51
|
Ruiz-Ojeda FJ, Méndez-Gutiérrez A, Aguilera CM, Plaza-Díaz J. Extracellular Matrix Remodeling of Adipose Tissue in Obesity and Metabolic Diseases. Int J Mol Sci 2019; 20:4888. [PMID: 31581657 PMCID: PMC6801592 DOI: 10.3390/ijms20194888] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/25/2019] [Accepted: 09/29/2019] [Indexed: 12/15/2022] Open
Abstract
The extracellular matrix (ECM) is a network of different proteins and proteoglycans that controls differentiation, migration, repair, survival, and development, and it seems that its remodeling is required for healthy adipose tissue expansion. Obesity drives an excessive lipid accumulation in adipocytes, which provokes immune cells infiltration, fibrosis (an excess of deposition of ECM components such as collagens, elastin, and fibronectin) and inflammation, considered a consequence of local hypoxia, and ultimately insulin resistance. To understand the mechanism of this process is a challenge to treat the metabolic diseases. This review is focused at identifying the putative role of ECM in adipose tissue, describing its structure and components, its main tissue receptors, and how it is affected in obesity, and subsequently the importance of an appropriate ECM remodeling in adipose tissue expansion to prevent metabolic diseases.
Collapse
Affiliation(s)
- Francisco Javier Ruiz-Ojeda
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain.
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain.
- RG Adipocytes and metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, 85764 Neuherberg, Munich, Germany.
| | - Andrea Méndez-Gutiérrez
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain.
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain.
- Institute of Nutrition and Food Technology "José Mataix", Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. 18016 Armilla, Granada, Spain.
- CIBEROBN (CIBER Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Concepción María Aguilera
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain.
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain.
- Institute of Nutrition and Food Technology "José Mataix", Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. 18016 Armilla, Granada, Spain.
- CIBEROBN (CIBER Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Julio Plaza-Díaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain.
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain.
- Institute of Nutrition and Food Technology "José Mataix", Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. 18016 Armilla, Granada, Spain.
| |
Collapse
|
52
|
DeVallance E, Branyan KW, Lemaster KC, Anderson R, Marshall KL, Olfert IM, Smith DM, Kelley EE, Bryner RW, Frisbee JC, Chantler PD. Exercise training prevents the perivascular adipose tissue-induced aortic dysfunction with metabolic syndrome. Redox Biol 2019; 26:101285. [PMID: 31374361 PMCID: PMC6669320 DOI: 10.1016/j.redox.2019.101285] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/22/2019] [Accepted: 07/25/2019] [Indexed: 12/20/2022] Open
Abstract
The aim of the study was to determine the effects of exercise training on improving the thoracic perivascular adipose tissue (tPVAT) phenotype (inflammation, oxidative stress, and proteasome function) in metabolic syndrome and its subsequent actions on aortic function. Methods Lean and obese (model of metabolic syndrome) Zucker rats (n=8/group) underwent 8-weeks of control conditions or treadmill exercise (70% of max speed, 1 h/day, 5 days/week). At the end of the intervention, the tPVAT was removed and conditioned media was made. The cleaned aorta was attached to a force transducer to assess endothelium-dependent and independent dilation in the presence or absence of tPVAT-conditioned media. tPVAT gene expression, inflammatory /oxidative phenotype, and proteasome function were assessed. Results The main findings were that Ex induced: (1) a beige-like, anti-inflammatory tPVAT phenotype; (2) a greater abundance of •NO in tPVAT; (3) a reduction in tPVAT oxidant production; and (4) an improved tPVAT proteasome function. Regarding aortic function, endothelium-dependent dilation was greater in exercised lean and obese groups vs. controls (p < 0.05). Lean control tPVAT improved aortic relaxation, whereas obese control tPVAT decreased aortic relaxation. In contrast, the obese Ex-tPVAT increased aortic dilation, whereas the lean Ex-tPVAT did not affect aortic dilation. Conclusion Overall, exercise had the most dramatic impact on the obese tPVAT reflecting a change towards an environment with less oxidant load, less inflammation and improved proteasome function. Such beneficial changes to the tPVAT micro-environment with exercise likely played a significant role in mediating the improvement in aortic function in metabolic syndrome following 8 weeks of exercise.
Collapse
Affiliation(s)
- Evan DeVallance
- Division of Exercise Physiology, WVU School of Medicine, Morgantown, WV, USA
| | - Kayla W Branyan
- Division of Exercise Physiology, WVU School of Medicine, Morgantown, WV, USA
| | - Kent C Lemaster
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Ray Anderson
- Department of Biochemistry, WVU School of Medicine, Morgantown, WV, USA
| | - Kent L Marshall
- Division of Exercise Physiology, WVU School of Medicine, Morgantown, WV, USA
| | - I Mark Olfert
- Division of Exercise Physiology, WVU School of Medicine, Morgantown, WV, USA
| | - David M Smith
- Department of Biochemistry, WVU School of Medicine, Morgantown, WV, USA
| | - Eric E Kelley
- Department of Physiology & Pharmacology, WVU School of Medicine, Morgantown, WV, USA
| | - Randy W Bryner
- Division of Exercise Physiology, WVU School of Medicine, Morgantown, WV, USA
| | - Jefferson C Frisbee
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada; Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Paul D Chantler
- Division of Exercise Physiology, WVU School of Medicine, Morgantown, WV, USA; Department of Neuroscience, WVU School of Medicine, Morgantown, WV, USA.
| |
Collapse
|
53
|
Liu R, Nikolajczyk BS. Tissue Immune Cells Fuel Obesity-Associated Inflammation in Adipose Tissue and Beyond. Front Immunol 2019; 10:1587. [PMID: 31379820 PMCID: PMC6653202 DOI: 10.3389/fimmu.2019.01587] [Citation(s) in RCA: 195] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/25/2019] [Indexed: 12/12/2022] Open
Abstract
Obesity-associated inflammation stems from a combination of cell-intrinsic changes of individual immune cell subsets and the dynamic crosstalk amongst a broad array of immune cells. Although much of the focus of immune cell contributions to metabolic disease has focused on adipose tissue-associated cells, these potent sources of inflammation inhabit other metabolic regulatory tissues, including liver and gut, and recirculate to promote systemic inflammation and thus obesity comorbidities. Tissue-associated immune cells, especially T cell subpopulations, have become a hotspot of inquiry based on their contributions to obesity, type 2 diabetes, non-alcoholic fatty liver diseases and certain types of cancers. The cell-cell interactions that take place under the stress of obesity are mediated by intracellular contact and cytokine production, and constitute a complicated network that drives the phenotypic alterations of immune cells and perpetuates a feed-forward loop of metabolic decline. Herein we discuss immune cell functions in various tissues and obesity-associated cancers from the viewpoint of inflammation. We also emphasize recent advances in the understanding of crosstalk amongst immune cell subsets under obese conditions, and suggest future directions for focused investigations with clinical relevance.
Collapse
Affiliation(s)
- Rui Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Barbara S. Nikolajczyk
- Department of Pharmacology and Nutritional Sciences, Barnstable Brown Diabetes and Obesity Research Center, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
54
|
Rubinow KB, Houston B, Wang S, Goodspeed L, Ogimoto K, Morton GJ, McCarty C, Braun RE, Page ST. Androgen receptor deficiency in monocytes/macrophages does not alter adiposity or glucose homeostasis in male mice. Asian J Androl 2019; 20:276-283. [PMID: 29205180 PMCID: PMC5952483 DOI: 10.4103/aja.aja_54_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Androgen deprivation in men leads to increased adiposity, but the mechanisms underlying androgen regulation of fat mass have not been fully defined. Androgen receptor (AR) is expressed in monocytes/macrophages, which are resident in key metabolic tissues and influence energy metabolism in surrounding cells. Male mice bearing a cell-specific knockout of the AR in monocytes/macrophages (M-ARKO) were generated to determine whether selective loss of androgen signaling in these cells would lead to altered body composition. Wild-type (WT) and M-ARKO mice (12–22 weeks of age, n = 12 per group) were maintained on a regular chow diet for 8 weeks and then switched to a high-fat diet for 8 additional weeks. At baseline and on both the regular chow and high-fat diets, no differences in lean mass or fat mass were observed between groups. Consistent with the absence of differential body weight or adiposity, no differences in food intake (3.0 ± 0.5 g per day for WT mice vs 2.8 ± 0.4 g per day for M-ARKO mice) or total energy expenditure (0.6 ± 0.1 Kcal h−1 for WT mice vs 0.5 ± 0.1 Kcal h−1 for M-ARKO mice) were evident between groups during high-fat feeding. Liver weight was greater in M-ARKO than that in WT mice (1.5 ± 0.1 g vs 1.3 ± 0.0 g, respectively, P = 0.02). Finally, M-ARKO mice did not exhibit impairments in glucose tolerance or insulin sensitivity relative to WT mice at any study time point. In aggregate, these findings suggest that AR signaling specifically in monocytes/macrophages does not contribute to the regulation of systemic energy balance, adiposity, or insulin sensitivity in male mice.
Collapse
Affiliation(s)
- Katya B Rubinow
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Barbara Houston
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Shari Wang
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Leela Goodspeed
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Kayoko Ogimoto
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Gregory J Morton
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | | | | | - Stephanie T Page
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
55
|
Role of innate immune cells in metabolism: from physiology to type 2 diabetes. Semin Immunopathol 2019; 41:531-545. [DOI: 10.1007/s00281-019-00736-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 03/15/2019] [Indexed: 12/19/2022]
|
56
|
Silva KR, Baptista LS. Adipose-derived stromal/stem cells from different adipose depots in obesity development. World J Stem Cells 2019; 11:147-166. [PMID: 30949294 PMCID: PMC6441940 DOI: 10.4252/wjsc.v11.i3.147] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 01/27/2019] [Accepted: 02/28/2019] [Indexed: 02/06/2023] Open
Abstract
The increasing prevalence of obesity is alarming because it is a risk factor for cardiovascular and metabolic diseases (such as type 2 diabetes). The occurrence of these comorbidities in obese patients can arise from white adipose tissue (WAT) dysfunctions, which affect metabolism, insulin sensitivity and promote local and systemic inflammation. In mammals, WAT depots at different anatomical locations (subcutaneous, preperitoneal and visceral) are highly heterogeneous in their morpho-phenotypic profiles and contribute differently to homeostasis and obesity development, depending on their ability to trigger and modulate WAT inflammation. This heterogeneity is likely due to the differential behavior of cells from each depot. Numerous studies suggest that adipose-derived stem/stromal cells (ASC; referred to as adipose progenitor cells, in vivo) with depot-specific gene expression profiles and adipogenic and immunomodulatory potentials are keys for the establishment of the morpho-functional heterogeneity between WAT depots, as well as for the development of depot-specific responses to metabolic challenges. In this review, we discuss depot-specific ASC properties and how they can contribute to the pathophysiology of obesity and metabolic disorders, to provide guidance for researchers and clinicians in the development of ASC-based therapeutic approaches.
Collapse
Affiliation(s)
- Karina Ribeiro Silva
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology, Duque de Caxias, RJ 25250-020, Brazil
- Post-Graduation Program of Biotechnology, National Institute of Metrology, Quality and Technology, Duque de Caxias, RJ 25250-020, Brazil
| | - Leandra Santos Baptista
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology, Duque de Caxias, RJ 25250-020, Brazil
- Post-Graduation Program of Biotechnology, National Institute of Metrology, Quality and Technology, Duque de Caxias, RJ 25250-020, Brazil
- Multidisciplinary Center for Biological Research (Numpex-Bio), Federal University of Rio de Janeiro Campus Duque de Caxias, Duque de Caxias, RJ 25245-390, Brazil
| |
Collapse
|
57
|
Kwon EY, Choi MS. Dietary Eriodictyol Alleviates Adiposity, Hepatic Steatosis, Insulin Resistance, and Inflammation in Diet-Induced Obese Mice. Int J Mol Sci 2019; 20:ijms20051227. [PMID: 30862092 PMCID: PMC6429409 DOI: 10.3390/ijms20051227] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 12/14/2022] Open
Abstract
The present study aimed to investigate the molecular mechanisms underlying the anti-obesity effect of flavonoid eriodictyol (ED) supplementation in mice fed with a high-fat diet (HFD). C57BL/6N mice were fed with normal diet (ND), HFD (40 kcal% fat), or HFD + 0.005% (w/w) ED for 16 weeks. In HFD-induced obese mice, dietary ED supplementation significantly alleviated dyslipidemia and adiposity by downregulating the expression of lipogenesis-related genes in white adipose tissue (WAT), while enhancing fecal lipid excretion. ED additionally improved hepatic steatosis and decreased the production of pro-inflammatory cytokines by downregulating the expression of hepatic enzymes and the genes involved in lipogenesis and upregulating the expression of hepatic fatty acid oxidation-related enzymes and genes. In addition, ED improved insulin resistance (IR) by suppressing hepatic gluconeogenesis, enhancing glucose utilization, and modulating the production and release of two incretin hormones, namely gastric inhibitory polypeptide (GIP) and glucagon-like peptide-1 (GLP-1). Taken together, the current findings indicated that ED can protect against diet-induced obesity and related metabolic disturbances, including dyslipidemia, inflammation, fatty liver disease, and IR in diet-induced obese mice.
Collapse
Affiliation(s)
- Eun-Young Kwon
- Department of Food Science and Nutrition, Kyungpook National University, 1370 San-Kyuk Dong Puk-Ku, Daegu 41566, Korea.
- Center for Food and Nutritional Genomics Research, Kyungpook National University, 1370 San-Kyuk Dong, Puk-Ku, Daegu 41566, Korea.
| | - Myung-Sook Choi
- Department of Food Science and Nutrition, Kyungpook National University, 1370 San-Kyuk Dong Puk-Ku, Daegu 41566, Korea.
- Center for Food and Nutritional Genomics Research, Kyungpook National University, 1370 San-Kyuk Dong, Puk-Ku, Daegu 41566, Korea.
| |
Collapse
|
58
|
Lee MJ, Pickering RT, Shibad V, Wu Y, Karastergiou K, Jager M, Layne MD, Fried SK. Impaired Glucocorticoid Suppression of TGFβ Signaling in Human Omental Adipose Tissues Limits Adipogenesis and May Promote Fibrosis. Diabetes 2019; 68:587-597. [PMID: 30530781 PMCID: PMC6385749 DOI: 10.2337/db18-0955] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 11/26/2018] [Indexed: 12/18/2022]
Abstract
Visceral obesity is associated with insulin resistance and higher risk of type 2 diabetes and metabolic diseases. A limited ability of adipose tissues to remodel through the recruitment and differentiation of adipose stem cells (ASCs) is associated with adipose tissue inflammation and fibrosis and the metabolic syndrome. We show that the lower adipogenesis of omental (Om) compared with abdominal subcutaneous (Abdsc) ASCs was associated with greater secretion of TGFβ ligands that acted in an autocrine/paracrine loop to activate SMAD2 and suppress adipogenesis. Inhibition of TGFβ signaling rescued Om ASC differentiation. In Abdsc ASCs, low concentrations of dexamethasone suppressed TGFβ signaling and enhanced adipogenesis, at least in part by increasing TGFBR3 protein that can sequester TGFβ ligands. Om ASCs were resistant to these dexamethasone effects; recombinant TGFBR3 increased their differentiation. Pericellular fibrosis, a hallmark of dysfunctional adipose tissue, was greater in Om and correlated with higher level of tissue TGFβ signaling activity and lower ASC differentiation. We conclude that glucocorticoids restrain cell-autonomous TGFβ signaling in ASCs to facilitate adipogenesis and healthy remodeling in Abdsc and these processes are impaired in Om. Therapies directed at overcoming glucocorticoid resistance in visceral adipose tissue may improve remodeling and help prevent metabolic complications of visceral obesity.
Collapse
Affiliation(s)
- Mi-Jeong Lee
- Diabetes Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Obesity Research Center, Boston University School of Medicine, Boston, MA
| | - R Taylor Pickering
- Diabetes Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Obesity Research Center, Boston University School of Medicine, Boston, MA
| | - Varuna Shibad
- Obesity Research Center, Boston University School of Medicine, Boston, MA
| | - Yuanyuan Wu
- Obesity Research Center, Boston University School of Medicine, Boston, MA
| | - Kalypso Karastergiou
- Diabetes Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Obesity Research Center, Boston University School of Medicine, Boston, MA
| | - Mike Jager
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| | - Matthew D Layne
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| | - Susan K Fried
- Diabetes Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
- Obesity Research Center, Boston University School of Medicine, Boston, MA
| |
Collapse
|
59
|
Hengist A, Perkin O, Gonzalez JT, Betts JA, Hewison M, Manolopoulos KN, Jones KS, Koulman A, Thompson D. Mobilising vitamin D from adipose tissue: The potential impact of exercise. NUTR BULL 2019; 44:25-35. [PMID: 34853551 PMCID: PMC8609434 DOI: 10.1111/nbu.12369] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Vitamin D is lipophilic and accumulates substantially in adipose tissue. Even without supplementation, the amount of vitamin D in the adipose of a typical adult is equivalent to several months of the daily reference nutrient intake (RNI). Paradoxically, despite the large amounts of vitamin D located in adipose tissue, individuals with obesity are often vitamin D deficient according to consensus measures of vitamin D status (serum 25-hydroxyvitamin D concentrations). Thus, it appears that vitamin D can become 'trapped' in adipose tissue, potentially due to insufficient lipolytic stimulation and/or due to tissue dysfunction/adaptation resulting from adipose expansion. Emerging evidence suggests that exercise may mobilise vitamin D from adipose (even in the absence of weight loss). If exercise helps to mobilise vitamin D from adipose tissue, then this could have important ramifications for practitioners and policymakers regarding the management of low circulating levels of vitamin D, as well as chronically low levels of physical activity, obesity and associated health conditions. This perspective led us to design a study to examine the impact of exercise on vitamin D status, vitamin D turnover and adipose tissue vitamin D content (the VitaDEx project). The VitaDEx project will determine whether increasing physical activity (via exercise) represents a potentially useful strategy to mobilise vitamin D from adipose tissue.
Collapse
Affiliation(s)
- A Hengist
- Department for Health University of Bath Bath UK
| | - O Perkin
- Department for Health University of Bath Bath UK
| | - J T Gonzalez
- Department for Health University of Bath Bath UK
| | - J A Betts
- Department for Health University of Bath Bath UK
| | - M Hewison
- Institute of Metabolism and Systems Research University of Birmingham Birmingham UK
| | - K N Manolopoulos
- Institute of Metabolism and Systems Research University of Birmingham Birmingham UK
| | - K S Jones
- NIHR BRC Nutritional Biomarker Laboratory University of Cambridge Cambridge UK
| | - A Koulman
- NIHR BRC Nutritional Biomarker Laboratory University of Cambridge Cambridge UK
| | - D Thompson
- Department for Health University of Bath Bath UK
| |
Collapse
|
60
|
Burhans MS, Hagman DK, Kuzma JN, Schmidt KA, Kratz M. Contribution of Adipose Tissue Inflammation to the Development of Type 2 Diabetes Mellitus. Compr Physiol 2018; 9:1-58. [PMID: 30549014 DOI: 10.1002/cphy.c170040] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The objective of this comprehensive review is to summarize and discuss the available evidence of how adipose tissue inflammation affects insulin sensitivity and glucose tolerance. Low-grade, chronic adipose tissue inflammation is characterized by infiltration of macrophages and other immune cell populations into adipose tissue, and a shift toward more proinflammatory subtypes of leukocytes. The infiltration of proinflammatory cells in adipose tissue is associated with an increased production of key chemokines such as C-C motif chemokine ligand 2, proinflammatory cytokines including tumor necrosis factor α and interleukins 1β and 6 as well as reduced expression of the key insulin-sensitizing adipokine, adiponectin. In both rodent models and humans, adipose tissue inflammation is consistently associated with excess fat mass and insulin resistance. In humans, associations with insulin resistance are stronger and more consistent for inflammation in visceral as opposed to subcutaneous fat. Further, genetic alterations in mouse models of obesity that reduce adipose tissue inflammation are-almost without exception-associated with improved insulin sensitivity. However, a dissociation between adipose tissue inflammation and insulin resistance can be observed in very few rodent models of obesity as well as in humans following bariatric surgery- or low-calorie-diet-induced weight loss, illustrating that the etiology of insulin resistance is multifactorial. Taken together, adipose tissue inflammation is a key factor in the development of insulin resistance and type 2 diabetes in obesity, along with other factors that likely include inflammation and fat accumulation in other metabolically active tissues. © 2019 American Physiological Society. Compr Physiol 9:1-58, 2019.
Collapse
Affiliation(s)
- Maggie S Burhans
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Derek K Hagman
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Jessica N Kuzma
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Kelsey A Schmidt
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Mario Kratz
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Epidemiology, University of Washington, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
61
|
Visceral fat and insulin resistance - what we know? Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2018; 163:19-27. [PMID: 30398218 DOI: 10.5507/bp.2018.062] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 10/02/2018] [Indexed: 02/08/2023] Open
Abstract
One of the most significant challenges of current medicine is the increasing prevalence of obesity worldwide that is accompanied by a wide range of chronic health complications and increased mortality. White adipose tissue actively contributes to metabolic regulation by production of a variety of hormones and cytokines, commonly referred to as adipokines. The spectrum and quantity of adipokines produced by the adipose tissue of obese patients is directly or indirectly involved in much obesity-related pathology (type 2 diabetes mellitus, cardiovascular disease, inflammatory response). One of the underlying mechanisms linking obesity, diabetes, and cardiovascular complications is subclinical inflammation, primarily arising in visceral adipose tissue. Adipocyte size, number and polarization of lymphocytes and infiltrated macrophages are closely related to metabolic and obesity-related diseases. The storage capacity of hypertrophic adipocytes in obese patients is limited. This results in chronic energy overload and leads to increased apoptosis of adipocytes that in turn stimulates the infiltration of visceral adipose tissue by immune cells, in particular macrophages. These cells produce many proinflammatory factors; while the overall production of anti-inflammatory cytokines and adipokines is decreased. The constant release of proinflammatory factors into the circulation then contributes to a subclinical systemic inflammation, which is directly linked to the metabolic and cardiovascular complications of obesity.
Collapse
|
62
|
Russo L, Lumeng CN. Properties and functions of adipose tissue macrophages in obesity. Immunology 2018; 155:407-417. [PMID: 30229891 DOI: 10.1111/imm.13002] [Citation(s) in RCA: 440] [Impact Index Per Article: 62.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/06/2018] [Accepted: 09/07/2018] [Indexed: 12/12/2022] Open
Abstract
The expansion of adipose tissue (AT) in obesity is accompanied by the accumulation of immune cells that contribute to a state of low-grade, chronic inflammation and dysregulated metabolism. Adipose tissue macrophages (ATMs) represent the most abundant class of leukocytes in AT and are involved in the regulation of several regulatory physiological processes, such as tissue remodeling and insulin sensitivity. With progressive obesity, ATMs are key mediators of meta-inflammation, insulin resistance and impairment of adipocyte function. While macrophage recruitment from blood monocytes is a critical component of the generation of AT inflammation, new studies have revealed a role for ATM proliferation in the early stages of obesity and in sustaining AT inflammation. In addition, studies have revealed a more complex range of macrophage activation states than the previous M1/M2 model, and the existence of different macrophage profiles between human and animal models. This review will summarize the current understanding of the regulatory mechanisms of ATM function in relation to obesity, type 2 diabetes, depot of origin, and to other leukocytes such as AT dendritic cells, with hopes of emphasizing the regulatory nodes that can potentially be targeted to prevent and treat obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Lucia Russo
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Carey N Lumeng
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA.,Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
63
|
Senders ML, Hernot S, Carlucci G, van de Voort JC, Fay F, Calcagno C, Tang J, Alaarg A, Zhao Y, Ishino S, Palmisano A, Boeykens G, Meerwaldt AE, Sanchez-Gaytan BL, Baxter S, Zendman L, Lobatto ME, Karakatsanis NA, Robson PM, Broisat A, Raes G, Lewis JS, Tsimikas S, Reiner T, Fayad ZA, Devoogdt N, Mulder WJM, Pérez-Medina C. Nanobody-Facilitated Multiparametric PET/MRI Phenotyping of Atherosclerosis. JACC Cardiovasc Imaging 2018; 12:2015-2026. [PMID: 30343086 PMCID: PMC6461528 DOI: 10.1016/j.jcmg.2018.07.027] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 06/11/2018] [Accepted: 07/12/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVES This study sought to develop an integrative positron emission tomography (PET) with magnetic resonance imaging (MRI) procedure for accurate atherosclerotic plaque phenotyping, facilitated by clinically approved and nanobody radiotracers. BACKGROUND Noninvasive characterization of atherosclerosis remains a challenge in clinical practice. The limitations of current diagnostic methods demonstrate that, in addition to atherosclerotic plaque morphology and composition, disease activity needs to be evaluated. METHODS We screened 3 nanobody radiotracers targeted to different biomarkers of atherosclerosis progression, namely vascular cell adhesion molecule (VCAM)-1, lectin-like oxidized low-density lipoprotein receptor (LOX)-1, and macrophage mannose receptor (MMR). The nanobodies, initially radiolabeled with copper-64 (64Cu), were extensively evaluated in Apoe–/– mice and atherosclerotic rabbits using a combination of in vivo PET/MRI readouts and ex vivo radioactivity counting, autoradiography, and histological analyses. RESULTS The 3 nanobody radiotracers accumulated in atherosclerotic plaques and displayed short circulation times due to fast renal clearance. The MMR nanobody was selected for labeling with gallium-68 (68Ga), a short-lived radioisotope with high clinical relevance, and used in an ensuing atherosclerosis progression PET/MRI study. Macrophage burden was longitudinally studied by 68Ga-MMR–PET, plaque burden by T2-weighted MRI, and neovascularization by dynamic contrast-enhanced (DCE) MRI. Additionally, inflammation and microcalcifications were evaluated by fluorine-18 (18F)-labeled fluorodeoxyglucose (18F-FDG) and 18F-sodium fluoride (18F-NaF) PET, respectively. We observed an increase in all the aforementioned measures as disease progressed, and the imaging signatures correlated with histopathological features. CONCLUSIONS We have evaluated nanobody-based radiotracers in rabbits and developed an integrative PET/MRI protocol that allows noninvasive assessment of different processes relevant to atherosclerosis progression. This approach allows the multiparametric study of atherosclerosis and can aid in early stage anti-atherosclerosis drug trials.
Collapse
Affiliation(s)
- Max L Senders
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Medical Biochemistry, Academic Medical Center, Amsterdam, the Netherlands
| | - Sophie Hernot
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Giuseppe Carlucci
- Bernard and Irene Schwarz Center for Biomedical Imaging, New York University, New York, New York; Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Jan C van de Voort
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Francois Fay
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Chemistry, York College of The City University of New York, New York, New York
| | - Claudia Calcagno
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jun Tang
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Amr Alaarg
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Biomaterials Science and Technology, Technical Medical Centre. University of Twente, Enschede, the Netherlands
| | - Yiming Zhao
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Seigo Ishino
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Anna Palmisano
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Unit of Clinical Research in Radiology, Experimental Imaging Center, San Raffaele Scientific Institute, Milan, Italy
| | - Gilles Boeykens
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Anu E Meerwaldt
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Brenda L Sanchez-Gaytan
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Samantha Baxter
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Laura Zendman
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Mark E Lobatto
- Department of Radiology, Academic Medical Center, Amsterdam, the Netherlands
| | - Nicolas A Karakatsanis
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Philip M Robson
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alexis Broisat
- Bioclinic Radiopharmaceutics Laboratory, Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche S 1039, Grenoble, France
| | - Geert Raes
- Research Group of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Laboratory of Myeloid Cell Immunology, Vlaams Instituut voor Biotechnologie Inflammation Research Center, Ghent, Belgium
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Radiology, Weill Cornell Medical College, New York, New York; Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sotirios Tsimikas
- Division of Cardiovascular Diseases, Sulpizio Cardiovascular Center, Department of Medicine, University of California-La Jolla, San Diego, California
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Zahi A Fayad
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Willem J M Mulder
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Medical Biochemistry, Academic Medical Center, Amsterdam, the Netherlands.
| | - Carlos Pérez-Medina
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
64
|
Luteolin Targets the Toll-Like Receptor Signaling Pathway in Prevention of Hepatic and Adipocyte Fibrosis and Insulin Resistance in Diet-Induced Obese Mice. Nutrients 2018; 10:nu10101415. [PMID: 30282902 PMCID: PMC6213163 DOI: 10.3390/nu10101415] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 09/22/2018] [Accepted: 09/24/2018] [Indexed: 12/18/2022] Open
Abstract
This study was to investigate the protective role of luteolin on inflammation-mediated metabolic diseases, focusing on the role of luteolin in the modulation of the Toll-like receptor (TLR) signaling pathway. C57BL/6J mice were fed a normal, high-fat, or high-fat + 0.005% (w/w) luteolin diet for 16 weeks. Luteolin improved chronic low-grade inflammation by modulating the TLR signaling pathway, resulting in reduced pro-inflammatory cytokines and macrophage accumulation. A positive relationship was detected between gene expressions of Tlr5, Map2k7, Mapk12, Mapk13, and Mapk9 and lipogenesis in epididymal white adipose tissue (eWAT) of luteolin-treated mice, which was linked to attenuation of hepatic lipotoxicity by increasing free fatty acid (FFA) flux to the WAT. Luteolin prevented fibrosis by decreasing extracellular matrix accumulation and cathepsin gene expressions, while enhancing the hepatic antioxidant system. Emr1 and Ccl7, important markers inducing low-grade inflammation, were affected by advanced age and greater body weight, which were normalized by luteolin treatment. Luteolin improved insulin resistance by normalizing pancreatic islet dysfunction and differentially modulating the plasma glucagon-like peptide-1 and gastric inhibitory polypeptide levels. Our results suggest that luteolin ameliorates diet-induced obesity and its comorbidities. Overall, this study provides novel insights into the effect of luteolin on the links among adiposopathy, insulin resistance, hepatic steatosis, and fibrosis.
Collapse
|
65
|
Enhanced ANGPTL2 expression in adipose tissues and its association with insulin resistance in obese women. Sci Rep 2018; 8:13976. [PMID: 30228336 PMCID: PMC6143523 DOI: 10.1038/s41598-018-32419-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 09/07/2018] [Indexed: 12/27/2022] Open
Abstract
Angiopoietin-like protein 2 has been proposed to be a key mediator linking obesity and insulin resistance. However, no detailed study of ANGPTL2 expression in human adipose tissues has yet been reported. To investigate the pattern and regulation of ANGPTL2 expression in human adipose tissues in obesity and its related diseases, we recruited 32 non-diabetic and 13 type 2 diabetic obese women and 32 normal-weight women. ANGPTL2 mRNA was expressed at a similar level in visceral and subcutaneous adipose tissues. Adipose tissue ANGPTL2 mRNA was much higher in obese patients. Adipose tissue ANGPTL2 mRNA and serum ANGPTL2 levels showed strong associations with metabolic parameters associated with insulin resistance. In adipose tissue, ANGPTL2 mRNA was closely correlated with the expression of genes involved in inflammation and ER stress. ANGPTL2 mRNA was principally expressed in adipocytes, and its expression was markedly higher in the adipocyte but non-adipocyte fraction of obese adipose tissues. Culture of human adipocytes under conditions mimicking the microenvironment of obese adipose tissue (especially, increased ER stress) stimulated ANGPTL2 gene expression and secretion. In addition, co-culture of adipocytes and macrophages suggested that ANGPTL2 excessively produced by adipocytes, may contribute inflammation and remodeling in obese adipose tissues, thereby promoting insulin resistance.
Collapse
|
66
|
Groell F, Jordan O, Borchard G. In vitro models for immunogenicity prediction of therapeutic proteins. Eur J Pharm Biopharm 2018; 130:128-142. [DOI: 10.1016/j.ejpb.2018.06.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 05/09/2018] [Accepted: 06/08/2018] [Indexed: 12/21/2022]
|
67
|
Sajja AP, Joshi AA, Teague HL, Dey AK, Mehta NN. Potential Immunological Links Between Psoriasis and Cardiovascular Disease. Front Immunol 2018; 9:1234. [PMID: 29910818 PMCID: PMC5992299 DOI: 10.3389/fimmu.2018.01234] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 05/16/2018] [Indexed: 12/12/2022] Open
Abstract
Preclinical and clinical research provide strong evidence that chronic, systemic inflammation plays a key role in development and progression of atherosclerosis. Indeed, chronic inflammatory diseases, such as psoriasis, are associated with accelerated atherosclerosis and increased risk of cardiovascular events. Contemporary research has demonstrated plausible mechanistic links between immune cell dysfunction and cardiometabolic disease in psoriasis. In this review, we describe the role of potential common immunological mechanisms underlying both psoriasis and atherogenesis. We primarily discuss innate and adaptive immune cell subsets and their contributions to psoriatic disease and cardiovascular morbidity. Emerging efforts should focus on understanding the interplay among immune cells, adipose tissue, and various biomarkers of immune dysfunction to provide direction for future targeted therapy.
Collapse
Affiliation(s)
| | | | | | | | - Nehal N. Mehta
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
68
|
Campbell JP, Turner JE. Debunking the Myth of Exercise-Induced Immune Suppression: Redefining the Impact of Exercise on Immunological Health Across the Lifespan. Front Immunol 2018; 9:648. [PMID: 29713319 PMCID: PMC5911985 DOI: 10.3389/fimmu.2018.00648] [Citation(s) in RCA: 386] [Impact Index Per Article: 55.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 03/15/2018] [Indexed: 12/17/2022] Open
Abstract
Epidemiological evidence indicates that regular physical activity and/or frequent structured exercise reduces the incidence of many chronic diseases in older age, including communicable diseases such as viral and bacterial infections, as well as non-communicable diseases such as cancer and chronic inflammatory disorders. Despite the apparent health benefits achieved by leading an active lifestyle, which imply that regular physical activity and frequent exercise enhance immune competency and regulation, the effect of a single bout of exercise on immune function remains a controversial topic. Indeed, to this day, it is perceived by many that a vigorous bout of exercise can temporarily suppress immune function. In the first part of this review, we deconstruct the key pillars which lay the foundation to this theory-referred to as the "open window" hypothesis-and highlight that: (i) limited reliable evidence exists to support the claim that vigorous exercise heightens risk of opportunistic infections; (ii) purported changes to mucosal immunity, namely salivary IgA levels, after exercise do not signpost a period of immune suppression; and (iii) the dramatic reductions to lymphocyte numbers and function 1-2 h after exercise reflects a transient and time-dependent redistribution of immune cells to peripheral tissues, resulting in a heightened state of immune surveillance and immune regulation, as opposed to immune suppression. In the second part of this review, we provide evidence that frequent exercise enhances-rather than suppresses-immune competency, and highlight key findings from human vaccination studies which show heightened responses to bacterial and viral antigens following bouts of exercise. Finally, in the third part of this review, we highlight that regular physical activity and frequent exercise might limit or delay aging of the immune system, providing further evidence that exercise is beneficial for immunological health. In summary, the over-arching aim of this review is to rebalance opinion over the perceived relationships between exercise and immune function. We emphasize that it is a misconception to label any form of acute exercise as immunosuppressive, and, instead, exercise most likely improves immune competency across the lifespan.
Collapse
Affiliation(s)
- John P Campbell
- Department for Health, University of Bath, Bath, United Kingdom
| | - James E Turner
- Department for Health, University of Bath, Bath, United Kingdom
| |
Collapse
|
69
|
Chen L, Wang L, Li Y, Wuang L, Liu Y, Pang N, Luo Y, He J, Zhang L, Chen N, Li R, Wu J. Transplantation of Normal Adipose Tissue Improves Blood Flow and Reduces Inflammation in High Fat Fed Mice With Hindlimb Ischemia. Front Physiol 2018; 9:197. [PMID: 29568274 PMCID: PMC5852102 DOI: 10.3389/fphys.2018.00197] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 02/23/2018] [Indexed: 01/18/2023] Open
Abstract
Background: Fat deposition is associated with peripheral arterial disease. Adipose tissue has recently been implicated in vascular remodeling and angiogenic activity. We hypothesized that the transplantation of adipose tissues from normal mice improves blood flow perfusion and neovascularization in high-fat diet fed mice. Methods: After 14 weeks of high-fat diet (HFD)-fed mice, unilateral hind limb ischemia was performed. Subcutaneous white adipose tissue (WAT) and brown adipose tissue (BAT) fat pads were harvested from normal EGFP mice, and subcutaneously transplanted over the region of the adductor muscles of HFD mice. Blood flow was measured using Laser Doppler Scanner. Vascular density, macrophages infiltration, and macrophage polarization were examined by RT-qPCR, and immunohistochemistry. Results: We found that the transplantation of WAT derived from normal mice improved functional blood flow in HFD-fed mice compared to mice transplanted with BAT and sham-treated mice. WAT transplantation increased the recruitment of pericytes associated with nascent blood vessels, but did not affect capillary formation. Furthermore, transplantation of WAT ameliorated HFD-induced insulin resistance, M2 macrophage predominance and the release of arteriogenic factors in ischemic muscles. Mice receiving WAT also displayed a marked reduction in several proinflammatory cytokines. In contrast, mice transplanted with BAT were glucose intolerant and demonstrated increased IL-6 levels in ischemic muscles. Conclusion: These results indicate that transplantation of adipose tissue elicits improvements in blood perfusion and beneficial effects on systemic glucose homeostasis and could be a promising therapeutic option for the treatment of diabetic peripheral arterial disease.
Collapse
Affiliation(s)
- Liyuan Chen
- Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Lin Wang
- Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yongjie Li
- Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Liqun Wuang
- Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yaofang Liu
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Ningbo Pang
- Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yulin Luo
- Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jing He
- Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Liping Zhang
- Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Ni Chen
- Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Rong Li
- Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jianbo Wu
- Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, China.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| |
Collapse
|
70
|
Muir LA, Kiridena S, Griffin C, DelProposto JB, Geletka L, Martinez-Santibañez G, Zamarron BF, Lucas H, Singer K, O' Rourke RW, Lumeng CN. Frontline Science: Rapid adipose tissue expansion triggers unique proliferation and lipid accumulation profiles in adipose tissue macrophages. J Leukoc Biol 2018; 103:615-628. [PMID: 29493813 DOI: 10.1002/jlb.3hi1017-422r] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/04/2018] [Accepted: 01/10/2018] [Indexed: 12/22/2022] Open
Abstract
Obesity-related changes in adipose tissue leukocytes, in particular adipose tissue macrophages (ATMs) and dendritic cells (ATDCs), are implicated in metabolic inflammation, insulin resistance, and altered regulation of adipocyte function. We evaluated stromal cell and white adipose tissue (WAT) expansion dynamics with high fat diet (HFD) feeding for 3-56 days, quantifying ATMs, ATDCs, endothelial cells (ECs), and preadipocytes (PAs) in visceral epididymal WAT and subcutaneous inguinal WAT. To better understand mechanisms of the early response to obesity, we evaluated ATM proliferation and lipid accumulation. ATMs, ATDCs, and ECs increased with rapid WAT expansion, with ATMs derived primarily from a CCR2-independent resident population. WAT expansion stimulated proliferation in resident ATMs and ECs, but not CD11c+ ATMs or ATDCs. ATM proliferation was unperturbed in Csf2- and Rag1-deficient mice with WAT expansion. Additionally, ATM apoptosis decreased with WAT expansion, and proliferation and apoptosis reverted to baseline with weight loss. Adipocytes reached maximal hypertrophy at 28 days of HFD, coinciding with a plateau in resident ATM accumulation and the appearance of lipid-laden CD11c+ ATMs in visceral epididymal WAT. ATM increases were proportional to tissue expansion and adipocyte hypertrophy, supporting adipocyte-mediated regulation of resident ATMs. The appearance of lipid-laden CD11c+ ATMs at peak adipocyte size supports a role in responding to ectopic lipid accumulation within adipose tissue. In contrast, ATDCs increase independently of proliferation and may be derived from circulating precursors. These changes precede and establish the setting in which large-scale adipose tissue infiltration of CD11c+ ATMs, inflammation, and adipose tissue dysfunction contributes to insulin resistance.
Collapse
Affiliation(s)
- Lindsey A Muir
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Samadhi Kiridena
- College of Literature Science and the Arts, University of Michigan, Ann Arbor, Michigan, USA
| | - Cameron Griffin
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jennifer B DelProposto
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Lynn Geletka
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Gabriel Martinez-Santibañez
- Cellular and Molecular Biology Graduate Program, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Brian F Zamarron
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Hannah Lucas
- College of Literature Science and the Arts, University of Michigan, Ann Arbor, Michigan, USA
| | - Kanakadurga Singer
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Robert W O' Rourke
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Surgery, Ann Arbor Veterans Administration Hospital, Ann Arbor, Michigan, USA
| | - Carey N Lumeng
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Cellular and Molecular Biology Graduate Program, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
71
|
Tanaka K, Sata M. Roles of Perivascular Adipose Tissue in the Pathogenesis of Atherosclerosis. Front Physiol 2018; 9:3. [PMID: 29487532 PMCID: PMC5816816 DOI: 10.3389/fphys.2018.00003] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 01/03/2018] [Indexed: 01/14/2023] Open
Abstract
Traditionally, it is believed that white adipose tissues serve as energy storage, heat insulation, and mechanical cushion, whereas non-shivering thermogenesis occurs in brown adipose tissue. Recent evidence revealed that adipose tissue secretes many types of cytokines, called as adipocytokines, which modulate glucose metabolism, lipid profile, appetite, fibrinolysis, blood pressure, and inflammation. Most of the arteries are surrounded by perivascular adipose tissue (PVAT). PVAT has been thought to be simply a structurally supportive tissue for vasculature. However, recent studies showed that PVAT influences vasodilation and vasocontraction, suggesting that PVAT regulates vascular tone and diameter. Adipocytokines secreted from PVAT appear to have direct access to the adjacent arterial wall by diffusion or via vasa vasorum. In fact, PVAT around atherosclerotic lesions and mechanically-injured arteries displayed inflammatory cytokine profiles, suggesting that PVAT functions to promote vascular lesion formation. Many clinical studies revealed that increased accumulation of epicardial adipose tissue (EAT), which surrounds coronary arteries, is associated with coronary artery disease. In this review article, we will summarize recent findings about potential roles of PVAT in the pathogenesis of atherosclerosis, particularly focusing on a series of basic and clinical studies from our laboratory.
Collapse
Affiliation(s)
- Kimie Tanaka
- Division for Health Service Promotion, The University of Tokyo, Tokyo, Japan
| | - Masataka Sata
- Department of Cardiovascular Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
72
|
Trim W, Turner JE, Thompson D. Parallels in Immunometabolic Adipose Tissue Dysfunction with Ageing and Obesity. Front Immunol 2018; 9:169. [PMID: 29479350 PMCID: PMC5811473 DOI: 10.3389/fimmu.2018.00169] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/19/2018] [Indexed: 12/12/2022] Open
Abstract
Ageing, like obesity, is often associated with alterations in metabolic and inflammatory processes resulting in morbidity from diseases characterised by poor metabolic control, insulin insensitivity, and inflammation. Ageing populations also exhibit a decline in immune competence referred to as immunosenescence, which contributes to, or might be driven by chronic, low-grade inflammation termed "inflammageing". In recent years, animal and human studies have started to uncover a role for immune cells within the stromal fraction of adipose tissue in driving the health complications that come with obesity, but relatively little work has been conducted in the context of immunometabolic adipose function in ageing. It is now clear that aberrant immune function within adipose tissue in obesity-including an accumulation of pro-inflammatory immune cell populations-plays a major role in the development of systemic chronic, low-grade inflammation, and limiting the function of adipocytes leading to an impaired fat handling capacity. As a consequence, these changes increase the chance of multiorgan dysfunction and disease onset. Considering the important role of the immune system in obesity-associated metabolic and inflammatory diseases, it is critically important to further understand the interplay between immunological processes and adipose tissue function, establishing whether this interaction contributes to age-associated immunometabolic dysfunction and inflammation. Therefore, the aim of this article is to summarise how the interaction between adipose tissue and the immune system changes with ageing, likely contributing to the age-associated increase in inflammatory activity and loss of metabolic control. To understand the potential mechanisms involved, parallels will be drawn to the current knowledge derived from investigations in obesity. We also highlight gaps in research and propose potential future directions based on the current evidence.
Collapse
Affiliation(s)
- William Trim
- Department for Health, University of Bath, Bath, United Kingdom
| | - James E Turner
- Department for Health, University of Bath, Bath, United Kingdom
| | - Dylan Thompson
- Department for Health, University of Bath, Bath, United Kingdom
| |
Collapse
|
73
|
Orr JS, Kennedy AJ, Hill AA, Anderson-Baucum EK, Hubler MJ, Hasty AH. CC-chemokine receptor 7 (CCR7) deficiency alters adipose tissue leukocyte populations in mice. Physiol Rep 2018; 4:4/18/e12971. [PMID: 27655794 PMCID: PMC5037919 DOI: 10.14814/phy2.12971] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 08/22/2016] [Indexed: 11/24/2022] Open
Abstract
The mechanism by which macrophages and other immune cells accumulate in adipose tissue (AT) has been an area of intense investigation over the past decade. Several different chemokines and their cognate receptors have been studied for their role as chemoattractants in promoting recruitment of immune cells to AT. However, it is also possible that chemoattractants known to promote clearance of immune cells from tissues to regional lymph nodes might be a critical component to overall AT immune homeostasis. In this study, we evaluated whether CCR7 influences AT macrophage (ATM) or T‐cell (ATT) accumulation. CCR7−/− and littermate wild‐type (WT) mice were placed on low‐fat diet (LFD) or high‐fat diet (HFD) for 16 weeks. CCR7 deficiency did not impact HFD‐induced weight gain, hepatic steatosis, or glucose intolerance. Although lean CCR7−/− mice had an increased proportion of alternatively activated ATMs, there were no differences in ATM accumulation or polarization between HFD‐fed CCR7−/− mice and their WT counterparts. However, CCR7 deficiency did lead to the preferential accumulation of CD8+ATT cells, which was further exacerbated by HFD feeding. Finally, expression of inflammatory cytokines/chemokines, such as Tnf, Il6, Il1β, Ccl2, and Ccl3, was equally elevated in AT by HFD feeding in CCR7−/− and WT mice, while Ifng and Il18 were elevated by HFD feeding in CCR7−/− but not in WT mice. Together, these data suggest that CCR7 plays a role in CD8+ATT cell egress, but does not influence ATM accumulation or the metabolic impact of diet‐induced obesity.
Collapse
Affiliation(s)
- Jeb S Orr
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Arion J Kennedy
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Andrea A Hill
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Emily K Anderson-Baucum
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Merla J Hubler
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
74
|
Al Dubayee MS, Alayed H, Almansour R, Alqaoud N, Alnamlah R, Obeid D, Alshahrani A, Zahra MM, Nasr A, Al-Bawab A, Aljada A. Differential Expression of Human Peripheral Mononuclear Cells Phenotype Markers in Type 2 Diabetic Patients and Type 2 Diabetic Patients on Metformin. Front Endocrinol (Lausanne) 2018; 9:537. [PMID: 30356719 PMCID: PMC6189318 DOI: 10.3389/fendo.2018.00537] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 08/23/2018] [Indexed: 01/22/2023] Open
Abstract
Background: Although peripheral blood mononuclear cells (PBMC) have been demonstrated to be in a pro-inflammatory state in obesity and type 2 Diabetes Mellitus (T2DM), characterization of circulating PBMC phenotypes in the obese and T2DM and the effect of Metformin on these phenotypes in humans is still ill-defined and remains to be determined. Methods: Thirty normal healthy adult volunteers of normal weight, 30 obese subjects, 20 obese newly diagnosed diabetics and 30 obese diabetics on Metformin were recruited for the study. Fasting blood samples were collected and PBMC were isolated from whole blood. Polarization markers (CD86, IL-6, TNFα, iNOS, CD36, CD11c, CD169, CD206, CD163, CD68, CD11b, CD16, and CD14) were measured by RT-qPCR. Gene expression fold changes were calculated using the 2-ΔΔCT method for RT-qPCR. Results: Obesity and T2DM are associated an increased CD68 marker in PBMC. mRNA expression of CD11b, CD11c, CD169, and CD163 were significantly reduced in PBMC from T2DM subjects whereas CD11c was significantly inhibited in PBMC from obese subjects. On the other hand, macrophage M1-like phenotype was observed in T2DM circulation as demonstrated by increased mRNA expression of CD16, IL-6, iNOS, TNFα, and CD36. There were no significant changes in CD14 and CD86 in the obese and T2DM when compared to the lean subjects. Metformin treatment in T2DM reverted CD11c, CD169, IL-6, iNOS, TNFα, and CD36 to levels comparable to lean subjects. CD206 mRNA expression was significantly upregulated in PBMC of T2DM while Metformin treatment inhibited CD206 expression levels. Conclusions: These data support the notion that PBMC in circulation in T2DM express different pattern of phenotypic markers than the patterns typically present in M1 and M2 like cells. These phenotypic markers could be representative of metabolically activated macrophages (MMe)-like cells. Metformin, on the other hand, reduces MMe-like cells in circulation.
Collapse
Affiliation(s)
- Mohammed S. Al Dubayee
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah Specialized Children Hospital, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Hind Alayed
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Rana Almansour
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Nora Alqaoud
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Rahaf Alnamlah
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Dana Obeid
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Awad Alshahrani
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Mahmoud M. Zahra
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Amre Nasr
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Ahmad Al-Bawab
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Ahmad Aljada
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- *Correspondence: Ahmad Aljada
| |
Collapse
|
75
|
Fernández-Alfonso MS, Somoza B, Tsvetkov D, Kuczmanski A, Dashwood M, Gil-Ortega M. Role of Perivascular Adipose Tissue in Health and Disease. Compr Physiol 2017; 8:23-59. [PMID: 29357124 DOI: 10.1002/cphy.c170004] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Perivascular adipose tissue (PVAT) is cushion of fat tissue surrounding blood vessels, which is phenotypically different from other adipose tissue depots. PVAT is composed of adipocytes and stromal vascular fraction, constituted by different populations of immune cells, endothelial cells, and adipose-derived stromal cells. It expresses and releases an important number of vasoactive factors with paracrine effects on vascular structure and function. In healthy individuals, these factors elicit a net anticontractile and anti-inflammatory paracrine effect aimed at meeting hemodynamic and metabolic demands of specific organs and regions of the body. Pathophysiological situations, such as obesity, diabetes or hypertension, induce changes in its amount and in the expression pattern of vasoactive factors leading to a PVAT dysfunction in which the beneficial paracrine influence of PVAT is shifted to a pro-oxidant, proinflammatory, contractile, and trophic environment leading to functional and structural cardiovascular alterations and cardiovascular disease. Many different PVATs surrounding a variety of blood vessels have been described and exhibit regional differences. Both protective and deleterious influence of PVAT differs regionally depending on the specific vascular bed contributing to variations in the susceptibility of arteries and veins to vascular disease. PVAT therefore, might represent a novel target for pharmacological intervention in cardiovascular disease. © 2018 American Physiological Society. Compr Physiol 8:23-59, 2018.
Collapse
Affiliation(s)
| | - Beatriz Somoza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | - Dmitry Tsvetkov
- Department of Anestesiology, Perioperative and Pain Medicine, HELIOS Klinikum, Berlin-Buch GmbH, Germany.,Institute of Experimental and Clinical Pharmacology and Toxicology, Department of Pharmacology and Experimental Therapy, Eberhard Karls University Hospitals and Clinics, and Interfaculty Center of Pharmacogenomics and Drug Research, Tübingen, Germany
| | - Artur Kuczmanski
- Department of Anestesiology, Perioperative and Pain Medicine, HELIOS Klinikum, Berlin-Buch GmbH, Germany
| | - Mick Dashwood
- Royal Free Hospital Campus, University College Medical School, London, United Kingdom
| | - Marta Gil-Ortega
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| |
Collapse
|
76
|
Integrated Immunomodulatory Mechanisms through which Long-Chain n-3 Polyunsaturated Fatty Acids Attenuate Obese Adipose Tissue Dysfunction. Nutrients 2017; 9:nu9121289. [PMID: 29186929 PMCID: PMC5748740 DOI: 10.3390/nu9121289] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/14/2017] [Accepted: 11/16/2017] [Indexed: 12/13/2022] Open
Abstract
Obesity is a global health concern with rising prevalence that increases the risk of developing other chronic diseases. A causal link connecting overnutrition, the development of obesity and obesity-associated co-morbidities is visceral adipose tissue (AT) dysfunction, characterized by changes in the cellularity of various immune cell populations, altered production of inflammatory adipokines that sustain a chronic state of low-grade inflammation and, ultimately, dysregulated AT metabolic function. Therefore, dietary intervention strategies aimed to halt the progression of obese AT dysfunction through any of the aforementioned processes represent an important active area of research. In this connection, fish oil-derived dietary long-chain n-3 polyunsaturated fatty acids (PUFA) in the form of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have been demonstrated to attenuate obese AT dysfunction through multiple mechanisms, ultimately affecting AT immune cellularity and function, adipokine production, and metabolic signaling pathways, all of which will be discussed herein.
Collapse
|
77
|
Kralova Lesna I, Petras M, Cejkova S, Kralova A, Fronek J, Janousek L, Thieme F, Tyll T, Poledne R. Cardiovascular disease predictors and adipose tissue macrophage polarization: Is there a link? Eur J Prev Cardiol 2017; 25:328-334. [PMID: 29154680 DOI: 10.1177/2047487317743355] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background The risk of cardiovascular disease is closely connected to adipose tissue inflammation. The links between cardiovascular risk predictors and pro and anti-inflammatory macrophages in human adipose tissue were analysed to gain an insight into the pathophysiology of cardiovascular disease. Design Subcutaneous and visceral adipose tissues were obtained from 79 subjects, 52 living kidney donors (during nephrectomy) and 27 patients with peripheral artery disease (during arterial tree reconstruction). Methods Macrophage subsets were isolated from adipose tissues and analysed by flow cytometry using CD14, CD16, CD36 and CD163 monoclonal antibodies. The mutually adjusted differences of phagocytic pro-inflammatory (CD14 + CD16 + CD36high), anti-inflammatory (CD14 + CD16-CD163+) and transitional subsets of macrophages were analysed in relation to cardiovascular predictors (sex, age, body mass index, smoking, hypercholesterolaemia, hypertension and statin treatment). Results Age, male sex and hypercholesterolaemia were closely positively associated with the phagocytic pro-inflammatory macrophage subset in visceral adipose tissues. Interestingly, the proportion of phagocytic pro-inflammatory macrophages was relevantly decreased by statin therapy. A strong positive association of body mass index to the phagocytic pro-inflammatory subset was found in subcutaneous adipose tissues only. A minor transitional subpopulation, CD14 + CD16 + CD36lowCD163+, increased with age in both adipose tissues. This transitional subpopulation was also negatively associated with obesity and hypercholesterolaemia in visceral adipose tissues. Conclusion An effect of cardiovascular risk predictors on adipose tissue macrophage subpopulations was revealed. Interestingly, while age, male sex and hypercholesterolaemia were connected with the pro-inflammatory macrophage subpopulation in visceral adipose tissues, body mass index had a prominent effect in subcutaneous adipose tissues only. A decreasing effect of statins on these pro-inflammatory macrophages was documented.
Collapse
Affiliation(s)
- Ivana Kralova Lesna
- 1 Laboratory for Atherosclerosis Research, Institute for Clinical and Experimental Medicine, Czech Republic
| | - Marek Petras
- 2 2nd Faculty of Medicine, Charles University, Czech Republic
| | - Sona Cejkova
- 1 Laboratory for Atherosclerosis Research, Institute for Clinical and Experimental Medicine, Czech Republic
| | - Anna Kralova
- 1 Laboratory for Atherosclerosis Research, Institute for Clinical and Experimental Medicine, Czech Republic
| | - Jiri Fronek
- 3 Department of Transplant Surgery, Institute for Clinical and Experimental Medicine, Czech Republic
| | - Libor Janousek
- 3 Department of Transplant Surgery, Institute for Clinical and Experimental Medicine, Czech Republic
| | - Filip Thieme
- 3 Department of Transplant Surgery, Institute for Clinical and Experimental Medicine, Czech Republic
| | - Tomas Tyll
- 4 Anaesthesiology, Resuscitation and Intensive Care Unit, Military University Hospital, Czech Republic
| | - Rudolf Poledne
- 1 Laboratory for Atherosclerosis Research, Institute for Clinical and Experimental Medicine, Czech Republic
| |
Collapse
|
78
|
Extracellular matrix remodeling and matrix metalloproteinase inhibition in visceral adipose during weight cycling in mice. Exp Cell Res 2017; 359:431-440. [DOI: 10.1016/j.yexcr.2017.08.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 07/20/2017] [Accepted: 08/17/2017] [Indexed: 11/23/2022]
|
79
|
CINKAJZLOVÁ A, LACINOVÁ Z, KLOUČKOVÁ J, KAVÁLKOVÁ P, TRACHTA P, KOSÁK M, KRÁTKÝ J, KASALICKÝ M, DOLEŽALOVÁ K, MRÁZ M, HALUZÍK M. An Alternatively Activated Macrophage Marker CD163 in Severely Obese Patients: the Influence of Very Low-Calorie Diet and Bariatric Surgery. Physiol Res 2017; 66:641-652. [DOI: 10.33549/physiolres.933522] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
CD163 is a marker of macrophages with anti-inflammatory properties and its soluble form (sCD163) is considered a prognostic predictor of several diseases including type 2 diabetes mellitus (T2DM). We explored sCD163 levels at baseline and after very low-calorie diet (VLCD) or bariatric surgery in 32 patients with obesity (20 undergoing VLCD and 12 bariatric surgery), 32 obese patients with T2DM (22 undergoing VLCD and 10 bariatric surgery), and 19 control subjects. We also assessed the changes of CD163 positive cells of monocyte-macrophage lineage in peripheral blood and subcutaneous adipose tissue (SAT) in subset of patients. Plasma sCD163 levels were increased in obese and T2DM subjects relative to control subjects (467.2±40.2 and 513.8±37.0 vs. 334.4±24.8 ng/ml, p=0.001) and decreased after both interventions. Obesity decreased percentage of CD163+CD14+ monocytes in peripheral blood compared to controls (78.9±1.48 vs. 86.2±1.31 %, p=0.003) and bariatric surgery decreased CD163+CD14+HLA-DR+ macrophages in SAT (19.4±2.32 vs. 11.3±0.90 %, p=0.004). Our data suggest that increased basal sCD163 levels are related to obesity and its metabolic complications. On the contrary, sCD163 or CD163 positive cell changes do not precisely reflect metabolic improvements after weight loss.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - M. HALUZÍK
- Centre for Experimental Medicine and Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
80
|
Caputo T, Gilardi F, Desvergne B. From chronic overnutrition to metaflammation and insulin resistance: adipose tissue and liver contributions. FEBS Lett 2017; 591:3061-3088. [DOI: 10.1002/1873-3468.12742] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 06/28/2017] [Accepted: 07/02/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Tiziana Caputo
- Center for Integrative Genomics; Lausanne Faculty of Biology and Medicine; University of Lausanne; Switzerland
| | - Federica Gilardi
- Center for Integrative Genomics; Lausanne Faculty of Biology and Medicine; University of Lausanne; Switzerland
| | - Béatrice Desvergne
- Center for Integrative Genomics; Lausanne Faculty of Biology and Medicine; University of Lausanne; Switzerland
| |
Collapse
|
81
|
Keuper M, Sachs S, Walheim E, Berti L, Raedle B, Tews D, Fischer-Posovszky P, Wabitsch M, Hrabě de Angelis M, Kastenmüller G, Tschöp MH, Jastroch M, Staiger H, Hofmann SM. Activated macrophages control human adipocyte mitochondrial bioenergetics via secreted factors. Mol Metab 2017; 6:1226-1239. [PMID: 29031722 PMCID: PMC5641636 DOI: 10.1016/j.molmet.2017.07.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 07/11/2017] [Accepted: 07/13/2017] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Obesity-associated WAT inflammation is characterized by the accumulation and local activation of macrophages (MΦs), and recent data from mouse studies suggest that macrophages are modifiers of adipocyte energy metabolism and mitochondrial function. As mitochondrial dysfunction has been associated with obesity and the metabolic syndrome in humans, herein we aimed to delineate how human macrophages may affect energy metabolism of white adipocytes. METHODS Human adipose tissue gene expression analysis for markers of macrophage activation and tissue inflammation (CD11c, CD40, CD163, CD206, CD80, MCP1, TNFα) in relationship to mitochondrial complex I (NDUFB8) and complex III (UQCRC2) was performed on subcutaneous WAT of 24 women (BMI 20-61 kg/m2). Guided by these results, the impact of secreted factors of LPS/IFNγ- and IL10/TGFβ-activated human macrophages (THP1, primary blood-derived) on mitochondrial function in human subcutaneous white adipocytes (SGBS, primary) was determined by extracellular flux analysis (Seahorse technology) and gene/protein expression. RESULTS Stepwise regression analysis of human WAT gene expression data revealed that a linear combination of CD40 and CD163 was the strongest predictor for mitochondrial complex I (NDUFB8) and complex III (UQCRC2) levels, independent of BMI. IL10/TGFβ-activated MΦs displayed high CD163 and low CD40 expression and secreted factors that decreased UQCRC2 gene/protein expression and ATP-linked respiration in human white adipocytes. In contrast, LPS/IFNγ-activated MΦs showed high CD40 and low CD163 expression and secreted factors that enhanced adipocyte mitochondrial activity resulting in a total difference of 37% in ATP-linked respiration of white adipocytes (p = 0.0024) when comparing the effect of LPS/IFNγ- vs IL10/TGFβ-activated MΦs. CONCLUSION Our data demonstrate that macrophages modulate human adipocyte energy metabolism via an activation-dependent paracrine mechanism.
Collapse
Affiliation(s)
- Michaela Keuper
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute of Experimental Genetics, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Neuherberg, Germany.
| | - Stephan Sachs
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Ellen Walheim
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute for Diabetes and Obesity, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Lucia Berti
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
| | - Bernhard Raedle
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute of Experimental Genetics, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Daniel Tews
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Pamela Fischer-Posovszky
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center, Ulm, Germany
| | - Martin Hrabě de Angelis
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute of Experimental Genetics, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Neuherberg, Germany; Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, Alte Akademie 8, 85354 Freising, Germany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Matthias H Tschöp
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute for Diabetes and Obesity, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Martin Jastroch
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute for Diabetes and Obesity, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Harald Staiger
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany; Institute of Pharmaceutical Sciences, Department of Pharmacy and Biochemistry, Eberhard Karls University Tübingen, Germany
| | - Susanna M Hofmann
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Medizinische Klinik und Poliklinik IV, Klinikum der LMU, 80336 München, Germany
| |
Collapse
|
82
|
Breasson L, Becattini B, Sardi C, Molinaro A, Zani F, Marone R, Botindari F, Bousquenaud M, Ruegg C, Wymann MP, Solinas G. PI3Kγ activity in leukocytes promotes adipose tissue inflammation and early-onset insulin resistance during obesity. Sci Signal 2017; 10:10/488/eaaf2969. [PMID: 28720716 DOI: 10.1126/scisignal.aaf2969] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The phosphoinositide 3-kinase γ (PI3Kγ) plays a major role in leukocyte recruitment during acute inflammation and has been proposed to inhibit classical macrophage activation by driving immunosuppressive gene expression. PI3Kγ plays an important role in diet-induced obesity and insulin resistance. In seeking to determine the underlying molecular mechanisms, we showed that PI3Kγ action in high-fat diet-induced inflammation and insulin resistance depended largely on its role in the control of adiposity, which was due to PI3Kγ activity in a nonhematopoietic cell type. However, PI3Kγ activity in leukocytes was required for efficient neutrophil recruitment to adipose tissue. Neutrophil recruitment was correlated with proinflammatory gene expression in macrophages in adipose tissue, which triggered insulin resistance early during the development of obesity. Our data challenge the concept that PI3Kγ is a general suppressor of classical macrophage activation and indicate that PI3Kγ controls macrophage gene expression by non-cell-autonomous mechanisms, the outcome of which is context-dependent.
Collapse
Affiliation(s)
- Ludovic Breasson
- Department of Medicine/Physiology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Barbara Becattini
- Department of Medicine/Physiology, University of Fribourg, 1700 Fribourg, Switzerland
| | | | | | - Fabio Zani
- Department of Medicine/Physiology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Romina Marone
- Cancer and Immunobiology Laboratory, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Fabrizio Botindari
- Cancer and Immunobiology Laboratory, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Mélanie Bousquenaud
- Department of Medicine/Physiology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Curzio Ruegg
- Department of Medicine/Physiology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Matthias P Wymann
- Cancer and Immunobiology Laboratory, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland.
| | - Giovanni Solinas
- Department of Medicine/Physiology, University of Fribourg, 1700 Fribourg, Switzerland.
| |
Collapse
|
83
|
Pathogenic Role of IL-17-Producing Immune Cells in Obesity, and Related Inflammatory Diseases. J Clin Med 2017; 6:jcm6070068. [PMID: 28708082 PMCID: PMC5532576 DOI: 10.3390/jcm6070068] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 07/03/2017] [Accepted: 07/04/2017] [Indexed: 12/14/2022] Open
Abstract
Obesity is associated with low-grade chronic inflammation. Indeed, adipose tissues (AT) in obese individuals are the former site of progressive infiltration by pro-inflammatory immune cells, which together with increased inflammatory adipokine secretion induce adipocyte insulin resistance. IL-17-producing T (Th17) cells are part of obese AT infiltrating cells, and are likely to be promoted by adipose tissue-derived mesenchymal stem cells, as previously reported by our team. Whereas Th17 cell are physiologically implicated in the neutralization of fungal and bacterial pathogens through activation of neutrophils, they may also play a pivotal role in the onset and/or progression of chronic inflammatory diseases, or cancer, in which obesity is recognized as a risk factor. In this review, we will highlight the pathogenic role of IL-17A producing cells in the mechanisms leading to inflammation in obesity and to progression of obesity-related inflammatory diseases.
Collapse
|
84
|
Chimin P, Andrade ML, Belchior T, Paschoal VA, Magdalon J, Yamashita AS, Castro É, Castoldi A, Chaves-Filho AB, Yoshinaga MY, Miyamoto S, Câmara NO, Festuccia WT. Adipocyte mTORC1 deficiency promotes adipose tissue inflammation and NLRP3 inflammasome activation via oxidative stress and de novo ceramide synthesis. J Lipid Res 2017; 58:1797-1807. [PMID: 28679588 DOI: 10.1194/jlr.m074518] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 06/13/2017] [Indexed: 12/20/2022] Open
Abstract
Mechanistic target of rapamycin complex (mTORC)1 activity is increased in adipose tissue of obese insulin-resistant mice, but its role in the regulation of tissue inflammation is unknown. Herein, we investigated the effects of adipocyte mTORC1 deficiency on adipose tissue inflammation and glucose homeostasis. For this, mice with adipocyte raptor deletion and controls fed a chow or a high-fat diet were evaluated for body mass, adiposity, glucose homeostasis, and adipose tissue inflammation. Despite reducing adiposity, adipocyte mTORC1 deficiency promoted hepatic steatosis, insulin resistance, and adipose tissue inflammation (increased infiltration of macrophages, neutrophils, and B lymphocytes; crown-like structure density; TNF-α, interleukin (IL)-6, and monocyte chemoattractant protein 1 expression; IL-1β protein content; lipid peroxidation; and de novo ceramide synthesis). The anti-oxidant, N-acetylcysteine, partially attenuated, whereas treatment with de novo ceramide synthesis inhibitor, myriocin, completely blocked adipose tissue inflammation and nucleotide oligomerization domain-like receptor pyrin domain-containing 3 (NLRP3)-inflammasome activation, but not hepatic steatosis and insulin resistance induced by adipocyte raptor deletion. Rosiglitazone treatment, however, completely abrogated insulin resistance induced by adipocyte raptor deletion. In conclusion, adipocyte mTORC1 deficiency induces adipose tissue inflammation and NLRP3-inflammasome activation by promoting oxidative stress and de novo ceramide synthesis. Such adipose tissue inflammation, however, is not an underlying cause of the insulin resistance displayed by these mice.
Collapse
Affiliation(s)
- Patricia Chimin
- Departments of Physiology and Biophysics University of Sao Paulo, Sao Paulo, Brazil 05508000.,Department of Physical Education, Physical Education and Sports Center, Londrina State University, Parana, Brazil 86051-990
| | - Maynara L Andrade
- Departments of Physiology and Biophysics University of Sao Paulo, Sao Paulo, Brazil 05508000
| | - Thiago Belchior
- Departments of Physiology and Biophysics University of Sao Paulo, Sao Paulo, Brazil 05508000
| | - Vivian A Paschoal
- Departments of Physiology and Biophysics University of Sao Paulo, Sao Paulo, Brazil 05508000
| | - Juliana Magdalon
- Departments of Physiology and Biophysics University of Sao Paulo, Sao Paulo, Brazil 05508000
| | - Alex S Yamashita
- Departments of Physiology and Biophysics University of Sao Paulo, Sao Paulo, Brazil 05508000
| | - Érique Castro
- Departments of Physiology and Biophysics University of Sao Paulo, Sao Paulo, Brazil 05508000
| | - Angela Castoldi
- Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil 05508000
| | - Adriano B Chaves-Filho
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Sao Paulo, Brazil 05508000
| | - Marcos Y Yoshinaga
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Sao Paulo, Brazil 05508000
| | - Sayuri Miyamoto
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Sao Paulo, Brazil 05508000
| | - Niels O Câmara
- Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil 05508000
| | - William T Festuccia
- Departments of Physiology and Biophysics University of Sao Paulo, Sao Paulo, Brazil 05508000
| |
Collapse
|
85
|
Gaborit B, Sengenes C, Ancel P, Jacquier A, Dutour A. Role of Epicardial Adipose Tissue in Health and Disease: A Matter of Fat? Compr Physiol 2017. [PMID: 28640452 DOI: 10.1002/cphy.c160034] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Epicardial adipose tissue (EAT) is a small but very biologically active ectopic fat depot that surrounds the heart. Given its rapid metabolism, thermogenic capacity, unique transcriptome, secretory profile, and simply measurability, epicardial fat has drawn increasing attention among researchers attempting to elucidate its putative role in health and cardiovascular diseases. The cellular crosstalk between epicardial adipocytes and cells of the vascular wall or myocytes is high and suggests a local role for this tissue. The balance between protective and proinflammatory/profibrotic cytokines, chemokines, and adipokines released by EAT seem to be a key element in atherogenesis and could represent a future therapeutic target. EAT amount has been found to predict clinical coronary outcomes. EAT can also modulate cardiac structure and function. Its amount has been associated with atrial fibrillation, coronary artery disease, and sleep apnea syndrome. Conversely, a beiging fat profile of EAT has been identified. In this review, we describe the current state of knowledge regarding the anatomy, physiology and pathophysiological role of EAT, and the factors more globally leading to ectopic fat development. We will also highlight the most recent findings on the origin of this ectopic tissue, and its association with cardiac diseases. © 2017 American Physiological Society. Compr Physiol 7:1051-1082, 2017.
Collapse
Affiliation(s)
- Bénédicte Gaborit
- NORT, Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France.,Endocrinology Metabolic Diseases, and Nutrition Department, Pole ENDO, APHM, Aix-Marseille Univ, Marseille, France
| | - Coralie Sengenes
- STROMALab, Université de Toulouse, EFS, ENVT, Inserm U1031, ERL CNRS 5311, CHU Rangueil, Toulouse, France
| | - Patricia Ancel
- NORT, Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France
| | - Alexis Jacquier
- CNRS UMR 7339, Centre de Résonance Magnétique Biologique et Médicale (CRMBM), Marseille, France.,Radiology department, CHU La Timone, Marseille, France
| | - Anne Dutour
- NORT, Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France.,Endocrinology Metabolic Diseases, and Nutrition Department, Pole ENDO, APHM, Aix-Marseille Univ, Marseille, France
| |
Collapse
|
86
|
Harasymowicz NS, Clement ND, Azfer A, Burnett R, Salter DM, Simpson AHWR. Regional Differences Between Perisynovial and Infrapatellar Adipose Tissue Depots and Their Response to Class II and Class III Obesity in Patients With Osteoarthritis. Arthritis Rheumatol 2017; 69:1396-1406. [PMID: 28320058 DOI: 10.1002/art.40102] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 03/14/2017] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Obesity is associated with an increased risk of developing osteoarthritis (OA), which is postulated to be secondary to adipose tissue-dependent inflammation. Periarticular adipose tissue depots are present in synovial joints, but the association of this tissue with OA has not been extensively explored. The aim of this study was to investigate differences in local adipose tissue depots in knees with OA and characterize the changes related to class II and class III obesity in patients with end-stage knee OA. METHODS Synovium and the infrapatellar fat pad (IPFP) were collected during total knee replacement from 69 patients with end-stage OA. Histologic changes, changes in gene and protein expression of adiponectin, peroxisome proliferator-activated receptor γ (PPARγ), and Toll-like receptor 4 (TLR-4), and immune cell infiltration into the adipose tissue were investigated. RESULTS IPFP and synovium adipose tissue depots differed significantly and were influenced by the patient's body mass index. Compared to adipocytes from the IPFP and synovium of lean patients, adipocytes from the IPFP of obese patients were significantly larger and the synovium of obese patients displayed marked fibrosis, increased macrophage infiltration, and higher levels of TLR4 gene expression. The adipose-related markers PPARγ in the IPFP and adiponectin and PPARγ in the synovium were expressed at lower levels in obese patients compared to lean patients. Furthermore, there were increased numbers of CD45+ hematopoietic cells, CD45+CD14+ total macrophages, and CD14+CD206+ M2-type macrophages in both the IPFP and synovial tissue of obese patients. CONCLUSION These differences suggest that IPFP and synovium may contain 2 different white adipose tissue depots and support the theory of inflammation-induced OA in patients with class II or III obesity. These findings warrant further investigation as a potentially reversible, or at least suppressible, cause of OA in obese patients.
Collapse
|
87
|
Rahamon SK, Fabian UA, Charles-Davies MA, Olaniyi JA, Fasanmade AA, Akinlade KS, Oyewole OE, Owolabi MO, Adebusuyi JR, Hassan OO, Ajobo BM, Ebesunun MO, Adigun K, Popoola OO, Omiyale W, Arinola OG, Agbedana EO. Changes in mediators of inflammation and pro-thrombosis after 12 months of dietary modification in adults with metabolic syndrome. Afr Health Sci 2017; 17:453-462. [PMID: 29062341 DOI: 10.4314/ahs.v17i2.20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE This study evaluated the effects of a 12-month dietary modification on indices of inflammation and pro-thrombosis in adults with metabolic syndrome (MS). MATERIALS AND METHODS This longitudinal study involved 252 adults with MS recruited from the Bodija market, Ibadan and its environs. Participants were placed on 20%, 30% and 50% calories obtained from protein, total fat and carbohydrate respectively and were followed up monthly for 12 months. Anthropometry and blood pressure were measured using standard methods. Fasting plasma glucose (FPG), total cholesterol (TC), triglycerides (TG), high density lipoprotein-cholesterol (HDL-C), fibrinogen, plasminogen activator inhibitor-1 (PAI-1)], interleukin-6 (IL-6) and interleukin-10 (IL-10) were measured using spectrophotometric methods and ELISA as appropriate. Data was analysed using ANCOVA, Student's t-test, Mann-Whitney U and Wilcoxon signed-rank tests. P-values less than 0.05 were considered significant. RESULTS After 6 months of dietary modification, there was a significant reduction in waist circumference (WC), while the levels of HDL-C, fibrinogen and PAI-1 were significantly increased when compared with the corresponding baseline values. However, WC and fibrinogen reduced significantly, while HDL-C and IL-10 significantly increased after 12 months of dietary modification as compared with the respective baseline values. CONCLUSION Long-term regular dietary modification may be beneficial in ameliorating inflammation and pro-thrombosis in metabolic syndrome.
Collapse
Affiliation(s)
- S K Rahamon
- Department of Chemical Pathology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - U A Fabian
- Department of Chemical Pathology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - M A Charles-Davies
- Department of Chemical Pathology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - J A Olaniyi
- Department of Haematology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - A A Fasanmade
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - K S Akinlade
- Department of Chemical Pathology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - O E Oyewole
- Department of Health Promotion and Education, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - M O Owolabi
- Department of Haematology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - J R Adebusuyi
- Department of Medical Social Services, University College Hospital, Ibadan, Nigeria
| | - O O Hassan
- Department of Medical Social Services, University College Hospital, Ibadan, Nigeria
| | - B M Ajobo
- Dietetics Department, University College Hospital, Ibadan, Nigeria
| | - M O Ebesunun
- Department of Chemical Pathology, College of Health Sciences, Olabisi Onabanjo University, Ago-Iwoye, Nigeria
| | - K Adigun
- Department of Family Medicine, University College Hospital, Ibadan, Nigeria
| | - O O Popoola
- Department of Chemical Pathology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - W Omiyale
- Department of Chemical Pathology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - O G Arinola
- Department of Chemical Pathology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - E O Agbedana
- Department of Chemical Pathology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
88
|
Cinkajzlová A, Mráz M, Haluzík M. Lymphocytes and macrophages in adipose tissue in obesity: markers or makers of subclinical inflammation? PROTOPLASMA 2017; 254:1219-1232. [PMID: 28150048 DOI: 10.1007/s00709-017-1082-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 01/25/2017] [Indexed: 05/17/2023]
Abstract
Obesity is accompanied by the development of chronic low-grade inflammation in adipose tissue. The presence of chronic inflammatory response along with metabolically harmful factors released by adipose tissue into the circulation is associated with several metabolic complications of obesity such as type 2 diabetes mellitus or accelerated atherosclerosis. The present review is focused on macrophages and lymphocytes and their possible role in low-grade inflammation in fat. Both macrophages and lymphocytes respond to obesity-induced adipocyte hypertrophy by their migration into adipose tissue. After activation and differentiation, they contribute to the development of local inflammatory response and modulation of endocrine function of adipose tissue. Despite intensive research, the exact role of lymphocytes and macrophages within adipose tissue is only partially clarified and various data obtained by different approaches bring ambiguous information with respect to their polarization and cytokine production. Compared to immunocompetent cells, the role of adipocytes in the obesity-related adipose tissue inflammation is often underestimated despite their abundant production of factors with immunomodulatory actions such as cytokines or adipokines such as leptin, adiponektin, and others. In summary, conflicting evidence together with only partial correlation of in vitro findings with true in vivo situation due to great heterogeneity and molecular complexity of tissue environment calls for intensive research in this rapidly evolving and important area.
Collapse
Affiliation(s)
- Anna Cinkajzlová
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University in Prague and General University Hospital, Prague, Czech Republic
- Centre of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Miloš Mráz
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University in Prague and General University Hospital, Prague, Czech Republic
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Martin Haluzík
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University in Prague and General University Hospital, Prague, Czech Republic.
- Centre of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
- Department of Obesitology, Institute of Endocrinology, Prague, Czech Republic.
| |
Collapse
|
89
|
Human adipose tissue accumulation is associated with pro-inflammatory changes in subcutaneous rather than visceral adipose tissue. Nutr Diabetes 2017; 7:e264. [PMID: 28394364 PMCID: PMC5436095 DOI: 10.1038/nutd.2017.15] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/22/2017] [Accepted: 02/26/2017] [Indexed: 12/12/2022] Open
Abstract
The importance of the involvement of adipose tissue macrophage subpopulations in obesity-related disorders is well known from different animal models, but human data are scarcer. Subcutaneous (n=44) and visceral (n=52) adipose tissues of healthy living kidney donors were obtained during living donor nephrectomy. Stromal vascular fractions were isolated and analysed by flow cytometry using CD14, CD16, CD36 and CD163 antibodies. Total macrophage numbers in subcutaneous adipose tissue increased (P=0.02) with body mass index (BMI), with a similar increase seen in the proportion of phagocytic CD14+CD16+CD36high macrophages (P<0.01). On the other hand, there was an inverse correlation between anti-inflammatory CD14+CD16-CD163+ macrophages (P<0.05) and BMI. These correlations disappeared after excluding obese subjects (BMI ⩾30 kg m-2) from the analysis. Interestingly, none of these subpopulations were significantly related to BMI in visceral adipose tissue. Obesity per se is associated with distinct, highly phagocytic macrophage accumulation in human subcutaneous adipose tissue.
Collapse
|
90
|
Tashiro K, Feng J, Wu SH, Mashiko T, Kanayama K, Narushima M, Uda H, Miyamoto S, Koshima I, Yoshimura K. Pathological changes of adipose tissue in secondary lymphoedema. Br J Dermatol 2017; 177:158-167. [PMID: 28000916 DOI: 10.1111/bjd.15238] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND The pathophysiology of lymphoedema is poorly understood. Current treatment options include compression therapy, resection, liposuction and lymphatic microsurgery, but determining the optimal treatment approach for each patient remains challenging. OBJECTIVES We characterized skin and adipose tissue alterations in the setting of secondary lymphoedema. METHODS Morphological and histopathological evaluations were conducted for 70 specimens collected from 26 female patients with lower-extremity secondary lymphoedema following surgical intervention for gynaecological cancers. Indocyanine green lymphography was performed for each patient to assess lymphoedema severity. RESULTS Macroscopic and ultrasound findings revealed that lymphoedema adipose tissue had larger lobules of adipose tissue, with these lobules surrounded by thick collagen fibres and interstitial lymphatic fluid. In lymphoedema specimens, adipocytes displayed hypertrophic changes and more collagen fibre deposits when examined using electron microscopy, whole-mount staining and immunohistochemistry. The number of capillary lymphatic channels was also found to be increased in the dermis of lymphoedema limbs. Crown-like structures (dead adipocytes surrounded by M1 macrophages) were less frequently seen in lymphoedema samples. Flow cytometry revealed that, among the cellular components of adipose tissue, adipose-derived stem/stromal cells and M2 macrophages were decreased in number in lymphoedema adipose tissue compared with normal controls. CONCLUSIONS These findings suggest that long-term lymphatic volume overload can induce chronic tissue inflammation, progressive fibrosis, impaired homeostasis, altered remodelling of adipose tissue, impaired regenerative capacity and immunological dysfunction. Further elucidation of the pathophysiological mechanisms underlying lymphoedema will lead to more reliable therapeutic strategies.
Collapse
Affiliation(s)
- K Tashiro
- Department of Plastic Surgery, University of Tokyo School of Medicine, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan.,Department of Plastic Surgery, National Cancer Center, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - J Feng
- Department of Plastic Surgery, University of Tokyo School of Medicine, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan.,Department of Plastic Surgery, Southern Medical University Nanfang Hospital, 1838 Guangzhou South Ave., Guangzhou, 510515, China
| | - S-H Wu
- Department of Plastic Surgery, University of Tokyo School of Medicine, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - T Mashiko
- Department of Plastic Surgery, University of Tokyo School of Medicine, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - K Kanayama
- Department of Plastic Surgery, University of Tokyo School of Medicine, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - M Narushima
- Department of Plastic Surgery, University of Tokyo School of Medicine, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - H Uda
- Department of Plastic Surgery, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - S Miyamoto
- Department of Plastic Surgery, National Cancer Center, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - I Koshima
- Department of Plastic Surgery, University of Tokyo School of Medicine, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - K Yoshimura
- Department of Plastic Surgery, University of Tokyo School of Medicine, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan.,Department of Plastic Surgery, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| |
Collapse
|
91
|
Silva KR, Côrtes I, Liechocki S, Carneiro JRI, Souza AAP, Borojevic R, Maya-Monteiro CM, Baptista LS. Characterization of stromal vascular fraction and adipose stem cells from subcutaneous, preperitoneal and visceral morbidly obese human adipose tissue depots. PLoS One 2017; 12:e0174115. [PMID: 28323901 PMCID: PMC5360317 DOI: 10.1371/journal.pone.0174115] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 03/04/2017] [Indexed: 12/14/2022] Open
Abstract
Background/Objectives The pathological condition of obesity is accompanied by a dysfunctional adipose tissue. We postulate that subcutaneous, preperitoneal and visceral obese abdominal white adipose tissue depots could have stromal vascular fractions (SVF) with distinct composition and adipose stem cells (ASC) that would differentially account for the pathogenesis of obesity. Methods In order to evaluate the distribution of SVF subpopulations, samples of subcutaneous, preperitoneal and visceral adipose tissues from morbidly obese women (n = 12, BMI: 46.2±5.1 kg/m2) were collected during bariatric surgery, enzymatically digested and analyzed by flow cytometry (n = 12). ASC from all depots were evaluated for morphology, surface expression, ability to accumulate lipid after induction and cytokine secretion (n = 3). Results A high content of preadipocytes was found in the SVF of subcutaneous depot (p = 0.0178). ASC from the three depots had similar fibroblastoid morphology with a homogeneous expression of CD34, CD146, CD105, CD73 and CD90. ASC from the visceral depot secreted the highest levels of IL-6, MCP-1 and G-CSF (p = 0.0278). Interestingly, preperitoneal ASC under lipid accumulation stimulus showed the lowest levels of all the secreted cytokines, except for adiponectin that was enhanced (p = 0.0278). Conclusions ASC from preperitoneal adipose tissue revealed the less pro-inflammatory properties, although it is an internal adipose depot. Conversely, ASC from visceral adipose tissue are the most pro-inflammatory. Therefore, ASC from subcutaneous, visceral and preperitoneal adipose depots could differentially contribute to the chronic inflammatory scenario of obesity.
Collapse
Affiliation(s)
- Karina Ribeiro Silva
- Post-graduation Program of Medical Clinics, Federal University of Rio de Janeiro – UFRJ, Rio de Janeiro, Brazil
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology – INMETRO, Duque de Caxias, Rio de Janeiro, Brazil
- * E-mail: (KRS); (LSB)
| | - Isis Côrtes
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology – INMETRO, Duque de Caxias, Rio de Janeiro, Brazil
| | - Sally Liechocki
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation - FIOCRUZ, Rio de Janeiro, Brazil
| | - João Regis Ivar Carneiro
- Nutrology Department, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro - UFRJ, Rio de Janeiro, Brazil
| | - Antônio Augusto Peixoto Souza
- Surgery Department, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Radovan Borojevic
- Post-graduation Program of Medical Clinics, Federal University of Rio de Janeiro – UFRJ, Rio de Janeiro, Brazil
- Center of Regenerative Medicine, Petrópolis Faculty of Medicine – FASE, Petrópolis, Rio de Janeiro, Brazil
| | | | - Leandra Santos Baptista
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology – INMETRO, Duque de Caxias, Rio de Janeiro, Brazil
- Nucleus of Multidisciplinary Research in Xerem - Biology (Numpex-Bio), Federal University of Rio de Janeiro - Xerém, Duque de Caxias, Rio de Janeiro, Brazil
- * E-mail: (KRS); (LSB)
| |
Collapse
|
92
|
Carneiro PJ, Clevelario AL, Padilha GA, Silva JD, Kitoko JZ, Olsen PC, Capelozzi VL, Rocco PRM, Cruz FF. Bosutinib Therapy Ameliorates Lung Inflammation and Fibrosis in Experimental Silicosis. Front Physiol 2017; 8:159. [PMID: 28360865 PMCID: PMC5350127 DOI: 10.3389/fphys.2017.00159] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 03/01/2017] [Indexed: 12/16/2022] Open
Abstract
Silicosis is an occupational lung disease for which no effective therapy exists. We hypothesized that bosutinib, a tyrosine kinase inhibitor, might ameliorate inflammatory responses, attenuate pulmonary fibrosis, and thus improve lung function in experimental silicosis. For this purpose, we investigated the potential efficacy of bosutinib in the treatment of experimental silicosis induced in C57BL/6 mice by intratracheal administration of silica particles. After 15 days, once disease was established, animals were randomly assigned to receive DMSO or bosutinib (1 mg/kg/dose in 0.1 mL 1% DMSO) by oral gavage, twice daily for 14 days. On day 30, lung mechanics and morphometry, total and differential cell count in alveolar septa and granuloma, levels of interleukin (IL)-1β, tumor necrosis factor (TNF)-α, interferon (IFN)-γ, IL-4, transforming growth factor (TGF)-β, and vascular endothelial growth factor in lung homogenate, M1 and M2 macrophages, total leukocytes, and T cells in BALF, lymph nodes, and thymus, and collagen fiber content in alveolar septa and granuloma were analyzed. In a separate in vitro experiment, RAW264.7 macrophages were exposed to silica particles in the presence or absence of bosutinib. After 24 h, gene expressions of arginase-1, IL-10, IL-12, inducible nitric oxide synthase (iNOS), metalloproteinase (MMP)-9, tissue inhibitor of metalloproteinase (TIMP)-1, and caspase-3 were evaluated. In vivo, in silicotic animals, bosutinib, compared to DMSO, decreased: (1) fraction area of collapsed alveoli, (2) size and number of granulomas, and mononuclear cell granuloma infiltration; (3) IL-1β, TNF-α, IFN-γ, and TGF-β levels in lung homogenates, (4) collagen fiber content in lung parenchyma, and (5) viscoelastic pressure and static lung elastance. Bosutinib also reduced M1 cell counts while increasing M2 macrophage population in both lung parenchyma and granulomas. Total leukocyte, regulatory T, CD4+, and CD8+ cell counts in the lung-draining lymph nodes also decreased with bosutinib therapy without affecting thymus cellularity. In vitro, bosutinib led to a decrease in IL-12 and iNOS and increase in IL-10, arginase-1, MMP-9, and TIMP-1. In conclusion, in the current model of silicosis, bosutinib therapy yielded beneficial effects on lung inflammation and remodeling, therefore resulting in lung mechanics improvement. Bosutinib may hold promise for silicosis; however, further studies are required.
Collapse
Affiliation(s)
- Priscila J Carneiro
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Amanda L Clevelario
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Gisele A Padilha
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Johnatas D Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Jamil Z Kitoko
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de Janeiro, Brazil; Laboratory of Clinical Bacteriology and Immunology, Department of Toxicological and Clinical Analysis, School of Pharmacy, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Priscilla C Olsen
- Laboratory of Clinical Bacteriology and Immunology, Department of Toxicological and Clinical Analysis, School of Pharmacy, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Vera L Capelozzi
- Laboratory of Pulmonary Genomics, Department of Pathology, School of Medicine, University of São Paulo São Paulo, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Fernanda F Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| |
Collapse
|
93
|
Hui X, Zhang M, Gu P, Li K, Gao Y, Wu D, Wang Y, Xu A. Adipocyte SIRT1 controls systemic insulin sensitivity by modulating macrophages in adipose tissue. EMBO Rep 2017; 18:645-657. [PMID: 28270525 DOI: 10.15252/embr.201643184] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 02/05/2017] [Accepted: 02/07/2017] [Indexed: 11/09/2022] Open
Abstract
Adipose tissue inflammation, characterized by augmented infiltration and altered polarization of macrophages, contributes to insulin resistance and its associated metabolic diseases. The NAD+-dependent deacetylase SIRT1 serves as a guardian against metabolic disorders in multiple tissues. To dissect the roles of SIRT1 in adipose tissues, metabolic phenotypes of mice with selective ablation of SIRT1 in adipocytes and myeloid cells were monitored. Compared to myeloid-specific SIRT1 depletion, mice with adipocyte-selective deletion of SIRT1 are more susceptible to diet-induced insulin resistance. The phenotypic changes in adipocyte-selective SIRT1 knockout mice are associated with an increased number of adipose-resident macrophages and their polarization toward the pro-inflammatory M1 subtype. Mechanistically, SIRT1 in adipocytes modulates expression and secretion of several adipokines, including adiponectin, MCP-1, and interleukin 4, which in turn alters recruitment and polarization of the macrophages in adipose tissues. In adipocytes, SIRT1 deacetylates the transcription factor NFATc1 and thereby enhances the binding of NFATc1 to the Il4 gene promoter. These findings suggest that adipocyte SIRT1 controls systemic glucose homeostasis and insulin sensitivity via the cross talk with adipose-resident macrophages.
Collapse
Affiliation(s)
- Xiaoyan Hui
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Mingliang Zhang
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Ping Gu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Hong Kong, China.,Department of Endocrinology, School of Medicine, Nanjing University Nanjing General Hospital of Nanjing Military Command, Nanjing, China
| | - Kuai Li
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yuan Gao
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Donghai Wu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China .,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China .,Department of Medicine, The University of Hong Kong, Hong Kong, China.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
94
|
Braune J, Weyer U, Hobusch C, Mauer J, Brüning JC, Bechmann I, Gericke M. IL-6 Regulates M2 Polarization and Local Proliferation of Adipose Tissue Macrophages in Obesity. THE JOURNAL OF IMMUNOLOGY 2017; 198:2927-2934. [PMID: 28193830 DOI: 10.4049/jimmunol.1600476] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 01/20/2017] [Indexed: 12/12/2022]
Abstract
Obesity is associated with chronic low-grade inflammation of adipose tissue (AT) and an increase of AT macrophages (ATMs) that is linked to the onset of type 2 diabetes. We have recently shown that focal sites of inflammation around dying adipocytes, so-called crown-like structures, exhibit a unique microenvironment for macrophage proliferation. Interestingly, locally proliferating macrophages were not classically activated (M1), but they exhibited a rather alternatively activated (M2) immune phenotype. In this study, we established organotypic cell cultures of AT explants to study the impact of cytokine treatment on local ATM proliferation, without the bias of early monocyte recruitment. We show that exposure of AT to Th2 cytokines, such as IL-4, IL-13, and GM-CSF, stimulates ATM proliferation, whereas Th1 cytokines, such as TNF-α, inhibit local ATM proliferation. Furthermore, AT from obese mice exhibits an increased sensitivity to IL-4 stimulation, indicated by an increased phosphorylation of STAT6. In line with this, gene expression of the IL-4 receptor (Il4ra) and its ligand IL-13 are elevated in AT of obese C57BL/6 mice. Most importantly, Il4ra expression and susceptibility to IL-4 or IL-13 treatment depend on IL-6 signaling, which seems to be the underlying mechanism of local ATM proliferation in obesity. We conclude that IL-6 acts as a Th2 cytokine in obesity by stimulating M2 polarization and local ATM proliferation, presumably due to upregulation of the IL-4 receptor α.
Collapse
Affiliation(s)
- Julia Braune
- Institute of Anatomy, Leipzig University, 04317 Leipzig, Germany
| | - Ulrike Weyer
- Institute of Anatomy, Leipzig University, 04317 Leipzig, Germany
| | | | - Jan Mauer
- Department of Pharmacology, Weill Cornell Medical College, Cornell University, New York, NY 10065; and
| | - Jens C Brüning
- Max Planck Institute for Metabolism Research, 50931 Cologne, Germany
| | - Ingo Bechmann
- Institute of Anatomy, Leipzig University, 04317 Leipzig, Germany
| | - Martin Gericke
- Institute of Anatomy, Leipzig University, 04317 Leipzig, Germany;
| |
Collapse
|
95
|
Zamarron BF, Mergian TA, Cho KW, Martinez-Santibanez G, Luan D, Singer K, DelProposto JL, Geletka LM, Muir LA, Lumeng CN. Macrophage Proliferation Sustains Adipose Tissue Inflammation in Formerly Obese Mice. Diabetes 2017; 66:392-406. [PMID: 28108608 PMCID: PMC5248991 DOI: 10.2337/db16-0500] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 10/29/2016] [Indexed: 12/12/2022]
Abstract
Obesity causes dramatic proinflammatory changes in the adipose tissue immune environment, but relatively little is known regarding how this inflammation responds to weight loss (WL). To understand the mechanisms by which meta-inflammation resolves during WL, we examined adipose tissue leukocytes in mice after withdrawal of a high-fat diet. After 8 weeks of WL, mice achieved similar weights and glucose tolerance values as age-matched lean controls but showed abnormal insulin tolerance. Despite fat mass normalization, total and CD11c+ adipose tissue macrophage (ATM) content remained elevated in WL mice for up to 6 months and was associated with persistent fibrosis in adipose tissue. ATMs in formerly obese mice demonstrated a proinflammatory profile, including elevated expression of interferon-γ, tumor necrosis factor-α, and interleukin-1β. T-cell-deficient Rag1-/- mice showed a degree of ATM persistence similar to that in WT mice, but with reduced inflammatory gene expression. ATM proliferation was identified as the predominant mechanism by which ATMs are retained in adipose tissue with WL. Our study suggests that WL does not completely resolve obesity-induced ATM activation, which may contribute to the persistent adipose tissue damage and reduced insulin sensitivity observed in formerly obese mice.
Collapse
Affiliation(s)
- Brian F Zamarron
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI
| | - Taleen A Mergian
- College of Literature Sciences and Arts, University of Michigan, Ann Arbor, MI
| | - Kae Won Cho
- Soonchunhyang Institute of Medi-bio Science, Soonchunhyang University, Cheonan-si, Chungcheongnam-do, Korea
| | | | - Danny Luan
- College of Literature Sciences and Arts, University of Michigan, Ann Arbor, MI
| | - Kanakadurga Singer
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI
| | - Jennifer L DelProposto
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI
| | - Lynn M Geletka
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI
| | - Lindsey A Muir
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI
| | - Carey N Lumeng
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI
- Department of Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
96
|
Piñeros AR, Campos LW, Fonseca DM, Bertolini TB, Gembre AF, Prado RQ, Alves-Filho JC, Ramos SG, Russo M, Bonato VLD. M2 macrophages or IL-33 treatment attenuate ongoing Mycobacterium tuberculosis infection. Sci Rep 2017; 7:41240. [PMID: 28128217 PMCID: PMC5269597 DOI: 10.1038/srep41240] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 12/19/2016] [Indexed: 11/09/2022] Open
Abstract
The protective effects of mycobacterial infections on lung allergy are well documented. However, the inverse relationship between tuberculosis and type 2 immunity is still elusive. Although type 1 immunity is essential to protection against Mycobacterium tuberculosis it might be also detrimental to the host due to the induction of extensive tissue damage. Here, we determined whether lung type 2 immunity induced by allergen sensitization and challenge could affect the outcome of M. tuberculosis infection. We used two different protocols in which sensitization and allergen challenge were performed before or after M. tuberculosis infection. We found an increased resistance to M. tuberculosis only when allergen exposure was given after, but not before infection. Infected mice exposed to allergen exhibited lower bacterial load and cellular infiltrates in the lungs. Enhanced resistance to infection after allergen challenge was associated with increased gene expression of alternatively activated macrophages (M2 macrophages) and IL-33 levels. Accordingly, either adoptive transfer of M2 macrophages or systemic IL-33 treatment was effective in attenuating M. tuberculosis infection. Notably, the enhanced resistance induced by allergen exposure was dependent on IL-33 receptor ST2. Our work indicates that IL-33 might be an alternative therapeutic treatment for severe tuberculosis.
Collapse
Affiliation(s)
- A R Piñeros
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - L W Campos
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - D M Fonseca
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - T B Bertolini
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - A F Gembre
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - R Q Prado
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - J C Alves-Filho
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - S G Ramos
- Department of Pathology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - M Russo
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - V L D Bonato
- Department of Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
97
|
Microparticles derived from obese adipose tissue elicit a pro-inflammatory phenotype of CD16 + , CCR5 + and TLR8 + monocytes. Biochim Biophys Acta Mol Basis Dis 2017; 1863:139-151. [DOI: 10.1016/j.bbadis.2016.09.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 08/30/2016] [Accepted: 09/20/2016] [Indexed: 12/11/2022]
|
98
|
Nakajima S, Koh V, Kua LF, So J, Davide L, Lim KS, Petersen SH, Yong WP, Shabbir A, Kono K. Accumulation of CD11c+CD163+ Adipose Tissue Macrophages through Upregulation of Intracellular 11β-HSD1 in Human Obesity. THE JOURNAL OF IMMUNOLOGY 2016; 197:3735-3745. [PMID: 27698011 DOI: 10.4049/jimmunol.1600895] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 09/01/2016] [Indexed: 12/31/2022]
Abstract
Adipose tissue (AT) macrophages (ATMs) are key players for regulation of AT homeostasis and obesity-related metabolic disorders. However, the phenotypes of human ATMs and regulatory mechanisms of their polarization have not been clearly described. In this study, we investigated human ATMs in both abdominal visceral AT and s.c. AT and proposed an 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1)-glucocorticoid receptor regulatory axis that might dictate M1/M2 polarization in ATMs. The accumulation of CD11c+CD163+ ATMs in both visceral AT and s.c. AT of obese individuals was confirmed at the cellular level and was found to be clearly correlated with body mass index and production of reactive oxygen species. Using our in vitro system where human peripheral blood monocytes (hPBMs) were cocultured with Simpson-Golabi-Behmel syndrome adipocytes, M1/M2 polarization was found to be dependent on 11β-HSD1, an intracellular glucocorticoid reactivating enzyme. Exposure of hPBMs to cortisol-induced expression of CD163 and RU-486, a glucocorticoid receptor antagonist, significantly abrogated CD163 expression through coculture of mature adipocytes with hPBMs. Moreover, 11β-HSD1 was expressed in crown ATMs in obese AT. Importantly, conditioned medium from coculture of adipocytes with hPBMs enhanced proliferation of human breast cancer MCF7 and MDA-MB-231 cells. In summary, the phenotypic switch of ATMs from M2 to mixed M1/M2 phenotype occurred through differentiation of adipocytes in obese individuals, and upregulation of intracellular 11β-HSD1 might play a role in the process.
Collapse
Affiliation(s)
- Shotaro Nakajima
- Center for Translational Medicine, Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599
| | - Vivien Koh
- Department of Hematology-Oncology, National University of Singapore, Singapore 119228
| | - Ley-Fang Kua
- Department of Hematology-Oncology, National University of Singapore, Singapore 119228
| | - Jimmy So
- Department of Surgery, National University of Singapore, Singapore 119228
| | - Lomanto Davide
- Department of Surgery, National University of Singapore, Singapore 119228
| | - Kee Siang Lim
- Center for Translational Medicine, Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599
| | - Sven Hans Petersen
- Center for Translational Medicine, Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599
| | - Wei-Peng Yong
- Center for Translational Medicine, Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599.,Department of Hematology-Oncology, National University of Singapore, Singapore 119228
| | - Asim Shabbir
- Department of Surgery, National University of Singapore, Singapore 119228
| | - Koji Kono
- Center for Translational Medicine, Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599; .,Department of Surgery, National University of Singapore, Singapore 119228.,Department of Organ Regulatory Surgery, Fukushima Medical University, Fukushima 960-1295, Japan; and.,Department of Advanced Cancer Immunotherapy, Fukushima Medical University, Fukushima 960-1295, Japan
| |
Collapse
|
99
|
Torres-Castro I, Arroyo-Camarena ÚD, Martínez-Reyes CP, Gómez-Arauz AY, Dueñas-Andrade Y, Hernández-Ruiz J, Béjar YL, Zaga-Clavellina V, Morales-Montor J, Terrazas LI, Kzhyshkowska J, Escobedo G. Human monocytes and macrophages undergo M1-type inflammatory polarization in response to high levels of glucose. Immunol Lett 2016; 176:81-9. [DOI: 10.1016/j.imlet.2016.06.001] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 05/27/2016] [Accepted: 06/03/2016] [Indexed: 12/17/2022]
|
100
|
Kralova Lesna I, Kralova A, Cejkova S, Fronek J, Petras M, Sekerkova A, Thieme F, Janousek L, Poledne R. Characterisation and comparison of adipose tissue macrophages from human subcutaneous, visceral and perivascular adipose tissue. J Transl Med 2016; 14:208. [PMID: 27400732 PMCID: PMC4940901 DOI: 10.1186/s12967-016-0962-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 06/27/2016] [Indexed: 12/21/2022] Open
Abstract
Background and aims Macrophages play important roles in adipose tissue inflammation and its consequences. Unfortunately, a detailed description of the macrophage phenotypes in different human adipose tissues is not available. Subjects and methods Subcutaneous, visceral and perivascular adipose tissues were obtained from 52 living kidney donors during live donor nephrectomy. Stromal vascular fractions were isolated, and the macrophage phenotypes were analyzed by flow cytometry using surface markers (CD14, CD16, CD36, and CD163). Results In addition to CD16 positivity, pro-inflammatory macrophages also display high scavenger receptor CD36 expression. The great majority of CD16 negative macrophages express the anti-inflammatory CD163 marker. The presence of pro-inflammatory macrophages was almost twice as high in visceral (p < 0.0001) and perivascular (p < 0.0001) adipose tissues than in subcutaneous tissue. This difference was substantially more pronounced in the postmenopausal women subgroup, consequentlly, the total difference was driven by this subgroup. Conclusion We obtained detailed information about M1 and M2 macrophage phenotypes in human adipose tissue. The visceral and perivascular adipose tissues had substantially higher pro-inflammatory characteristics than the subcutaneous tissue. The higher proportion of pro-inflammatory macrophages in the visceral adipose tissue of postmenopausal women might be related to an increased cardiovascular risk.
Collapse
Affiliation(s)
- Ivana Kralova Lesna
- Laboratory for Atherosclerosis Research, Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | - Anna Kralova
- Laboratory for Atherosclerosis Research, Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Sona Cejkova
- Laboratory for Atherosclerosis Research, Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jiri Fronek
- Transplant Surgery Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Marek Petras
- 2nd Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Alena Sekerkova
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Filip Thieme
- Transplant Surgery Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Libor Janousek
- Transplant Surgery Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Rudolf Poledne
- Laboratory for Atherosclerosis Research, Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|