1
|
Li G, Meex RCR, Goossens GH. The role of tissue oxygenation in obesity-related cardiometabolic complications. Rev Endocr Metab Disord 2025; 26:19-30. [PMID: 39298040 PMCID: PMC11790814 DOI: 10.1007/s11154-024-09910-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/08/2024] [Indexed: 09/21/2024]
Abstract
Obesity is a complex, multifactorial, chronic disease that acts as a gateway to a range of other diseases. Evidence from recent studies suggests that changes in oxygen availability in the microenvironment of metabolic organs may exert an important role in the development of obesity-related cardiometabolic complications. In this review, we will first discuss results from observational and controlled laboratory studies that examined the relationship between reduced oxygen availability and obesity-related metabolic derangements. Next, the effects of alterations in oxygen partial pressure (pO2) in the adipose tissue, skeletal muscle and the liver microenvironment on physiological processes in these key metabolic organs will be addressed, and how this might relate to cardiometabolic complications. Since many obesity-related chronic diseases, including type 2 diabetes mellitus, cardiovascular diseases, chronic kidney disease, chronic obstructive pulmonary disease and obstructive sleep apnea, are characterized by changes in pO2 in the tissue microenvironment, a better understanding of the metabolic impact of altered tissue oxygenation can provide valuable insights into the complex interplay between environmental and biological factors involved in the pathophysiology of metabolic impairments. This may ultimately contribute to the development of novel strategies to prevent and treat obesity-related cardiometabolic diseases.
Collapse
Affiliation(s)
- Geng Li
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, PO Box 616, Maastricht, 6200 MD, The Netherlands
| | - Ruth C R Meex
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, PO Box 616, Maastricht, 6200 MD, The Netherlands
| | - Gijs H Goossens
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, PO Box 616, Maastricht, 6200 MD, The Netherlands.
| |
Collapse
|
2
|
Cruvinel JM, Greene ES, Read RW, Lee K, Dridi S, Chen PR. Research note: Increased lipid accumulation within broiler preadipocytes during differentiation in vitro at atmospheric oxygen tension. Poult Sci 2025; 104:104531. [PMID: 39566173 PMCID: PMC11617452 DOI: 10.1016/j.psj.2024.104531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/28/2024] [Accepted: 11/07/2024] [Indexed: 11/22/2024] Open
Abstract
In the broiler industry, intensive genetic selection has been placed on muscle growth which has undesirably led to increased fat accretion. Models of chicken preadipocyte differentiation in vitro have conventionally used incubators without the ability to control oxygen (O2) tension; thus, the cells are exposed to atmospheric (∼20-21%) O2, which is supraphysiological compared to the O2 tension within adipose tissue. The objective of this study was to investigate embryonic broiler preadipocyte differentiation at different O2 tensions, including atmospheric (20%), physiological (5%), and hypoxic (1%). Culture at 1% O2 resulted in increased abundance of HIF1α, a canonical protein stabilized during hypoxia, thus confirming effectiveness of the treatment. Increased accumulation of lipid was observed in preadipocytes cultured in adipogenic differentiation medium compared to the control medium. When considering oxygen tension, lipid accumulation was increased in preadipocytes that were cultured in differentiation medium at 20% O2 compared to 5% or 1% O2. Furthermore, abundance of transcripts related to fatty acid transport and adipogenesis, fatty acid binding protein 4 (FABP4) and peroxisome proliferator-activated receptor gamma (PPARγ), were increased in differentiated preadipocytes cultured at 20% O2 compared to 5% or 1% O2. Abundance of transcripts related to lipid synthesis and oxidation, acyl-CoA synthetase long chain family member 1 (ACSL1) and carnitine palmitoyltransferase 1A (CPT1A), were increased in the differentiation cultures compared to the control cultures. Abundance of glutathione peroxidase 4 (GPX4) was increased in all the differentiation cultures compared to the controls, regardless of oxygen tension; however, differences in the abundance of other antioxidant enzymes were not observed. Overall, exposure to atmospheric oxygen tension promotes lipid accumulation within chicken preadipocytes, which may need to be considered when developing in vitro models of this process.
Collapse
Affiliation(s)
| | - Elizabeth S Greene
- University of Arkansas, Center of Excellence for Poultry Science, Fayetteville, AR 72701, USA
| | - Rachel W Read
- Division of Animal Sciences, University of Missouri, Columbia 65211, USA
| | - Kichoon Lee
- Department of Animal Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Sami Dridi
- University of Arkansas, Center of Excellence for Poultry Science, Fayetteville, AR 72701, USA
| | - Paula R Chen
- USDA-ARS, Plant Genetics Research Unit, Columbia, MO 65211, USA.
| |
Collapse
|
3
|
Wang X, Sun H, Cheng G, Ge J. Reduction of oxidative stress response and protection of liver and renal cell functions by reduced glutathione in lower limb arterial ischemia-reperfusion in New Zealand white rabbits with high triglyceride levels. Heliyon 2024; 10:e33258. [PMID: 39022000 PMCID: PMC11252971 DOI: 10.1016/j.heliyon.2024.e33258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/21/2024] [Accepted: 06/18/2024] [Indexed: 07/20/2024] Open
Abstract
Objective Acute liver and kidney injury is the most common complication after aortic surgery, which seriously affects the survival and safety of perioperative patients. The presence of chronic preoperative liver and renal insufficiency, presence of preoperative blood inflammation indicators, duration of intraoperative extracorporeal circulation, and volume of red blood cell transfusion are the main influencing factors for acute postoperative liver and kidney injuries. In recent years, with the research progress on oxidative stress, a growing body of evidence has demonstrated that oxidative stress may cause tissue damage after ischemia-reperfusion (IR). However, the impact of the oxidative stress of distal tissues caused by IR on liver and renal cells after arterial surgeries has not yet been elucidated. Methods New Zealand white rabbits were used for the experiments and were divided into three groups. Among them, two groups were fed high-fat feed to establish a white rabbit model of hypertriglyceridemia, whereas the control group was provided with ordinary feed. In the experiment, white rabbits were subjected to occlusion of the infrarenal aorta abdominalis to simulate IR of the lower limbs. The effects of high triglyceride levels after the arterial IR of the lower limbs were investigated using the contents of reactive oxygen species (ROS) and malondialdehyde (MDA), a fat metabolite, in ischemic muscle tissues and blood tissues. One of the groups receiving high-fat feed received intervention with reduced glutathione (GSH) before IR of the lower limbs. Pathological studies were performed to identify the expression levels of inflammatory factors and inflammatory cells in liver and renal cells as well as cell apoptosis. The effects of GSH administration before IR on reducing the oxidative stress in adipose tissues and alleviating liver and kidney damage after stress response were investigated. Results After IR, the increases in ROS and MDA in ischemic muscle tissues and blood tissues were higher in white rabbits with high triglyceride levels than in those that only received ordinary feed or received intervention with GSH. In addition, for white rabbits with high triglyceride levels, the TNF-α expression levels in the liver increased after IR. Moreover, a considerable increase in the expression of TNF-α, IL-6, macrophages, and T lymphocytes were observed in renal cells. A large number of inflammatory cells and the formation of immune complexes were also noted in the glomeruli; in addition, cell apoptosis was promoted. Conclusion This study showed that high triglyceride levels enhanced the oxidative stress response and increased ROS production in New Zealand white rabbits after arterial IR of the lower limbs. High ROS levels activated the expression of inflammatory factors and inflammatory cells in the liver and kidney, which affected cell functions and promoted apoptosis. At high triglyceride levels, GSH downregulated ROS production in oxidative stress after IR, thereby protecting liver and kidney functions.
Collapse
Affiliation(s)
- Xiaochen Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, Anhui, 230001, PR China
| | - Hailei Sun
- Department of Cardiovascular Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, Anhui, 230001, PR China
| | - Guangcun Cheng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, Anhui, 230001, PR China
| | - Jianjun Ge
- Department of Cardiovascular Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, Anhui, 230001, PR China
| |
Collapse
|
4
|
Ailizire A, Wang X, Ma Y, Yan X, Li S, Wu Z, Du W. How hypoxia affects microbiota metabolism in mice. Front Microbiol 2023; 14:1244519. [PMID: 37840721 PMCID: PMC10569469 DOI: 10.3389/fmicb.2023.1244519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 08/25/2023] [Indexed: 10/17/2023] Open
Abstract
Objective To investigate the relationship between gut microbiota and the fecal metabolites of hypoxic environments in mice. Methods High-fat diet-induced obese mice (n = 20) and normal diet-fed mice (n = 20) were randomly divided into four groups: high altitude obese group (HOB), high altitude normal weight group (HN), low altitude obese group LOB (LOB), and low altitude normal weight group (LN). Fecal samples from each group were 16S rRNA gene sequenced, and five samples from each of the four groups above were selected for non-targeted fecal metabolomics analysis using liquid chromatography-mass spectrometry. The relationship between gut microbiota and fecal metabolites was analyzed using SIMCA 14.1, MetaboAnalyst 5.0 and R 4.1.11. Results (A) Body weight was significantly lower in the hypoxic obesity group than in the normoxic obesity group. (B) Differences in α-diversity and β-diversity were found in the fecal gut microbiota of mice of different body weights and altitude, and the diversity of gut microbiota was higher in the normal group than in the obese group; the results of the comparison between the two groups showed that Faecalibaculum, Romboutsia, Lactobacillus, and A2 were associated with obesity; Romboutsia was associated with hypoxia. (C) The metabolic profiles of fecal metabolites differed between groups: gut microbiota were associated with nucleotide and amino acid metabolism in the same body groups, while gut microbiota were associated with lipid and amino acid metabolism in the same oxygen concentration groups. Conclusion (a) Gut microbiota diversity was reduced in obese groups. Romboutsia was the dominant microbiota in the hypoxia group. (b) Gut microbiota were associated with nucleotide and amino acid metabolism in the same body weight groups, while they were associated with lipid and amino acid metabolism in the same altitude groups.
Collapse
Affiliation(s)
- Ainiwaer Ailizire
- Department of Public Health, Qinghai University School of Medicine, Xining, China
| | - Xiaojing Wang
- Department of Proctology, Qinghai Provincial Traditional Chinese Medicine Hospital, Xining, China
| | - Yan Ma
- Research Center for High Altitude Medicine, Qinghai University School of Medicine, Xining, China
- Key Laboratory for Application of High Altitude Medicine in Qinghai Province, Qinghai University, Xining, China
| | - Xin Yan
- Department of Public Health, Qinghai University School of Medicine, Xining, China
| | - Shiqi Li
- Department of Public Health, Qinghai University School of Medicine, Xining, China
| | - Ziyi Wu
- Department of Public Health, Qinghai University School of Medicine, Xining, China
| | - Wenqi Du
- Department of Public Health, Qinghai University School of Medicine, Xining, China
- Research Center for High Altitude Medicine, Qinghai University School of Medicine, Xining, China
- Key Laboratory for Application of High Altitude Medicine in Qinghai Province, Qinghai University, Xining, China
| |
Collapse
|
5
|
Willar B, Tran KV, Fitzgibbons TP. Epicardial adipocytes in the pathogenesis of atrial fibrillation: An update on basic and translational studies. Front Endocrinol (Lausanne) 2023; 14:1154824. [PMID: 37020587 PMCID: PMC10067711 DOI: 10.3389/fendo.2023.1154824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/06/2023] [Indexed: 04/07/2023] Open
Abstract
Epicardial adipose tissue (EAT) is an endocrine organ containing a host of cell types and undoubtedly serving a multitude of important physiologic functions. Aging and obesity cause hypertrophy of EAT. There is great interest in the possible connection between EAT and cardiovascular disease, in particular, atrial fibrillation (AF). Increased EAT is independently associated with AF and adverse events after AF ablation (e.g., recurrence of AF, and stroke). In general, the amount of EAT correlates with BMI or visceral adiposity. Yet on a molecular level, there are similarities and differences between epicardial and abdominal visceral adipocytes. In comparison to subcutaneous adipose tissue, both depots are enriched in inflammatory cells and chemokines, even in normal conditions. On the other hand, in comparison to visceral fat, epicardial adipocytes have an increased rate of fatty acid release, decreased size, and increased vascularity. Several studies have described an association between fibrosis of EAT and fibrosis of the underlying atrial myocardium. Others have discovered paracrine factors released from EAT that could possibly mediate this association. In addition to the adjacent atrial cardiomyocytes, EAT contains a robust stromal-vascular fraction and surrounds the ganglionic plexi of the cardiac autonomic nervous system (cANS). The importance of the cANS in the pathogenesis of atrial fibrillation is well known, and it is quite likely that there is feedback between EAT and the cANS. This complex interplay may be crucial to the maintenance of normal sinus rhythm or the development of atrial fibrillation. The extent the adipocyte is a microcosm of metabolic health in the individual patient may determine which is the predominant rhythm.
Collapse
|
6
|
Hypoxia as a Double-Edged Sword to Combat Obesity and Comorbidities. Cells 2022; 11:cells11233735. [PMID: 36496995 PMCID: PMC9736735 DOI: 10.3390/cells11233735] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/14/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
The global epidemic of obesity is tightly associated with numerous comorbidities, such as type II diabetes, cardiovascular diseases and the metabolic syndrome. Among the key features of obesity, some studies have suggested the abnormal expansion of adipose-tissue-induced local endogenous hypoxic, while other studies indicated endogenous hyperoxia as the opposite trend. Endogenous hypoxic aggravates dysfunction in adipose tissue and stimulates secretion of inflammatory molecules, which contribute to obesity. In contrast, hypoxic exposure combined with training effectively generate exogenous hypoxic to reduce body weight and downregulate metabolic risks. The (patho)physiological effects in adipose tissue are distinct from those of endogenous hypoxic. We critically assess the latest advances on the molecular mediators of endogenous hypoxic that regulate the dysfunction in adipose tissue. Subsequently we propose potential therapeutic targets in adipose tissues and the small molecules that may reverse the detrimental effect of local endogenous hypoxic. More importantly, we discuss alterations of metabolic pathways in adipose tissue and the metabolic benefits brought by hypoxic exercise. In terms of therapeutic intervention, numerous approaches have been developed to treat obesity, nevertheless durability and safety remain the major concern. Thus, a combination of the therapies that suppress endogenous hypoxic with exercise plans that augment exogenous hypoxic may accelerate the development of more effective and durable medications to treat obesity and comorbidities.
Collapse
|
7
|
Lempesis IG, Hoebers N, Essers Y, Jocken JWE, Rouschop KMA, Blaak EE, Manolopoulos KN, Goossens GH. Physiological Oxygen Levels Differentially Regulate Adipokine Production in Abdominal and Femoral Adipocytes from Individuals with Obesity Versus Normal Weight. Cells 2022; 11:cells11223532. [PMID: 36428961 PMCID: PMC9688196 DOI: 10.3390/cells11223532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/28/2022] [Accepted: 11/06/2022] [Indexed: 11/09/2022] Open
Abstract
Adipose tissue (AT) inflammation may increase obesity-related cardiometabolic complications. Altered AT oxygen partial pressure (pO2) may impact the adipocyte inflammatory phenotype. Here, we investigated the effects of physiological pO2 levels on the inflammatory phenotype of abdominal (ABD) and femoral (FEM) adipocytes derived from postmenopausal women with normal weight (NW) or obesity (OB). Biopsies were collected from ABD and FEM subcutaneous AT in eighteen postmenopausal women (aged 50-65 years) with NW (BMI 18-25 kg/m2, n = 9) or OB (BMI 30-40 kg/m2, n = 9). We compared the effects of prolonged exposure to different physiological pO2 levels on adipokine expression and secretion in differentiated human multipotent adipose-derived stem cells. Low physiological pO2 (5% O2) significantly increased leptin gene expression/secretion in ABD and FEM adipocytes derived from individuals with NW and OB compared with high physiological pO2 (10% O2) and standard laboratory conditions (21% O2). Gene expression/secretion of IL-6, DPP-4, and MCP-1 was reduced in differentiated ABD and FEM adipocytes from individuals with OB but not NW following exposure to low compared with high physiological pO2 levels. Low physiological pO2 decreases gene expression and secretion of several proinflammatory factors in ABD and FEM adipocytes derived from individuals with OB but not NW.
Collapse
Affiliation(s)
- Ioannis G. Lempesis
- Institute of Metabolism and Systems Research (IMSR), College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK
- Correspondence: (I.G.L.); (G.H.G.)
| | - Nicole Hoebers
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Yvonne Essers
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Johan W. E. Jocken
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Kasper M. A. Rouschop
- Radiotherapy, GROW School for Oncology & Reproduction, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Ellen E. Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
| | - Konstantinos N. Manolopoulos
- Institute of Metabolism and Systems Research (IMSR), College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK
| | - Gijs H. Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6229 ER Maastricht, The Netherlands
- Correspondence: (I.G.L.); (G.H.G.)
| |
Collapse
|
8
|
Docosahexaenoic Acid Counteracts the Hypoxic-Induced Inflammatory and Metabolic Alterations in 3T3-L1 Adipocytes. Nutrients 2022; 14:nu14214600. [PMID: 36364860 PMCID: PMC9659308 DOI: 10.3390/nu14214600] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/15/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022] Open
Abstract
Background: Hypoxia is caused by the excessive expansion of the white adipose tissue (AT) and is associated with obesity-related conditions such as insulin resistance, inflammation, and oxidative stress. Docosahexaenoic acid (DHA) is an omega-3 fatty acid reported to have beneficial health effects. However, the effects of DHA in AT against hypoxia-induced immune-metabolic perturbations in adipocytes exposed to low O2 tension are not well known. Consequently, this study aimed to evaluate the impact of DHA on markers of inflammation, metabolism, apoptosis, and oxidative stress in 3T3-L1 cell adipocytes exposed to low O2 tension (1% O2) induced hypoxia. Methods: The apoptosis and reactive oxygen species (ROS) rates were evaluated. Metabolic parameters such as lactate, FFA, glycerol release, glucose uptake, and ATP content were assessed by a fluorometer. The expression of HIF-1, GLUT1 and the secretion of adipocytokines such as leptin, adiponectin, and pro-inflammatory markers was evaluated. Results: DHA-treated hypoxic cells showed significantly decreased basal free fatty acid release, lactate production, and enhanced glucose consumption. In addition, DHA-treatment of hypoxic cells caused a significant reduction in the apoptosis rate and ROS production with decreased lipid peroxidation. Moreover, DHA-treatment of hypoxic cells caused a decreased secretion of pro-inflammatory markers (IL-6, MCP-1) and leptin and increased adiponectin secretion compared with hypoxic cells. Furthermore, DHA-treatment of hypoxic cells caused significant reductions in the expression of genes related to hypoxia (HIF-1, HIF-2), anaerobic metabolism (GLUT1 and Ldha), ATP production (ANT2), and fat metabolism (FASN and PPARY). Conclusion: This study suggests that DHA can exert potential anti-obesity effects by reducing the secretion of inflammatory adipokines, oxidative stress, lipolysis, and apoptosis.
Collapse
|
9
|
Liang H, Yan J, Song K. Comprehensive lipidomic analysis reveals regulation of glyceride metabolism in rat visceral adipose tissue by high-altitude chronic hypoxia. PLoS One 2022; 17:e0267513. [PMID: 35522648 PMCID: PMC9075645 DOI: 10.1371/journal.pone.0267513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/10/2022] [Indexed: 11/18/2022] Open
Abstract
Adipose tissue plays a central role in energy substrate homeostasis and is a key regulator of lipid flow throughout these processes. As hypoxia affects lipid metabolism in adipose tissue, we aimed to investigate the effects of high-altitude chronic hypoxia on lipid metabolism in the adipose tissue of rats using a lipidomic analysis approach. Visceral adipose tissues from rats housed in a high-altitude hypoxia environment representing 4,300 m with 14.07% oxygen (hypoxia group) and from rats housed in a low-altitude normoxia environment representing 41 m with 20.95% oxygen (normoxia group) for 8 weeks were analyzed using an ultra-performance liquid chromatography-Orbitrap mass spectrometry system. After 8 weeks, the body weight and visceral adipose tissue weight of the hypoxia group were significantly decreased compared to those of the normoxia group (p < 0.05). The area and diameter of visceral adipose cells in the hypoxia group were significantly smaller than those of visceral adipose cells in the normoxia group (p < 0.05). The results of lipidomic analysis showed a total of 21 lipid classes and 819 lipid species. The total lipid concentration of the hypoxia group was lower than that in the normoxia group (p < 0.05). Concentrations of diacylglycerols and triacylglycerols in the hypoxia group were significantly lower than those in the normoxia group (p < 0.05). Using univariate and multivariate analyses, we identified 74 lipids that were significantly altered between the normoxia and hypoxia groups. These results demonstrate that high-altitude chronic hypoxia changes the metabolism of visceral adipose glycerides, which may potentially modulate other metabolic processes.
Collapse
Affiliation(s)
- Hong Liang
- Department of Basic Medical Sciences, Medical College, Qinghai University, Xining, PR, China
| | - Jun Yan
- Cardiovascular Medicine Department, Xuzhou Medical University affiliated Hospital, Xuzhou, PR China
| | - Kang Song
- Endocrinology Department, Qinghai Provincial People’s Hospital, Xining, PR, China
- Qinghai University affiliated Provincial People’s Hospital, Xining, PR, China
- * E-mail:
| |
Collapse
|
10
|
Mahat B, Mauger JF, Imbeault P. Effects of different oxygen tensions on differentiated human preadipocytes lipid storage and mobilisation. Arch Physiol Biochem 2021; 127:37-43. [PMID: 31055955 DOI: 10.1080/13813455.2019.1609995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Adipose tissue expansion has been suggested to impair oxygen (O2) diffusion in the adipose tissue and cause hypoxia. This study aimed at characterising the effects of hypoxia on adipocyte lipid storage and mobilisation functions. Human preadipocytes were exposed to different O2 tensions (3, 10 and 21%) either acutely for 24 h after differentiation (acute exposure) or during differentiation (14d, chronic hypoxia). Lipoprotein lipase (LPL) activity was decreased dose-dependently by both acute and chronic hypoxia (p < .05). Acute exposure to 3, and 10% O2 stimulated the expression of lipid storage gene, while chronic exposure to 3% O2 inhibited the expression of genes involved in lipid storage and mobilisation (p < .05). Acute hypoxia dose-dependently stimulated basal lipolysis. Conversely, chronic hypoxia did not affect basal lipolysis but significantly decreased isoproterenol-stimulated lipolysis (p < .05). In conclusion, the effects of hypoxia on human adipocyte lipid storage and mobilisation functions are complex but could favour ectopic fat deposition.
Collapse
Affiliation(s)
- Bimit Mahat
- Behavioral and Metabolic Research Unit, School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Canada
| | - Jean-François Mauger
- Behavioral and Metabolic Research Unit, School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Canada
| | - Pascal Imbeault
- Behavioral and Metabolic Research Unit, School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, Canada
- Institut du savoir Montfort, Hôpital Montfort, Ottawa, Canada
| |
Collapse
|
11
|
Wu G, Liu Y, Feng W, An X, Lin W, Tang C. Hypoxia-Induced Adipose Lipolysis Requires Fibroblast Growth Factor 21. Front Pharmacol 2020; 11:1279. [PMID: 32922298 PMCID: PMC7456904 DOI: 10.3389/fphar.2020.01279] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/03/2020] [Indexed: 02/05/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21) is a recently discovered hepatokine that regulates lipid and glucose metabolism and is upregulated in response to numerous physiological and pathological stimuli. Herein, we demonstrate that both physical and chemical hypoxia increase the systemic and hepatic expression of FGF21 in mice; by contrast, hypoxia induces a reduction of FGF21 expression in hepatocytes, indicating that hypoxia-induced FGF21 expression is differentially regulated in intact animals and in hepatocytes. Furthermore, we demonstrate that hypoxia treatment increases hormone-sensitive lipase-mediated adipose tissue lipolysis in mice, which is reduced in Fgf21 knockout mice, thereby implying that FGF21 plays a critical role in hypoxia-related adipose lipolysis. Adipose tissue lipolysis causes an increase in the amount of circulating free fatty acids, which leads to the activation of peroxisome proliferators-activated receptor alpha and an increased expression of FGF21 in hepatocytes. We further show that hypoxia-induced elevation of reactive oxygen species, but not the hypoxia-inducible factor, is responsible for the lipolysis and FGF21 expression. In conclusion, our data clearly demonstrate that FGF21 plays a critical role in hypoxia-induced adipose lipolysis, which induces hepatic expression of FGF21. Clarification of hypoxia-regulated FGF21 regulation will enhance our understanding of the pathophysiology of hypoxia-related diseases, such as sleep disorders and metabolic diseases.
Collapse
Affiliation(s)
- Guicheng Wu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China.,Department of Hepatology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Yanlong Liu
- School of Mental Health, Wenzhou Medical University, Wenzhou, China.,Zhuji Institute of Biomedicine, School of Pharmaceutical Sciences, Wenzhou Medical University, Shaoxing, China
| | - Wenke Feng
- Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, United States
| | - Xuan An
- Department of Hepatology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Wenhui Lin
- Department of Cardiology, Affiliated Wenling Hospital of Wenzhou Medical University, Wenling, China
| | - Chengwei Tang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Gastroenterology & Hepatology, State Key Laboratory of Biotherapy, Chengdu, China
| |
Collapse
|
12
|
Dai H, Yuan Y, Yin S, Zhang Y, Han Y, Sun L, Li T, Xu J, Sheng L, Gong Y, Li Y. Metoprolol Inhibits Profibrotic Remodeling of Epicardial Adipose Tissue in a Canine Model of Chronic Obstructive Sleep Apnea. J Am Heart Assoc 2020; 8:e011155. [PMID: 30686096 PMCID: PMC6405574 DOI: 10.1161/jaha.118.011155] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Whether chronic obstructive sleep apnea ( OSA ) could promote epicardial adipose tissue ( EAT ) secretion of profibrotic adipokines, and thereby contribute to atrial fibrosis, and the potential therapeutic effects of metoprolol remain unknown. Methods and Results A chronic OSA canine model was established by repeatedly clamping the endotracheal tube for and then reopening it for 4 hours every other day for 12 weeks. In a metoprolol treatment group, metoprolol succinate was administered daily for 12 weeks. The EAT infiltration and left atrial fibrosis were examined. The expressions of adipokines secreted by EAT and hypoxic 3T3-L1 adipocytes were detected. The changes in collagen synthesis, transforming growth factor-β1 expression, and cell differentiation and proliferation in cardiac fibroblasts induced by hypoxic 3T3-L1 adipocyte-derived conditioned medium were further analyzed. Chronic OSA induced infiltration of EAT into the left atrium. OSA enhanced the profibrotic effect of EAT on the adjacent atrial myocardium. Moreover, OSA induced profibrotic cytokine secretion from EAT . We also found that hypoxia induced adipokine secretion in cultured adipocytes, and the medium conditioned by the hypoxic adipocytes increased collagen and transforming growth factor-β1 protein expression and cell proliferation of cardiac fibroblasts. More importantly, metoprolol attenuated infiltration of EAT and alleviated the profibrotic effect of EAT by inhibiting adipokine secretion. Metoprolol also inhibited hypoxia-induced adipokine secretion in adipocytes and thereby blocked the hypoxic adipocyte-derived conditioned medium-induced fibrotic response of cardiac fibroblasts. Conclusions Chronic OSA enhanced the profibrotic effect of EAT on the neighboring atrial myocardium by stimulating the secretion of profibrotic adipokines from EAT , which was significantly attenuated by metoprolol. This study gives insights into mechanisms underlying OSA -induced atrial fibrillation and also provides experimental evidence for the protective effects of metoprolol.
Collapse
Affiliation(s)
- Hui Dai
- 1 Department of Cardiology the First Affiliated Hospital Harbin Medical University Harbin China.,2 Department of Emergency Xinhua Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Yue Yuan
- 1 Department of Cardiology the First Affiliated Hospital Harbin Medical University Harbin China
| | - Shuangli Yin
- 1 Department of Cardiology the First Affiliated Hospital Harbin Medical University Harbin China
| | - Yun Zhang
- 1 Department of Cardiology the First Affiliated Hospital Harbin Medical University Harbin China
| | - Yu Han
- 1 Department of Cardiology the First Affiliated Hospital Harbin Medical University Harbin China
| | - Li Sun
- 1 Department of Cardiology the First Affiliated Hospital Harbin Medical University Harbin China
| | - Tiankai Li
- 1 Department of Cardiology the First Affiliated Hospital Harbin Medical University Harbin China
| | - Jicheng Xu
- 1 Department of Cardiology the First Affiliated Hospital Harbin Medical University Harbin China
| | - Li Sheng
- 1 Department of Cardiology the First Affiliated Hospital Harbin Medical University Harbin China
| | - Yongtai Gong
- 1 Department of Cardiology the First Affiliated Hospital Harbin Medical University Harbin China
| | - Yue Li
- 1 Department of Cardiology the First Affiliated Hospital Harbin Medical University Harbin China
| |
Collapse
|
13
|
Lempesis IG, Meijel RLJ, Manolopoulos KN, Goossens GH. Oxygenation of adipose tissue: A human perspective. Acta Physiol (Oxf) 2020; 228:e13298. [PMID: 31077538 PMCID: PMC6916558 DOI: 10.1111/apha.13298] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/03/2019] [Accepted: 05/08/2019] [Indexed: 12/13/2022]
Abstract
Obesity is a complex disorder of excessive adiposity, and is associated with adverse health effects such as cardiometabolic complications, which are to a large extent attributable to dysfunctional white adipose tissue. Adipose tissue dysfunction is characterized by adipocyte hypertrophy, impaired adipokine secretion, a chronic low‐grade inflammatory status, hormonal resistance and altered metabolic responses, together contributing to insulin resistance and related chronic diseases. Adipose tissue hypoxia, defined as a relative oxygen deficit, in obesity has been proposed as a potential contributor to adipose tissue dysfunction, but studies in humans have yielded conflicting results. Here, we will review the role of adipose tissue oxygenation in the pathophysiology of obesity‐related complications, with a specific focus on human studies. We will provide an overview of the determinants of adipose tissue oxygenation, as well as the role of adipose tissue oxygenation in glucose homeostasis, lipid metabolism and inflammation. Finally, we will discuss the putative effects of physiological and experimental hypoxia on adipose tissue biology and whole‐body metabolism in humans. We conclude that several lines of evidence suggest that alteration of adipose tissue oxygenation may impact metabolic homeostasis, thereby providing a novel strategy to combat chronic metabolic diseases in obese humans.
Collapse
Affiliation(s)
- Ioannis G. Lempesis
- College of Medical and Dental Sciences, Institute of Metabolism and Systems Research (IMSR) University of Birmingham Birmingham UK
- Centre for Endocrinology, Diabetes and Metabolism Birmingham Health Partners Birmingham UK
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism Maastricht University Medical Centre Maastricht the Netherlands
| | - Rens L. J. Meijel
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism Maastricht University Medical Centre Maastricht the Netherlands
| | - Konstantinos N. Manolopoulos
- College of Medical and Dental Sciences, Institute of Metabolism and Systems Research (IMSR) University of Birmingham Birmingham UK
- Centre for Endocrinology, Diabetes and Metabolism Birmingham Health Partners Birmingham UK
| | - Gijs H. Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism Maastricht University Medical Centre Maastricht the Netherlands
| |
Collapse
|
14
|
Laria AE, Messineo S, Arcidiacono B, Varano M, Chiefari E, Semple RK, Rocha N, Russo D, Cuda G, Gaspari M, Brunetti A, Foti DP. Secretome Analysis of Hypoxia-Induced 3T3-L1 Adipocytes Uncovers Novel Proteins Potentially Involved in Obesity. Proteomics 2019; 18:e1700260. [PMID: 29466620 DOI: 10.1002/pmic.201700260] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 01/27/2018] [Indexed: 12/17/2022]
Abstract
In the obese state, as adipose tissue expands, adipocytes become hypoxic and dysfunctional, leading to changes in the pattern of adipocyte-secreted proteins. To better understand the role of hypoxia in the mechanisms linked to obesity, we comparatively analyzed the secretome of murine differentiated 3T3-L1 adipocytes exposed to normoxia or hypoxia for 24 h. Proteins secreted into the culture media were precipitated by trichloroacetic acid and then digested with trypsin. The peptides were labeled with dimethyl labeling and analyzed by reversed phase nanoscale liquid chromatography coupled to a quadrupole Orbitrap mass spectrometer. From a total of 1508 identified proteins, 109 were differentially regulated, of which 108 were genuinely secreted. Factors significantly downregulated in hypoxic conditions included adiponectin, a known adipokine implicated in metabolic processes, as well as thrombospondin-1 and -2, and matrix metalloproteinase-11, all multifunctional proteins involved in extracellular matrix (ECM) homeostasis. Findings were validated by Western blot analysis. Expression studies of the relative genes were performed in parallel experiments in vitro, in differentiated 3T3-L1 adipocytes, and in vivo, in fat tissues from obese versus lean mice. Our observations are compatible with the concept that hypoxia may be an early trigger for both adipose cell dysfunction and ECM remodeling.
Collapse
Affiliation(s)
- Anna Elisa Laria
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Sebastiano Messineo
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Biagio Arcidiacono
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Mariaconcetta Varano
- Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Eusebio Chiefari
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Robert K Semple
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge Metabolic Research Laboratories, Cambridge, UK
- The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
- University of Edinburgh Centre for Cardiovascular Science, Edinburgh, UK
| | - Nuno Rocha
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge Metabolic Research Laboratories, Cambridge, UK
- The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Diego Russo
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Giovanni Cuda
- Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Marco Gaspari
- Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Antonio Brunetti
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Daniela P Foti
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| |
Collapse
|
15
|
Keeley TP, Mann GE. Defining Physiological Normoxia for Improved Translation of Cell Physiology to Animal Models and Humans. Physiol Rev 2019; 99:161-234. [PMID: 30354965 DOI: 10.1152/physrev.00041.2017] [Citation(s) in RCA: 216] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The extensive oxygen gradient between the air we breathe (Po2 ~21 kPa) and its ultimate distribution within mitochondria (as low as ~0.5-1 kPa) is testament to the efforts expended in limiting its inherent toxicity. It has long been recognized that cell culture undertaken under room air conditions falls short of replicating this protection in vitro. Despite this, difficulty in accurately determining the appropriate O2 levels in which to culture cells, coupled with a lack of the technology to replicate and maintain a physiological O2 environment in vitro, has hindered addressing this issue thus far. In this review, we aim to address the current understanding of tissue Po2 distribution in vivo and summarize the attempts made to replicate these conditions in vitro. The state-of-the-art techniques employed to accurately determine O2 levels, as well as the issues associated with reproducing physiological O2 levels in vitro, are also critically reviewed. We aim to provide the framework for researchers to undertake cell culture under O2 levels relevant to specific tissues and organs. We envisage that this review will facilitate a paradigm shift, enabling translation of findings under physiological conditions in vitro to disease pathology and the design of novel therapeutics.
Collapse
Affiliation(s)
- Thomas P Keeley
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, King's College London , London , United Kingdom
| | - Giovanni E Mann
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, King's College London , London , United Kingdom
| |
Collapse
|
16
|
Hypoxia Restrains Lipid Utilization via Protein Kinase A and Adipose Triglyceride Lipase Downregulation through Hypoxia-Inducible Factor. Mol Cell Biol 2019; 39:MCB.00390-18. [PMID: 30397073 DOI: 10.1128/mcb.00390-18] [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] [Received: 08/01/2018] [Accepted: 10/22/2018] [Indexed: 12/22/2022] Open
Abstract
Oxygen is a key molecule for efficient energy production in living organisms. Although aerobic organisms have adaptive processes to survive in low-oxygen environments, it is poorly understood how lipolysis, the first step of energy production from stored lipid metabolites, would be modulated during hypoxia. Here, we demonstrate that fasting-induced lipolysis is downregulated by hypoxia through the hypoxia-inducible factor (HIF) signaling pathway. In Caenorhabditis elegans and mammalian adipocytes, hypoxia suppressed protein kinase A (PKA)-stimulated lipolysis, which is evolutionarily well conserved. During hypoxia, the levels of PKA activity and adipose triglyceride lipase (ATGL) protein were downregulated, resulting in attenuated fasting-induced lipolysis. In worms, HIF stabilization was sufficient to moderate the suppressive effect of hypoxia on lipolysis through ATGL and PKA inhibition. These data suggest that HIF activation under hypoxia plays key roles in the suppression of lipolysis, which might preserve energy resources in both C. elegans and mammalian adipocytes.
Collapse
|
17
|
Oikonomou E, Kostopoulou E, Rojas-Gil AP, Georgiou G, Spiliotis BE. Adipocyte aquaporin 7 (AQP7) expression in lean children and children with obesity. Possible involvement in molecular mechanisms of childhood obesity. J Pediatr Endocrinol Metab 2018; 31:1081-1089. [PMID: 30226208 DOI: 10.1515/jpem-2018-0281] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 08/13/2018] [Indexed: 12/13/2022]
Abstract
Background Aquaporin 7 (AQP7), a water/glycerol transporting protein, regulates adipocyte glycerol efflux and influences lipid and glucose homeostasis. Altered AQP7 expression in adults leads to impaired glycerol dynamics, adipocyte hypertrophy and it predisposes them to obesity and diabetes. To assess its possible involvement in childhood obesity, this study investigated the expression of adipocyte AQP7 in cultured adipocytes of children. Methods Primary in vitro differentiated adipocyte cultures were developed from surgical biopsies of subcutaneous abdominal adipose tissue from 61 (46 prepubertal, 15 pubertal) lean children (body mass index [BMI] <85%) and 41 (22 prepubertal, 19 pubertal) children with obesity (BMI >95%). AQP7 expression was studied by reverse transcription polymerase chain reaction and Western immunoblotting and insulin by enzyme-linked immunosorbent assay. Results AQP7 messenger RNA (mRNA) was increased in the younger obese prepubertal (YOP) children but decreased in the obese adolescents (OA) (p=0.014) who also had increased insulin and homeostatic model assessment - insulin resistance (HOMA-IR). Lean pubertal (LP) children and YOP had increased 41 kDa AQP7 protein expression (p=0.001 and p=0.005, respectively). The OA who expressed 34 kDa AQP7 had lower triglyceride (TG) levels than those who did not express it (p=0.013). In the lean children, TG were negatively correlated with 34 kDa AQP7 (p=0.033). Conclusions The lower AQP7 mRNA expression in the OA may reflect a predisposition to adipocyte hypertrophy and metabolic dysfunction, as in the adults, whereas the YOP may be protected from this. The increased 41 kDa AQP7 protein expression in the LP may reflect the increased energy requirements of puberty for glycerol while in the YOP it may also be protective against the development of adipocyte hypertrophy.
Collapse
Affiliation(s)
- Eleni Oikonomou
- Research Laboratory of the Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Patras School of Medicine, Patras, Greece
| | - Eirini Kostopoulou
- Research Laboratory of the Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Patras School of Medicine, Patras, Greece
| | - Andrea Paola Rojas-Gil
- Research Laboratory of the Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Patras School of Medicine, Patras, Greece.,Faculty of Human Movement and Quality of Life Sciences, Department of Nursing, University of Peloponnese, Sparta, Lakonias, Greece
| | - George Georgiou
- Department of Paediatric Surgery, Karamandaneio Children's Hospital, Patras, Greece
| | - Bessie E Spiliotis
- Research Laboratory of the Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Patras School of Medicine, Patras, Greece
| |
Collapse
|
18
|
Vogel MAA, Jocken JWE, Sell H, Hoebers N, Essers Y, Rouschop KMA, Cajlakovic M, Blaak EE, Goossens GH. Differences in Upper and Lower Body Adipose Tissue Oxygen Tension Contribute to the Adipose Tissue Phenotype in Humans. J Clin Endocrinol Metab 2018; 103:3688-3697. [PMID: 30020463 DOI: 10.1210/jc.2018-00547] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/12/2018] [Indexed: 12/18/2022]
Abstract
CONTEXT AND OBJECTIVES Upper and lower body adipose tissue (AT) exhibits opposing associations with obesity-related cardiometabolic diseases. Recent studies have suggested that altered AT oxygen tension (pO2) may contribute to AT dysfunction. Here, we compared in vivo abdominal (ABD) and femoral (FEM) subcutaneous AT pO2 in women who are overweight and have obesity, and investigated the effects of physiological AT pO2 on human adipocyte function. DESIGN ABD and FEM subcutaneous AT pO2 and AT blood flow (ATBF) were assessed in eight [BMI (body mass index) 34.4 ± 1.6 kg/m2] postmenopausal women who were overweight with obesity and impaired glucose metabolism. ABD and FEM AT biopsy specimens were collected to determine adipocyte morphology and AT gene expression. Moreover, the effects of prolonged exposure (14 days) to physiological AT pO2 on adipokine expression/secretion, mitochondrial respiration, and glucose uptake were investigated in differentiated human multipotent adipose-derived stem cells. RESULTS AT pO2 was higher in ABD than FEM AT (62.7 ± 6.6 vs 50.0 ± 4.5 mm Hg, P = 0.013), whereas ATBF was comparable between depots. Maximal uncoupled oxygen consumption rates were substantially lower in ABD than FEM adipocytes for all pO2 conditions. Low physiological pO2 (5% O2) decreased proinflammatory gene expression, increased basal glucose uptake, and altered adipokine secretion in ABD and FEM adipocytes. CONCLUSIONS We demonstrated for the first time, to our knowledge, that AT pO2 is higher in ABD than FEM subcutaneous AT in women who are overweight/with obesity, partly due to a lower oxygen consumption rate in ABD adipocytes. Moreover, low physiological pO2 decreased proinflammatory gene expression and improved the metabolic phenotype in differentiated human adipocytes, whereas more heterogeneous effects on adipokine secretion were found.
Collapse
Affiliation(s)
- Max A A Vogel
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - Johan W E Jocken
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - Henrike Sell
- Paul-Langerhans-Group for Integrative Physiology, German Diabetes Center, Dusseldorf, Germany
| | - Nicole Hoebers
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - Yvonne Essers
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - Kasper M A Rouschop
- Maastricht Radiation Oncology (MaastRO) Laboratory, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Merima Cajlakovic
- Joanneum Research Forschungsgesellschaft mbH, MATERIALS-Institute for Surface Technologies and Photonic, Sensors and Functional Printing, Weiz, Austria
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| |
Collapse
|
19
|
Weiszenstein M, Shimoda LA, Koc M, Seda O, Polak J. Inhibition of Lipolysis Ameliorates Diabetic Phenotype in a Mouse Model of Obstructive Sleep Apnea. Am J Respir Cell Mol Biol 2017; 55:299-307. [PMID: 26978122 DOI: 10.1165/rcmb.2015-0315oc] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Obstructive sleep apnea (OSA) is associated with insulin resistance, glucose intolerance, and type 2 diabetes. Causal mechanisms mediating this association are not well defined; however, augmented lipolysis in adipose might be involved. Here, we investigated the effect of acipimox treatment (lipolysis inhibitor) on glucose tolerance and insulin sensitivity in mice exposed to intermittent hypoxia (IH). C57BL6/J mice were exposed for 14 days to IH or control conditions. IH was created by decreasing the fraction of inspired oxygen from 20.9 to 6.5%, 60 times/h. Control exposure was air (fraction of inspired oxygen, 20.9%) delivered at an identical flow rate. Acipimox was provided in drinking water (0.5 g/ml) during exposures. After exposures, intraperitoneal insulin (0.5 IU/kg) and glucose (1 g/kg) tolerance tests were performed, and primary adipocytes were isolated for lipolysis experiments. IH elevated fasting glucose by 51% and worsened glucose tolerance and insulin sensitivity by 33 and 102%, respectively. In parallel, IH increased spontaneous lipolysis by 264%, and reduced epididymal fat mass by 15% and adipocyte size by 8%. Acipimox treatment prevented IH-induced lipolysis and increased epididymal fat mass and adipocyte size by 19 and 10%, respectively. Acipimox fully prevented IH-induced impairments in fasting glycemia, glucose tolerance, and insulin sensitivity. For all reported results, P less than 0.05 was considered significant. Augmented lipolysis contributes to insulin resistance and glucose intolerance observed in mice exposed to IH. Acipimox treatment ameliorated the metabolic consequences of IH and might represent a novel treatment option for patients with obstructive sleep apnea.
Collapse
Affiliation(s)
- Martin Weiszenstein
- 1 Center for Research on Diabetes, Metabolism, and Nutrition, Third Faculty of Medicine.,2 Center of Toxicology and Health Safety, National Institute of Public Health, Prague, Czech Republic
| | - Larissa A Shimoda
- 3 Division of Pulmonary and Critical Care Medicine, Asthma and Allergy Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Michal Koc
- 4 Sports Medicine Department, Third Faculty of Medicine, and
| | - Ondrej Seda
- 5 Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic; and
| | - Jan Polak
- 1 Center for Research on Diabetes, Metabolism, and Nutrition, Third Faculty of Medicine.,2 Center of Toxicology and Health Safety, National Institute of Public Health, Prague, Czech Republic.,3 Division of Pulmonary and Critical Care Medicine, Asthma and Allergy Center, Johns Hopkins School of Medicine, Baltimore, Maryland.,6 Second Internal Medicine Department, Vinohrady Teaching Hospital, Prague, Czech Republic
| |
Collapse
|
20
|
Llobet L, Bayona-Bafaluy MP, Pacheu-Grau D, Torres-Pérez E, Arbones-Mainar JM, Navarro MÁ, Gómez-Díaz C, Montoya J, López-Gallardo E, Ruiz-Pesini E. Pharmacologic concentrations of linezolid modify oxidative phosphorylation function and adipocyte secretome. Redox Biol 2017; 13:244-254. [PMID: 28600981 PMCID: PMC5466587 DOI: 10.1016/j.redox.2017.05.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 05/29/2017] [Accepted: 05/30/2017] [Indexed: 11/16/2022] Open
Abstract
The oxidative phosphorylation system is important for adipocyte differentiation. Therefore, xenobiotics inhibitors of the oxidative phosphorylation system could affect adipocyte differentiation and adipokine secretion. As adipokines impact the overall health status, these xenobiotics may have wide effects on human health. Some of these xenobiotics are widely used therapeutic drugs, such as ribosomal antibiotics. Because of its similarity to the bacterial one, mitochondrial translation system is an off-target for these compounds. To study the influence of the ribosomal antibiotic linezolid on adipokine production, we analyzed its effects on adipocyte secretome. Linezolid, at therapeutic concentrations, modifies the levels of apolipoprotein E and several adipokines and proteins related with the extracellular matrix. This antibiotic also alters the global methylation status of human adipose tissue-derived stem cells and, therefore, its effects are not limited to the exposure period. Besides their consequences on other tissues, xenobiotics acting on the adipocyte oxidative phosphorylation system alter apolipoprotein E and adipokine production, secondarily contributing to their systemic effects. Linezolid decreases oxidative phosphorylation system capacity. Linezolid reduces adipocyte differentiation from human adipose-derived stem cells. Linezolid modifies APOE, adipokine and extracellular matrix proteins levels. Linezolid changes DNA methylation of human adipose tissue-derived stem cells. Xenobiotics, acting on adipocyte oxidative phosphorylation, affect human health.
Collapse
Affiliation(s)
- Laura Llobet
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, C/ Miguel Servet 177, 50013 Zaragoza, Spain; Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Universidad de Zaragoza, C/ Miguel Servet 177, 50013 Zaragoza, Spain; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Universidad de Zaragoza, C/ Miguel Servet 177, 50013 Zaragoza, Spain.
| | - M Pilar Bayona-Bafaluy
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, C/ Miguel Servet 177, 50013 Zaragoza, Spain; Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Universidad de Zaragoza, C/ Miguel Servet 177, 50013 Zaragoza, Spain.
| | - David Pacheu-Grau
- Department of Cellular Biochemistry, University Medical Center, Humboldtalle 23, 37073 Göttingen, Germany.
| | - Elena Torres-Pérez
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Universidad de Zaragoza, C/ Miguel Servet 177, 50013 Zaragoza, Spain; Unidad de Investigación Traslacional, Instituto Aragones de Ciencias de la Salud (IACS), Hospital Universitario Miguel Servet, Paseo de Isabel la Católica 1-3, 50009 Zaragoza, Spain.
| | - José M Arbones-Mainar
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Universidad de Zaragoza, C/ Miguel Servet 177, 50013 Zaragoza, Spain; Unidad de Investigación Traslacional, Instituto Aragones de Ciencias de la Salud (IACS), Hospital Universitario Miguel Servet, Paseo de Isabel la Católica 1-3, 50009 Zaragoza, Spain; Centro de Investigaciones Biomédicas en Red Fisiopatología de la Obesidad y Nutrición (CIBERObn), Hospital Universitario Miguel Servet, Paseo de Isabel la Católica 1-3, 50009 Zaragoza, Spain.
| | - M Ángeles Navarro
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, C/ Miguel Servet 177, 50013 Zaragoza, Spain; Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Universidad de Zaragoza, C/ Miguel Servet 177, 50013 Zaragoza, Spain; Centro de Investigaciones Biomédicas en Red Fisiopatología de la Obesidad y Nutrición (CIBERObn), Hospital Universitario Miguel Servet, Paseo de Isabel la Católica 1-3, 50009 Zaragoza, Spain.
| | - Covadonga Gómez-Díaz
- Servicio de Otorrinolaringología, Hospital Universitario Miguel Servet, Paseo de Isabel la Católica 1-3, 50009 Zaragoza, Spain.
| | - Julio Montoya
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, C/ Miguel Servet 177, 50013 Zaragoza, Spain; Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Universidad de Zaragoza, C/ Miguel Servet 177, 50013 Zaragoza, Spain; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Universidad de Zaragoza, C/ Miguel Servet 177, 50013 Zaragoza, Spain.
| | - Ester López-Gallardo
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, C/ Miguel Servet 177, 50013 Zaragoza, Spain; Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Universidad de Zaragoza, C/ Miguel Servet 177, 50013 Zaragoza, Spain; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Universidad de Zaragoza, C/ Miguel Servet 177, 50013 Zaragoza, Spain.
| | - Eduardo Ruiz-Pesini
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, C/ Miguel Servet 177, 50013 Zaragoza, Spain; Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Universidad de Zaragoza, C/ Miguel Servet 177, 50013 Zaragoza, Spain; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Universidad de Zaragoza, C/ Miguel Servet 177, 50013 Zaragoza, Spain; Fundación ARAID, Universidad de Zaragoza, C/ Miguel Servet 177, 50013 Zaragoza, Spain.
| |
Collapse
|
21
|
Lu Y, Feng L, Xie M, Zhang L, Xu J, He Z, You T. Hypoxic Living and Exercise Training Alter Adipose Tissue Leptin/Leptin Receptor in Rats. Front Physiol 2016; 7:554. [PMID: 27932989 PMCID: PMC5120121 DOI: 10.3389/fphys.2016.00554] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 11/03/2016] [Indexed: 01/23/2023] Open
Abstract
Background: Hypobaric hypoxia results in weight loss in obese individuals, and exercise training is advocated for the treatment of obesity and its related metabolic dysfunctions. The purpose of this study was to investigate the effects of hypoxic living and exercise training on obesity and adipose tissue leptin/leptin receptor in dietary-induced obese rats. Methods: One hundred and thirty high-fat diet fed Sprague-Dawley rats were assigned into one of the following groups (n = 10 each): control, sedentary hypoxic living for 1–4 weeks (SH1, SH2, SH3, and SH4), living, and exercise training in normoxic conditions for 1–4 weeks (TN1, TN2, TN3, and TN4), and living and exercise training in hypoxic conditions for 1–4 weeks (TN1, TN2, TN3, and TN4). Epididymal adipose tissue expression levels of leptin and leptin receptor were determined Results: Compared to hypoxic living and living and exercise training in normoxic conditions, living and exercise training in hypoxic conditions for 3–4 weeks resulted in lower Lee index (P < 0.05–0.01), and higher expression of leptin and leptin receptor (P < 0.05–0.01) in adipose tissue. Conclusion: In a rodent model of altitude training, living, and exercise training in hypoxic conditions resulted in greater alterations in obesity and adipose tissue leptin/leptin receptor than hypoxic living alone and living and exercise training in normoxic conditions.
Collapse
Affiliation(s)
- Yingli Lu
- China Institute of Sport Science Beijing, China
| | | | - Minhao Xie
- China Institute of Sports Medicine Beijing, China
| | - Li Zhang
- China Institute of Sport Science Beijing, China
| | - Jianfang Xu
- China Institute of Sport Science Beijing, China
| | - Zihong He
- China Institute of Sport Science Beijing, China
| | - Tongjian You
- Department of Exercise and Health Sciences, College of Nursing and Health Sciences, University of Massachusetts Boston Boston, MA, USA
| |
Collapse
|
22
|
Wang X, Buechler NL, Yoza BK, McCall CE, Vachharajani V. Adiponectin treatment attenuates inflammatory response during early sepsis in obese mice. J Inflamm Res 2016; 9:167-174. [PMID: 27785087 PMCID: PMC5063563 DOI: 10.2147/jir.s119021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Morbid obesity increases the cost of care in critically ill patients. Sepsis is the leading cause of death in noncoronary intensive care units. Circulating cell-endothelial cell interactions in microcirculation are the rate-determining factors in any inflammation; obesity increases these interactions further. Adiponectin deficiency is implicated in increased cardiovascular risk in obese patients. We have shown that adiponectin deficiency increases microvascular dysfunction in early sepsis. In the present study, we investigated the effect of adiponectin replacement on nutritionally obese mice with early sepsis. METHODS We used cecal ligation and puncture model of sepsis in mice with diet-induced obesity (DIO) vs control diet (CTRL), with or without adiponectin treatment. We studied leukocyte/platelet adhesion in the cerebral microcirculation in early sepsis. We also studied the effect of adiponectin on free fatty acid (FFA)-fed and lipopolysaccharide-stimulated bone marrow-derived macrophages (BMDM) for mechanistic studies. RESULTS Leukocyte and platelet adhesion increased in the cerebral microcirculation of DIO and CTRL mice with early sepsis vs. sham; moreover cell adhesion in DIO-sepsis group was significantly higher than in the CTRL-sepsis group. Adiponectin replacement decreased leukocyte/platelet adhesion in CTRL and DIO mice. In FFA-fed BMDM, adiponectin treatment decreased tumor necrosis factor-alpha mRNA expression and increased sirtuin-1 (SIRT1) mRNA expression. Furthermore, using BMDM from SIRT1 knockout mice, we showed that the adiponectin treatment decreased inflammatory response in FFA-fed BMDM via SIRT1-dependent and -independent pathways. CONCLUSION Adiponectin replacement attenuates microvascular inflammation in DIO-sepsis mice. Mechanistically, adiponectin treatment in FFA-fed mouse macrophages attenuates inflammatory response via SIRT1-dependent and -independent pathways.
Collapse
Affiliation(s)
- XianFeng Wang
- Department of Anesthesiology, Medicine and Surgery, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Nancy L Buechler
- Department of Anesthesiology, Medicine and Surgery, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Barbara K Yoza
- Department of Anesthesiology, Medicine and Surgery, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Charles E McCall
- Department of Anesthesiology, Medicine and Surgery, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Vidula Vachharajani
- Department of Anesthesiology, Medicine and Surgery, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
23
|
Cell Models and Their Application for Studying Adipogenic Differentiation in Relation to Obesity: A Review. Int J Mol Sci 2016; 17:ijms17071040. [PMID: 27376273 PMCID: PMC4964416 DOI: 10.3390/ijms17071040] [Citation(s) in RCA: 259] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/23/2016] [Accepted: 06/24/2016] [Indexed: 02/08/2023] Open
Abstract
Over the last several years, the increasing prevalence of obesity has favored an intense study of adipose tissue biology and the precise mechanisms involved in adipocyte differentiation and adipogenesis. Adipocyte commitment and differentiation are complex processes, which can be investigated thanks to the development of diverse in vitro cell models and molecular biology techniques that allow for a better understanding of adipogenesis and adipocyte dysfunction associated with obesity. The aim of the present work was to update the different animal and human cell culture models available for studying the in vitro adipogenic differentiation process related to obesity and its co-morbidities. The main characteristics, new protocols, and applications of the cell models used to study the adipogenesis in the last five years have been extensively revised. Moreover, we depict co-cultures and three-dimensional cultures, given their utility to understand the connections between adipocytes and their surrounding cells in adipose tissue.
Collapse
|
24
|
Ekambaram P, Parasuraman P, Jayachandran T. Differential regulation of pro- and antiapoptotic proteins in fish adipocytes during hypoxic conditions. FISH PHYSIOLOGY AND BIOCHEMISTRY 2016; 42:919-934. [PMID: 26744268 DOI: 10.1007/s10695-015-0185-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 12/21/2015] [Indexed: 06/05/2023]
Abstract
Worldwide, the frequencies and magnitudes of hypoxic events in estuarine waters have increased considerably over the past two decades. Fish populations are suitable indicators for the assessment of quality of aquatic ecosystems and often comprise a variety of adaptation systems by triggering oxidants, antioxidants and hypoxia-responsive signaling proteins. Signaling pathway may lead to cell survival or cell death which is fine-tuned by both positive and negative factors, which includes hypoxia-inducible factor-1α (HIF1α), heat-shock protein-70 (HSP70), phospho-c-Jun N-terminal kinase 1/2 (p-JNK1/2) and apoptosis signal-regulating kinase-1 (ASK1). In the present study, we attempt to determine stress-mediated signaling changes and molecular mechanism behind the cell survival by comparing adipocytes of fish from field hypoxic condition and laboratory-induced hypoxic condition (in vitro hypoxia). Comparison of field and laboratory studies in fish adipocytes showed differential expression of HIF1α, HSP70, p-JNK1/2 and ASK1 with altered oxidants and antioxidants. Further, the results also suggest that in vitro hypoxic conditions mimic field hypoxic conditions. Trends of hypoxia response were same in in vitro hypoxia of control adipocytes as in Ennore estuary, and hypoxia response was more pronounced in the test adipocytes under in vitro hypoxic condition. Results of the present work suggest that hypoxia is the major crusade of water pollutants affecting fish by differential regulation of pro- and antiapoptotic proteins probably through HSP70. This may play a vital role by providing cytoprotection in pollutant-induced stressed fish adipocytes substantiated by the in vitro hypoxic studies.
Collapse
Affiliation(s)
- Padmini Ekambaram
- P.G. Department of Biochemistry, Bharathi Women's College, Affiliated to University of Madras, Chennai, Tamil Nadu, 600108, India.
| | - Parimala Parasuraman
- P.G. Department of Biochemistry, Bharathi Women's College, Affiliated to University of Madras, Chennai, Tamil Nadu, 600108, India
| | - Tharani Jayachandran
- P.G. Department of Biochemistry, Bharathi Women's College, Affiliated to University of Madras, Chennai, Tamil Nadu, 600108, India
| |
Collapse
|
25
|
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: 24] [Impact Index Per Article: 2.4] [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
|
26
|
Llobet L, Toivonen JM, Montoya J, Ruiz-Pesini E, López-Gallardo E. Xenobiotics that affect oxidative phosphorylation alter differentiation of human adipose-derived stem cells at concentrations that are found in human blood. Dis Model Mech 2015; 8:1441-55. [PMID: 26398948 PMCID: PMC4631789 DOI: 10.1242/dmm.021774] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 09/03/2015] [Indexed: 12/17/2022] Open
Abstract
Adipogenesis is accompanied by differentiation of adipose tissue-derived stem cells to adipocytes. As part of this differentiation, biogenesis of the oxidative phosphorylation system occurs. Many chemical compounds used in medicine, agriculture or other human activities affect oxidative phosphorylation function. Therefore, these xenobiotics could alter adipogenesis. We have analyzed the effects on adipocyte differentiation of some xenobiotics that act on the oxidative phosphorylation system. The tested concentrations have been previously reported in human blood. Our results show that pharmaceutical drugs that decrease mitochondrial DNA replication, such as nucleoside reverse transcriptase inhibitors, or inhibitors of mitochondrial protein synthesis, such as ribosomal antibiotics, diminish adipocyte differentiation and leptin secretion. By contrast, the environmental chemical pollutant tributyltin chloride, which inhibits the ATP synthase of the oxidative phosphorylation system, can promote adipocyte differentiation and leptin secretion, leading to obesity and metabolic syndrome as postulated by the obesogen hypothesis. Summary: Some medical drugs and environmental chemical pollutants acting on the oxidative phosphorylation system can alter adipocyte differentiation and adipogenesis and, thus, have important consequences for human health.
Collapse
Affiliation(s)
- Laura Llobet
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50013-Zaragoza, Spain Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, 50013-Zaragoza, Spain CIBER de Enfermedades Raras (CIBERER), Universidad de Zaragoza, 50013-Zaragoza, Spain
| | - Janne M Toivonen
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50013-Zaragoza, Spain Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, 50013-Zaragoza, Spain
| | - Julio Montoya
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50013-Zaragoza, Spain Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, 50013-Zaragoza, Spain CIBER de Enfermedades Raras (CIBERER), Universidad de Zaragoza, 50013-Zaragoza, Spain
| | - Eduardo Ruiz-Pesini
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50013-Zaragoza, Spain Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, 50013-Zaragoza, Spain CIBER de Enfermedades Raras (CIBERER), Universidad de Zaragoza, 50013-Zaragoza, Spain Fundación ARAID, Universidad de Zaragoza, 50013-Zaragoza, Spain
| | - Ester López-Gallardo
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50013-Zaragoza, Spain Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, 50013-Zaragoza, Spain CIBER de Enfermedades Raras (CIBERER), Universidad de Zaragoza, 50013-Zaragoza, Spain
| |
Collapse
|
27
|
Netzer N, Gatterer H, Faulhaber M, Burtscher M, Pramsohler S, Pesta D. Hypoxia, Oxidative Stress and Fat. Biomolecules 2015; 5:1143-50. [PMID: 26061760 PMCID: PMC4496714 DOI: 10.3390/biom5021143] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Revised: 05/19/2015] [Accepted: 05/19/2015] [Indexed: 12/17/2022] Open
Abstract
Metabolic disturbances in white adipose tissue in obese individuals contribute to the pathogenesis of insulin resistance and the development of type 2 diabetes mellitus. Impaired insulin action in adipocytes is associated with elevated lipolysis and increased free fatty acids leading to ectopic fat deposition in liver and skeletal muscle. Chronic adipose tissue hypoxia has been suggested to be part of pathomechanisms causing dysfunction of adipocytes. Hypoxia can provoke oxidative stress in human and animal adipocytes and reduce the production of beneficial adipokines, such as adiponectin. However, time-dose responses to hypoxia relativize the effects of hypoxic stress. Long-term exposure of fat cells to hypoxia can lead to the production of beneficial substances such as leptin. Knowledge of time-dose responses of hypoxia on white adipose tissue and the time course of generation of oxidative stress in adipocytes is still scarce. This paper reviews the potential links between adipose tissue hypoxia, oxidative stress, mitochondrial dysfunction, and low-grade inflammation caused by adipocyte hypertrophy, macrophage infiltration and production of inflammatory mediators.
Collapse
Affiliation(s)
- Nikolaus Netzer
- Department of Sport Science, Faculty for Sports Science and Psychology, University of Innsbruck, Innsbruck 6020, Austria.
- Hermann Buhl Institute for Hypoxia and Sleep Medicine Research, Bad Aibling 83043, Germany.
- Department Medicine, Division Sports Medicine, University Hospitals Ulm, Ulm 89081, Germany.
| | - Hannes Gatterer
- Department of Sport Science, Faculty for Sports Science and Psychology, University of Innsbruck, Innsbruck 6020, Austria.
| | - Martin Faulhaber
- Department of Sport Science, Faculty for Sports Science and Psychology, University of Innsbruck, Innsbruck 6020, Austria.
| | - Martin Burtscher
- Department of Sport Science, Faculty for Sports Science and Psychology, University of Innsbruck, Innsbruck 6020, Austria.
| | - Stephan Pramsohler
- Hermann Buhl Institute for Hypoxia and Sleep Medicine Research, Bad Aibling 83043, Germany.
| | - Dominik Pesta
- Department of Sport Science, Faculty for Sports Science and Psychology, University of Innsbruck, Innsbruck 6020, Austria.
| |
Collapse
|
28
|
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: 9.9] [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
|
29
|
Xiong Y, Qu Z, Chen N, Gong H, Song M, Chen X, Du J, Xu C. The local corticotropin-releasing hormone receptor 2 signalling pathway partly mediates hypoxia-induced increases in lipolysis via the cAMP-protein kinase A signalling pathway in white adipose tissue. Mol Cell Endocrinol 2014; 392:106-14. [PMID: 24859650 DOI: 10.1016/j.mce.2014.05.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 04/19/2014] [Accepted: 05/13/2014] [Indexed: 12/27/2022]
Abstract
Our objective was to investigate the mechanisms by which the endogenous CRHR2 in white adipose tissue (WAT) regulates metabolic activities associated with lipogenesis and lipolysis under continuous exposure to hypoxia. We found that hypobaric hypoxia at a simulated altitude of 5000 m significantly reduced the body weight, food intake, and WAT mass of rats. Hypoxia also accelerated lipolysis and suppressed lipogenesis in WAT. Pretreatment with astressin 2B, a selective CRHR2 antagonist, partly but significantly attenuated the hypoxia-induced reductions in body weight and WAT mass by blocking the cAMP-protein kinase A (PKA)-hormone-sensitive lipase (HSL)/perilipin signalling pathway. Astressin 2B treatment failed to attenuate hypoxia induced lipogenic inhibition. In conclusion, activation of endogenous WAT Ucn2/3 autocrine/paracrine pathway was involved in hypoxia induced lipolysis via CRHR2 - cAMP-PKA signalling pathway. This study provides the novel understanding of local CRHR2 signaling pathway playing important role in WAT loss and lipid metabolism under hypoxia.
Collapse
Affiliation(s)
- Yanlei Xiong
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS), School of Basic Medicine, Peking Union Medical College (PUMC), Beijing, People's Republic of China
| | - Zhuan Qu
- Division of Neurobiology and Physiology, Department of Basic Medical Sciences, School of Medicine, Key Laboratory of Medical Neurobiology of the Ministry of Health, Zhejiang University, Hangzhou, People's Republic of China
| | - Nan Chen
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS), School of Basic Medicine, Peking Union Medical College (PUMC), Beijing, People's Republic of China
| | - Hui Gong
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS), School of Basic Medicine, Peking Union Medical College (PUMC), Beijing, People's Republic of China
| | - Mintao Song
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS), School of Basic Medicine, Peking Union Medical College (PUMC), Beijing, People's Republic of China
| | - Xuequn Chen
- Division of Neurobiology and Physiology, Department of Basic Medical Sciences, School of Medicine, Key Laboratory of Medical Neurobiology of the Ministry of Health, Zhejiang University, Hangzhou, People's Republic of China
| | - Jizeng Du
- Division of Neurobiology and Physiology, Department of Basic Medical Sciences, School of Medicine, Key Laboratory of Medical Neurobiology of the Ministry of Health, Zhejiang University, Hangzhou, People's Republic of China.
| | - Chengli Xu
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS), School of Basic Medicine, Peking Union Medical College (PUMC), Beijing, People's Republic of China.
| |
Collapse
|
30
|
Rodrigues T, Matafome P, Seiça R. A vascular piece in the puzzle of adipose tissue dysfunction: mechanisms and consequences. Arch Physiol Biochem 2014; 120:1-11. [PMID: 24063516 DOI: 10.3109/13813455.2013.838971] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
In the last years, several studies unravelled many aspects of adipose tissue pathophysiology in metabolic diseases. Some studies suggested hypoxia as one of such aspects, despite the exact mechanisms and pathophysiological significance is still partially unknown. Adipose tissue was shown to be hypoxic in obesity, mainly resulting from adipocyte hypertrophy, leading to increased activation of inflammatory pathways. In animal and cell models, hypoxia-induced inflammation was shown to lead to endocrine alterations and dysmetabolism. However, recent evidences suggest that instead of a simple low oxygenation theory, adipose tissue microvasculature may be regulated by a series of factors, including vasoactive factors like angiotensin II, angiogenesis and glycation, among others. This review summarizes the current knowledge about the role of these factors in the regulation of adipose tissue irrigation and the functional consequences of adipose tissue microvascular dysfunction.
Collapse
Affiliation(s)
- Tiago Rodrigues
- Laboratory of Physiology, Faculty of Medicine, Institute of Biomedical Imaging and Life Sciences (IBILI), University of Coimbra , Portugal
| | | | | |
Collapse
|
31
|
Das SS, Hayashi H, Sato T, Yamada R, Hiratsuka M, Hirasawa N. Regulation of dipeptidyl peptidase 4 production in adipocytes by glucose. Diabetes Metab Syndr Obes 2014; 7:185-94. [PMID: 24920931 PMCID: PMC4043708 DOI: 10.2147/dmso.s62610] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE Type 1 and 2 diabetes are characterized by elevated blood glucose levels and increased dipeptidyl peptidase 4 (DPP4) activity levels in the serum. However, previous studies reported a negative correlation between glucose concentrations and DPP4 levels. The purpose of this study was to elucidate the connection between glucose and DPP4 in adipocytes under physiological and diabetic conditions, because DPP4 is an adipokine. METHODS Blood glucose and serum DPP4 levels were measured, and adipocytes were collected from mice under normal, high-fat diet fed, and diabetic conditions. The adipocytes obtained were incubated for 24 hours in medium containing 5.5 or 25 mM glucose, and 3T3-L1 preadipocytes were differentiated under 5.5 or 25 mM glucose. Adipocytes from mice and 3T3-L1 were stimulated by tumor necrosis factor-α (TNF-α) for 24 hours. The levels of released and intracellular DPP4 were determined by enzyme-linked immunosorbent assay. RESULTS Mice fed high-fat diet had lower serum DPP4 levels in the first and second week than controls. However, this difference gradually disappeared over 6 weeks. The differentiation of 3T3-L1 adipocytes under 25 mM glucose produced lower DPP4 levels than those differentiated under 5.5 mM; this was also observed in isolated adipocytes from mice. However, these effects of glucose were lost in adipocytes from diabetic mice, and an increase in total DPP4 levels was observed. The stimulation of adipocytes with TNF-α increased the release of DPP4 irrespective of glucose concentration. CONCLUSION The production of DPP4 in adipocytes was negatively regulated by 25 mM glucose under physiological conditions, but not in diabetic mice. Our results suggest that the observed increase in serum DPP4 levels may be attributed to increased production of DPP4 in adipocytes and an enhancement in TNF-α-induced release.
Collapse
Affiliation(s)
- Siddhartha Shankar Das
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Hiroto Hayashi
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Taiki Sato
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Ren Yamada
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Masahiro Hiratsuka
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Noriyasu Hirasawa
- Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
- Correspondence: Noriyasu Hirasawa, Laboratory of Pharmacotherapy of Life-Style Related Diseases, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba Aramaki, Aoba-Ku, Sendai, Miyagi, Japan 982-0841, Tel +81 22 795 5915, Fax +81 22 795 5504, Email
| |
Collapse
|
32
|
Rosenow A, Noben JP, Bouwman FG, Mariman ECM, Renes J. Hypoxia-mimetic effects in the secretome of human preadipocytes and adipocytes. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2761-71. [PMID: 24140569 DOI: 10.1016/j.bbapap.2013.10.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 09/16/2013] [Accepted: 10/03/2013] [Indexed: 10/26/2022]
Abstract
White adipose tissue (WAT) regulates energy metabolism by secretion of proteins with endocrine and paracrine effects. Dysregulation of the secretome of obesity-associated enlarged WAT may lead to obesity-related disorders. This can be caused by hypoxia as a result of poorly vascularized WAT. The effect of hypoxia on the secretome of human (pre)adipocytes is largely unknown. Therefore, we investigated the effect of CoCl2, a hypoxia mimetic, on the secretome of human SGBS (pre)adipocytes by a proteomics approach combined with bioinformatic analysis. In addition, regulation of protein secretion was examined by protein turnover experiments. As such, secretome changes were particularly associated with protein down-regulation and extracellular matrix protein dysregulation. The observed up-regulation of collagens in adipocytes may be essential for cell survival while down-regulation of collagens in preadipocytes may indicate a disturbed differentiation process. These CoCl2-induced changes reflect WAT dysfunction that ultimately may lead to obesity-associated complications. In addition, 9 novel adipocyte secreted proteins were identified from which 6 were regulated by CoCl2. Mass spectrometry data have been deposited to the ProteomeXchange with identifier PXD000162.
Collapse
Affiliation(s)
- Anja Rosenow
- NUTRIM School for Nutrition, Toxicology and Metabolism, Department of Human Biology, Maastricht University, Maastricht, The Netherlands.
| | | | | | | | | |
Collapse
|