci Rep 2016; 6:25616. [PMID: 27161953 PMCID: PMC4861907 DOI: 10.1038/srep25616] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 04/20/2016] [Indexed: 12/18/2022] Open
Abstract
Ultraviolet (UV) exposure to the human skin reduces triglycerides contents and lipid synthesis in the subcutaneous (SC) fat. Because adiponectin and leptin are the most abundant adipokines from the SC fat, we aim to investigate how they interact with UV exposure and skin aging. The expressions of adiponectin and leptin were significantly decreased in SC fat of sun-exposed forearm skin, in comparison with that of sun-protected buttock skin of the same elderly individuals, indicating that chronic UV exposure decreases both adipokines. Acute UV irradiation also decreased the expressions of adiponectin and leptin in SC fat. The expressions of adiponectin receptor 1/2 and leptin receptor were significantly decreased in the dermis as well as in SC fat. Moreover, while exogenous adiponectin and leptin administration prevented UV- and TNF-α induced matrix metalloproteinase (MMP)-1 expression, they also increased UV- and TNF-α induced reduction of type 1 procollagen production. Silencing of adiponectin, leptin or their receptors led to an increased MMP-1 and a decreased type 1 procollagen expression, which was reversed by treatment with recombinant human adiponectin or leptin. In conclusion, UV exposure decreases the expression of adiponectin and leptin, leading to the exacerbation of photoaging by stimulating MMP-1 expression and inhibiting procollagen synthesis.
Collapse
Affiliation(s)
- Eun Ju Kim
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.,Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Republic of Korea
| | - Yeon Kyung Kim
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.,Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Republic of Korea
| | - Min-Kyoung Kim
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.,Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Republic of Korea
| | - Sungsoo Kim
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.,Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Republic of Korea
| | - Jin Yong Kim
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.,Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Republic of Korea
| | - Dong Hun Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.,Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Republic of Korea
| | - Jin Ho Chung
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.,Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Republic of Korea.,Institute on Aging, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
617
|
Wojciechowicz T, Skrzypski M, Szczepankiewicz D, Hertig I, Kołodziejski PA, Billert M, Strowski MZ, Nowak KW. Original Research: Orexins A and B stimulate proliferation and differentiation of porcine preadipocytes. Exp Biol Med (Maywood) 2016; 241:1786-95. [PMID: 27190275 DOI: 10.1177/1535370216649261] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 04/18/2016] [Indexed: 12/17/2022] Open
Abstract
Orexin A (OXA) and B (OXB) are neuropeptides which regulate appetite, energy expenditure, and arousal via G-protein coupled receptors termed as OXR1 and OXR2. The aim of this study was to characterize the effects of OXA and OXB on proliferation and differentiation of porcine preadipocytes. Porcine preadipocytes express both OXRs. OXA and OXB enhance porcine preadipocyte proliferation by 54.8% or 63.2 %, respectively. OXA and OXB potentiate differentiation of porcine preadipocytes, as judged by the increased lipid accumulation and expression of proadipogenic genes. Cellular lipid content after exposure of preadipocytes for six days to 100 nM OXA or OXB increased by 82.2% or 59.2%, respectively. OXA and OXB suppressed glycerol release by 23.9% or 24.9% in preadipocytes differentiated for six days. OXA (100 nM) increased peroxisome proliferator-activated receptor gamma (PPARγ) expression in cells differentiated for 24 h by 100.5%. PPARγ expression was also stimulated in preadipocytes differentiated in the presence of 10 nM (58.3%) or 100 nM OXA (50.6%) for three days. OXB potentiated PPARγ mRNA expression at 1 nM (59%), 10 nM (53.2%), and 100 nM (73.9%) in cells differentiated for three days. OXA increased CCAAT/enhancer binding protein alpha expression in preadipocytes differentiated for six days by 65%. OXB stimulated CCAAT/enhancer binding protein beta expression in preadipocytes differentiated for three days at 10 nM (149.5%) as well as 100 nM (207.2%). Lipoprotein lipase mRNA expression increased in cells treated with 10 nM OXA by 152.6% and 100 nM OXA by 162%. Lipoprotein lipase expression increased by 134% at 100 nM OXB. Furthermore, OXA (100 nM) and OXB (100 nM) increased leptin mRNA expression in preadipocytes differentiated for three days by 49.9% or 71.3%, respectively. These data indicate that orexin receptors may be relevant in the context of white adipose tissue formation.
Collapse
Affiliation(s)
- Tatiana Wojciechowicz
- Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, Poznań 60-637, Poland
| | - Marek Skrzypski
- Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, Poznań 60-637, Poland
| | - Dawid Szczepankiewicz
- Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, Poznań 60-637, Poland
| | - Iwona Hertig
- Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, Poznań 60-637, Poland
| | - Paweł A Kołodziejski
- Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, Poznań 60-637, Poland
| | - Maria Billert
- Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, Poznań 60-637, Poland
| | - Mathias Z Strowski
- Department of Hepatology and Gastroenterology and Interdisciplinary Centre of Metabolism: Endocrinology, Diabetes and Metabolism, Charite-University Medicine Berlin, Berlin 13353, Germany Department of Gastroenterology, Medical Clinic, Elblandklinik, Meissen 01662, Germany
| | - Krzysztof W Nowak
- Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, Poznań 60-637, Poland
| |
Collapse
|
618
|
Chusyd DE, Wang D, Huffman DM, Nagy TR. Relationships between Rodent White Adipose Fat Pads and Human White Adipose Fat Depots. Front Nutr 2016; 3:10. [PMID: 27148535 PMCID: PMC4835715 DOI: 10.3389/fnut.2016.00010] [Citation(s) in RCA: 235] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/26/2016] [Indexed: 01/09/2023] Open
Abstract
The objective of this review was to compare and contrast the physiological and metabolic profiles of rodent white adipose fat pads with white adipose fat depots in humans. Human fat distribution and its metabolic consequences have received extensive attention, but much of what has been tested in translational research has relied heavily on rodents. Unfortunately, the validity of using rodent fat pads as a model of human adiposity has received less attention. There is a surprisingly lack of studies demonstrating an analogous relationship between rodent and human adiposity on obesity-related comorbidities. Therefore, we aimed to compare known similarities and disparities in terms of white adipose tissue (WAT) development and distribution, sexual dimorphism, weight loss, adipokine secretion, and aging. While the literature supports the notion that many similarities exist between rodents and humans, notable differences emerge related to fat deposition and function of WAT. Thus, further research is warranted to more carefully define the strengths and limitations of rodent WAT as a model for humans, with a particular emphasis on comparable fat depots, such as mesenteric fat.
Collapse
Affiliation(s)
- Daniella E Chusyd
- Department of Nutrition Science, University of Alabama at Birmingham , Birmingham, AL , USA
| | - Donghai Wang
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Derek M Huffman
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Tim R Nagy
- Department of Nutrition Science, University of Alabama at Birmingham , Birmingham, AL , USA
| |
Collapse
|
619
|
Ido Y. Diabetic complications within the context of aging: Nicotinamide adenine dinucleotide redox, insulin C-peptide, sirtuin 1-liver kinase B1-adenosine monophosphate-activated protein kinase positive feedback and forkhead box O3. J Diabetes Investig 2016; 7:448-58. [PMID: 27181414 PMCID: PMC4931191 DOI: 10.1111/jdi.12485] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 01/17/2016] [Accepted: 01/18/2016] [Indexed: 12/31/2022] Open
Abstract
Recent research in nutritional control of aging suggests that cytosolic increases in the reduced form of nicotinamide adenine dinucleotide and decreasing nicotinamide adenine dinucleotide metabolism plays a central role in controlling the longevity gene products sirtuin 1 (SIRT1), adenosine monophosphate‐activated protein kinase (AMPK) and forkhead box O3 (FOXO3). High nutrition conditions, such as the diabetic milieu, increase the ratio of reduced to oxidized forms of cytosolic nicotinamide adenine dinucleotide through cascades including the polyol pathway. This redox change is associated with insulin resistance and the development of diabetic complications, and might be counteracted by insulin C‐peptide. My research and others' suggest that the SIRT1–liver kinase B1–AMPK cascade creates positive feedback through nicotinamide adenine dinucleotide synthesis to help cells cope with metabolic stress. SIRT1 and AMPK can upregulate liver kinase B1 and FOXO3, key factors that help residential stem cells cope with oxidative stress. FOXO3 directly changes epigenetics around transcription start sites, maintaining the health of stem cells. ‘Diabetic memory’ is likely a result of epigenetic changes caused by high nutritional conditions, which disturb the quiescent state of residential stem cells and impair tissue repair. This could be prevented by restoring SIRT1–AMPK positive feedback through activating FOXO3.
Collapse
Affiliation(s)
- Yasuo Ido
- Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
620
|
Kirkland JL. Translating the Science of Aging into Therapeutic Interventions. Cold Spring Harb Perspect Med 2016; 6:a025908. [PMID: 26931808 DOI: 10.1101/cshperspect.a025908] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Life and health span have been extended in experimental animals using drugs that are potentially translatable into humans. Considerable effort is needed beyond the usual steps in drug development to devise the models, and realistic preclinical and clinical trial strategies are required to advance these agents into clinical application. It will be important to focus on subjects who already have symptoms or are at imminent risk of developing disorders related to fundamental aging processes, to use short-term, clinically relevant outcomes, as opposed to long-term outcomes, such as health span or life span, and to validate endpoint measures so they are acceptable to regulatory agencies. Funding is a roadblock, as is shortage of investigators with combined expertise in the basic biology of aging, clinical geriatrics, and investigational new drug clinical trials. Strategies for developing a path from the bench to the bedside are reviewed for interventions that target fundamental aging mechanisms.
Collapse
Affiliation(s)
- James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota 55905
| |
Collapse
|
621
|
|
622
|
Aging and adipose tissue: potential interventions for diabetes and regenerative medicine. Exp Gerontol 2016; 86:97-105. [PMID: 26924669 DOI: 10.1016/j.exger.2016.02.013] [Citation(s) in RCA: 225] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 02/22/2016] [Accepted: 02/24/2016] [Indexed: 12/15/2022]
Abstract
Adipose tissue dysfunction occurs with aging and has systemic effects, including peripheral insulin resistance, ectopic lipid deposition, and inflammation. Fundamental aging mechanisms, including cellular senescence and progenitor cell dysfunction, occur in adipose tissue with aging and may serve as potential therapeutic targets in age-related disease. In this review, we examine the role of adipose tissue in healthy individuals and explore how aging leads to adipose tissue dysfunction, redistribution, and changes in gene regulation. Adipose tissue plays a central role in longevity, and interventions restricted to adipose tissue may impact lifespan. Conversely, obesity may represent a state of accelerated aging. We discuss the potential therapeutic potential of targeting basic aging mechanisms, including cellular senescence, in adipose tissue, using type II diabetes and regenerative medicine as examples. We make the case that aging should not be neglected in the study of adipose-derived stem cells for regenerative medicine strategies, as elderly patients make up a large portion of individuals in need of such therapies.
Collapse
|
623
|
Berryman DE, Henry B, Hjortebjerg R, List EO, Kopchick JJ. Developments in our understanding of the effects of growth hormone on white adipose tissue from mice: implications to the clinic. Expert Rev Endocrinol Metab 2016; 11:197-207. [PMID: 28435436 PMCID: PMC5397118 DOI: 10.1586/17446651.2016.1147950] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Adipose tissue (AT) is a well-established target of growth hormone (GH) and is altered in clinical conditions associated with excess, deficiency and absence of GH action. Due to the difficulty in collecting AT from clinical populations, genetically modified mice have been useful in better understanding how GH affects this tissue. Recent findings in mice would suggest that the impact of GH on AT is beyond alterations of lipolysis, lipogenesis or proliferation/ differentiation. AT depot-specific alterations in immune cells, extracellular matrix, adipokines, and senescence indicate an expanded role for GH in AT physiology. This mouse data will guide additional studies necessary to evaluate the therapeutic potential and safety of GH for conditions associated with altering AT, such as obesity. In this review, we introduce several relatively new intricacies of GH's effect on AT, focusing on recent studies in mice. Finally, we summarize the clinical implications of these findings.
Collapse
Affiliation(s)
- Darlene E Berryman
- Executive Director, The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, (740) 593-9661 - phone, (740) 593-4795 - fax
| | - Brooke Henry
- 108 Konneker Research Labs, Ohio University, (740) 593-9665
| | - Rikke Hjortebjerg
- Medical Research Laboratory, Department of Clinical Medicine, Aarhus University, Noerrebrogade 44, 8000 Aarhus C, Denmark, +45 6166 8045 - phone, +45 7846 2150 - fax
| | - Edward O List
- Senior Scientist, 218 Konneker Research Labs, Edison Biotechnology Institute, Ohio University, (740) 593-4620 - phone, (740) 593-4795 - fax
| | - John J Kopchick
- Distinguished Professor, Goll Ohio Eminent Scholar, 172 Water Tower Drive, Ohio University, (740) 593-4534 - phone, (740) 593-4795 - fax
| |
Collapse
|
624
|
Takahashi K, Takisawa S, Shimokado K, Kono N, Arai H, Ishigami A. Age-related changes of vitamin E: α-tocopherol levels in plasma and various tissues of mice and hepatic α-tocopherol transfer protein. Eur J Nutr 2016; 56:1317-1327. [DOI: 10.1007/s00394-016-1182-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/06/2016] [Indexed: 10/22/2022]
|
625
|
Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan. Nature 2016; 530:184-9. [PMID: 26840489 PMCID: PMC4845101 DOI: 10.1038/nature16932] [Citation(s) in RCA: 2002] [Impact Index Per Article: 222.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 12/21/2015] [Indexed: 01/08/2023]
Abstract
Cellular senescence, a stress-induced irreversible growth arrest often characterized by expression of p16(Ink4a) (encoded by the Ink4a/Arf locus, also known as Cdkn2a) and a distinctive secretory phenotype, prevents the proliferation of preneoplastic cells and has beneficial roles in tissue remodelling during embryogenesis and wound healing. Senescent cells accumulate in various tissues and organs over time, and have been speculated to have a role in ageing. To explore the physiological relevance and consequences of naturally occurring senescent cells, here we use a previously established transgene, INK-ATTAC, to induce apoptosis in p16(Ink4a)-expressing cells of wild-type mice by injection of AP20187 twice a week starting at one year of age. We show that compared to vehicle alone, AP20187 treatment extended median lifespan in both male and female mice of two distinct genetic backgrounds. The clearance of p16(Ink4a)-positive cells delayed tumorigenesis and attenuated age-related deterioration of several organs without apparent side effects, including kidney, heart and fat, where clearance preserved the functionality of glomeruli, cardio-protective KATP channels and adipocytes, respectively. Thus, p16(Ink4a)-positive cells that accumulate during adulthood negatively influence lifespan and promote age-dependent changes in several organs, and their therapeutic removal may be an attractive approach to extend healthy lifespan.
Collapse
|
626
|
van der Heijden RA, Sheedfar F, Morrison MC, Hommelberg PPH, Kor D, Kloosterhuis NJ, Gruben N, Youssef SA, de Bruin A, Hofker MH, Kleemann R, Koonen DPY, Heeringa P. High-fat diet induced obesity primes inflammation in adipose tissue prior to liver in C57BL/6j mice. Aging (Albany NY) 2016; 7:256-68. [PMID: 25979814 PMCID: PMC4429090 DOI: 10.18632/aging.100738] [Citation(s) in RCA: 212] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Metabolic inflammation in adipose tissue and the liver is frequently observed as a result of diet-induced obesity in human and rodent studies. Although the adipose tissue and the liver are both prone to become chronically inflamed with prolonged obesity, their individual contribution to the development of metabolic inflammation remains speculative. Thus, we aimed to elucidate the sequence of inflammatory events in adipose and hepatic tissues to determine their contribution to the development of metabolic inflammation and insulin resistance (IR) in diet-induced obesity. To confirm our hypothesis that adipose tissue (AT) inflammation is initiated prior to hepatic inflammation, C57BL/6J male mice were fed a low-fat diet (LFD; 10% kcal fat) or high-fat diet (HFD; 45% kcal fat) for either 24, 40 or 52 weeks. Lipid accumulation and inflammation was measured in AT and liver. Glucose tolerance was assessed and plasma levels of glucose, insulin, leptin and adiponectin were measured at various time points throughout the study. With HFD, C57BL/6j mice developed a progressive obese phenotype, accompanied by IR at 24 and 40 weeks of HFD, but IR was attenuated after 52 weeks of HFD. AT inflammation was present after 24 weeks of HFD, as indicated by the increased presence of crown-like structures and up-regulation of pro-inflammatory genes Tnf, Il1β, Mcp1 and F4/80. As hepatic inflammation was not detected until 40 weeks of HFD, we show that AT inflammation is established prior to the development of hepatic inflammation. Thus, AT inflammation is likely to have a greater contribution to the development of IR compared to hepatic inflammation.
Collapse
Affiliation(s)
- Roel A van der Heijden
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Section Medical Biology, Groningen, The Netherlands
| | - Fareeba Sheedfar
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Section Molecular Genetics, Groningen, The Netherlands.,Radboud University Medical Center, Department of Physiology, Nijmegen, The Netherlands
| | - Martine C Morrison
- Department of Metabolic Health Research, Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Pascal P H Hommelberg
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Section Molecular Genetics, Groningen, The Netherlands
| | - Danny Kor
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Section Medical Biology, Groningen, The Netherlands
| | - Niels J Kloosterhuis
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Section Molecular Genetics, Groningen, The Netherlands
| | - Nanda Gruben
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Section Molecular Genetics, Groningen, The Netherlands
| | - Sameh A Youssef
- Dutch Molecular Pathology Center, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Alain de Bruin
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Section Molecular Genetics, Groningen, The Netherlands.,Dutch Molecular Pathology Center, Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Marten H Hofker
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Section Medical Biology, Groningen, The Netherlands
| | - Robert Kleemann
- Department of Metabolic Health Research, Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Debby P Y Koonen
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Section Molecular Genetics, Groningen, The Netherlands
| | - Peter Heeringa
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Section Medical Biology, Groningen, The Netherlands
| |
Collapse
|
627
|
Jura M, Kozak LP. Obesity and related consequences to ageing. AGE (DORDRECHT, NETHERLANDS) 2016; 38:23. [PMID: 26846415 PMCID: PMC5005878 DOI: 10.1007/s11357-016-9884-3] [Citation(s) in RCA: 291] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 01/26/2016] [Indexed: 04/17/2023]
Abstract
Obesity has become a major public health problem. Given the current increase in life expectancy, the prevalence of obesity also raises steadily among older age groups. The increase in life expectancy is often accompanied with additional years of susceptibility to chronic ill health associated with obesity in the elderly. Both obesity and ageing are conditions leading to serious health problems and increased risk for disease and death. Ageing is associated with an increase in abdominal obesity, a major contributor to insulin resistance and the metabolic syndrome. Obesity in the elderly is thus a serious concern and comprehension of the key mechanisms of ageing and age-related diseases has become a necessary matter. Here, we aimed to identify similarities underlying mechanisms related to both obesity and ageing. We bring together evidence that age-related changes in body fat distribution and metabolism might be key factors of a vicious cycle that can accelerate the ageing process and onset of age-related diseases.
Collapse
Affiliation(s)
- Magdalena Jura
- Institute of Animal Reproduction and Food Research, Polish Academy of Science, ul. Tuwima 10, 10-748, Olsztyn, Poland.
| | - Leslie P Kozak
- Institute of Animal Reproduction and Food Research, Polish Academy of Science, ul. Tuwima 10, 10-748, Olsztyn, Poland.
| |
Collapse
|
628
|
Challenges and opportunities for stem cell therapy in patients with chronic kidney disease. Kidney Int 2016; 89:767-78. [PMID: 26924058 DOI: 10.1016/j.kint.2015.11.023] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/10/2015] [Accepted: 11/18/2015] [Indexed: 02/07/2023]
Abstract
Chronic kidney disease (CKD) is a global health care burden affecting billions of individuals worldwide. The kidney has limited regenerative capacity from chronic insults, and for the most common causes of CKD, no effective treatment exists to prevent progression to end-stage kidney failure. Therefore, novel interventions, such as regenerative cell-based therapies, need to be developed for CKD. Given the risk of allosensitization, autologous transplantation of cells to boost regenerative potential is preferred. Therefore, verification of cell function and vitality in CKD patients is imperative. Two cell types have been most commonly applied in regenerative medicine. Endothelial progenitor cells contribute to neovasculogenesis primarily through paracrine angiogenic activity and partly by differentiation into mature endothelial cells in situ. Mesenchymal stem cells also exert paracrine effects, including proangiogenic, anti-inflammatory, and antifibrotic activity. However, in CKD, multiple factors may contribute to reduced cell function, including older age, coexisting cardiovascular disease, diabetes, chronic inflammatory states, and uremia, which may limit the effectiveness of an autologous cell-based therapy approach. This Review highlights current knowledge on stem and progenitor cell function and vitality, aspects of the uremic milieu that may serve as a barrier to therapy, and novel methods to improve stem cell function for potential transplantation.
Collapse
|
629
|
Stout MB, Steyn FJ, Jurczak MJ, Camporez JPG, Zhu Y, Hawse JR, Jurk D, Palmer AK, Xu M, Pirtskhalava T, Evans GL, de Souza Santos R, Frank AP, White TA, Monroe DG, Singh RJ, Casaclang-Verzosa G, Miller JD, Clegg DJ, LeBrasseur NK, von Zglinicki T, Shulman GI, Tchkonia T, Kirkland JL. 17α-Estradiol Alleviates Age-related Metabolic and Inflammatory Dysfunction in Male Mice Without Inducing Feminization. J Gerontol A Biol Sci Med Sci 2016; 72:3-15. [PMID: 26809497 PMCID: PMC5155656 DOI: 10.1093/gerona/glv309] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 12/15/2015] [Indexed: 12/20/2022] Open
Abstract
Aging is associated with visceral adiposity, metabolic disorders, and chronic low-grade inflammation. 17α-estradiol (17α-E2), a naturally occurring enantiomer of 17β-estradiol (17β-E2), extends life span in male mice through unresolved mechanisms. We tested whether 17α-E2 could alleviate age-related metabolic dysfunction and inflammation. 17α-E2 reduced body mass, visceral adiposity, and ectopic lipid deposition without decreasing lean mass. These declines were associated with reductions in energy intake due to the activation of hypothalamic anorexigenic pathways and direct effects of 17α-E2 on nutrient-sensing pathways in visceral adipose tissue. 17α-E2 did not alter energy expenditure or excretion. Fasting glucose, insulin, and glycosylated hemoglobin were also reduced by 17α-E2, and hyperinsulinemic-euglycemic clamps revealed improvements in peripheral glucose disposal and hepatic glucose production. Inflammatory mediators in visceral adipose tissue and the circulation were reduced by 17α-E2. 17α-E2 increased AMPKα and reduced mTOR complex 1 activity in visceral adipose tissue but not in liver or quadriceps muscle, which is in contrast to the generalized systemic effects of caloric restriction. These beneficial phenotypic changes occurred in the absence of feminization or cardiac dysfunction, two commonly observed deleterious effects of exogenous estrogen administration. Thus, 17α-E2 holds potential as a novel therapeutic for alleviating age-related metabolic dysfunction through tissue-specific effects.
Collapse
Affiliation(s)
- Michael B Stout
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Frederik J Steyn
- Center for Clinical Research and School of Biomedical Sciences, University of Queensland, Herston, Australia
| | - Michael J Jurczak
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pennsylvania
| | | | - Yi Zhu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - John R Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Diana Jurk
- Institutes for Cell & Molecular Biosciences and Ageing, Newcastle University
| | - Allyson K Palmer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Ming Xu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Tamar Pirtskhalava
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Glenda L Evans
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Roberta de Souza Santos
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Beverly Hills, California
| | - Aaron P Frank
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Beverly Hills, California
| | - Thomas A White
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - David G Monroe
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Ravinder J Singh
- Department of Laboratory Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Jordan D Miller
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Deborah J Clegg
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Beverly Hills, California
| | | | | | - Gerald I Shulman
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut
| | - Tamara Tchkonia
- 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.
| |
Collapse
|
630
|
Martín V, Castilla J, Godoy P, Delgado-Rodríguez M, Soldevila N, Fernández-Villa T, Molina AJ, Astray J, Castro A, González-Candelas F, Mayoral JM, Quintana JM, Domínguez Á. High Body Mass Index as a Risk Factor for Hospitalization Due to Influenza: A Case-Control Study. Arch Bronconeumol 2016; 52:299-307. [PMID: 26809749 DOI: 10.1016/j.arbres.2015.11.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 10/22/2015] [Accepted: 11/09/2015] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Obesity has emerged as a significant independent predictor of severity in pandemic influenzaA (H1N1)pdm09. The aim of this study was to investigate the association between body mass index (BMI) and the risk of hospitalization due to influenza. METHODS Hospitalized patients (n=755) with laboratory-confirmed influenza were individually matched by age, admission/visit date, and province with an outpatient (n=783) with laboratory-confirmed influenza and an outpatient control (n=950). We compared the BMI using conditional logistic regression adjusted for potential confounding factors (aOR). The population attributable fraction (PAF) was calculated. RESULTS A higher BMI was associated with an increased risk of hospitalization compared to both outpatient cases (aOR=1.11; 95%CI: 1.07-1.16) and outpatient controls (aOR=1.04; 95%CI: 1.01-1.07). Compared with normal weight, obesity type I, obesity type II and obesity type III was associated with a greater likelihood of hospitalization compared with outpatient cases (aOR=1.85, 95%CI: 1.05-3.26; aOR=5.24, 95%CI: 1.94-14.15 and aOR=44.38, 95%CI: 4.47-440.5). Compared with normal weight, obesity type II and obesity type III was associated with a greater likelihood of hospitalization compared with outpatient controls (aOR=4.37, 95%CI: 1.79-10.69 and aOR=4.95, 95%CI: 1.45-16.87). In persons without influenza vaccination, all categories of BMI≥30kg/m(2) were associated with a greater likelihood of hospitalization compared with normal weight in both outpatient cases and outpatient controls. The PAF of hospitalization by influenza due to BMI ranged from 21.9% to 8.5%; in the case of unvaccinated against influenza between 20.5% to 16.9%. CONCLUSION A high BMI is associated with an increased risk of hospitalization due to influenza. High percentage of hospital admissions are attributable to their BMI, especially in non vaccinated.
Collapse
Affiliation(s)
- Vicente Martín
- Grupo de Investigación interacciones gen-ambiente y salud, Universidad de León, León, España; CIBER Epidemiología y Salud Pública, Madrid, España.
| | - Jesús Castilla
- CIBER Epidemiología y Salud Pública, Madrid, España; Instituto de Salud Pública de Navarra, Pamplona, España
| | - Pere Godoy
- CIBER Epidemiología y Salud Pública, Madrid, España; Departament de Salut, Generalitat de Catalunya, Barcelona, España
| | - Miguel Delgado-Rodríguez
- CIBER Epidemiología y Salud Pública, Madrid, España; División de Medicina Preventiva y Salud Pública, Universidad de Jaén, Jaén, España
| | | | - Tania Fernández-Villa
- Grupo de Investigación interacciones gen-ambiente y salud, Universidad de León, León, España
| | - Antonio J Molina
- Grupo de Investigación interacciones gen-ambiente y salud, Universidad de León, León, España
| | - Jenaro Astray
- Subdirección de Vigilancia, Comunidad de Madrid, Madrid, España
| | - Ady Castro
- CIBER Enfermedades Respiratorias, Madrid, España
| | - Fernando González-Candelas
- CIBER Epidemiología y Salud Pública, Madrid, España; Unidad Mixta Genómica y Salud CSISP (FISABIO), Universitat de València, Valencia, España
| | | | - José María Quintana
- CIBER Epidemiología y Salud Pública, Madrid, España; Fundación Vasca de Innovación e Investigación Sanitarias, Sondika, Vizcaya, España
| | - Ángela Domínguez
- CIBER Epidemiología y Salud Pública, Madrid, España; Departament de Salut Pública, Universitat de Barcelona, Barcelona, España
| | | |
Collapse
|
631
|
Abstract
For years the human microbiota has been implicated in the etiology of colorectal cancer (CRC). However, identifying the molecular mechanisms for how aneuploidy and chromosomal instability (CIN) arise in sporadic and colitis-associated CRC has been difficult. In this Addendum we review recent work from our laboratory that explore mechanisms by which intestinal commensals polarize colon macrophages to an M1 phenotype to generate a bystander effect (BSE) that leads to mutations, spindle malfunction, cell cycle arrest, tetraploidy, and aneuploidy in epithelial cells. BSE represents the application of a phenomenon initially described in the radiation biology field. The result of commensal-driven BSE on colon epithelial cells is aneuploidy, chromosomal instability (CIN), expression of stem cell and tumor stem cell markers and, ultimately, malignant transformation. Our findings provide a conceptual framework for integrating the microbiota with aging, cyclooxygenase (COX)-2, and inflammation as risk factors for CRC.
Collapse
Affiliation(s)
- Xingmin Wang
- Department of Radiation Oncology; University of Oklahoma Health Sciences Center; Oklahoma City, OK USA
| | - Mark M Huycke
- Department of Medicine; University of Oklahoma Health Sciences Center; Oklahoma City, OK USA,The Muchmore Laboratories for Infectious Diseases Research; Oklahoma City VA Health Care System; Oklahoma City, OK USA,Correspondence to: Mark M Huycke;
| |
Collapse
|
632
|
Pole A, Dimri M, P. Dimri G. Oxidative stress, cellular senescence and ageing. AIMS MOLECULAR SCIENCE 2016. [DOI: 10.3934/molsci.2016.3.300] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
633
|
Lin L, Lee JH, Buras ED, Yu K, Wang R, Smith CW, Wu H, Sheikh-Hamad D, Sun Y. Ghrelin receptor regulates adipose tissue inflammation in aging. Aging (Albany NY) 2016; 8:178-91. [PMID: 26837433 PMCID: PMC4761721 DOI: 10.18632/aging.100888] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 01/20/2016] [Indexed: 12/30/2022]
Abstract
Aging is commonly associated with low-grade adipose inflammation, which is closely linked to insulin resistance. Ghrelin is the only circulating orexigenic hormone which is known to increase obesity and insulin resistance. We previously reported that the expression of the ghrelin receptor, growth hormone secretagogue receptor (GHS-R), increases in adipose tissues during aging, and old Ghsr(-/-) mice exhibit a lean and insulin-sensitive phenotype. Macrophages are major mediators of adipose tissue inflammation, which consist of pro-inflammatory M1 and anti-inflammatory M2 subtypes. Here, we show that in aged mice, GHS-R ablation promotes macrophage phenotypical shift toward anti-inflammatory M2. Old Ghsrp(-/-) mice have reduced macrophage infiltration, M1/M2 ratio, and pro-inflammatory cytokine expression in white and brown adipose tissues. We also found that peritoneal macrophages of old Ghsrp(-/-) mice produce higher norepinephrine, which is in line with increased alternatively-activated M2 macrophages. Our data further reveal that GHS-R has cell-autonomous effects in macrophages, and GHS-R antagonist suppresses lipopolysaccharide (LPS)-induced inflammatory responses in macrophages. Collectively, our studies demonstrate that ghrelin signaling has an important role in macrophage polarization and adipose tissue inflammation during aging. GHS-R antagonists may serve as a novel and effective therapeutic option for age-associated adipose tissue inflammation and insulin resistance.
Collapse
Affiliation(s)
- Ligen Lin
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Jong Han Lee
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Eric D. Buras
- Department of Internal Medicine at University of Michigan Health System, Ann Arbor, MI 48109, USA
| | - Kaijiang Yu
- Department of Intensive Care Unit, the Third Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
| | - Ruitao Wang
- Department of Intensive Care Unit, the Third Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
| | - C. Wayne Smith
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Huaizhu Wu
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - David Sheikh-Hamad
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yuxiang Sun
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
634
|
Lewis-Smith H, Diedrichs PC, Rumsey N, Harcourt D. A systematic review of interventions on body image and disordered eating outcomes among women in midlife. Int J Eat Disord 2016; 49:5-18. [PMID: 26607999 DOI: 10.1002/eat.22480] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 10/02/2015] [Accepted: 10/06/2015] [Indexed: 01/27/2023]
Abstract
OBJECTIVE Body dissatisfaction and disordered eating are widely recognized as issues that warrant attention among women in midlife, particularly the development and delivery of effective interventions. This article systematically reviews existing research on interventions among midlife women on body image and disordered eating outcomes, in order to inform intervention delivery and provide strategic directions for future research. METHOD Fourteen electronic databases were searched for articles published between 1992 and 2015 that evaluated interventions with nonclinical samples of women (M age 35-55 years) in controlled trials with at least one body image measure. Data were extracted and evaluated, and the methodological quality of studies was assessed using the Cochrane Collaboration tool for assessing risk of bias. RESULTS From 7,475 records identified, nine articles evaluating 11 interventions met the inclusion criteria. Seven interventions significantly improved body image at post-test (d's = 0.19-2.22), with significant improvements on disordered eating achieved by two of these interventions (d's = 0.90-1.72). Sustained improvements were achieved by three interventions that employed a multisession, therapeutically based, group intervention format; two with sustained body image and disordered eating improvements, and one with sustained body image improvements only (d's = 0.55-1.21; 2 weeks to 6 months). Methodological quality varied between studies. DISCUSSION To date, three interventions have demonstrated sustained improvements and are indicated for practitioners aiming to improve body image and disordered eating among women in midlife. Replication and more rigorous randomised controlled trials are required to enhance the methodological quality of intervention studies in this field.
Collapse
Affiliation(s)
- Helena Lewis-Smith
- Centre for Appearance Research, University of the West of England, Bristol, United Kingdom
| | - Phillippa C Diedrichs
- Centre for Appearance Research, University of the West of England, Bristol, United Kingdom
| | - Nichola Rumsey
- Centre for Appearance Research, University of the West of England, Bristol, United Kingdom
| | - Diana Harcourt
- Centre for Appearance Research, University of the West of England, Bristol, United Kingdom
| |
Collapse
|
635
|
Kao ES, Yang MY, Hung CH, Huang CN, Wang CJ. Polyphenolic extract from Hibiscus sabdariffa reduces body fat by inhibiting hepatic lipogenesis and preadipocyte adipogenesis. Food Funct 2016; 7:171-82. [DOI: 10.1039/c5fo00714c] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Diets high in fat lead to excess lipid accumulation in adipose tissue, which is a crucial factor in the development of obesity, hepatitis, and hyperlipidemia.
Collapse
Affiliation(s)
- Erl-Shyh Kao
- Department of Beauty Science and Graduate Institute of Beauty Science Technology
- Chienkuo Technology University
- Changhua 500
- Taiwan
| | - Mon-Yuan Yang
- Institute of Biochemistry and Biotechnology
- Chung Shan Medical University
- Taichung 402
- Taiwan
| | - Chia-Hung Hung
- Institute of Biochemistry and Biotechnology
- Chung Shan Medical University
- Taichung 402
- Taiwan
| | - Chien-Ning Huang
- Institute of Medicine
- Chung Shan Medical University
- Taichung 402
- Taiwan
- Department of Internal Medicine
| | - Chau-Jong Wang
- Institute of Biochemistry and Biotechnology
- Chung Shan Medical University
- Taichung 402
- Taiwan
- Department of Medical Research
| |
Collapse
|
636
|
Jahansouz C. Adipocyte Dysfunction, Inflammation, and Insulin Resistance in Obesity. METABOLIC SYNDROME AND DIABETES 2016:61-80. [DOI: 10.1007/978-1-4939-3220-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
637
|
Xu M, Palmer AK, Ding H, Weivoda MM, Pirtskhalava T, White TA, Sepe A, Johnson KO, Stout MB, Giorgadze N, Jensen MD, LeBrasseur NK, Tchkonia T, Kirkland JL. Targeting senescent cells enhances adipogenesis and metabolic function in old age. eLife 2015; 4:e12997. [PMID: 26687007 PMCID: PMC4758946 DOI: 10.7554/elife.12997] [Citation(s) in RCA: 438] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 12/18/2015] [Indexed: 12/22/2022] Open
Abstract
Senescent cells accumulate in fat with aging. We previously found genetic clearance of senescent cells from progeroid INK-ATTAC mice prevents lipodystrophy. Here we show that primary human senescent fat progenitors secrete activin A and directly inhibit adipogenesis in non-senescent progenitors. Blocking activin A partially restored lipid accumulation and expression of key adipogenic markers in differentiating progenitors exposed to senescent cells. Mouse fat tissue activin A increased with aging. Clearing senescent cells from 18-month-old naturally-aged INK-ATTAC mice reduced circulating activin A, blunted fat loss, and enhanced adipogenic transcription factor expression within 3 weeks. JAK inhibitor suppressed senescent cell activin A production and blunted senescent cell-mediated inhibition of adipogenesis. Eight weeks-treatment with ruxolitinib, an FDA-approved JAK1/2 inhibitor, reduced circulating activin A, preserved fat mass, reduced lipotoxicity, and increased insulin sensitivity in 22-month-old mice. Our study indicates targeting senescent cells or their products may alleviate age-related dysfunction of progenitors, adipose tissue, and metabolism. DOI:http://dx.doi.org/10.7554/eLife.12997.001 The likelihood of developing metabolic diseases such as diabetes increases with age. This is, in part, because the cells within fat and other tissues become less sensitive to the hormone insulin as people and other animals get older. Also, the stem cells that give rise to new, insulin-responsive fat cells become dysfunctional with increasing age. This is related to the accumulation of “senescent” cells, which, unlike normal fat cell progenitors, release molecules that are toxic to nearby and distant cells. Xu, Palmer et al. have now investigated if senescent cells interfere with the activity of stem cells from human fat tissue, and if getting rid of these senescent cells might restore the normal activity and insulin responsiveness of aged fat tissue. The experiments revealed that human senescent fat cell progenitors release a protein called activin A, which impedes the normal function of stem cells and fat tissue. Additionally, older mice had higher levels of activin A in both their blood and fat tissue than young mice. Xu, Palmer et al. then analyzed older mice that had been engineered to have senescent fat cells that could be triggered to essentially kill themselves when the mice were treated with a drug. Eliminating the senescent cells from these mice led to lower levels of activin A and more fat tissue (due to improved stem cell capacity to become fully functional fat cells) that expressed genes required for insulin responsiveness. This showed that senescent cells are a cause of age-related fat tissue loss and metabolic disease in older mice. Next, Xu, Palmer et al. treated older mice with drugs called JAK inhibitors, which they found reduce the production of activin A by senescent cells isolated from fat tissue. After two months of treatment, the levels of activin A in the blood and in fat tissue were indeed reduced. The fat tissue in treated mice also showed fewer features associated with the development of diabetes than the fat tissue of untreated mice. As such, these results paralleled those after selectively eliminating the senescent cells. Together these findings suggest that JAK inhibitors or drugs (called senolytics) that selectively eliminate senescent cells may have clinical benefits in treating age-related conditions such as diabetes and stem cell dysfunction. DOI:http://dx.doi.org/10.7554/eLife.12997.002
Collapse
Affiliation(s)
- Ming Xu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, United States
| | - Allyson K Palmer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, United States
| | - Husheng Ding
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, United States
| | - Megan M Weivoda
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, United States
| | - Tamar Pirtskhalava
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, United States
| | - Thomas A White
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, United States
| | - Anna Sepe
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, United States
| | - Kurt O Johnson
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, United States
| | - Michael B Stout
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, United States
| | - Nino Giorgadze
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, United States
| | - Michael D Jensen
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, United States
| | - Nathan K LeBrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, United States
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, United States
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, United States
| |
Collapse
|
638
|
Bouchi R, Takeuchi T, Akihisa M, Ohara N, Nakano Y, Nishitani R, Murakami M, Fukuda T, Fujita M, Minami I, Mihara M, Yoshimoto T, Ogawa Y. Increased visceral adiposity with normal weight is associated with the prevalence of non-alcoholic fatty liver disease in Japanese patients with type 2 diabetes. J Diabetes Investig 2015; 7:607-14. [PMID: 27182043 PMCID: PMC4931213 DOI: 10.1111/jdi.12443] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 10/28/2015] [Accepted: 11/03/2015] [Indexed: 12/22/2022] Open
Abstract
AIMS/INTRODUCTION To investigate the impact of increased visceral adiposity with normal weight (OB[-]VA[+]) on the prevalence of non-alcoholic fatty liver disease in patients with type 2 diabetes. MATERIALS AND METHODS This was a cross-sectional study of 140 Japanese patients with type 2 diabetes (mean age 65 ± 11 year; 44.6% women). Visceral fat area (VFA; cm(2) ) and liver attenuation index (LAI) were assessed by abdominal computed tomography. The patients were divided into four groups by VFA and body mass index (BMI; kg/m(2) ) as follows: BMI <25 kg/m(2) and VFA <100 cm(2) (OB[-]VA[-]), BMI ≥25 kg/m(2) and VFA <100 cm(2) (OB[+]VA[-]), BMI <25 kg/m(2) and VFA ≥100 cm(2) (OB[-]VA[+]), and BMI ≥25 kg/m(2) and VFA ≥100 cm(2) (OB[+]VA[+]). Multivariate linear regression and logistic regression analysis were carried out to determine the impact of OB(-)VA(+) on LAI. RESULTS In the present study, 25.0% were OB(-)VA(+) patients, where the LAI levels were lower (1.09 ± 0.22) than those in OB(-)VA(-) patients (1.23 ± 0.15), and were equivalent to those in OB(+)VA(+) patients (1.03 ± 0.26). In multivariate linear regression analysis, OB(-)VA(+) was independently associated with LAI (standardized β-0.212, P = 0.014). In multivariate logistic regression analysis, OB(-)VA(+) was a significant predictor of LAI <0.9 (odds ratio 5.88, 95% confidence interval 1.03-33.52, P = 0.046). CONCLUSIONS The present study provides evidence that increased visceral adiposity with normal weight is a strong predictor for the prevalence of non-alcoholic fatty liver disease in Japanese patients with type 2 diabetes.
Collapse
Affiliation(s)
- Ryotaro Bouchi
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takato Takeuchi
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Momoko Akihisa
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Norihiko Ohara
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yujiro Nakano
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Rie Nishitani
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masanori Murakami
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tatsuya Fukuda
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masamichi Fujita
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Isao Minami
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masatomo Mihara
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takanobu Yoshimoto
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshihiro Ogawa
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Japan Agency for Medical Research and Development, CREST, Tokyo, Japan
| |
Collapse
|
639
|
Adeniran-Catlett AE, Weinstock LD, Bozal FK, Beguin E, Caraballo AT, Murthy SK. Accelerated adipogenic differentiation of hMSCs in a microfluidic shear stimulation platform. Biotechnol Prog 2015; 32:440-6. [PMID: 26587686 DOI: 10.1002/btpr.2211] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 11/17/2015] [Indexed: 01/16/2023]
Abstract
The use of transplanted adipose tissue to repair crucial defects is clinically interesting for surgical reconstruction. Terminally differentiated adipocytes are utilized to promote the healthy regeneration of defective tissue. Use of differentiated mesenchymal stem cells, capable of differentiation into adipocytes, is advantageous because of their regenerative properties. Conventionally, the differentiation of hMSCs toward adipocytes occurs through chemical stimulation. We designed a microfluidic system, consisting of plastic tubing and a syringe pump, to create an environment of shear to accelerate this differentiation process. This system employed a flow rate equivalent to the accelerated flow rates found within the arterial system in order to promote and activate intracellular and extracellular proteins associated with the adipogenic lineage. Confirmation of sustained viability following shear exposure was obtained using a fluorescent live-dead assay. Visualization of intracellular lipid accumulation was achieved via Oil Red O staining. When placed into culture, shear stimulated hMSCs were further induced toward brown adipose tissue, as evidenced by a greater quantity of lipid triglycerides, relative to unstimulated hMSCs. qRT-PCR analysis validated the phenotypic changes observed when the hMSCs were later cultured in adipogenic differentiation media. Additionally, increased fold change for adipogenic markers such as LPL1, CFL1, and SSP1 were observed as a result of shear stimulation. The significance of this work lies in the demonstration that transient fluid shear exposure of hMSCs in suspension can influence differentiation into adipocytes. © 2015 American Institute of Chemical Engineers Biotechnol. Prog., 32:440-446, 2016.
Collapse
Affiliation(s)
| | | | - Fazli K Bozal
- Biochemistry Program, Northeastern University, Boston, MA
| | - Estelle Beguin
- Dept. of Chemical Engineering, Northeastern University, Boston, MA
| | | | - Shashi K Murthy
- Dept. of Chemical Engineering, Northeastern University, Boston, MA.,Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA
| |
Collapse
|
640
|
Kocher NJ, Kunchala S, Reynolds C, Lehman E, Nie S, Raman JD. Adherent perinephric fat at minimally invasive partial nephrectomy is associated with adverse peri-operative outcomes and malignant renal histology. BJU Int 2015; 117:636-41. [PMID: 26573951 DOI: 10.1111/bju.13378] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES To predict adherent perinephric fat (APF) at minimally invasive partial nephrectomy (MIPN) using the Mayo adhesive probability (MAP) score and to determine the impact of MAP score and APF on MIPN outcomes. PATIENTS AND METHODS A total of 245 patients undergoing MIPN were included in the study. The presence of APF was determined through keywords in operating notes, and radiographic data were obtained from preoperative cross-sectional imaging. Posterior fat thickness was measured between the renal capsule and the posterior abdominal wall at the level of the renal vein. Perinephric stranding was graded on a 0-3 severity scale. RESULTS The study included 123 men and 122 women, with a median age of 55 years, body mass index of 31.7, tumour size of 2.7 cm and nephrometry score of 6. The median posterior fat thickness was 1.79 cm and MAP score 2.63. In all, 26 patients (10.6%) had evidence of APF at the time of renal surgery. Factors predictive of APF included increasing age (P = 0.001), male gender (P = 0.045), perinephric stranding (P = 0.002), posterior fat thickness (P < 0.001) and MAP score (P < 0.001). APF was associated with adverse pathological and peri-operative outcomes including malignant renal histology (P = 0.04), longer operating time (P = 0.005) and greater estimated blood loss (EBL; P = 0.025). CONCLUSIONS Specific clinical and radiographic factors predict APF at MIPN. The presence of APF is associated with adverse peri-operative outcomes including longer operating time and greater EBL. APF was also associated with renal malignancy on final pathology, but further studies are necessary to elucidate the precise underlying mechanism.
Collapse
Affiliation(s)
- Neil J Kocher
- Department of Surgery, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Sudhir Kunchala
- Department of Surgery, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Christopher Reynolds
- Department of Surgery, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Erik Lehman
- Department of Public Health Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Sarah Nie
- Department of Surgery, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Jay D Raman
- Department of Surgery, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| |
Collapse
|
641
|
Oliva-Olivera W, Leiva Gea A, Lhamyani S, Coín-Aragüez L, Alcaide Torres J, Bernal-López MR, García-Luna PP, Morales Conde S, Fernández-Veledo S, El Bekay R, Tinahones FJ. Differences in the Osteogenic Differentiation Capacity of Omental Adipose-Derived Stem Cells in Obese Patients With and Without Metabolic Syndrome. Endocrinology 2015; 156:4492-501. [PMID: 26372179 PMCID: PMC4655209 DOI: 10.1210/en.2015-1413] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Multiple studies have suggested that the reduced differentiation capacity of multipotent adipose tissue-derived mesenchymal stem cells (ASCs) in obese subjects could compromise their use in cell therapy. Our aim was to assess the osteogenic potential of omental ASCs and to examine the status of the isolated CD34(negative)-enriched fraction of omental-derived ASCs from subjects with different metabolic profiles. Omental ASCs from normal-weight subjects and subjects with or without metabolic syndrome were isolated, and the osteogenic potential of omental ASCs was evaluated. Additionally, osteogenic and clonogenic potential, proliferation rate, mRNA expression levels of proteins involved in redox balance, and fibrotic proteins were examined in the CD34(negative)-enriched fraction of omental-derived ASCs. Both the omental ASCs and the CD34(negative)-enriched fraction of omental ASCs from subjects without metabolic syndrome have a greater osteogenic potential than those from subjects with metabolic syndrome. The alkaline phosphatase and osteonectin mRNA were negatively correlated with nicotinamide adenine dinucleotide phosphate oxidase-2 mRNA and the mRNA expression levels of the fibrotic proteins correlated positively with nicotinamide adenine dinucleotide phosphate oxidase-5 mRNA and the homeostasis model assessment. Although the population doubling time was significantly higher in subjects with a body mass index of 25 kg/m(2) or greater, only the CD34(negative)-enriched omental ASC fraction in the subjects with metabolic syndrome had a higher population doubling time than the normal-weight subjects. The osteogenic, clonogenic, fibrotic potential, and proliferation rate observed in vitro suggest that omental ASCs from subjects without metabolic syndrome are more suitable for therapeutic osteogenic applications than those from subjects with metabolic syndrome.
Collapse
Affiliation(s)
- Wilfredo Oliva-Olivera
- Department of Clinical Endocrinology and Nutrition (W.O.-O., S.L., L.C.-A., J.A.T., R.E.B., F.J.T.), Institute of Biomedical Research of Malaga, Hospital Complex of Malaga (Virgen de la Victoria), University of Malaga, and Department of Orthopedic Surgery and Traumatology (A.L.G.), Virgen de la Victoria University Hospital and University of Malaga, and Department of Internal Medicine (M.R.B.-L.), Regional University Hospital of Malaga, Institute of Biomedical Research of Malaga, 29010 Malaga, Spain; Centro de Investigación Biomédica en Red of Obesity Physiopathology and Nutrition (W.O.-O., S.L., L..C.-A., J.A.T., M.R.B.-L., R.E.B., F.J.T.), and Centro de Investigación Biomédica en Red of Diabetes and Associated Metabolic Disorders (S.F.-V.), Institute of Health Salud Carlos III, 28029 Madrid, Spain; Department of Endocrinology and Nutrition (P.P.G.L.) and Unit of Innovation in Minimally Invasive Surgery (S.M.C.), Department of Surgery, Virgen del Rocío University Hospital, 41013 Seville, Spain; and University Hospital of Tarragona Joan XXIII Institut d'Investigació (S.F.-V.), Sanitaria Pere Virgili, Universitat Rovirai Virgili, 43003 Tarragona, Spain
| | - Antonio Leiva Gea
- Department of Clinical Endocrinology and Nutrition (W.O.-O., S.L., L.C.-A., J.A.T., R.E.B., F.J.T.), Institute of Biomedical Research of Malaga, Hospital Complex of Malaga (Virgen de la Victoria), University of Malaga, and Department of Orthopedic Surgery and Traumatology (A.L.G.), Virgen de la Victoria University Hospital and University of Malaga, and Department of Internal Medicine (M.R.B.-L.), Regional University Hospital of Malaga, Institute of Biomedical Research of Malaga, 29010 Malaga, Spain; Centro de Investigación Biomédica en Red of Obesity Physiopathology and Nutrition (W.O.-O., S.L., L..C.-A., J.A.T., M.R.B.-L., R.E.B., F.J.T.), and Centro de Investigación Biomédica en Red of Diabetes and Associated Metabolic Disorders (S.F.-V.), Institute of Health Salud Carlos III, 28029 Madrid, Spain; Department of Endocrinology and Nutrition (P.P.G.L.) and Unit of Innovation in Minimally Invasive Surgery (S.M.C.), Department of Surgery, Virgen del Rocío University Hospital, 41013 Seville, Spain; and University Hospital of Tarragona Joan XXIII Institut d'Investigació (S.F.-V.), Sanitaria Pere Virgili, Universitat Rovirai Virgili, 43003 Tarragona, Spain
| | - Said Lhamyani
- Department of Clinical Endocrinology and Nutrition (W.O.-O., S.L., L.C.-A., J.A.T., R.E.B., F.J.T.), Institute of Biomedical Research of Malaga, Hospital Complex of Malaga (Virgen de la Victoria), University of Malaga, and Department of Orthopedic Surgery and Traumatology (A.L.G.), Virgen de la Victoria University Hospital and University of Malaga, and Department of Internal Medicine (M.R.B.-L.), Regional University Hospital of Malaga, Institute of Biomedical Research of Malaga, 29010 Malaga, Spain; Centro de Investigación Biomédica en Red of Obesity Physiopathology and Nutrition (W.O.-O., S.L., L..C.-A., J.A.T., M.R.B.-L., R.E.B., F.J.T.), and Centro de Investigación Biomédica en Red of Diabetes and Associated Metabolic Disorders (S.F.-V.), Institute of Health Salud Carlos III, 28029 Madrid, Spain; Department of Endocrinology and Nutrition (P.P.G.L.) and Unit of Innovation in Minimally Invasive Surgery (S.M.C.), Department of Surgery, Virgen del Rocío University Hospital, 41013 Seville, Spain; and University Hospital of Tarragona Joan XXIII Institut d'Investigació (S.F.-V.), Sanitaria Pere Virgili, Universitat Rovirai Virgili, 43003 Tarragona, Spain
| | - Leticia Coín-Aragüez
- Department of Clinical Endocrinology and Nutrition (W.O.-O., S.L., L.C.-A., J.A.T., R.E.B., F.J.T.), Institute of Biomedical Research of Malaga, Hospital Complex of Malaga (Virgen de la Victoria), University of Malaga, and Department of Orthopedic Surgery and Traumatology (A.L.G.), Virgen de la Victoria University Hospital and University of Malaga, and Department of Internal Medicine (M.R.B.-L.), Regional University Hospital of Malaga, Institute of Biomedical Research of Malaga, 29010 Malaga, Spain; Centro de Investigación Biomédica en Red of Obesity Physiopathology and Nutrition (W.O.-O., S.L., L..C.-A., J.A.T., M.R.B.-L., R.E.B., F.J.T.), and Centro de Investigación Biomédica en Red of Diabetes and Associated Metabolic Disorders (S.F.-V.), Institute of Health Salud Carlos III, 28029 Madrid, Spain; Department of Endocrinology and Nutrition (P.P.G.L.) and Unit of Innovation in Minimally Invasive Surgery (S.M.C.), Department of Surgery, Virgen del Rocío University Hospital, 41013 Seville, Spain; and University Hospital of Tarragona Joan XXIII Institut d'Investigació (S.F.-V.), Sanitaria Pere Virgili, Universitat Rovirai Virgili, 43003 Tarragona, Spain
| | - Juan Alcaide Torres
- Department of Clinical Endocrinology and Nutrition (W.O.-O., S.L., L.C.-A., J.A.T., R.E.B., F.J.T.), Institute of Biomedical Research of Malaga, Hospital Complex of Malaga (Virgen de la Victoria), University of Malaga, and Department of Orthopedic Surgery and Traumatology (A.L.G.), Virgen de la Victoria University Hospital and University of Malaga, and Department of Internal Medicine (M.R.B.-L.), Regional University Hospital of Malaga, Institute of Biomedical Research of Malaga, 29010 Malaga, Spain; Centro de Investigación Biomédica en Red of Obesity Physiopathology and Nutrition (W.O.-O., S.L., L..C.-A., J.A.T., M.R.B.-L., R.E.B., F.J.T.), and Centro de Investigación Biomédica en Red of Diabetes and Associated Metabolic Disorders (S.F.-V.), Institute of Health Salud Carlos III, 28029 Madrid, Spain; Department of Endocrinology and Nutrition (P.P.G.L.) and Unit of Innovation in Minimally Invasive Surgery (S.M.C.), Department of Surgery, Virgen del Rocío University Hospital, 41013 Seville, Spain; and University Hospital of Tarragona Joan XXIII Institut d'Investigació (S.F.-V.), Sanitaria Pere Virgili, Universitat Rovirai Virgili, 43003 Tarragona, Spain
| | - Maria Rosa Bernal-López
- Department of Clinical Endocrinology and Nutrition (W.O.-O., S.L., L.C.-A., J.A.T., R.E.B., F.J.T.), Institute of Biomedical Research of Malaga, Hospital Complex of Malaga (Virgen de la Victoria), University of Malaga, and Department of Orthopedic Surgery and Traumatology (A.L.G.), Virgen de la Victoria University Hospital and University of Malaga, and Department of Internal Medicine (M.R.B.-L.), Regional University Hospital of Malaga, Institute of Biomedical Research of Malaga, 29010 Malaga, Spain; Centro de Investigación Biomédica en Red of Obesity Physiopathology and Nutrition (W.O.-O., S.L., L..C.-A., J.A.T., M.R.B.-L., R.E.B., F.J.T.), and Centro de Investigación Biomédica en Red of Diabetes and Associated Metabolic Disorders (S.F.-V.), Institute of Health Salud Carlos III, 28029 Madrid, Spain; Department of Endocrinology and Nutrition (P.P.G.L.) and Unit of Innovation in Minimally Invasive Surgery (S.M.C.), Department of Surgery, Virgen del Rocío University Hospital, 41013 Seville, Spain; and University Hospital of Tarragona Joan XXIII Institut d'Investigació (S.F.-V.), Sanitaria Pere Virgili, Universitat Rovirai Virgili, 43003 Tarragona, Spain
| | - Pedro Pablo García-Luna
- Department of Clinical Endocrinology and Nutrition (W.O.-O., S.L., L.C.-A., J.A.T., R.E.B., F.J.T.), Institute of Biomedical Research of Malaga, Hospital Complex of Malaga (Virgen de la Victoria), University of Malaga, and Department of Orthopedic Surgery and Traumatology (A.L.G.), Virgen de la Victoria University Hospital and University of Malaga, and Department of Internal Medicine (M.R.B.-L.), Regional University Hospital of Malaga, Institute of Biomedical Research of Malaga, 29010 Malaga, Spain; Centro de Investigación Biomédica en Red of Obesity Physiopathology and Nutrition (W.O.-O., S.L., L..C.-A., J.A.T., M.R.B.-L., R.E.B., F.J.T.), and Centro de Investigación Biomédica en Red of Diabetes and Associated Metabolic Disorders (S.F.-V.), Institute of Health Salud Carlos III, 28029 Madrid, Spain; Department of Endocrinology and Nutrition (P.P.G.L.) and Unit of Innovation in Minimally Invasive Surgery (S.M.C.), Department of Surgery, Virgen del Rocío University Hospital, 41013 Seville, Spain; and University Hospital of Tarragona Joan XXIII Institut d'Investigació (S.F.-V.), Sanitaria Pere Virgili, Universitat Rovirai Virgili, 43003 Tarragona, Spain
| | - Salvador Morales Conde
- Department of Clinical Endocrinology and Nutrition (W.O.-O., S.L., L.C.-A., J.A.T., R.E.B., F.J.T.), Institute of Biomedical Research of Malaga, Hospital Complex of Malaga (Virgen de la Victoria), University of Malaga, and Department of Orthopedic Surgery and Traumatology (A.L.G.), Virgen de la Victoria University Hospital and University of Malaga, and Department of Internal Medicine (M.R.B.-L.), Regional University Hospital of Malaga, Institute of Biomedical Research of Malaga, 29010 Malaga, Spain; Centro de Investigación Biomédica en Red of Obesity Physiopathology and Nutrition (W.O.-O., S.L., L..C.-A., J.A.T., M.R.B.-L., R.E.B., F.J.T.), and Centro de Investigación Biomédica en Red of Diabetes and Associated Metabolic Disorders (S.F.-V.), Institute of Health Salud Carlos III, 28029 Madrid, Spain; Department of Endocrinology and Nutrition (P.P.G.L.) and Unit of Innovation in Minimally Invasive Surgery (S.M.C.), Department of Surgery, Virgen del Rocío University Hospital, 41013 Seville, Spain; and University Hospital of Tarragona Joan XXIII Institut d'Investigació (S.F.-V.), Sanitaria Pere Virgili, Universitat Rovirai Virgili, 43003 Tarragona, Spain
| | - Sonia Fernández-Veledo
- Department of Clinical Endocrinology and Nutrition (W.O.-O., S.L., L.C.-A., J.A.T., R.E.B., F.J.T.), Institute of Biomedical Research of Malaga, Hospital Complex of Malaga (Virgen de la Victoria), University of Malaga, and Department of Orthopedic Surgery and Traumatology (A.L.G.), Virgen de la Victoria University Hospital and University of Malaga, and Department of Internal Medicine (M.R.B.-L.), Regional University Hospital of Malaga, Institute of Biomedical Research of Malaga, 29010 Malaga, Spain; Centro de Investigación Biomédica en Red of Obesity Physiopathology and Nutrition (W.O.-O., S.L., L..C.-A., J.A.T., M.R.B.-L., R.E.B., F.J.T.), and Centro de Investigación Biomédica en Red of Diabetes and Associated Metabolic Disorders (S.F.-V.), Institute of Health Salud Carlos III, 28029 Madrid, Spain; Department of Endocrinology and Nutrition (P.P.G.L.) and Unit of Innovation in Minimally Invasive Surgery (S.M.C.), Department of Surgery, Virgen del Rocío University Hospital, 41013 Seville, Spain; and University Hospital of Tarragona Joan XXIII Institut d'Investigació (S.F.-V.), Sanitaria Pere Virgili, Universitat Rovirai Virgili, 43003 Tarragona, Spain
| | - Rajaa El Bekay
- Department of Clinical Endocrinology and Nutrition (W.O.-O., S.L., L.C.-A., J.A.T., R.E.B., F.J.T.), Institute of Biomedical Research of Malaga, Hospital Complex of Malaga (Virgen de la Victoria), University of Malaga, and Department of Orthopedic Surgery and Traumatology (A.L.G.), Virgen de la Victoria University Hospital and University of Malaga, and Department of Internal Medicine (M.R.B.-L.), Regional University Hospital of Malaga, Institute of Biomedical Research of Malaga, 29010 Malaga, Spain; Centro de Investigación Biomédica en Red of Obesity Physiopathology and Nutrition (W.O.-O., S.L., L..C.-A., J.A.T., M.R.B.-L., R.E.B., F.J.T.), and Centro de Investigación Biomédica en Red of Diabetes and Associated Metabolic Disorders (S.F.-V.), Institute of Health Salud Carlos III, 28029 Madrid, Spain; Department of Endocrinology and Nutrition (P.P.G.L.) and Unit of Innovation in Minimally Invasive Surgery (S.M.C.), Department of Surgery, Virgen del Rocío University Hospital, 41013 Seville, Spain; and University Hospital of Tarragona Joan XXIII Institut d'Investigació (S.F.-V.), Sanitaria Pere Virgili, Universitat Rovirai Virgili, 43003 Tarragona, Spain
| | - Francisco José Tinahones
- Department of Clinical Endocrinology and Nutrition (W.O.-O., S.L., L.C.-A., J.A.T., R.E.B., F.J.T.), Institute of Biomedical Research of Malaga, Hospital Complex of Malaga (Virgen de la Victoria), University of Malaga, and Department of Orthopedic Surgery and Traumatology (A.L.G.), Virgen de la Victoria University Hospital and University of Malaga, and Department of Internal Medicine (M.R.B.-L.), Regional University Hospital of Malaga, Institute of Biomedical Research of Malaga, 29010 Malaga, Spain; Centro de Investigación Biomédica en Red of Obesity Physiopathology and Nutrition (W.O.-O., S.L., L..C.-A., J.A.T., M.R.B.-L., R.E.B., F.J.T.), and Centro de Investigación Biomédica en Red of Diabetes and Associated Metabolic Disorders (S.F.-V.), Institute of Health Salud Carlos III, 28029 Madrid, Spain; Department of Endocrinology and Nutrition (P.P.G.L.) and Unit of Innovation in Minimally Invasive Surgery (S.M.C.), Department of Surgery, Virgen del Rocío University Hospital, 41013 Seville, Spain; and University Hospital of Tarragona Joan XXIII Institut d'Investigació (S.F.-V.), Sanitaria Pere Virgili, Universitat Rovirai Virgili, 43003 Tarragona, Spain
| |
Collapse
|
642
|
JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. Proc Natl Acad Sci U S A 2015; 112:E6301-10. [PMID: 26578790 DOI: 10.1073/pnas.1515386112] [Citation(s) in RCA: 595] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Chronic, low grade, sterile inflammation frequently accompanies aging and age-related diseases. Cellular senescence is associated with the production of proinflammatory chemokines, cytokines, and extracellular matrix (ECM) remodeling proteases, which comprise the senescence-associated secretory phenotype (SASP). We found a higher burden of senescent cells in adipose tissue with aging. Senescent human primary preadipocytes as well as human umbilical vein endothelial cells (HUVECs) developed a SASP that could be suppressed by targeting the JAK pathway using RNAi or JAK inhibitors. Conditioned medium (CM) from senescent human preadipocytes induced macrophage migration in vitro and inflammation in healthy adipose tissue and preadipocytes. When the senescent cells from which CM was derived had been treated with JAK inhibitors, the resulting CM was much less proinflammatory. The administration of JAK inhibitor to aged mice for 10 wk alleviated both adipose tissue and systemic inflammation and enhanced physical function. Our findings are consistent with a possible contribution of senescent cells and the SASP to age-related inflammation and frailty. We speculate that SASP inhibition by JAK inhibitors may contribute to alleviating frailty. Targeting the JAK pathway holds promise for treating age-related dysfunction.
Collapse
|
643
|
Molligan J, Mitchell R, Bhasin P, Lakhani A, Schon L, Zhang Z. Implantation of Autologous Adipose Tissue-Derived Mesenchymal Stem Cells in Foot Fat Pad in Rats. Foot Ankle Int 2015; 36:1344-51. [PMID: 26085579 DOI: 10.1177/1071100715591092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND The foot fat pad (FFP) bears body weight and may become a source of foot pain during aging. This study investigated the regenerative effects of autologous adipose tissue-derived mesenchymal stem cells (AT-MSCs) in the FFP of rats. METHODS Fat tissue was harvested from a total of 30 male Sprague-Dawley rats for isolation of AT-MSCs. The cells were cultured, adipogenic differentiation was induced for 1 week, and the AT-MSCs were labeled with fluorescent dye before injection. AT-MSCs (5 × 10(4) in 50 µL of saline) were injected into the second infradigital pad in the right hindfoot of the rat of origin. Saline only (50 µL) was injected into the corresponding fat pad in the left hind paw of each rat. Rats (n = 10) were euthanized at 1, 2, and 3 weeks, and the second infradigital fat pads were dissected for histologic examination. RESULTS The fluorescence-labeled AT-MSCs were present in the foot pads throughout the 3-week experimental period. On histologic testing, the area of fat pad units (FPUs) in the fat pads that received AT-MSC injections was greater than that in the control fat pads. Although the thickness of septae was not changed by AT-MSC injections, the density of elastic fibers in the septae was increased in the fat pads with implanted AT-MSCs. CONCLUSION In this short-term study, the implanted AT-MSCs largely survived and might have stimulated the expansion of individual FPUs and increased the density of elastic fibers in the FFP in this rat model. CLINICAL RELEVANCE These data support the development of stem cell therapies for age-associated degeneration in FFP in humans.
Collapse
Affiliation(s)
- Jeremy Molligan
- Orthobiologic Laboratory, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Reed Mitchell
- Orthobiologic Laboratory, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Priya Bhasin
- Orthobiologic Laboratory, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Aliya Lakhani
- Orthobiologic Laboratory, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Lew Schon
- Orthobiologic Laboratory, MedStar Union Memorial Hospital, Baltimore, MD, USA
| | - Zijun Zhang
- Orthobiologic Laboratory, MedStar Union Memorial Hospital, Baltimore, MD, USA
| |
Collapse
|
644
|
Reinier F, Zoledziewska M, Hanna D, Smith JD, Valentini M, Zara I, Berutti R, Sanna S, Oppo M, Cusano R, Satta R, Montesu MA, Jones C, Cerimele D, Nickerson DA, Angius A, Cucca F, Cottoni F, Crisponi L. Mandibular hypoplasia, deafness, progeroid features and lipodystrophy (MDPL) syndrome in the context of inherited lipodystrophies. Metabolism 2015; 64:1530-40. [PMID: 26350127 DOI: 10.1016/j.metabol.2015.07.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 07/10/2015] [Accepted: 07/23/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Lipodystrophies are a large heterogeneous group of genetic or acquired disorders characterized by generalized or partial fat loss, usually associated with metabolic complications such as diabetes mellitus, hypertriglyceridemia and hepatic steatosis. Many efforts have been made in the last years in identifying the genetic etiologies of several lipodystrophy forms, although some remain to be elucidated. METHODS We report here the clinical description of a woman with a rare severe lipodystrophic and progeroid syndrome associated with hypertriglyceridemia and diabetes whose genetic bases have been clarified through whole-exome sequencing (WES) analysis. RESULTS This article reports the 5th MDPL (Mandibular hypoplasia, deafness, progeroid features, and lipodystrophy syndrome) patient with the same de novo p.S605del mutation in POLD1. We provided further genetic evidence that this is a disease-causing mutation along with a plausible molecular mechanism responsible for this recurring event. Moreover we overviewed the current classification of the inherited forms of lipodystrophy, along with their underlying molecular basis. CONCLUSIONS Progress in the identification of lipodystrophy genes will help in better understanding the role of the pathways involved in the complex physiology of fat. This will lead to new targets towards develop innovative therapeutic strategies for treating the disorder and its metabolic complications, as well as more common forms of adipose tissue redistribution as observed in the metabolic syndrome and type 2 diabetes.
Collapse
Affiliation(s)
- Frederic Reinier
- Centre for Advanced Studies, Research and Development in Sardinia (CRS4), Pula, Italy; Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Magdalena Zoledziewska
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Monserrato, Italy
| | - David Hanna
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Josh D Smith
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Maria Valentini
- Centre for Advanced Studies, Research and Development in Sardinia (CRS4), Pula, Italy
| | - Ilenia Zara
- Centre for Advanced Studies, Research and Development in Sardinia (CRS4), Pula, Italy
| | - Riccardo Berutti
- Centre for Advanced Studies, Research and Development in Sardinia (CRS4), Pula, Italy
| | - Serena Sanna
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Monserrato, Italy
| | - Manuela Oppo
- Centre for Advanced Studies, Research and Development in Sardinia (CRS4), Pula, Italy; Dipartimento di Scienze Biomediche, Università di Sassari, Sassari, Italy
| | - Roberto Cusano
- Centre for Advanced Studies, Research and Development in Sardinia (CRS4), Pula, Italy
| | - Rosanna Satta
- Dipartimento di Scienze Chirurgiche, Microchirurgiche e Mediche-Dermatologia-Università di Sassari, Italy
| | - Maria Antonietta Montesu
- Dipartimento di Scienze Chirurgiche, Microchirurgiche e Mediche-Dermatologia-Università di Sassari, Italy
| | - Chris Jones
- Centre for Advanced Studies, Research and Development in Sardinia (CRS4), Pula, Italy
| | - Decio Cerimele
- Dipartimento di Scienze Chirurgiche, Microchirurgiche e Mediche-Dermatologia-Università di Sassari, Italy
| | | | - Andrea Angius
- Centre for Advanced Studies, Research and Development in Sardinia (CRS4), Pula, Italy; Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Monserrato, Italy
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Monserrato, Italy; Dipartimento di Scienze Biomediche, Università di Sassari, Sassari, Italy
| | - Francesca Cottoni
- Dipartimento di Scienze Chirurgiche, Microchirurgiche e Mediche-Dermatologia-Università di Sassari, Italy
| | - Laura Crisponi
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerche (CNR), Monserrato, Italy.
| |
Collapse
|
645
|
Aging-related inflammation in osteoarthritis. Osteoarthritis Cartilage 2015; 23:1966-71. [PMID: 26521742 PMCID: PMC4630808 DOI: 10.1016/j.joca.2015.01.008] [Citation(s) in RCA: 359] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 01/05/2015] [Accepted: 01/09/2015] [Indexed: 02/07/2023]
Abstract
It is well accepted that aging is an important contributing factor to the development of osteoarthritis (OA). The mechanisms responsible appear to be multifactorial and may include an age-related pro-inflammatory state that has been termed "inflamm-aging." Age-related inflammation can be both systemic and local. Systemic inflammation can be promoted by aging changes in adipose tissue that result in increased production of cytokines such as interleukin (IL)-6 and tumor necrosis factor-α (TNFα). Numerous studies have shown an age-related increase in blood levels of IL-6 that has been associated with decreased physical function and frailty. Importantly, higher levels of IL-6 have been associated with an increased risk of knee OA progression. However, knockout of IL-6 in male mice resulted in worse age-related OA rather than less OA. Joint tissue cells, including chondrocytes and meniscal cells, as well as the neighboring infrapatellar fat in the knee joint, can be a local source of inflammatory mediators that increase with age and contribute to OA. An increased production of pro-inflammatory mediators that include cytokines and chemokines, as well as matrix-degrading enzymes important in joint tissue destruction, can be the result of cell senescence and the development of the senescence-associated secretory phenotype (SASP). Further studies are needed to better understand the basis for inflamm-aging and its role in OA with the hope that this work will lead to new interventions targeting inflammation to reduce not only joint tissue destruction but also pain and disability in older adults with OA.
Collapse
|
646
|
Different adipose tissue depots: Metabolic implications and effects of surgical removal. ACTA ACUST UNITED AC 2015; 62:458-64. [DOI: 10.1016/j.endonu.2015.05.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 05/25/2015] [Accepted: 05/29/2015] [Indexed: 01/18/2023]
|
647
|
Walston JD. Connecting Age-Related Biological Decline to Frailty and Late-Life Vulnerability. NESTLÉ NUTRITION INSTITUTE WORKSHOP SERIES 2015; 83:1-10. [PMID: 26485518 DOI: 10.1159/000382052] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Frailty is an important construct in aging which allows for the identification of the most vulnerable subset of older adults. At least two conceptual models of frailty have been developed that have in turn facilitated the development of multiple frailty screening tools. This has enabled the study of populations of frail and nonfrail older adults, and facilitated the risk assessment for adverse health outcomes. In addition, using the syndromic approach to frailty, numerous biological hypotheses have been tested, which have identified chronic inflammatory pathway activation, hypothalamic-pituitary-adrenal axis activation, and sympathetic nervous system activity as important in the development of frailty. In addition, age-related molecular changes related to autophagy, mitochondrial decline, apoptosis, senescent cell development, and necroptosis likely contribute to the heterogeneous phenotype of frailty. The recent development of a frail mouse model with chronic inflammatory pathway activation has helped to facilitate further whole organism biological discoveries. The following article attempts to create an understanding of the connections between these age-related biological changes and frailty.
Collapse
|
648
|
Fu Y, Huebner JL, Kraus VB, Griffin TM. Effect of Aging on Adipose Tissue Inflammation in the Knee Joints of F344BN Rats. J Gerontol A Biol Sci Med Sci 2015; 71:1131-40. [PMID: 26450946 DOI: 10.1093/gerona/glv151] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 08/10/2015] [Indexed: 01/28/2023] Open
Abstract
The infrapatellar fat pad (IFP) secretes inflammatory mediators in osteoarthritic knees, but the effect of aging on IFP inflammation is unknown. We tested the hypothesis that aging increases basal and interleukin-1β (IL-1β)-stimulated IFP inflammation in 10-, 20-, and 30-month-old male F344BN F1-hybrid rats. IFPs were cultured ex vivo for 24 hours and treated ±1ng/mL IL-1β to simulate injury-induced inflammation. IFP inflammation was evaluated by measuring secreted cytokine concentrations and by quantitative expression of immunoregulatory and pro- and anti-adipogenic genes. With age, osteoarthritis pathology increased and IFP mass decreased. Although adipocyte size did not change with age, variation in adipocyte size was positively associated with synovial thickness independent of age whereas associations with cartilage damage were age dependent. In the absence of IL-1β, aging was associated with a significant increase in IFP secretion of tumor necrosis factor α by 67% and IL-13 by 35% and a reduction in the expression of immunoregulatory M2 macrophage genes. However, following an IL-1β challenge, adipogenesis markers decreased and pro- and anti-inflammatory cytokines increased independent of age. The lone exception was leptin, which decreased >70% with age. Thus, although aging promotes osteoarthritis risk by increasing basal inflammation, our findings also revealed a potentially protective effect of aging by decreasing IL-1β-stimulated leptin production.
Collapse
Affiliation(s)
- Yao Fu
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation. Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center
| | | | - Virginia B Kraus
- Duke Molecular Physiology Institute and Division of Rheumatology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Timothy M Griffin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation. Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center. Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center.
| |
Collapse
|
649
|
Sertié RAL, Caminhotto RDO, Andreotti S, Campaña AB, de Proença ARG, de Castro NC, Lima FB. Metabolic adaptations in the adipose tissue that underlie the body fat mass gain in middle-aged rats. AGE (DORDRECHT, NETHERLANDS) 2015; 37:87. [PMID: 26307156 PMCID: PMC5005828 DOI: 10.1007/s11357-015-9826-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 08/07/2015] [Indexed: 06/04/2023]
Abstract
Little is known about adipocyte metabolism during aging process and whether this can influence body fat redistribution and systemic metabolism. To better understand this phenomenon, two animal groups were studied: young-14 weeks old-and middle-aged-16 months old. Periepididymal (PE) and subcutaneous (SC) adipocytes were isolated and tested for their capacities to perform lipolysis and to incorporate D-[U-(14)C]-glucose, D-[U-(14)C]-lactate, and [9,10(n)-(3)H]-oleic acid into lipids. Additionally, the morphometric characteristics of the adipose tissues, glucose tolerance tests, and biochemical determinations (fasting glucose, triglycerides, insulin) in blood were performed. The middle-aged rats showed adipocyte (PE and SC) hypertrophy and glucose intolerance, although there were no significant changes in fasting glycemia and insulin. Furthermore, PE tissue revealed elevated rates (+50 %) of lipolysis during beta-adrenergic-stimulation. There was also an increase (+62 %) in the baseline rate of glucose incorporation into lipids in the PE adipocytes, while these PE cells were almost unresponsive to insulin stimulation and less responsive (a 34 % decrease) in the SC tissue. Also, the capacity of oleic acid esterification was elevated in baseline state and with insulin stimulus in the PE tissue (+90 and 82 %, respectively). Likewise, spontaneous incorporation of lactate into lipids in the PE and SC tissues was higher (+100 and 11 %, respectively) in middle-aged rats. We concluded that adipocyte metabolism of middle-aged animals seems to strongly favor cellular hypertrophy and increased adipose mass, particularly the intra-abdominal PE fat pad. In discussion, we have interpreted all these results as a metabolic adaptations to avoid the spreading of fat that can reach tissues beyond adipose protecting them against ectopic fat accumulation. However, these adaptations may have the potential to lead to future metabolic dysfunctions seen in the senescence.
Collapse
Affiliation(s)
- Rogério Antonio Laurato Sertié
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP Brazil
| | - Rennan de Oliveira Caminhotto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP Brazil
| | - Sandra Andreotti
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP Brazil
| | - Amanda Baron Campaña
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP Brazil
| | | | - Natalie Carolina de Castro
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP Brazil
| | - Fábio Bessa Lima
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP Brazil
| |
Collapse
|
650
|
Brown FF, Bigley AB, Ross JC, LaVoy EC, Simpson RJ, Galloway SDR. T-lymphocyte populations following a period of high volume training in female soccer players. Physiol Behav 2015; 152:175-81. [PMID: 26432452 DOI: 10.1016/j.physbeh.2015.09.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 09/26/2015] [Accepted: 09/28/2015] [Indexed: 11/27/2022]
Abstract
PURPOSE To investigate the T-lymphocyte response to a period of increased training volume in trained females compared to habitual activity in female controls. METHODS Thirteen trained female (19.8 ± 1.9 yrs) soccer players were monitored during a two-week long high volume training period (increased by 39%) and thirteen female untrained (20.5 ± 2.2 yrs) controls were monitored during two-weeks of habitual activity. Blood lymphocytes, collected at rest, were isolated before and after the two-week period. Isolated lymphocytes were assessed for the cell surface expression of the co-receptor CD28, a marker of T-lymphocyte naivety, and CD57 a marker used to identify highly-differentiated T-lymphocytes. Co-expression of these markers was identified on helper CD4(+) and cytotoxic CD8(+) T-lymphocytes. In addition a further population of γδ(+) T-lymphocytes were identified. Plasma was used to determine Cytomegalovirus (CMV) serostatus. RESULTS No difference was observed in the T-lymphocyte populations following the two-week period of increased volume training. At baseline the number of total CD3(+), cytotoxic CD8(+), naïve (CD8(+) CD28(+) CD57(-)), intermediate (CD8(+) CD28(+) CD57(+)) T-lymphocytes and the number and proportion of γδ(+) T-lymphocytes were greater in the trained compared to the untrained females (p<0.05). The proportion of CD4(+)T-lymphocytes was greater in the untrained compared to the trained (p<0.05), in turn the CD4(+):CD8(+) ratio was also greater in the untrained females (p<0.05). Inclusion of percentage body fat as a covariate removed the main effect of training status in all T-lymphocyte sub-populations, with the exception of the γδ(+) T-lymphocyte population. 8% of the untrained group was defined as positive for CMV whereas 23% of the trained group was positive for CMV. However, CMV was not a significant covariate in the analysis of T-lymphocyte proportions. CONCLUSION The period of high volume training had no effect on T-lymphocyte populations in trained females. However, baseline training status differences were evident between groups. This indicates that long-term exercise training, as opposed to short-term changes in exercise volume, appears to elicit discernible changes in the composition of the blood T-lymphocyte pool.
Collapse
Affiliation(s)
- F F Brown
- Health and Exercise Sciences Research Group, University of Stirling, Stirling FK9 4LA, United Kingdom.
| | - A B Bigley
- Health and Exercise Sciences Research Group, University of Stirling, Stirling FK9 4LA, United Kingdom
| | - J C Ross
- Health and Exercise Sciences Research Group, University of Stirling, Stirling FK9 4LA, United Kingdom
| | - E C LaVoy
- Health and Exercise Sciences Research Group, University of Stirling, Stirling FK9 4LA, United Kingdom
| | - R J Simpson
- Health and Exercise Sciences Research Group, University of Stirling, Stirling FK9 4LA, United Kingdom
| | - S D R Galloway
- Health and Exercise Sciences Research Group, University of Stirling, Stirling FK9 4LA, United Kingdom
| |
Collapse
|