51
|
Mehrpouya-Bahrami P, Miranda K, Singh NP, Zumbrun EE, Nagarkatti M, Nagarkatti PS. Role of microRNA in CB1 antagonist-mediated regulation of adipose tissue macrophage polarization and chemotaxis during diet-induced obesity. J Biol Chem 2019; 294:7669-7681. [PMID: 30910812 DOI: 10.1074/jbc.ra118.005094] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 03/18/2019] [Indexed: 12/12/2022] Open
Abstract
Although cannabinoid receptor 1 (CB1) antagonists have been shown to attenuate diet-induced obesity (DIO) and associated inflammation, the precise molecular mechanisms involved are not clear. In the current study, we investigated the role of microRNA (miR) in the regulation of adipose tissue macrophage (ATM) phenotype following treatment of DIO mice with the CB1 antagonist SR141716A. DIO mice were fed high-fat diet (HFD) for 12 weeks and then treated daily with SR141716A (10 mg/kg) for 4 weeks while continuing HFD. Treated mice experienced weight loss, persistent reduction in fat mass, improvements in metabolic profile, and decreased adipose inflammation. CB1 blockade resulted in down-regulation of several miRs in ATMs, including the miR-466 family and miR-762. Reduced expression of the miR-466 family led to induction of anti-inflammatory M2 transcription factors KLF4 and STAT6, whereas down-regulation of miR-762 promoted induction of AGAP-2, a negative regulator of the neuroimmune retention cues, Netrin-1 and its coreceptor UNC5B. Furthermore, treatment of primary macrophages with SR141716A up-regulated KLF4 and STAT6, reduced secretion of Netrin-1, and increased migration toward the lymph node chemoattractant CCL19. These studies demonstrate for the first time that CB1 receptor blockade attenuates DIO-associated inflammation through alterations in ATM miR expression that promote M2 ATM polarization and macrophage egress from adipose tissue. The current study also identifies additional novel therapeutic targets for diet-induced obesity and metabolic disorder.
Collapse
Affiliation(s)
- Pegah Mehrpouya-Bahrami
- From the Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina 29208
| | - Kathryn Miranda
- From the Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina 29208
| | - Narendra P Singh
- From the Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina 29208
| | - Elizabeth E Zumbrun
- From the Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina 29208
| | - Mitzi Nagarkatti
- From the Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina 29208
| | - Prakash S Nagarkatti
- From the Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina 29208
| |
Collapse
|
52
|
Atawia RT, Bunch KL, Toque HA, Caldwell RB, Caldwell RW. Mechanisms of obesity-induced metabolic and vascular dysfunctions. FRONT BIOSCI-LANDMRK 2019; 24:890-934. [PMID: 30844720 PMCID: PMC6689231 DOI: 10.2741/4758] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Obesity has reached epidemic proportions and its prevalence is climbing. Obesity is characterized by hypertrophied adipocytes with a dysregulated adipokine secretion profile, increased recruitment of inflammatory cells, and impaired metabolic homeostasis that eventually results in the development of systemic insulin resistance, a phenotype of type 2 diabetes. Nitric oxide synthase (NOS) is an enzyme that converts L-arginine to nitric oxide (NO), which functions to maintain vascular and adipocyte homeostasis. Arginase is a ureohydrolase enzyme that competes with NOS for L-arginine. Arginase activity/expression is upregulated in obesity, which results in diminished bioavailability of NO, impairing both adipocyte and vascular endothelial cell function. Given the emerging role of NO in the regulation of adipocyte physiology and metabolic capacity, this review explores the interplay between arginase and NO, and their effect on the development of metabolic disorders, cardiovascular diseases, and mitochondrial dysfunction in obesity. A comprehensive understanding of the mechanisms involved in the development of obesity-induced metabolic and vascular dysfunction is necessary for the identification of more effective and tailored therapeutic avenues for their prevention and treatment.
Collapse
Affiliation(s)
- Reem T Atawia
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University. Augusta, GA 30904, USA
| | - Katharine L Bunch
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University. Augusta, GA 30904, USA
| | - Haroldo A Toque
- Department of Pharmacology and Toxicology,and Vascular Biology Center, Medical College of Georgia, Augusta University. Augusta, GA 30904, USA
| | - Ruth B Caldwell
- Vascular Biology Center, Medical College of Georgia, Augusta University. Augusta, GA 30904, USA
| | - Robert W Caldwell
- Vascular Biology Center, Medical College of Georgia, Augusta University. Augusta, GA 30904,USA,
| |
Collapse
|
53
|
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: 158] [Impact Index Per Article: 26.3] [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
|
54
|
Tardelli LP, Breda L, Marques LF, Gomes Carvalho Lima NC, Furtado de Camargo T, Scherer BR, Moreira NF, Dias JF, Dalia RA, Thomazini BF, Corezolla do Amaral ME, Alves AA. High lipid and low carbohydrate content diet, immediately after weaning, causes hepatic injury, systemic oxidative stress and diminishment of lipids in white adipose tissue. JOURNAL OF NUTRITION & INTERMEDIARY METABOLISM 2018. [DOI: 10.1016/j.jnim.2018.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
55
|
Rodríguez CP, González MC, Aguilar-Salinas CA, Nájera-Medina O. Peripheral Lymphocytes, Obesity, and Metabolic Syndrome in Young Adults: An Immunometabolism Study. Metab Syndr Relat Disord 2018; 16:342-349. [PMID: 29957122 DOI: 10.1089/met.2018.0005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Obesity is characterized by a low-intensity chronic inflammatory process in which immune system cells interact in a complex network, which affects systemic metabolic processes. This raises interest in analyzing possible changes in the proportions of immune system cells in individuals with obesity with and without metabolic syndrome (MS), in relation to their body composition. METHODS Circulating cells were analyzed with flow cytometry in young adults: monocytes, granulocytes, lymphocytes (T, B, and natural killer [NK]), TCD4+CD62-, TCD8+CD28-, and naive and memory cells of TCD3+ and TCD4+. Body composition was obtained by bioelectrical impedance analysis and dual-energy X-ray absorptiometry, and metabolic parameters. RESULTS A total of 169 persons were evaluated: 20% presented normal body mass index (BMI); 49% was overweight, and 31% had obesity; 28% had MS. It was observed that with an increase in BMI and visceral adipose tissue increase (VATI), body composition and biochemical variables were negatively altered. With regard to cell subpopulations, total lymphocytes increased and granulocytes and NK lymphocytes decreased in patients with MS and VATI. Memory cells increased with BMI and VATI. In individuals with MS, monocytes, and NK lymphocytes comprised a negative association with VAT, fat mass, and skeletal muscle mass (SMM). In individuals with MS and VATI, a negative correlation was observed between monocytes and SMM. CONCLUSIONS Significant changes were detected in the subpopulations of lymphocytes, suggesting that weight gain, SMM, and VAT accumulation gave rise to immunological changes at the peripheral level, and the presence of increased memory cells could be related to low-intensity chronic inflammation.
Collapse
Affiliation(s)
- Carmen Paulina Rodríguez
- 1 División de Ciencias Biológicas y de la Salud, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa , Ciudad de México, México.,2 Posgrado en Biología Experimental, Universidad Autónoma Metropolitana-Iztapalapa , Ciudad de México, México
| | - María Cristina González
- 1 División de Ciencias Biológicas y de la Salud, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa , Ciudad de México, México
| | - Carlos A Aguilar-Salinas
- 3 Departamento de Endocrinología y Metabolismo, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán , Ciudad de México, México
| | - Oralia Nájera-Medina
- 4 División de Ciencias Biológicas y de la Salud, Departamento de Atención a la Salud, Universidad Autónoma Metropolitana-Xochimilco , Ciudad de México, México
| |
Collapse
|
56
|
Guzik TJ, Skiba DS, Touyz RM, Harrison DG. The role of infiltrating immune cells in dysfunctional adipose tissue. Cardiovasc Res 2018; 113:1009-1023. [PMID: 28838042 PMCID: PMC5852626 DOI: 10.1093/cvr/cvx108] [Citation(s) in RCA: 281] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 07/05/2017] [Indexed: 12/15/2022] Open
Abstract
Adipose tissue (AT) dysfunction, characterized by loss of its homeostatic functions, is a hallmark of non-communicable diseases. It is characterized by chronic low-grade inflammation and is observed in obesity, metabolic disorders such as insulin resistance and diabetes. While classically it has been identified by increased cytokine or chemokine expression, such as increased MCP-1, RANTES, IL-6, interferon (IFN) gamma or TNFα, mechanistically, immune cell infiltration is a prominent feature of the dysfunctional AT. These immune cells include M1 and M2 macrophages, effector and memory T cells, IL-10 producing FoxP3+ T regulatory cells, natural killer and NKT cells and granulocytes. Immune composition varies, depending on the stage and the type of pathology. Infiltrating immune cells not only produce cytokines but also metalloproteinases, reactive oxygen species, and chemokines that participate in tissue remodelling, cell signalling, and regulation of immunity. The presence of inflammatory cells in AT affects adjacent tissues and organs. In blood vessels, perivascular AT inflammation leads to vascular remodelling, superoxide production, endothelial dysfunction with loss of nitric oxide (NO) bioavailability, contributing to vascular disease, atherosclerosis, and plaque instability. Dysfunctional AT also releases adipokines such as leptin, resistin, and visfatin that promote metabolic dysfunction, alter systemic homeostasis, sympathetic outflow, glucose handling, and insulin sensitivity. Anti-inflammatory and protective adiponectin is reduced. AT may also serve as an important reservoir and possible site of activation in autoimmune-mediated and inflammatory diseases. Thus, reciprocal regulation between immune cell infiltration and AT dysfunction is a promising future therapeutic target.
Collapse
Affiliation(s)
- Tomasz J Guzik
- British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK.,Translational Medicine Laboratory, Department of Internal Medicine, Jagiellonian University, Collegium Medicum, Krakow, Poland
| | - Dominik S Skiba
- British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK.,Translational Medicine Laboratory, Department of Internal Medicine, Jagiellonian University, Collegium Medicum, Krakow, Poland
| | - Rhian M Touyz
- British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - David G Harrison
- British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK.,Department of Clinical Pharmacology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
57
|
Zhang S, Cao H, Li Y, Jing Y, Liu S, Ye C, Wang H, Yu S, Peng C, Hui L, Wang YC, Zhang H, Guo F, Zhai Q, Wang H, Huang R, Zhang L, Jiang J, Liu W, Ying H. Metabolic benefits of inhibition of p38α in white adipose tissue in obesity. PLoS Biol 2018; 16:e2004225. [PMID: 29750781 PMCID: PMC5965899 DOI: 10.1371/journal.pbio.2004225] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 05/23/2018] [Accepted: 04/09/2018] [Indexed: 01/22/2023] Open
Abstract
p38 has long been known as a central mediator of protein kinase A (PKA) signaling in brown adipocytes, which positively regulate the transcription of uncoupling protein 1 (UCP-1). However, the physiological role of p38 in adipose tissues, especially the white adipose tissue (WAT), is largely unknown. Here, we show that mice lacking p38α in adipose tissues display a lean phenotype, improved metabolism, and resistance to diet-induced obesity. Surprisingly, ablation of p38α causes minimal effects on brown adipose tissue (BAT) in adult mice, as evident from undetectable changes in UCP-1 expression, mitochondrial function, body temperature (BT), and energy expenditure. In contrast, genetic ablation of p38α in adipose tissues not only markedly facilitates the browning in WAT upon cold stress but also prevents diet-induced obesity. Consistently, pharmaceutical inhibition of p38α remarkably enhances the browning of WAT and has metabolic benefits. Furthermore, our data suggest that p38α deficiency promotes white-to-beige adipocyte reprogramming in a cell-autonomous manner. Mechanistically, inhibition of p38α stimulates the UCP-1 transcription through PKA and its downstream cAMP-response element binding protein (CREB), which form a positive feedback loop that functions to reinforce the white-to-beige phenotypic switch during cold exposure. Together, our study reveals that inhibition of p38α is able to promote WAT browning and confer metabolic benefits. Our study also indicates that p38α in WAT represents an exciting pharmacological target to combat obesity and metabolic diseases. The functional brown adipose tissue (BAT) identified in human adults consists of not only classic brown adipocytes but also brown-like adipocytes (beige adipocytes), both of which are important for energy homeostasis. Due to the same ability to convert fat into heat as brown adipocytes, beige adipocytes have been considered as a novel pharmacological target to combat obesity. Growing evidence suggests that promoting the development and formation of beige adipocytes in white adipose tissue (WAT), also called the browning of WAT, is able to prevent diet-induced obesity and improve metabolism in rodents. Thus, understanding the molecular basis for the regulation of browning in WAT may help us to develop new strategies to counteract obesity and metabolic diseases. In this study, adipocyte-specific p38α knockout (Fp38αKO) mice are generated that display a lean phenotype, improved metabolism, and resistance to diet-induced obesity. Interestingly, we found that adipocyte p38α deficiency facilitates the browning in WAT. Then, we show that pharmaceutical inhibition of p38α enhances the browning in WAT and has metabolic benefits. We propose that inhibiting p38α in WAT, possibly combined with cold exposure, could constitute an exciting pharmacological target to combat obesity and metabolic diseases.
Collapse
Affiliation(s)
- Shengjie Zhang
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hongchao Cao
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yan Li
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yanyan Jing
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shengnan Liu
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Cheng Ye
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hui Wang
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shuxian Yu
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chengyuan Peng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Lijian Hui
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yu-cheng Wang
- Shanghai Xuhui Central Hospital, Shanghai Clinical Center, Chinese Academy of Sciences, Shanghai, China
| | - Haibing Zhang
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Feifan Guo
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiwei Zhai
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hui Wang
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China
| | - Ruimin Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Ling Zhang
- Department of Head and Neck Surgery, Fudan University Cancer Center and Department of Oncology, Fudan University, Shanghai Medical College, Shanghai, China
| | - Jingjing Jiang
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Liu
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hao Ying
- CAS Key laboratory of nutrition, metabolism and food safety, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China
- * E-mail:
| |
Collapse
|
58
|
Kazeminasab F, Marandi SM, Ghaedi K, Safaeinejad Z, Esfarjani F, Nasr-Esfahani MH. A comparative study on the effects of high-fat diet and endurance training on the PGC-1α-FNDC5/irisin pathway in obese and nonobese male C57BL/6 mice. Appl Physiol Nutr Metab 2018; 43:651-662. [PMID: 29365291 DOI: 10.1139/apnm-2017-0614] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The present study was performed to clarify how a combined exercise/diet treatment could affect the expression level of the muscle fibronectin type III domain containing 5 (Fndc5) with respect to body fat mass. Male C57BL/6 mice were divided into 2 groups including low-fat (LF) and high-fat (HF) diets for 12 weeks. Then, LF fed (nonobese) and HF fed mice (obese) were divided into the following 4 groups: HF-Exercise, HF-Sedentary, LF-Exercise, and LF-Sedentary. The exercise group exercised on a motor-driven treadmill for 45 min/day, 5 days/week for 8 weeks. Mice were sacrificed 24 h after the final exercise session. Gastrocnemius muscle and the visceral adipose tissue were excised and frozen for the assessment of proliferator-activated receptor gamma coactivator 1 alpha (Pgc-1α) and Fndc5 messenger RNA (mRNA) and protein levels. Data indicated that protein level of muscle PGC-1α was decreased in HF versus LF groups and in obese versus nonobese mice. Moreover, Fndc5 mRNA levels were increased in the muscle tissue of HF versus LF groups and in obese versus nonobese mice. Also, in the gastrocnemius skeletal muscle, protein levels of FNDC5 were significantly higher in the HF fed mice, as compared with their low-fat fed counterparts, similar to what was observed for exercised versus sedentary mice. Overall, we found that the HF diet increased Fndc5 transcript levels in the skeletal muscle, but exercise had a minimal effect on the transcript level of Fndc5, whereas endurance training increased the protein content of FNDC5 in the skeletal muscle.
Collapse
Affiliation(s)
- Fatemeh Kazeminasab
- a Department of Exercise Physiology, Faculty of Sport Sciences, University of Isfahan, Hezar Jerib Ave., Azadi Sq., PO box 81799-54359, Isfahan, Iran
| | - Sayed Mohammad Marandi
- a Department of Exercise Physiology, Faculty of Sport Sciences, University of Isfahan, Hezar Jerib Ave., Azadi Sq., PO box 81799-54359, Isfahan, Iran
| | - Kamran Ghaedi
- b Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran.,c Department of Cellular Biotechnology, Cell Sciences Research Center, Royan Institute for Biotechnology, ACECR, Royan St., Salman St., Khorasgan, PO box 816513-1378, Isfahan, Iran
| | - Zahra Safaeinejad
- c Department of Cellular Biotechnology, Cell Sciences Research Center, Royan Institute for Biotechnology, ACECR, Royan St., Salman St., Khorasgan, PO box 816513-1378, Isfahan, Iran
| | - Fahimeh Esfarjani
- a Department of Exercise Physiology, Faculty of Sport Sciences, University of Isfahan, Hezar Jerib Ave., Azadi Sq., PO box 81799-54359, Isfahan, Iran
| | - Mohammad Hossein Nasr-Esfahani
- c Department of Cellular Biotechnology, Cell Sciences Research Center, Royan Institute for Biotechnology, ACECR, Royan St., Salman St., Khorasgan, PO box 816513-1378, Isfahan, Iran
| |
Collapse
|
59
|
Huo D, Liu S, Zhang C, He J, Zhou Z, Zhang H, Hu Y. Hypoxia-Targeting, Tumor Microenvironment Responsive Nanocluster Bomb for Radical-Enhanced Radiotherapy. ACS NANO 2017; 11:10159-10174. [PMID: 28992409 DOI: 10.1021/acsnano.7b04737] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Although ultrasmall metal nanoparticles (NPs) have been used as radiosensitizers to enhance the local damage to tumor tissues while reducing injury to the surrounding organs, their rapid clearance from the circulatory system and the presence of hypoxia within the tumor continue to hamper their further application in radiotherapy (RT). In this study, we report a size tunable nanocluster bomb with a initial size of approximately 33 nm featuring a long half-life during blood circulation and destructed to release small hypoxia microenvironment-targeting NPs (∼5 nm) to achieve deep tumor penetration. Hypoxic profiles of solid tumors were precisely imaged using NP-enhanced computed tomography (CT) with higher spatial resolution. Once irradiated with a 1064 nm laser, CT-guided, local photothermal ablation of the tumor and production of radical species could be achieved simultaneously. The induced radical species alleviated the hypoxia-induced resistance and sensitized the tumor to the killing efficacy of radiation in Akt-mTOR pathway-dependent manner. The therapeutic outcome was assessed in animal models of orthotopical breast cancer and pancreatic cancer, supporting the feasibility of our combinational treatment in hypoxic tumor management.
Collapse
Affiliation(s)
- Da Huo
- Department of Radiology, Drum Tower Hospital, School of Medicine, Nanjing University , Nanjing, Jiangsu 210008, China
- Collaborative Innovation Center of Chemistry for Life Sciences, College of Engineering and Applied Sciences, Nanjing University , Nanjing, Jiangsu 210093, China
| | - Sen Liu
- Collaborative Innovation Center of Chemistry for Life Sciences, College of Engineering and Applied Sciences, Nanjing University , Nanjing, Jiangsu 210093, China
| | - Chao Zhang
- Collaborative Innovation Center of Chemistry for Life Sciences, College of Engineering and Applied Sciences, Nanjing University , Nanjing, Jiangsu 210093, China
| | - Jian He
- Department of Radiology, Drum Tower Hospital, School of Medicine, Nanjing University , Nanjing, Jiangsu 210008, China
| | - Zhengyang Zhou
- Department of Radiology, Drum Tower Hospital, School of Medicine, Nanjing University , Nanjing, Jiangsu 210008, China
| | - Hao Zhang
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University , Nanjing, Jiangsu 210029, China
| | - Yong Hu
- Department of Radiology, Drum Tower Hospital, School of Medicine, Nanjing University , Nanjing, Jiangsu 210008, China
- Collaborative Innovation Center of Chemistry for Life Sciences, College of Engineering and Applied Sciences, Nanjing University , Nanjing, Jiangsu 210093, China
| |
Collapse
|
60
|
Perrini S, Cignarelli A, Quaranta VN, Falcone VA, Kounaki S, Porro S, Ciavarella A, Ficarella R, Barbaro M, Genchi VA, Nigro P, Carratù P, Natalicchio A, Laviola L, Resta O, Giorgino F. Correction of intermittent hypoxia reduces inflammation in obese subjects with obstructive sleep apnea. JCI Insight 2017; 2:94379. [PMID: 28878129 DOI: 10.1172/jci.insight.94379] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 07/25/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND In obese subjects with obstructive sleep apnea (OSA), chronic intermittent hypoxia (CIH) may be linked to systemic and adipose tissue inflammation. METHODS We obtained abdominal subcutaneous adipose tissue biopsies from OSA and non-OSA obese (BMI > 35) subjects at baseline and after 24 weeks (T1) of weight-loss intervention plus continuous positive airway pressure (c-PAP) or weight-loss intervention alone, respectively. OSA subjects were grouped according to good (therapeutic) or poor (subtherapeutic) adherence to c-PAP. RESULTS At baseline, anthropometric and metabolic parameters, serum cytokines, and adipose tissue mRNA levels of obesity-associated chemokines and inflammatory markers were not different in OSA and non-OSA subjects. At T1, body weight was significantly reduced in all groups. Serum concentrations of IL-2, IL-4, IL-6, MCP-1, PDGFβ, and VEGFα were reduced by therapeutic c-PAP in OSA subjects and remained unaltered in non-OSA and subtherapeutic c-PAP groups. Similarly, adipose tissue mRNA levels of macrophage-specific (CD68, CD36) and ER stress (ATF4, CHOP, ERO-1) gene markers, as well as of IL-6, PDGFβ, and VEGFα, were decreased only in the therapeutic c-PAP group. CONCLUSION CIH does not represent an additional factor increasing systemic and adipose tissue inflammation in morbid obesity. However, in subjects with OSA, an effective c-PAP therapy improves systemic and obesity-associated inflammatory markers. FUNDING Ministero dell'Università e della Ricerca and Progetti di Rilevante Interesse Nazionale.
Collapse
Affiliation(s)
- Sebastio Perrini
- Department of Emergency and Organ Transplantation - Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, and
| | - Angelo Cignarelli
- Department of Emergency and Organ Transplantation - Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, and
| | - Vitaliano Nicola Quaranta
- Department of Basic Medical Sciences, Neurosciences and Sense Organs - Section of Respiratory Disease, University of Bari Aldo Moro, Bari, Italy
| | - Vito Antonio Falcone
- Department of Basic Medical Sciences, Neurosciences and Sense Organs - Section of Respiratory Disease, University of Bari Aldo Moro, Bari, Italy
| | - Stella Kounaki
- Department of Emergency and Organ Transplantation - Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, and
| | - Stefania Porro
- Department of Emergency and Organ Transplantation - Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, and
| | - Alessandro Ciavarella
- Department of Emergency and Organ Transplantation - Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, and
| | - Romina Ficarella
- Department of Emergency and Organ Transplantation - Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, and
| | - Maria Barbaro
- Department of Emergency and Organ Transplantation - Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, and
| | - Valentina Annamaria Genchi
- Department of Emergency and Organ Transplantation - Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, and
| | - Pasquale Nigro
- Department of Emergency and Organ Transplantation - Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, and
| | - Pierluigi Carratù
- Department of Basic Medical Sciences, Neurosciences and Sense Organs - Section of Respiratory Disease, University of Bari Aldo Moro, Bari, Italy
| | - Annalisa Natalicchio
- Department of Emergency and Organ Transplantation - Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, and
| | - Luigi Laviola
- Department of Emergency and Organ Transplantation - Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, and
| | - Onofrio Resta
- Department of Basic Medical Sciences, Neurosciences and Sense Organs - Section of Respiratory Disease, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Giorgino
- Department of Emergency and Organ Transplantation - Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, and
| |
Collapse
|
61
|
Laiglesia LM, Lorente-Cebrián S, López-Yoldi M, Lanas R, Sáinz N, Martínez JA, Moreno-Aliaga MJ. Maresin 1 inhibits TNF-alpha-induced lipolysis and autophagy in 3T3-L1 adipocytes. J Cell Physiol 2017; 233:2238-2246. [PMID: 28703289 DOI: 10.1002/jcp.26096] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 07/11/2017] [Indexed: 12/30/2022]
Abstract
Obesity is associated with high levels of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), which promotes inflammation in adipose tissue. The omega-3 PUFAs, and their derived lipid mediators, such as Maresin 1 (MaR1) have anti-inflammatory effects on adipose tissue. This study aimed to analyze if MaR1 may counteract alterations induced by TNF-α on lipolysis and autophagy in mature 3T3-L1 adipocytes. Our data revealed that MaR1 (1-100 nM) inhibited the TNF-α-induced glycerol release after 48 hr, which may be related to MaR1 ability of preventing the decrease in lipid droplet-coating protein perilipin and G0/G1 Switch 2 protein expression. MaR1 also reversed the decrease in total hormone sensitive lipase (total HSL), and the ratio of phosphoHSL at Ser-565/total HSL, while preventing the increased ratio of phosphoHSL at Ser-660/total HSL and phosphorylation of extracellular signal-regulated kinase 1/2 induced by TNF-α. Moreover, MaR1 counteracted the cytokine-induced decrease of p62 protein, a key autophagy indicator, and also prevented the induction of LC3II/LC3I, an important autophagosome formation marker. Current data suggest that MaR1 may ameliorate TNF-α-induced alterations on lipolysis and autophagy in adipocytes. This may also contribute to the beneficial actions of MaR1 on adipose tissue and insulin sensitivity in obesity.
Collapse
Affiliation(s)
- Laura M Laiglesia
- Department Nutrition Food Science Physiology, University of Navarra, Pamplona, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Spain
| | - Silvia Lorente-Cebrián
- Department Nutrition Food Science Physiology, University of Navarra, Pamplona, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Miguel López-Yoldi
- Department Nutrition Food Science Physiology, University of Navarra, Pamplona, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Spain
| | - Raquel Lanas
- Department Nutrition Food Science Physiology, University of Navarra, Pamplona, Spain
| | - Neira Sáinz
- Centre for Nutrition Research, University of Navarra, Pamplona, Spain
| | - Jose Alfredo Martínez
- Department Nutrition Food Science Physiology, University of Navarra, Pamplona, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.,CIBERobn, Physiopathology of Obesity and Nutrition, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Maria J Moreno-Aliaga
- Department Nutrition Food Science Physiology, University of Navarra, Pamplona, Spain.,Centre for Nutrition Research, University of Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.,CIBERobn, Physiopathology of Obesity and Nutrition, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
62
|
Short JD, Tavakoli S, Nguyen HN, Carrera A, Farnen C, Cox LA, Asmis R. Dyslipidemic Diet-Induced Monocyte "Priming" and Dysfunction in Non-Human Primates Is Triggered by Elevated Plasma Cholesterol and Accompanied by Altered Histone Acetylation. Front Immunol 2017; 8:958. [PMID: 28878765 PMCID: PMC5572238 DOI: 10.3389/fimmu.2017.00958] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 07/26/2017] [Indexed: 12/13/2022] Open
Abstract
Monocytes and the recruitment of monocyte-derived macrophages into sites of inflammation play a key role in atherogenesis and other chronic inflammatory diseases linked to cardiometabolic syndrome and obesity. Previous studies from our group have shown that metabolic stress promotes monocyte priming, i.e., enhanced adhesion and accelerated chemotaxis of monocytes in response to chemokines, both in vitro and in dyslipidemic LDLR-/- mice. We also showed that metabolic stress-induced monocyte dysfunction is, at least to a large extent caused by the S-glutathionylation, inactivation, and subsequent degradation of mitogen-activated protein kinase phosphatase 1. Here, we analyzed the effects of a Western-style, dyslipidemic diet (DD), which was composed of high levels of saturated fat, cholesterol, and simple sugars, on monocyte (dys)function in non-human primates (NHPs). We found that similar to mice, a DD enhances monocyte chemotaxis in NHP within 4 weeks, occurring concordantly with the onset of hypercholesterolemia but prior to changes in triglycerides, blood glucose, monocytosis, or changes in monocyte subset composition. In addition, we identified transitory decreases in the acetylation of histone H3 at the lysine residues 18 and 23 in metabolically primed monocytes, and we found that monocyte priming was correlated with the acetylation of histone H3 at lysine 27 after an 8-week DD regimen. Our data show that metabolic stress promotes monocyte priming and hyper-chemotactic responses in NHP. The histone modifications accompanying monocyte priming in primates suggest a reprogramming of the epigenetic landscape, which may lead to dysregulated responses and functionalities in macrophages derived from primed monocytes that are recruited to sites of inflammation.
Collapse
Affiliation(s)
- John D Short
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Sina Tavakoli
- Department of Radiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Huynh Nga Nguyen
- Department of Biochemistry and Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Ana Carrera
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Chelbee Farnen
- Department of Molecular Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Laura A Cox
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX, United States.,Southwest National Primate Research Center, San Antonio, TX, United States
| | - Reto Asmis
- Department of Radiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.,Department of Biochemistry and Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.,Department of Clinical Laboratory Sciences, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
63
|
Dynamic thiol/disulphide homeostasis as a novel indicator of oxidative stress in obese children and its relationship with inflammatory-cardiovascular markers. Anatol J Cardiol 2017; 18:361-369. [PMID: 28761018 PMCID: PMC5731286 DOI: 10.14744/anatoljcardiol.2017.7740] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Objective: Childhood obesity is an important cause of cardiovascular risk with chronic inflammation. Oxidative stress may contribute to the pathogenesis of obesity-related cardiovascular pathologies. We aimed to evaluate thiol/disulphide homeostasis as a novel and sensitive marker of oxidative stress and to evaluate its relationship with some inflammatory and cardiovascular markers in obese children. Methods: In this case-controlled study, 65 children with exogenous obesity and 64 healthy children, as a control group, were included. In both groups, thiol/disulphide homeostasis parameters and inflammatory (white blood cells, platelets, mean corpuscular volume, neutrophil/lymphocyte ratio, and high-sensitivity C-reactive protein) and cardiovascular (epicardial adipose tissue thickness and left ventricular mass index) markers were studied. Correlation analyses of thiol/disulphide homeostasis parameters with body mass index standard deviation scores (BMI SDS) and inflammatory and cardiovascular markers were performed. Receiver-operating characteristic analysis was performed to determine the sensitivity, specificity, and optimal cut-off values of thiol/disulphide homeostasis parameters. Results: Native thiol, total thiol, and native thiol/total thiol ratios (antioxidant parameters) were lower (p<0.05) and disulphide/native thiol and disulphide/total thiol ratios (oxidant parameters) were higher in the obese group than in the control group (p<0.01). A positive correlation of oxidant parameters with BMI SDS and inflammatory markers was found. However, a negative correlation of antioxidant parameters with BMI SDS and inflammatory markers was found. The specificities of disulphide/native thiol and disulphide/total thiol ratios were higher in the obese group. Conclusion: The impairment in thiol/disulphide homeostasis, which is indicative of oxidative stress, is associated with inflammation in obesity. In addition, cardiovascular involvement may also contribute to this impairment.
Collapse
|
64
|
Avila-George K, Ramos-Olivares K, Vasquez-Munoz K, Villanueva-Morales V, Reyes-Farias M, Quintero P, Garcia L, Garcia-Diaz DF. Chemically induced hypoxia promotes differential outcomes over preadipocyte- or adipocyte-macrophage communication. Arch Physiol Biochem 2017; 123:175-181. [PMID: 28276712 DOI: 10.1080/13813455.2017.1285318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Expansion of white adipose tissue induce insufficient vascularization, driving hypoxia and low-grade inflammation. Resident preadipocytes are thought to be involved. We evaluated the effects of hypoxia over preadipocytes and adipocytes, to determine which cellular type impacts the most over macrophages activation. 3T3-L1 cells were either differentiated, or maintained undifferentiated. Each group was subjected to the presence or absence of chemical hypoxia (200 μM CoCl2) for 24 h. Conditioned media were used as treatment for murine RAW264.7 macrophages for 24 h. Gene expression of HIF-1α and TNF-α, and the release of several markers were assessed. It was observed that culture media from hypoxic preadipocytes induced greater expression of inflammatory markers and NO release than culture media from hypoxic adipocytes, by macrophages. Gene expression correlated closer with inflammatory markers release specially on macrophages treated with conditioned media from preadipocytes. Hence, the present work highlights the importance of preadipocytes on inflammatory conditions in vitro.
Collapse
Affiliation(s)
- K Avila-George
- a Department of Nutrition , Faculty of Medicine, University of Chile , Santiago , Chile
| | - K Ramos-Olivares
- a Department of Nutrition , Faculty of Medicine, University of Chile , Santiago , Chile
| | - K Vasquez-Munoz
- a Department of Nutrition , Faculty of Medicine, University of Chile , Santiago , Chile
| | - V Villanueva-Morales
- a Department of Nutrition , Faculty of Medicine, University of Chile , Santiago , Chile
| | - M Reyes-Farias
- a Department of Nutrition , Faculty of Medicine, University of Chile , Santiago , Chile
| | - P Quintero
- b Department of Gastroenterology , Faculty of Medicine, Pontifical Catholic University of Chile , Santiago , Chile , and
| | - L Garcia
- c Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, University of Chile , Santiago , Chile
| | - D F Garcia-Diaz
- a Department of Nutrition , Faculty of Medicine, University of Chile , Santiago , Chile
| |
Collapse
|
65
|
Ramakrishnan VM, Boyd NL. The Adipose Stromal Vascular Fraction as a Complex Cellular Source for Tissue Engineering Applications. TISSUE ENGINEERING PART B-REVIEWS 2017; 24:289-299. [PMID: 28316259 DOI: 10.1089/ten.teb.2017.0061] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
A major challenge in tissue engineering is the generation of sufficient volumes of viable tissue for organ transplant. The development of a stable, mature vasculature is required to sustain the metabolic and functional activities of engineered tissues. Adipose stromal vascular fraction (SVF) cells are an easily accessible, heterogeneous cell system comprised of endothelial cells, macrophages, pericytes, and various stem cell populations. Collectively, SVF has been shown to spontaneously form vessel-like networks in vitro and robust, patent, and functional vasculatures in vivo. Capitalizing on this ability, we and others have demonstrated adipose SVF's utility in generating and augmenting engineered liver, cardiac, and vascular tissues, to name a few. This review highlights the scientific origins of SVF, the use of SVF as a clinically relevant vascular source, various SVF constituents and their roles, and practical considerations associated with isolating SVF for various tissue engineering applications.
Collapse
Affiliation(s)
- Venkat M Ramakrishnan
- Cardiovascular Innovation Institute, Department of Physiology, University of Louisville School of Medicine , Louisville, Kentucky
| | - Nolan L Boyd
- Cardiovascular Innovation Institute, Department of Physiology, University of Louisville School of Medicine , Louisville, Kentucky
| |
Collapse
|
66
|
Cayres SU, Agostinete RR, de Moura Mello Antunes B, Lira FS, Fernandes RA. Impact of physical exercise/activity on vascular structure and inflammation in pediatric populations: A literature review. J SPEC PEDIATR NURS 2016; 21:99-108. [PMID: 27250102 DOI: 10.1111/jspn.12149] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 04/07/2016] [Accepted: 04/25/2016] [Indexed: 02/02/2023]
Abstract
PURPOSE To describe the effects of physical exercise/activity on the vascular architecture of children and adolescents, as well as to identify the effects of inflammation and sedentary behaviors on this relationship. METHODS Potentially relevant articles were identified in the databases MEDLINE and PubMed covering the period from 2000 to 2015. No language restrictions were applied. RESULTS Thirteen articles were found that included obese boys and girls in their samples (aged 9-19). Six interventional studies assessed inflammation and in five of these, physical exercise decreased inflammation. In 10 studies, vascular architecture was affected by physical exercise/activity. CONCLUSIONS The impact of physical exercise on vascular architecture and inflammation seems relevant, but has been mainly investigated in obese groups. PRACTICAL IMPLICATIONS Health professionals should act together in organized interventions in schools, targeting the promotion of higher physical activity levels in children and adolescents.
Collapse
Affiliation(s)
- Suziane Ungari Cayres
- Suziane Ungari Cayres, MSc, is PhD Student in Post-Graduate Program in Movement Sciences, São Paulo State University, Rio Claro, São Paulo, Brazil
| | - Ricardo Ribeiro Agostinete
- Ricardo Ribeiro Agostinete is Master's Degree Student in the Post-Graduate Program in Physiotherapy, São Paulo State University, Presidente Prudente, São Paulo, Brazil
| | - Barbara de Moura Mello Antunes
- Barbara de Moura Mello Antunes, MSc, is PhD Student in the Post-Graduate Program in Movement Sciences, São Paulo State University, Rio Claro, São Paulo, Brazil
| | - Fabio Santos Lira
- Fabio Santos de Lira, PhD, is Assistant Professor, Exercise and Immunometabolism Research Group, Department of Physical Education, São Paulo State University, UNESP, Presidente Prudente, São Paulo, Brazil
| | - Rômulo Araújo Fernandes
- Romulo Araújo Fernandes, PhD, is Assistant Professor, Laboratory of Investigation in Exercise, Department of Physical Education, Sao Paulo State University, São Paulo, Brazil
| |
Collapse
|
67
|
Vijayakanthi N, Greally JM, Rastogi D. Pediatric Obesity-Related Asthma: The Role of Metabolic Dysregulation. Pediatrics 2016; 137:peds.2015-0812. [PMID: 27244776 PMCID: PMC4845863 DOI: 10.1542/peds.2015-0812] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/19/2015] [Indexed: 12/15/2022] Open
Abstract
The burden of obesity-related asthma among children, particularly among ethnic minorities, necessitates an improved understanding of the underlying disease mechanisms. Although obesity is an independent risk factor for asthma, not all obese children develop asthma. Several recent studies have elucidated mechanisms, including the role of diet, sedentary lifestyle, mechanical fat load, and adiposity-mediated inflammation that may underlie the obese asthma pathophysiology. Here, we review these recent studies and emerging scientific evidence that suggest metabolic dysregulation may play a role in pediatric obesity-related asthma. We also review the genetic and epigenetic factors that may underlie susceptibility to metabolic dysregulation and associated pulmonary morbidity among children. Lastly, we identify knowledge gaps that need further exploration to better define pathways that will allow development of primary preventive strategies for obesity-related asthma in children.
Collapse
Affiliation(s)
| | - John M. Greally
- Departments of Pediatrics,,Genetics, and,Medicine, Albert Einstein College of Medicine, Bronx, New York
| | | |
Collapse
|
68
|
Abstract
Inflammation originating from the adipose tissue is considered to be one of the main driving forces for the development of insulin resistance and type 2 diabetes in obese individuals. Although a plethora of different immune cells shapes adipose tissue inflammation, this review is specifically focused on the contribution of macrophages that reside in adipose tissue in lean and obese conditions. Both conventional and tissue-specific functions of adipose tissue macrophages (ATMs) in lean and obese adipose tissue are discussed and linked with metabolic and inflammatory changes that occur during the development of obesity. Furthermore, we will address various circulating and adipose tissue-derived triggers that may be involved in shaping the ATM phenotype and underlie ATM function in lean and obese conditions. Finally, we will highlight how these changes affect adipose tissue inflammation and may be targeted for therapeutic interventions to improve insulin sensitivity in obese individuals.
Collapse
Affiliation(s)
- Lily Boutens
- Department of Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
- Nutrition, Metabolism and Genomics Group, Wageningen University, Bomenweg 2, 6703 HD, Wageningen, the Netherlands
| | - Rinke Stienstra
- Department of Medicine, Radboud University Medical Center, Nijmegen, the Netherlands.
- Nutrition, Metabolism and Genomics Group, Wageningen University, Bomenweg 2, 6703 HD, Wageningen, the Netherlands.
| |
Collapse
|
69
|
Pachón-Peña G, Serena C, Ejarque M, Petriz J, Duran X, Oliva-Olivera W, Simó R, Tinahones FJ, Fernández-Veledo S, Vendrell J. Obesity Determines the Immunophenotypic Profile and Functional Characteristics of Human Mesenchymal Stem Cells From Adipose Tissue. Stem Cells Transl Med 2016; 5:464-75. [PMID: 26956208 DOI: 10.5966/sctm.2015-0161] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 11/19/2015] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED Adipose tissue is a major source of mesenchymal stem cells (MSCs), which possess a variety of properties that make them ideal candidates for regenerative and immunomodulatory therapies. Here, we compared the immunophenotypic profile of human adipose-derived stem cells (hASCs) from lean and obese individuals, and explored its relationship with the apparent altered plasticity of hASCs. We also hypothesized that persistent hypoxia treatment of cultured hASCs may be necessary but not sufficient to drive significant changes in mature adipocytes. hASCs were obtained from subcutaneous adipose tissue of healthy, adult, female donors undergoing abdominal plastic surgery: lean (n=8; body mass index [BMI]: 23±1 kg/m2) and obese (n=8; BMI: 35±5 kg/m2). Cell surface marker expression, proliferation and migration capacity, and adipogenic differentiation potential of cultured hASCs at two different oxygen conditions were studied. Compared with lean-derived hASCs, obese-derived hASCs demonstrated increased proliferation and migration capacity but decreased lipid droplet accumulation, correlating with a higher expression of human leukocyte antigen (HLA)-II and cluster of differentiation (CD) 106 and lower expression of CD29. Of interest, adipogenic differentiation modified CD106, CD49b, HLA-ABC surface protein expression, which was dependent on the donor's BMI. Additionally, low oxygen tension increased proliferation and migration of lean but not obese hASCs, which correlated with an altered CD36 and CD49b immunophenotypic profile. In summary, the differences observed in proliferation, migration, and differentiation capacity in obese hASCs occurred in parallel with changes in cell surface markers, both under basal conditions and during differentiation. Therefore, obesity is an important determinant of stem cell function independent of oxygen tension. SIGNIFICANCE The obesity-related hypoxic environment may have latent effects on human adipose tissue-derived mesenchymal stem cells (hASCs) with potential consequences in mature cells. This study explores the immunophenotypic profile of hASCs obtained from lean and obese individuals and its potential relationship with the altered plasticity of hASCs observed in obesity. In this context, an altered pattern of cell surface marker expression in obese-derived hASCs in both undifferentiated and differentiated stages is demonstrated. Differences in proliferation, migration, and differentiation capacity of hASCs from obese adipose tissue correlated with alterations in cell surface expression. Remarkably, altered plasticity observed in obese-derived hASCs was maintained in the absence of hypoxia, suggesting that these cells might be obesity conditioned.
Collapse
Affiliation(s)
- Gisela Pachón-Peña
- Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili Universitat Rovira i Virgili, Tarragona, Spain CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Carolina Serena
- Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili Universitat Rovira i Virgili, Tarragona, Spain CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Miriam Ejarque
- Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili Universitat Rovira i Virgili, Tarragona, Spain CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Jordi Petriz
- Institut de Recerca Contra la Leucemia Josep Carreras, Barcelona, Spain
| | - Xevi Duran
- Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili Universitat Rovira i Virgili, Tarragona, Spain CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - W Oliva-Olivera
- Hospital Universitario Virgen de la Victoria, Instituto de Investigaciones Biomédicas de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain CIBER de Fisiopatología de Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Rafael Simó
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Francisco J Tinahones
- Hospital Universitario Virgen de la Victoria, Instituto de Investigaciones Biomédicas de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain CIBER de Fisiopatología de Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Sonia Fernández-Veledo
- Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili Universitat Rovira i Virgili, Tarragona, Spain CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Joan Vendrell
- Hospital Universitari de Tarragona Joan XXIII, Institut d´Investigació Sanitària Pere Virgili Universitat Rovira i Virgili, Tarragona, Spain CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
70
|
Muir LA, Neeley CK, Meyer KA, Baker NA, Brosius AM, Washabaugh AR, Varban OA, Finks JF, Zamarron BF, Flesher CG, Chang JS, DelProposto JB, Geletka L, Martinez-Santibanez G, Kaciroti N, Lumeng CN, O'Rourke RW. Adipose tissue fibrosis, hypertrophy, and hyperplasia: Correlations with diabetes in human obesity. Obesity (Silver Spring) 2016; 24:597-605. [PMID: 26916240 PMCID: PMC4920141 DOI: 10.1002/oby.21377] [Citation(s) in RCA: 236] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Revised: 09/19/2015] [Accepted: 09/26/2015] [Indexed: 12/16/2022]
Abstract
OBJECTIVE The relationship between adipose tissue fibrosis, adipocyte hypertrophy, and preadipocyte hyperplasia in the context of obesity and the correlation of these tissue-based phenomena with systemic metabolic disease are poorly defined. The goal of this study was to clarify the relationship between adipose tissue fibrosis, adipocyte hypertrophy, and preadipocyte hyperplasia in human obesity and determine the correlation of these adipose-tissue based phenomena with diabetes. METHODS Visceral and subcutaneous adipose tissues from humans with obesity collected during bariatric surgery were studied with QRTPCR, immunohistochemistry, and flow cytometry for expression of collagens and fibrosis-related proteins, adipocyte size, and preadipocyte frequency. Results were correlated with clinical characteristics including diabetes status. RESULTS Fibrosis was decreased, hypertrophy was increased, and preadipocyte frequency and fibrotic gene expression were decreased in adipose tissues from diabetic subjects compared to non-diabetic subjects. These differences were greater in visceral compared to subcutaneous adipose tissue. CONCLUSIONS These data are consistent with the hypothesis that adipose tissue fibrosis in the context of human obesity limits adipocyte hypertrophy and is associated with a reciprocal increase in adipocyte hyperplasia, with beneficial effects on systemic metabolism. These findings suggest adipose tissue fibrosis as a potential target for manipulation of adipocyte metabolism.
Collapse
Affiliation(s)
- Lindsey A. Muir
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Kevin A. Meyer
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Nicki A. Baker
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Alice M. Brosius
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Oliver A. Varban
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jonathan F. Finks
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Brian F. Zamarron
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Carmen G. Flesher
- Undergraduate Research Opportunity Program, University of Michigan, Ann Arbor, MI, USA
| | - Joshua S. Chang
- Undergraduate Research Opportunity Program, University of Michigan, Ann Arbor, MI, USA
| | - Jennifer B. DelProposto
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lynn Geletka
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gabriel Martinez-Santibanez
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
- Graduate Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Niko Kaciroti
- Center for Human Growth and Development, University of Michigan, Ann Arbor, MI, USA
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Carey N. Lumeng
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
- Graduate Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Robert W. O'Rourke
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Surgery, Ann Arbor Veteran's Administration Hospital, Ann Arbor, MI, USA
| |
Collapse
|
71
|
Acosta JR, Douagi I, Andersson DP, Bäckdahl J, Rydén M, Arner P, Laurencikiene J. Increased fat cell size: a major phenotype of subcutaneous white adipose tissue in non-obese individuals with type 2 diabetes. Diabetologia 2016; 59:560-70. [PMID: 26607638 DOI: 10.1007/s00125-015-3810-6] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 10/26/2015] [Indexed: 01/01/2023]
Abstract
AIMS/HYPOTHESIS We aimed to elucidate the impact of fat cell size and inflammatory status of adipose tissue on the development of type 2 diabetes in non-obese individuals. METHODS We characterised subcutaneous abdominal adipose tissue by examining stromal cell populations by 13 colour flow cytometry, measuring expression of adipogenesis genes in the progenitor cell fraction and determining lipolysis and adipose secretion of inflammatory proteins in 14 non-obese men with type 2 diabetes and 13 healthy controls matched for age, sex, body weight and total fat mass. RESULTS Individuals with diabetes had larger fat cells than the healthy controls but stromal cell population frequencies, adipose lipolysis and secretion of inflammatory proteins did not differ between the two groups. However, in the entire cohort fat cell size correlated positively with the ratio of M1/M2 macrophages, TNF-α secretion, lipolysis and insulin resistance. Expression of genes encoding regulators of adipogenesis and adipose morphology (BMP4, CEBPα [also known as CEBPA], PPARγ [also known as PPARG] and EBF1) correlated negatively with fat cell size. CONCLUSIONS/INTERPRETATION We show that a major phenotype of white adipose tissue in non-obese individuals with type 2 diabetes is adipocyte hypertrophy, which may be mediated by an impaired adipogenic capacity in progenitor cells. Consequently, this could have an impact on adipose tissue inflammation, release of fatty acids, ectopic fat deposition and insulin sensitivity.
Collapse
Affiliation(s)
- Juan R Acosta
- Lipid laboratory, Novum, D4, Department of Medicine Huddinge, Karolinska Institutet, Hälsovägen 7, 14186, Stockholm, Sweden
| | - Iyadh Douagi
- Center of Hematology and Regenerative Medicine, Novum, D4, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Daniel P Andersson
- Lipid laboratory, Novum, D4, Department of Medicine Huddinge, Karolinska Institutet, Hälsovägen 7, 14186, Stockholm, Sweden
| | - Jesper Bäckdahl
- Lipid laboratory, Novum, D4, Department of Medicine Huddinge, Karolinska Institutet, Hälsovägen 7, 14186, Stockholm, Sweden
| | - Mikael Rydén
- Lipid laboratory, Novum, D4, Department of Medicine Huddinge, Karolinska Institutet, Hälsovägen 7, 14186, Stockholm, Sweden
| | - Peter Arner
- Lipid laboratory, Novum, D4, Department of Medicine Huddinge, Karolinska Institutet, Hälsovägen 7, 14186, Stockholm, Sweden
| | - Jurga Laurencikiene
- Lipid laboratory, Novum, D4, Department of Medicine Huddinge, Karolinska Institutet, Hälsovägen 7, 14186, Stockholm, Sweden.
| |
Collapse
|
72
|
Vehapoglu A, Turkmen S, Goknar N, Özer ÖF. Reduced antioxidant capacity and increased subclinical inflammation markers in prepubescent obese children and their relationship with nutritional markers and metabolic parameters. Redox Rep 2016; 21:271-80. [PMID: 26865084 DOI: 10.1080/13510002.2015.1133035] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE There are associations between some inflammatory and oxidative markers and obesity in adults, but whether prepubescent children of different weights also have such markers has not been studied. We investigated multiple inflammatory markers and levels of erythrocyte oxidant/antioxidant enzymes in prepubescent children of different weights. METHODS Children aged 2-11 years were divided into three groups: 80 were underweight, 90 were obese but otherwise healthy, and 80 were healthy age- and sex-matched children of normal-weight. We analyzed inflammatory markers and the total oxidant status, total antioxidant status (TAS), and total thiol level were also determined, and the oxidative stress index was calculated as an indicator of the degree of oxidative stress. RESULTS The obese group exhibited higher levels of fasting glucose, insulin, total cholesterol, triglycerides, the homeostatic model assessment of insulin resistance (HOMA-IR), and the homeostatic model assessment of β-cell function (HOMA-β), C-reactive protein (CRP), neutrophils, and neutrophil/lymphocyte ratio (NLR), as well as lower TAS and total thiol levels than the other two groups (all P < 0.001). Moreover, TAS and total thiols were negatively correlated with age in the obese group (r = -0.212, P = 0.001; r = -0.231, P < 0.001, respectively). CRP levels in plasma were positively correlated with the body mass index (BMI), insulin and glucose levels, HOMA-IR, HOMA-β, WBC and neutrophil counts, and the NLR, and were negatively correlated with TAS and total thiol levels in the overall studied population. DISCUSSION The coexistence of increased obesity-related subclinical inflammation and decreased antioxidant capacity can be observed even in prepubescence, and may eventually increase the risk of long-term vascular damage.
Collapse
Affiliation(s)
- Aysel Vehapoglu
- a Department of Pediatrics, Faculty of Medicine , Bezmialem Vakıf University , Istanbul , Turkey
| | - Serdar Turkmen
- b Department of Biochemistry , Gaziosmanpaşa Taksim Training and Research Hospital , Istanbul , Turkey
| | - Nilufer Goknar
- a Department of Pediatrics, Faculty of Medicine , Bezmialem Vakıf University , Istanbul , Turkey
| | - Ömer Faruk Özer
- c Department of Biochemistry, School of Medicine , Bezmialem Vakıf University , Istanbul , Turkey
| |
Collapse
|
73
|
González-Muniesa P, Garcia-Gerique L, Quintero P, Arriaza S, Lopez-Pascual A, Martinez JA. Effects of Hyperoxia on Oxygen-Related Inflammation with a Focus on Obesity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:8957827. [PMID: 26697142 PMCID: PMC4678090 DOI: 10.1155/2016/8957827] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/29/2015] [Accepted: 08/19/2015] [Indexed: 12/22/2022]
Abstract
Several studies have shown a pathological oxygenation (hypoxia/hyperoxia) on the adipose tissue in obese subjects. Additionally, the excess of body weight is often accompanied by a state of chronic low-degree inflammation. The inflammation phenomenon is a complex biological response mounted by tissues to combat injurious stimuli in order to maintain cell homeostasis. Furthermore, it is believed that the abnormal oxygen partial pressure occurring in adipose tissue is involved in triggering inflammatory processes. In this context, oxygen is used in modern medicine as a treatment for several diseases with inflammatory components. Thus, hyperbaric oxygenation has demonstrated beneficial effects, apart from improving local tissue oxygenation, on promoting angiogenesis, wound healing, providing neuroprotection, facilitating glucose uptake, appetite, and others. Nevertheless, an excessive hyperoxia exposure can lead to deleterious effects such as oxidative stress, pulmonary edema, and maybe inflammation. Interestingly, some of these favorable outcomes occur under high and low oxygen concentrations. Hereby, we review a potential therapeutic approach to the management of obesity as well as the oxygen-related inflammation accompanying expanded adipose tissue, based on elevated oxygen concentrations. To conclude, we highlight at the end of this review some areas that need further clarification.
Collapse
Affiliation(s)
- Pedro González-Muniesa
- Centre for Nutrition Research, Department of Nutrition, Food Sciences and Physiology, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Navarra's Health Research Institute (IDISNA), 31008 Pamplona, Spain
| | - Laura Garcia-Gerique
- Centre for Nutrition Research, Department of Nutrition, Food Sciences and Physiology, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain
| | - Pablo Quintero
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Libertador Bernardo O'Higgins 340, Santiago, Chile
| | - Suyen Arriaza
- Centre for Nutrition Research, Department of Nutrition, Food Sciences and Physiology, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain
| | - Amaya Lopez-Pascual
- Centre for Nutrition Research, Department of Nutrition, Food Sciences and Physiology, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain
| | - J. Alfredo Martinez
- Centre for Nutrition Research, Department of Nutrition, Food Sciences and Physiology, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Navarra's Health Research Institute (IDISNA), 31008 Pamplona, Spain
| |
Collapse
|
74
|
Snodgrass RG, Boß M, Zezina E, Weigert A, Dehne N, Fleming I, Brüne B, Namgaladze D. Hypoxia Potentiates Palmitate-induced Pro-inflammatory Activation of Primary Human Macrophages. J Biol Chem 2015; 291:413-24. [PMID: 26578520 DOI: 10.1074/jbc.m115.686709] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Indexed: 12/17/2022] Open
Abstract
Pro-inflammatory cytokines secreted by adipose tissue macrophages (ATMs) contribute to chronic low-grade inflammation and obesity-induced insulin resistance. Recent studies have shown that adipose tissue hypoxia promotes an inflammatory phenotype in ATMs. However, our understanding of how hypoxia modulates the response of ATMs to free fatty acids within obese adipose tissue is limited. We examined the effects of hypoxia (1% O2) on the pro-inflammatory responses of human monocyte-derived macrophages to the saturated fatty acid palmitate. Compared with normoxia, hypoxia significantly increased palmitate-induced mRNA expression and protein secretion of IL-6 and IL-1β. Although palmitate-induced endoplasmic reticulum stress and nuclear factor κB pathway activation were not enhanced by hypoxia, hypoxia increased the activation of JNK and p38 mitogen-activated protein kinase signaling in palmitate-treated cells. Inhibition of JNK blocked the hypoxic induction of pro-inflammatory cytokine expression, whereas knockdown of hypoxia-induced transcription factors HIF-1α and HIF-2α alone or in combination failed to reduce IL-6 and only modestly reduced IL-1β gene expression in palmitate-treated hypoxic macrophages. Enhanced pro-inflammatory cytokine production and JNK activity under hypoxia were prevented by inhibiting reactive oxygen species generation. In addition, silencing of dual-specificity phosphatase 16 increased normoxic levels of IL-6 and IL-1β and reduced the hypoxic potentiation in palmitate-treated macrophages. The secretome of hypoxic palmitate-treated macrophages promoted IL-6 and macrophage chemoattractant protein 1 expression in primary human adipocytes, which was sensitive to macrophage JNK inhibition. Our results reveal that the coexistence of hypoxia along with free fatty acids exacerbates macrophage-mediated inflammation.
Collapse
Affiliation(s)
| | - Marcel Boß
- From the Institute of Biochemistry I and
| | | | | | | | - Ingrid Fleming
- Institute for Vascular Signaling, Center for Molecular Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | | | | |
Collapse
|
75
|
Ceperuelo-Mallafré V, Ejarque M, Serena C, Duran X, Montori-Grau M, Rodríguez MA, Yanes O, Núñez-Roa C, Roche K, Puthanveetil P, Garrido-Sánchez L, Saez E, Tinahones FJ, Garcia-Roves PM, Gómez-Foix AM, Saltiel AR, Vendrell J, Fernández-Veledo S. Adipose tissue glycogen accumulation is associated with obesity-linked inflammation in humans. Mol Metab 2015; 5:5-18. [PMID: 26844203 PMCID: PMC4703799 DOI: 10.1016/j.molmet.2015.10.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 10/02/2015] [Accepted: 10/09/2015] [Indexed: 01/14/2023] Open
Abstract
Objective Glycogen metabolism has emerged as a mediator in the control of energy homeostasis and studies in murine models reveal that adipose tissue might contain glycogen stores. Here we investigated the physio(patho)logical role of glycogen in human adipose tissue in the context of obesity and insulin resistance. Methods We studied glucose metabolic flux of hypoxic human adipoctyes by nuclear magnetic resonance and mass spectrometry-based metabolic approaches. Glycogen synthesis and glycogen content in response to hypoxia was analyzed in human adipocytes and macrophages. To explore the metabolic effects of enforced glycogen deposition in adipocytes and macrophages, we overexpressed PTG, the only glycogen-associated regulatory subunit (PP1-GTS) reported in murine adipocytes. Adipose tissue gene expression analysis was performed on wild type and homozygous PTG KO male mice. Finally, glycogen metabolism gene expression and glycogen accumulation was analyzed in adipose tissue, mature adipocytes and resident macrophages from lean and obese subjects with different degrees of insulin resistance in 2 independent cohorts. Results We show that hypoxia modulates glucose metabolic flux in human adipocytes and macrophages and promotes glycogenesis. Enforced glycogen deposition by overexpression of PTG re-orients adipocyte secretion to a pro-inflammatory response linked to insulin resistance and monocyte/lymphocyte migration. Furthermore, glycogen accumulation is associated with inhibition of mTORC1 signaling and increased basal autophagy flux, correlating with greater leptin release in glycogen-loaded adipocytes. PTG-KO mice have reduced expression of key inflammatory genes in adipose tissue and PTG overexpression in M0 macrophages induces a pro-inflammatory and glycolytic M1 phenotype. Increased glycogen synthase expression correlates with glycogen deposition in subcutaneous adipose tissue of obese patients. Glycogen content in subcutaneous mature adipocytes is associated with BMI and leptin expression. Conclusion Our data establish glycogen mishandling in adipose tissue as a potential key feature of inflammatory-related metabolic stress in human obesity. Hypoxia redirects extracellular glucose to glycogen synthesis in human adipocytes. Glycogen modifies the endocrine function of adipocytes and induces insulin resistance. Glycogen stimulates leptin secretion through an autophagy-dependent mechanism. Enforced glycogen accumulation in macrophages promotes M1 polarization. Obesity is associated with higher GS expression and glycogen stores in adipose tissue.
Collapse
Affiliation(s)
- Victòria Ceperuelo-Mallafré
- Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Miriam Ejarque
- Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Carolina Serena
- Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Xavier Duran
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Montori-Grau
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Miguel Angel Rodríguez
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Centre for Omic Sciences (COS), Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona, Spain
| | - Oscar Yanes
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Department of Electronic Engineering, Universitat Rovira i Virgili, Tarragona, Spain
| | - Catalina Núñez-Roa
- Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Kelly Roche
- Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Lourdes Garrido-Sánchez
- Hospital Universitario Virgen de la Victoria, Instituto de Investigaciones Biomédicas de Málaga (IBIMA), Universidad de Málaga, IBIMA, Spain
- CIBER de Fisiopatología de Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Enrique Saez
- Department of Chemical Physiology and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Francisco J. Tinahones
- Hospital Universitario Virgen de la Victoria, Instituto de Investigaciones Biomédicas de Málaga (IBIMA), Universidad de Málaga, IBIMA, Spain
- CIBER de Fisiopatología de Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Pablo M. Garcia-Roves
- Departamento de Ciencias Fisiológicas II, Facultad de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Anna Ma Gómez-Foix
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Alan R. Saltiel
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Joan Vendrell
- Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Corresponding authors. Research Unit, University Hospital of Tarragona Joan XXIII, c/o Dr. Mallafré Guasch, 4, 43007 Tarragona. Spain. Tel.: +34 977 29 58 00; fax: +34 977 29 58 23.
| | - Sonia Fernández-Veledo
- Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
- Corresponding authors. Research Unit, University Hospital of Tarragona Joan XXIII, c/o Dr. Mallafré Guasch, 4, 43007 Tarragona. Spain. Tel.: +34 977 29 58 00; fax: +34 977 29 58 23.
| |
Collapse
|
76
|
DeClercq V, d'Eon B, McLeod RS. Fatty acids increase adiponectin secretion through both classical and exosome pathways. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:1123-33. [DOI: 10.1016/j.bbalip.2015.04.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 04/06/2015] [Accepted: 04/13/2015] [Indexed: 11/26/2022]
|
77
|
Aumueller E, Remely M, Baeck H, Hippe B, Brath H, Haslberger AG. Interleukin-6 CpG Methylation and Body Weight Correlate Differently in Type 2 Diabetes Patients Compared to Obese and Lean Controls. JOURNAL OF NUTRIGENETICS AND NUTRIGENOMICS 2015; 8:26-35. [PMID: 26067576 DOI: 10.1159/000381714] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 03/17/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Diabetes mellitus type 2 (DMT2) is accompanied by systemic low-grade inflammation with elevated levels of interleukin-6 (IL-6), which is encoded by a gene (IL-6) previously shown to be regulated by DNA methylation. We investigated seven CpG sites in IL-6 in individuals with DMT2, obese individuals and lean controls. Further, the DMT2 group received the glucagon-like peptide 1 agonist liraglutide. METHODS Blood samples were taken at the beginning of the study and after 4 months. The DNA methylation was assessed using pyrosequencing. RESULTS Methylation levels at the CpG sites -664, -628 and +13 at the first sampling time point (T1) and at -666 and -664 at the second sampling time point (T2) correlated negatively with initial body weight in the DMT2 group. We found positive correlations for the obese and the lean control group. In the obese group, CpG +27 methylation at T1 correlated with initial body weight (r = 0.685; p = 0.014). In the lean group, CpG -664 at T1 (r = 0.874; p = 0.005) and CpG -628 at T2 (r = 0.632; p = 0.050) correlated with initial body weight. CONCLUSION These findings are an informative basis for further studies to elucidate epigenetic mechanisms underlying DMT2. Additionally, our results might provide starting points for the development of biomarkers for prevention and therapy strategies.
Collapse
Affiliation(s)
- Eva Aumueller
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria
| | | | | | | | | | | |
Collapse
|
78
|
Arriarán S, Agnelli S, Sabater D, Remesar X, Fernández-López JA, Alemany M. Evidences of basal lactate production in the main white adipose tissue sites of rats. Effects of sex and a cafeteria diet. PLoS One 2015; 10:e0119572. [PMID: 25741703 PMCID: PMC4351194 DOI: 10.1371/journal.pone.0119572] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 01/15/2015] [Indexed: 12/25/2022] Open
Abstract
Female and male adult Wistar rats were fed standard chow or a simplified cafeteria diet for one month. Then, the rats were killed and the white adipose tissue (WAT) in four sites: perigonadal, retroperitoneal, mesenteric and subcutaneous (inguinal) were sampled and frozen. The complete WAT weight in each site was measured. Gene expression analysis of key lipid and glucose metabolism enzymes were analyzed, as well as tissue and plasma lactate and the activity of lactate dehydrogenase. Lactate gradients between WAT and plasma were estimated. The influence of sex and diet (and indirectly WAT mass) on lactate levels and their relationships with lactate dehydrogenase activity and gene expressions were also measured. A main conclusion is the high production of lactate by WAT, practically irrespective of site, diet or sex. Lactate production is a direct correlate of lactate dehydrogenase activity in the tissue. Furthermore, lactate dehydrogenase activity is again directly correlated with the expression of the genes Ldha and Ldhb for this enzyme. In sum, the ability to produce lactate by WAT is not directly dependent of WAT metabolic state. We postulate that, in WAT, a main function of the lactate dehydrogenase path may be that of converting excess available glucose to 3C fragments, as a way to limit tissue self-utilization as substrate, to help control glycaemia and/or providing short chain substrates for use as energy source elsewhere. More information must be gathered before a conclusive role of WAT in the control of glycaemia, and the full existence of a renewed glucose-lactate-fatty acid cycle is definitely established.
Collapse
Affiliation(s)
- Sofía Arriarán
- Department of Nutrition and Food Science, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Silvia Agnelli
- Department of Nutrition and Food Science, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - David Sabater
- Department of Nutrition and Food Science, Faculty of Biology, University of Barcelona, Barcelona, Spain
- CIBER Obesity and Nutrition, Barcelona, Spain
| | - Xavier Remesar
- Department of Nutrition and Food Science, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona, Barcelona, Spain
- CIBER Obesity and Nutrition, Barcelona, Spain
| | - José Antonio Fernández-López
- Department of Nutrition and Food Science, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona, Barcelona, Spain
- CIBER Obesity and Nutrition, Barcelona, Spain
- * E-mail:
| | - Marià Alemany
- Department of Nutrition and Food Science, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona, Barcelona, Spain
- CIBER Obesity and Nutrition, Barcelona, Spain
| |
Collapse
|
79
|
Abstract
The prevalence of diabetes mellitus and obesity continues to increase globally. Diabetic vascular complications are the main chronic diabetic complications and associated with mortality and disability. Angiogenesis is a key pathological characteristic of diabetic microvascular complications. However, there are two tissue-specific paradoxical changes in the angiogenesis in diabetic microvascular complications: an excessive uncontrolled formation of premature blood vessels in some tissues, such as the retina, and a deficiency in the formation of small blood vessels in peripheral tissues, such as the skin. This review will discuss the paradoxical phenomena of angiogenesis and its underlying mechanism in obesity, diabetes and diabetic complications.
Collapse
Affiliation(s)
| | - Jian-xing Ma
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
80
|
Goossens GH, Blaak EE. Adipose tissue dysfunction and impaired metabolic health in human obesity: a matter of oxygen? Front Endocrinol (Lausanne) 2015; 6:55. [PMID: 25964776 PMCID: PMC4408910 DOI: 10.3389/fendo.2015.00055] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 04/06/2015] [Indexed: 12/23/2022] Open
Affiliation(s)
- Gijs H. Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, Netherlands
- *Correspondence: Gijs H. Goossens
| | - Ellen E. Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, Netherlands
| |
Collapse
|
81
|
Varlamov O, Chu M, Cornea A, Sampath H, Roberts CT. Cell-autonomous heterogeneity of nutrient uptake in white adipose tissue of rhesus macaques. Endocrinology 2015; 156:80-9. [PMID: 25356825 PMCID: PMC4272393 DOI: 10.1210/en.2014-1699] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Phenotypic diversity may play an adaptive role by providing graded biological responses to fluctuations in environmental stimuli. We used single-cell imaging of the metabolizable fluorescent fatty acid analog 4,4-difluoro-4-bora-3a,4a-diaza-s-indacene (BODIPY)-C12 and fluorescent 2-[N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl) amino]-2-deoxy-D-glucose (2-NBDG) to explore cellular heterogeneity in nutrient uptake in white adipose tissue (WAT) explants of rhesus macaques. Surprisingly, WAT displayed a striking cell size-independent mosaic pattern, in that adjacent adipocytes varied with respect to insulin-stimulated BODIPY-C12 and 2-NBDG uptake. Relative free fatty acid (FFA) transport activity correlated with the cellular levels of FFA transporter protein-1 and the scavenger receptor CD36 in individual adipocytes. In vitro incubation of WAT explants for 24 hours caused partial desynchronization of cellular responses, suggesting that adipocytes may slowly alter their differential nutrient uptake activity. In vitro-differentiated human adipocytes also exhibited a mosaic pattern of BODIPY-C12 uptake. WAT from animals containing a homogeneous population of large adipocytes was nonmosaic, in that every adipocyte exhibited a similar level of BODIPY-C12 fluorescence, suggesting that the development of obesity is associated with the loss of heterogeneity in WAT. Hence, for the first time, we demonstrate an intrinsic heterogeneity in FFA and glucose transport activity in WAT.
Collapse
Affiliation(s)
- Oleg Varlamov
- Divisions of Diabetes, Obesity, and Metabolism and Developmental and Reproductive Science (O.V., C.T.R.), and Division of Neuroscience (A.C.), Oregon National Primate Research Center, Beaverton, Oregon 97006; and Division of Endocrinology, Diabetes, and Clinical Nutrition, Department of Medicine (M.C., C.T.R.) and Center for Research Occupational and Environmental Toxicology (H.S.), Oregon Health and Science University, Portland, Oregon 97239
| | | | | | | | | |
Collapse
|
82
|
Priyanka A, Anusree SS, Nisha VM, Raghu KG. Curcumin improves hypoxia induced dysfunctions in 3T3-L1 adipocytes by protecting mitochondria and down regulating inflammation. Biofactors 2014; 40:513-23. [PMID: 25110893 DOI: 10.1002/biof.1175] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 07/04/2014] [Accepted: 07/16/2014] [Indexed: 12/19/2022]
Abstract
Obesity induced metabolic syndrome is increasing worldwide at an alarming rate. It is characterized by excessive expansion of white adipose tissue which leads to hypoxia and impairs normal metabolism. Recent studies reveal that hypoxia could be one of the factors for inflammation, insulin resistance and other obesity related complications. There is a high demand for anti-obese phytoceuticals to control and manage the complications resulting from obesity. In this study, we investigated how hypoxia affect the physiological functions of 3T3-L1 adipocytes emphasizing on oxidative stress, inflammation, and mitochondrial functions. We also evaluated the protective role of various doses of curcumin, a well-known dietary antioxidant, on hypoxia induced alterations. The results revealed that hypoxia significantly altered the vital parameters of adipocyte biology like HIF 1α expression (103.47% ↑), lactate, and glycerol release (184.34% and 69.1% ↑, respectively), reactive oxygen species production (432.53% ↑), lipid and protein oxidation (376.6% and 566.6% ↑, respectively), reduction in antioxidant enzymes (superoxide dismutase and catalase) status, secretion of inflammatory markers (TNF α, IL 6, IL 1β, and IFN γ), and mitochondrial functions (mitochondrial mass, membrane potential, permeability transition pore integrity, and superoxide generation). Curcumin substantially protected adipocytes from toxic effects of hypoxia in a dose dependent manner by protecting mitochondria and down regulating inflammation. Acriflavine is used as a positive control. A detailed investigation is required for the development of curcumin as an effective nutraceutical against obesity.
Collapse
Affiliation(s)
- Ariyapalli Priyanka
- Agroprocessing and Natural Products Division, Council of Scientific and Industrial Research-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, 695019, Kerala, India
| | | | | | | |
Collapse
|
83
|
Esser N, Legrand-Poels S, Piette J, Scheen AJ, Paquot N. Inflammation as a link between obesity, metabolic syndrome and type 2 diabetes. Diabetes Res Clin Pract 2014; 105:141-50. [PMID: 24798950 DOI: 10.1016/j.diabres.2014.04.006] [Citation(s) in RCA: 1260] [Impact Index Per Article: 126.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 04/07/2014] [Indexed: 12/11/2022]
Abstract
It is recognized that a chronic low-grade inflammation and an activation of the immune system are involved in the pathogenesis of obesity-related insulin resistance and type 2 diabetes. Systemic inflammatory markers are risk factors for the development of type 2 diabetes and its macrovascular complications. Adipose tissue, liver, muscle and pancreas are themselves sites of inflammation in presence of obesity. An infiltration of macrophages and other immune cells is observed in these tissues associated with a cell population shift from an anti-inflammatory to a pro-inflammatory profile. These cells are crucial for the production of pro-inflammatory cytokines, which act in an autocrine and paracrine manner to interfere with insulin signaling in peripheral tissues or induce β-cell dysfunction and subsequent insulin deficiency. Particularly, the pro-inflammatory interleukin-1β is implicated in the pathogenesis of type 2 diabetes through the activation of the NLRP3 inflammasome. The objectives of this review are to expose recent data supporting the role of the immune system in the pathogenesis of insulin resistance and type 2 diabetes and to examine various mechanisms underlying this relationship. If type 2 diabetes is an inflammatory disease, anti-inflammatory therapies could have a place in prevention and treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Nathalie Esser
- Virology and Immunology Unit, GIGA-Research, University of Liege, Liege, Belgium; Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, University Hospital of Liege, Liege, Belgium.
| | - Sylvie Legrand-Poels
- Virology and Immunology Unit, GIGA-Research, University of Liege, Liege, Belgium
| | - Jacques Piette
- Virology and Immunology Unit, GIGA-Research, University of Liege, Liege, Belgium
| | - André J Scheen
- Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, University Hospital of Liege, Liege, Belgium
| | - Nicolas Paquot
- Virology and Immunology Unit, GIGA-Research, University of Liege, Liege, Belgium; Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, University Hospital of Liege, Liege, Belgium
| |
Collapse
|
84
|
Martinez-Santibañez G, Lumeng CNK. Macrophages and the regulation of adipose tissue remodeling. Annu Rev Nutr 2014; 34:57-76. [PMID: 24850386 DOI: 10.1146/annurev-nutr-071812-161113] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The ability of adipose tissue to adapt to a changing nutrient environment is critical to the maintenance of metabolic control. Nutrient excess and deficiency alter the shape of adipose tissue drastically and trigger many events that are collectively known as adipose tissue remodeling. Remodeling of adipose tissue involves more than adipocytes and is controlled by an extensive network of stromal cells and extracellular matrix proteins. Prominent players in this process are adipose tissue macrophages, which are a specialized leukocyte present in lean and obese states that contributes to adipose tissue inflammation. The interest in adipose tissue remodeling has been accelerated by the current epidemic of obesity and the chronic generation of signals that lead to expansion of adipose tissue. It is clear that evidence of dysfunctional remodeling events is a hallmark of obesity associated with metabolic disease. This review summarizes and highlights the recent work in this area and provides a framework in which to consider how adipose tissue macrophages contribute to the remodeling events in lean and obese states. Advancing our understanding of the involvement of macrophages in adipose tissue remodeling will promote one aspect of the new field of "immunometabolism," which connects control systems developed for regulation of immunity with those that control metabolism. It will also provide insight into how physiologic and pathophysiologic remodeling differs in adipose tissue and identify potential nodes for intervention to break the link between obesity and disease.
Collapse
|
85
|
Abstract
Hypoxia develops in white adipose tissue in obese mice, resulting in changes in adipocyte function that may underpin the dysregulation that leads to obesity-associated disorders. Whether hypoxia occurs in adipose tissue in human obesity is unclear, with recent studies contradicting earlier reports that this was the case. Adipocytes, both murine and human, exhibit extensive functional changes in culture in response to hypoxia, which alters the expression of up to 1,300 genes. These include genes encoding key adipokines such as leptin, interleukin (IL)-6, vascular endothelial growth factor (VEGF), and matrix metalloproteinase-2 (MMP-2), which are upregulated, and adiponectin, which is downregulated. Hypoxia also inhibits the expression of genes linked to oxidative metabolism while stimulating the expression of genes associated with glycolysis. Glucose uptake and lactate release by adipocytes are both stimulated by hypoxia, and insulin sensitivity falls. Preadipocytes and macrophages in adipose tissue also respond to hypoxia. The hypoxia-signaling pathway may provide a new target for the treatment of obesity-associated disorders.
Collapse
Affiliation(s)
- Paul Trayhurn
- Obesity Biology Research Unit, Institute of Ageing and Chronic Diseases, University of Liverpool, Liverpool L69 3GA United Kingdom, and Clore Laboratory, University of Buckingham, Buckingham MK18 1EG, United Kingdom;
| |
Collapse
|
86
|
Frazier TP, McLachlan JB, Gimble JM, Tucker HA, Rowan BG. Human adipose-derived stromal/stem cells induce functional CD4+CD25+FoxP3+CD127- regulatory T cells under low oxygen culture conditions. Stem Cells Dev 2014; 23:968-77. [PMID: 24405386 DOI: 10.1089/scd.2013.0152] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Human adipose tissue stromal/stem cells (ASCs) are known to induce proliferation of resting T cells under ambient (21%) O2 conditions; however, ASCs exist physiologically under lower oxygen (5% O2) conditions in adipose tissue. The effects of low oxygen levels on ASC immunomodulation of T cells are unknown. In this study, we show that ASCs stimulated proliferation of naive CD4(+) T cells and the percentage of CD25(+) T cells was significantly increased under both low and ambient O2. Forkhead box P3 (FoxP3) and transforming growth factor beta (TGF-β) mRNA expression were significantly increased when ASCs were cocultured with CD4(+) T cells under low compared with ambient O2 conditions. The low O2-induced regulatory T cells (iTregs) exhibited functionality when added to mixed lymphocyte reactions as demonstrated by inhibition of peripheral blood mononuclear cell proliferation, and by >300-fold increase in FoxP3 mRNA, and >2-fold increase in TGF-β mRNA expression. These results demonstrate that under physiologically relevant low O2 conditions, direct contact of human ASCs with naive CD4(+) T cells induced functional iTregs.
Collapse
Affiliation(s)
- Trivia P Frazier
- 1 Department of Structural and Cellular Biology, Tulane University , New Orleans, Louisiana
| | | | | | | | | |
Collapse
|
87
|
The influence of deep hypothermia on inflammatory status, tissue hypoxia and endocrine function of adipose tissue during cardiac surgery. Cryobiology 2014; 68:269-75. [PMID: 24548542 DOI: 10.1016/j.cryobiol.2014.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 12/10/2013] [Accepted: 02/06/2014] [Indexed: 01/04/2023]
Abstract
Changes in endocrine function of adipose tissue during surgery, such as excessive production of proinflammatory cytokines, can significantly alter metabolic response to surgery and worsen its outcomes and prognosis of patients. Therapeutic hypothermia has been used to prevent damage connected with perioperative ischemia and hypoperfusion. The aim of our study was to explore the influence of deep hypothermia on systemic and local inflammation, adipose tissue hypoxia and adipocytokine production. We compared serum concentrations of proinflammatory markers (CRP, IL-6, IL-8, sIL-2R, sTNFRI, PCT) and mRNA expression of selected genes involved in inflammatory reactions (IL-6, TNF-α, MCP-1, MIF) and adaptation to hypoxia and oxidative stress (HIF1-α, MT3, GLUT1, IRS1, GPX1, BCL-2) in subcutaneous and visceral adipose tissue and in isolated adipocytes of patients undergoing cardiosurgical operation with hypothermic period. Deep hypothermia significantly delayed the onset of surgery-related systemic inflammatory response. The relative gene expression of the studied genes was not altered during the hypothermic period, but was significantly changed in six out of ten studied genes (IL-6, MCP-1, TNF-α, HIF1-α, GLUT1, GPX1) at the end of surgery. Our results show that deep hypothermia suppresses the development of systemic inflammatory response, delays the onset of local adipose tissue inflammation and thus may protect against excessive expression of proinflammatory and hypoxia-related factors in patients undergoing elective cardiac surgery procedure.
Collapse
|
88
|
Immune cells and metabolic dysfunction. Semin Immunopathol 2013; 36:13-25. [PMID: 24212254 DOI: 10.1007/s00281-013-0403-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 10/15/2013] [Indexed: 02/06/2023]
Abstract
Throughout evolution, effective nutrient sensing and control of systemic energy homeostasis have relied on a close physical and functional interaction between immune and metabolically active cells. However, in today's obesogenic environment, this fine-tuned immunometabolic interface is perturbed. As a consequence, chronic inflammatory conditions and aberrant activation of immune cells have emerged as key features of obesity-related metabolic disorders, including insulin resistance, cardiovascular complications, and type 2 diabetes, whereas a major research focus has been placed on the adipocyte-macrophage interaction in the context of metabolic dysfunction; recent studies have not only expanded the scope of relevant immune cells in this setting but also highlight the impact of distinct metabolic organs, including the liver, on immunometabolic control, metabolic disease development, and potential anti-inflammatory therapeutic options in obesity-driven pathologies. This review will thus summarize recent progress in this emerging area of metabolic research.
Collapse
|
89
|
Carson BP, Del Bas JM, Moreno-Navarrete JM, Fernandez-Real JM, Mora S. The rab11 effector protein FIP1 regulates adiponectin trafficking and secretion. PLoS One 2013; 8:e74687. [PMID: 24040321 PMCID: PMC3770573 DOI: 10.1371/journal.pone.0074687] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 08/07/2013] [Indexed: 12/28/2022] Open
Abstract
Adiponectin is an adipokine secreted by white adipocytes involved in regulating insulin sensitivity in peripheral tissues. Secretion of adiponectin in adipocytes relies on the endosomal system, however, the intracellular machinery involved in mediating adiponectin release is unknown. We have previously reported that intracellular adiponectin partially compartmentalizes with rab 5 and rab11, markers for the early/sorting and recycling compartments respectively. Here we have examined the role of several rab11 downstream effector proteins (rab11 FIPs) in regulating adiponectin trafficking and secretion. Overexpression of wild type rab11 FIP1, FIP3 and FIP5 decreased the amount of secreted adiponectin expressed in HEK293 cells, whereas overexpression of rab11 FIP2 or FIP4 had no effect. Furthermore shRNA-mediated depletion of FIP1 enhanced adiponectin release whereas knock down of FIP5 decreased adiponectin secretion. Knock down of FIP3 had no effect. In 3T3L1 adipocytes, endogenous FIP1 co-distributed intracellularly with endogenous adiponectin and FIP1 depletion enhanced adiponectin release without altering insulin-mediated trafficking of the glucose transporter Glut4. While adiponectin receptors internalized with transferrin receptors, there were no differences in transferrin receptor recycling between wild type and FIP1 depleted adipocytes. Consistent with its inhibitory role, FIP1 expression was decreased during adipocyte differentiation, by treatment with thiazolidinediones, and with increased BMI in humans. In contrast, FIP1 expression increased upon exposure of adipocytes to TNFα. In all, our findings identify FIP1 as a novel protein involved in the regulation of adiponectin trafficking and release.
Collapse
Affiliation(s)
- Brian P. Carson
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, the University of Liverpool, Liverpool, United Kingdom
| | - Josep Maria Del Bas
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, the University of Liverpool, Liverpool, United Kingdom
| | | | | | - Silvia Mora
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, the University of Liverpool, Liverpool, United Kingdom
- * E-mail:
| |
Collapse
|
90
|
O'Rourke RW, Meyer KA, Gaston G, White AE, Lumeng CN, Marks DL. Hexosamine biosynthesis is a possible mechanism underlying hypoxia's effects on lipid metabolism in human adipocytes. PLoS One 2013; 8:e71165. [PMID: 23967162 PMCID: PMC3743867 DOI: 10.1371/journal.pone.0071165] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 06/26/2013] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION Hypoxia regulates adipocyte metabolism. Hexosamine biosynthesis is implicated in murine 3T3L1 adipocyte differentiation and is a possible underlying mechanism for hypoxia's effects on adipocyte metabolism. METHODS Lipid metabolism was studied in human visceral and subcutaneous adipocytes in in vitro hypoxic culture with adipophilic staining, glycerol release, and palmitate oxidation assays. Gene expression and hexosamine biosynthesis activation was studied with QRTPCR, immunofluorescence microscopy, and Western blotting. RESULTS Hypoxia inhibits lipogenesis and induces basal lipolysis in visceral and subcutaneous human adipocytes. Hypoxia induces fatty acid oxidation in visceral adipocytes but had no effect on fatty acid oxidation in subcutaneous adipocytes. Hypoxia inhibits hexosamine biosynthesis in adipocytes. Inhibition of hexosamine biosynthesis with azaserine attenuates lipogenesis and induces lipolysis in adipocytes in normoxic conditions, while promotion of hexosamine biosynthesis with glucosamine in hypoxic conditions slightly increases lipogenesis. CONCLUSIONS Hypoxia's net effect on human adipocyte lipid metabolism would be expected to impair adipocyte buffering capacity and contribute to systemic lipotoxicity. Our data suggest that hypoxia may mediate its effects on lipogenesis and lipolysis through inhibition of hexosamine biosynthesis. Hexosamine biosynthesis represents a target for manipulation of adipocyte metabolism.
Collapse
Affiliation(s)
- Robert W O'Rourke
- Department of Surgery, Oregon Health and Science University, Portland, Oregon, United States of America.
| | | | | | | | | | | |
Collapse
|
91
|
Marzetti E, Calvani R, Cesari M, Buford TW, Lorenzi M, Behnke BJ, Leeuwenburgh C. Mitochondrial dysfunction and sarcopenia of aging: from signaling pathways to clinical trials. Int J Biochem Cell Biol 2013; 45:2288-301. [PMID: 23845738 DOI: 10.1016/j.biocel.2013.06.024] [Citation(s) in RCA: 386] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 06/20/2013] [Accepted: 06/26/2013] [Indexed: 12/12/2022]
Abstract
Sarcopenia, the age-related loss of muscle mass and function, imposes a dramatic burden on individuals and society. The development of preventive and therapeutic strategies against sarcopenia is therefore perceived as an urgent need by health professionals and has instigated intensive research on the pathophysiology of this syndrome. The pathogenesis of sarcopenia is multifaceted and encompasses lifestyle habits, systemic factors (e.g., chronic inflammation and hormonal alterations), local environment perturbations (e.g., vascular dysfunction), and intramuscular specific processes. In this scenario, derangements in skeletal myocyte mitochondrial function are recognized as major factors contributing to the age-dependent muscle degeneration. In this review, we summarize prominent findings and controversial issues on the contribution of specific mitochondrial processes - including oxidative stress, quality control mechanisms and apoptotic signaling - on the development of sarcopenia. Extramuscular alterations accompanying the aging process with a potential impact on myocyte mitochondrial function are also discussed. We conclude with presenting methodological and safety considerations for the design of clinical trials targeting mitochondrial dysfunction to treat sarcopenia. Special emphasis is placed on the importance of monitoring the effects of an intervention on muscle mitochondrial function and identifying the optimal target population for the trial. This article is part of a Directed Issue entitled: Molecular basis of muscle wasting.
Collapse
Affiliation(s)
- Emanuele Marzetti
- Department of Geriatrics, Neurosciences and Orthopedics, Catholic University of the Sacred Heart School of Medicine, Rome 00168, Italy.
| | | | | | | | | | | | | |
Collapse
|
92
|
Cizza G, Piaggi P, Lucassen EA, de Jonge L, Walter M, Mattingly MS, Kalish H, Csako G, Rother KI. Obstructive sleep apnea is a predictor of abnormal glucose metabolism in chronically sleep deprived obese adults. PLoS One 2013; 8:e65400. [PMID: 23734252 PMCID: PMC3667085 DOI: 10.1371/journal.pone.0065400] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 05/01/2013] [Indexed: 12/12/2022] Open
Abstract
Context Sleep abnormalities, including obstructive sleep apnea (OSA), have been associated with insulin resistance. Objective To determine the relationship between sleep, including OSA, and glucose parameters in a prospectively assembled cohort of chronically sleep-deprived obese subjects. Design Cross-sectional evaluation of a prospective cohort study. Setting Tertiary Referral Research Clinical Center. Main Outcome Measure(s) Sleep duration and quality assessed by actigraphy, sleep diaries and questionnaires, OSA determined by a portable device; glucose metabolism assessed by oral glucose tolerance test (oGTT), and HbA1c concentrations in 96 obese individuals reporting sleeping less than 6.5 h on a regular basis. Results Sixty % of subjects had an abnormal respiratory disturbance index (RDI≥5) and 44% of these subjects had abnormal oGTT results. Severity of OSA as assessed by RDI score was associated with fasting glucose (R = 0.325, p = 0.001) and fasting insulin levels (ρ = 0.217, p = 0.033). Subjects with moderate to severe OSA (RDI>15) had higher glucose concentrations at 120 min than those without OSA (RDI<5) (p = 0.017). Subjects with OSA also had significantly higher concentrations of plasma ACTH (p = 0.009). Several pro-inflammatory cytokines were higher in subjects with OSA (p<0.050). CRP levels were elevated in this sample, suggesting increased cardiovascular risk. Conclusions OSA is associated with impaired glucose metabolism in obese, sleep deprived individuals. Since sleep apnea is common and frequently undiagnosed, health care providers should be aware of its occurrence and associated risks. Trial Registration This study was conducted under the NIDDK protocol 06-DK-0036 and is listed in ClinicalTrials.gov NCT00261898
Collapse
Affiliation(s)
- Giovanni Cizza
- Section on Neuroendocrinology of Obesity, National Institute of Diabetes, Digestive, and Kidney Disease, Bethesda, Maryland, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Mathur SK, Jain P, Mathur P, Punjabi P, Agarwal A, Sharma A. Transcriptomic analysis of visceral adipose from healthy and diabetic obese subjects. Indian J Endocrinol Metab 2013; 17:446-450. [PMID: 23869300 PMCID: PMC3712374 DOI: 10.4103/2230-8210.111639] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Understanding the role of visceral fat accumulation in the occurrence and progression of metabolic syndrome is of considerable interest. In order to understand the difference between visceral tissue biology of healthy and unhealthy obese individuals, we have used microarray profiling to compare genome-wide expression differences between visceral adipose tissue biopsies obtained from obese diabetics, and those from age and body mass index (BMI) matched normal glucose tolerance subjects. Whereas genes upregulated in diabetics showed enrichment of natural killer cell mediated cytotoxicity, the downregulated genes showed enrichment of biosynthesis of unsaturated fatty acids. Given the known inhibitory effect of unsaturated fatty acids on inflammation and natural killer cell number or activity, our results suggest that visceral inflammation resulting from decreased levels of unsaturated fatty acids may underlie progression of diabetes in obese individuals.
Collapse
Affiliation(s)
| | - Priyanka Jain
- National Research Center on Plant Biotechnology, Indian Agricultural Research Institute, Pusa, India
| | - Prashant Mathur
- Department of Pharmacology, S.M.S. Medical College, Jaipur, Rajasthan, India
| | - Poonam Punjabi
- Department of Physiology, S.M.S. Medical College, Jaipur, Rajasthan, India
| | - Atima Agarwal
- Invitrogen BioServices India Pvt. Ltd., Gurgaon, Haryana, India
| | - Abhay Sharma
- Functional Genomic Unit, CSIR-Institute of Genomics and Integrative Biology, Council of Scientific and Industrial Research, New Delhi, India
| |
Collapse
|
94
|
Serglycin is a novel adipocytokine highly expressed in epicardial adipose tissue. Biochem Biophys Res Commun 2013; 432:105-10. [PMID: 23376071 DOI: 10.1016/j.bbrc.2013.01.078] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Accepted: 01/18/2013] [Indexed: 11/22/2022]
|
95
|
Abstract
The rise in the incidence of obesity has led to a major interest in the biology of white adipose tissue. The tissue is a major endocrine and signaling organ, with adipocytes, the characteristic cell type, secreting a multiplicity of protein factors, the adipokines. Increases in the secretion of a number of adipokines occur in obesity, underpinning inflammation in white adipose tissue and the development of obesity-associated diseases. There is substantial evidence, particularly from animal studies, that hypoxia develops in adipose tissue as the tissue mass expands, and the reduction in Po(2) is considered to underlie the inflammatory response. Exposure of white adipocytes to hypoxic conditions in culture induces changes in the expression of >1,000 genes. The secretion of a number of inflammation-related adipokines is upregulated by hypoxia, and there is a switch from oxidative metabolism to anaerobic glycolysis. Glucose utilization is increased in hypoxic adipocytes with corresponding increases in lactate production. Importantly, hypoxia induces insulin resistance in fat cells and leads to the development of adipose tissue fibrosis. Many of the responses of adipocytes to hypoxia are initiated at Po(2) levels above the normal physiological range for adipose tissue. The other cell types within the tissue also respond to hypoxia, with the differentiation of preadipocytes to adipocytes being inhibited and preadipocytes being transformed into leptin-secreting cells. Overall, hypoxia has pervasive effects on the function of adipocytes and appears to be a key factor in adipose tissue dysfunction in obesity.
Collapse
Affiliation(s)
- Paul Trayhurn
- Obesity Biology Research Unit, Institute of Ageing and Chronic Diseases, University of Liverpool, Liverpool, UK
| |
Collapse
|
96
|
Villalpando-Arteaga EV, Mendieta-Condado E, Esquivel-Solís H, Canales-Aguirre AA, Gálvez-Gastélum FJ, Mateos-Díaz JC, Rodríguez-González JA, Márquez-Aguirre AL. Hibiscus sabdariffa L. aqueous extract attenuates hepatic steatosis through down-regulation of PPAR-γ and SREBP-1c in diet-induced obese mice. Food Funct 2013; 4:618-26. [PMID: 23389749 DOI: 10.1039/c3fo30270a] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The growing incidence of obesity is a worldwide public health problem leading to a risk factor for non-alcoholic fatty liver disease, which extends from steatosis to steatohepatitis and cirrhosis. We investigated whether the aqueous extract of Hibiscus sabdariffa L. (Hs) reduces body weight gain and protects the liver by improving lipid metabolism in high fat diet-induced obese C57BL/6NHsd mice. We found that oral administration of the Hs extract reduced fat tissue accumulation, diminished body weight gain and normalized the glycemic index as well as reduced dyslipidemia compared to the obese mice group that did not receive Hs treatment. In addition, Hs treatment attenuated liver steatosis, down-regulated SREBP-1c and PPAR-γ, blocked the increase of IL-1, TNF-α mRNA and lipoperoxidation and increased catalase mRNA. Our results suggest that the anti-obesity, anti-lipidemic and hepatoprotective effects of the Hs extract are related to the regulation of PPAR-γ and SREBP-1c in the liver.
Collapse
Affiliation(s)
- Edgar Vinicio Villalpando-Arteaga
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Av. Normalistas 800, Guadalajara, Jalisco 44270, Mexico
| | | | | | | | | | | | | | | |
Collapse
|
97
|
Sarcopenia, obesity, and natural killer cell immune senescence in aging: altered cytokine levels as a common mechanism. Aging (Albany NY) 2013; 4:535-46. [PMID: 22935594 PMCID: PMC3461341 DOI: 10.18632/aging.100482] [Citation(s) in RCA: 216] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human aging is characterized by both physical and physiological frailty. A key feature of frailty, sarcopenia is the age-associated decline in skeletal muscle mass, strength, and endurance that characterize even the healthy elderly. Increases in adiposity, particularly in visceral adipose tissue, are almost universal in aging individuals and can contribute to sarcopenia and insulin resistance by increasing levels of inflammatory cytokines known collectively as adipokines. Aging also is associated with declines in adaptive and innate immunity, known as immune senescence, which are risk factors for cancer and all-cause mortality. The cytokine interleukin-15 (IL-15) is highly expressed in skeletal muscle tissue and declines in aging rodent models. IL-15 inhibits fat deposition and insulin resistance, is anabolic for skeletal muscle in certain situations, and is required for the development and survival of natural killer (NK) lymphocytes. We review the effect that adipokines and myokines have on NK cells, with special emphasis on IL-15. We posit that increased adipokine and decreased IL-15 levels during aging constitute a common mechanism for sarcopenia, obesity, and immune senescence.
Collapse
|
98
|
Johnson AR, Milner JJ, Makowski L. The inflammation highway: metabolism accelerates inflammatory traffic in obesity. Immunol Rev 2013; 249:218-38. [PMID: 22889225 DOI: 10.1111/j.1600-065x.2012.01151.x] [Citation(s) in RCA: 416] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
As humans evolved, perhaps the two strongest selection determinants of survival were a robust immune response able to clear bacterial, viral, and parasitic infection and an ability to efficiently store nutrients to survive times when food sources were scarce. These traits are not mutually exclusive. It is now apparent that critical proteins necessary for regulating energy metabolism, such as peroxisome proliferator-activated receptors, Toll-like receptors, and fatty acid-binding proteins, also act as links between nutrient metabolism and inflammatory pathway activation in immune cells. Obesity in humans is a symptom of energy imbalance: the scale has been tipped such that energy intake exceeds energy output and may be a result, in part, of evolutionary selection toward a phenotype characterized by efficient energy storage. As discussed in this review, obesity is a state of low-grade, chronic inflammation that promotes the development of insulin resistance and diabetes. Ironically, the formation of systemic and/or local, tissue-specific insulin resistance upon inflammatory cell activation may actually be a protective mechanism that co-evolved to repartition energy sources within the body during times of stress during infection. However, the point has been reached where a once beneficial adaptive trait has become detrimental to the health of the individual and an immense public health and economic burden. This article reviews the complex relationship between obesity, insulin resistance/diabetes, and inflammation, and although the liver, brain, pancreas, muscle, and other tissues are relevant, we focus specifically on how the obese adipose microenvironment can promote immune cell influx and sustain damaging inflammation that can lead to the onset of insulin resistance and diabetes. Finally, we address how substrate metabolism may regulate the immune response and discuss how fuel uptake and metabolism may be a targetable approach to limit or abrogate obesity-induced inflammation.
Collapse
Affiliation(s)
- Amy R Johnson
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | |
Collapse
|
99
|
Maneschi E, Vignozzi L, Morelli A, Mello T, Filippi S, Cellai I, Comeglio P, Sarchielli E, Calcagno A, Mazzanti B, Vettor R, Vannelli GB, Adorini L, Maggi M. FXR activation normalizes insulin sensitivity in visceral preadipocytes of a rabbit model of MetS. J Endocrinol 2013; 218:215-31. [PMID: 23750014 DOI: 10.1530/joe-13-0109] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Insulin resistance is the putative key underlying mechanism linking adipose tissue (AT) dysfunction with liver inflammation and steatosis in metabolic syndrome (MetS). We have recently demonstrated that the selective farnesoid X receptor (FXR) agonist obeticholic acid (OCA) ameliorates insulin resistance and the metabolic profile with a marked reduction in the amount of visceral AT (VAT) in a high-fat diet (HFD)-induced rabbit model of MetS. These effects were mediated by the activation of FXR, since treatment with the selective TGR5 agonist INT-777 was not able to ameliorate the metabolic parameters evaluated. Herein, we report the effects of in vivo OCA dosing on the liver, the VAT, and the adipogenic capacity of VAT preadipocytes (rPADs) isolated from rabbits on a HFD compared with those on a control diet. VAT and liver were studied by immunohistochemistry, Western blot analysis, and RT-PCR. rPADs were exposed to a differentiating mixture to evaluate adipogenesis. Adipocyte size, hypoxia, and the expression of perilipin and cytosolic insulin-regulated glucose transporter GLUT4 (SLC2A4) were significantly increased in VAT isolated from the HFD rabbits, and normalized by OCA. The expression of steatosis and inflammation markers was increased in the liver of the HFD rabbits and normalized by OCA. rPADs isolated from the HFD rabbits were less sensitive to insulin, as demonstrated by the decreased insulin-induced glucose uptake, triglyceride synthesis, and adipogenic capacity, as well as by the impaired fusion of lipid droplets. OCA treatment preserved all the aforementioned metabolic functions. In conclusion, OCA dosing in a MetS rabbit model ameliorates liver and VAT functions. This could reflect the ability of OCA to restore insulin sensitivity in AT unable to finalize its storage function, counteracting MetS-induced metabolic alterations and pathological AT deposition.
Collapse
Affiliation(s)
- Elena Maneschi
- Sexual Medicine and Andrology Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Pieraccini 6, Florence, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Maneschi E, Morelli A, Filippi S, Cellai I, Comeglio P, Mazzanti B, Mello T, Calcagno A, Sarchielli E, Vignozzi L, Saad F, Vettor R, Vannelli GB, Maggi M. Testosterone treatment improves metabolic syndrome-induced adipose tissue derangements. J Endocrinol 2012; 215:347-62. [PMID: 23045189 DOI: 10.1530/joe-12-0333] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We recently demonstrated that testosterone dosing ameliorated the metabolic profile and reduced visceral adipose tissue (VAT) in a high-fat diet (HFD)-induced rabbit model of metabolic syndrome (MetS). We studied the effects of HFD and in vivo testosterone dosing on VAT function and the adipogenic capacity of rabbit preadipocytes isolated from VAT of regular diet (RD), HFD, and testosterone-treated HFD rabbits. VAT was studied by immunohistochemistry, western blot, and RT-PCR. Isolated rPADs were exposed to adipocyte differentiating mixture (DIM) to evaluate adipogenic potential. Adipocyte size was significantly increased in HFD VAT compared with RD, indicating adipocyte dysfunction, which was normalized by testosterone dosing. Accordingly, perilipin, an anti-lipolytic protein, was significantly increased in HFD VAT, when compared with other groups. HFD VAT was hypoxic, while testosterone dosing normalized VAT oxygenation. In VAT, androgen receptor expression was positively associated with mRNA expression of GLUT4 (SLC2A4) (insulin-regulated glucose transporter) and STAMP2 (STEAP4) (androgen-dependent gene required for insulin signaling). In testosterone-treated HFD VAT, STAMP2 mRNA was significantly increased when compared with the other groups. Moreover, GLUT4 membrane translocation was significantly reduced in HFD VAT, compared with RD, and increased by testosterone. In DIM-exposed preadipocytes from HFD, triglyceride accumulation, adipocyte-specific genes, insulin-stimulated triglyceride synthesis, glucose uptake, and GLUT4 membrane translocation were reduced compared with preadipocytes from RD and normalized by in vivo testosterone dosing. In conclusion, testosterone dosing in a MetS animal model positively affects VAT functions. This could reflect the ability of testosterone in restoring insulin sensitivity in VAT, thus counteracting metabolic alterations.
Collapse
Affiliation(s)
- Elena Maneschi
- Sexual Medicine and Andrology Unit, Department of Clinical Physiopathology, University of Florence, Viale Pieraccini, 6, Florence 50139, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|