1
|
Zhang Q, Yang Z, Ou X, Zhang M, Qin X, Wu G. The role of immunity in insulin resistance in patients with polycystic ovary syndrome. Front Endocrinol (Lausanne) 2025; 15:1464561. [PMID: 39911236 PMCID: PMC11797073 DOI: 10.3389/fendo.2024.1464561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 12/31/2024] [Indexed: 02/07/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent disorder of the endocrine system with significant clinical implications, often leading to health complications related to adipose tissue accumulation, including obesity, insulin resistance (IR), metabolic syndrome, and type 2 diabetes mellitus. While the precise pathogenesis of PCOS remains unclear, it is now recognized that genetic, endocrine, and metabolic dysregulations all contribute significantly to its onset. The immunopathogenesis of PCOS has not been extensively explored, but there is growing speculation that immune system abnormalities may play a pivotal role. This chronic inflammatory state is exacerbated by factors such as obesity and hyperinsulinemia. Therefore, this review aims to elucidate the interplay between IR in PCOS patients, the controlled immune response orchestrated by immune cells and immunomodulatory molecules, and their interactions with adipocytes, hyperandrogenemia, chronic inflammation, and metabolic homeostasis.
Collapse
Affiliation(s)
- Qixuan Zhang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhe Yang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiangyang Ou
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mengying Zhang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiangyu Qin
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gengxiang Wu
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
2
|
Huang Y, Chen L, Li L, Qi Y, Tong H, Wu H, Xu J, Leng L, Cheema S, Sun G, Xia Z, McGuire J, Rodrigues B, Young LH, Bucala R, Qi D. Downregulation of adipose LPL by PAR2 contributes to the development of hypertriglyceridemia. JCI Insight 2024; 9:e173240. [PMID: 38973609 PMCID: PMC11383372 DOI: 10.1172/jci.insight.173240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 05/22/2024] [Indexed: 07/09/2024] Open
Abstract
Lipoprotein lipase (LPL) hydrolyzes circulating triglycerides (TGs), releasing fatty acids (FA) and promoting lipid storage in white adipose tissue (WAT). However, the mechanisms regulating adipose LPL and its relationship with the development of hypertriglyceridemia are largely unknown. WAT from obese humans exhibited high PAR2 expression, which was inversely correlated with the LPL gene. Decreased LPL expression was also inversely correlated with elevated plasma TG levels, suggesting that adipose PAR2 might regulate hypertriglyceridemia by downregulating LPL. In mice, aging and high palmitic acid diet (PD) increased PAR2 expression in WAT, which was associated with a high level of macrophage migration inhibitory factor (MIF). MIF downregulated LPL expression and activity in adipocytes by binding with CXCR2/4 receptors and inhibiting Akt phosphorylation. In a MIF overexpression model, high-circulating MIF levels suppressed adipose LPL, and this suppression was associated with increased plasma TGs but not FA. Following PD feeding, adipose LPL expression and activity were significantly reduced, and this reduction was reversed in Par2-/- mice. Recombinant MIF infusion restored high plasma MIF levels in Par2-/- mice, and the levels decreased LPL and attenuated adipocyte lipid storage, leading to hypertriglyceridemia. These data collectively suggest that downregulation of adipose LPL by PAR2/MIF may contribute to the development of hypertriglyceridemia.
Collapse
Affiliation(s)
- Yiheng Huang
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Liujun Chen
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lisha Li
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Yadan Qi
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Haibin Tong
- College of Life and Environment Sciences, Wenzhou University, Wenzhou, Zhejiang, China
| | - Hong Wu
- Institute of Cardiovascular Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jinjie Xu
- Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Lin Leng
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Guang Sun
- Faculty of Medicine, Memorial University, St. John’s, Newfoundland, Canada
| | - Zhengyuan Xia
- Guangdong Medical University, Zhanjiang, Guangdong, China
| | - John McGuire
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Brian Rodrigues
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lawrence H. Young
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Dake Qi
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Faculty of Medicine, Memorial University, St. John’s, Newfoundland, Canada
| |
Collapse
|
3
|
Huang Y, Cui D, Chen L, Tong H, Wu H, Muller GK, Qi Y, Wang S, Xu J, Gao X, Fifield KE, Wang L, Xia Z, Vanderluit JL, Liu S, Leng L, Sun G, McGuire J, Young LH, Bucala R, Qi D. A pref-1-controlled non-inflammatory mechanism of insulin resistance. iScience 2023; 26:106923. [PMID: 37283810 PMCID: PMC10239698 DOI: 10.1016/j.isci.2023.106923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/21/2023] [Accepted: 05/15/2023] [Indexed: 06/08/2023] Open
Abstract
While insulin resistance (IR) is associated with inflammation in white adipose tissue, we report a non-inflammatory adipose mechanism of high fat-induced IR mediated by loss of Pref-1. Pref-1, released from adipose Pref-1+ cells with characteristics of M2 macrophages, endothelial cells or progenitors, inhibits MIF release from both Pref-1+ cells and adipocytes by binding with integrin β1 and inhibiting the mobilization of p115. High palmitic acid induces PAR2 expression in Pref-1+ cells, downregulating Pref-1 expression and release in an AMPK-dependent manner. The loss of Pref-1 increases adipose MIF secretion contributing to non-inflammatory IR in obesity. Treatment with Pref-1 blunts the increase in circulating plasma MIF levels and subsequent IR induced by a high palmitic acid diet. Thus, high levels of fatty acids suppress Pref-1 expression and secretion, through increased activation of PAR2, resulting in an increase in MIF secretion and a non-inflammatory adipose mechanism of IR.
Collapse
Affiliation(s)
- Yiheng Huang
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Donghong Cui
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liujun Chen
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Haibin Tong
- College of Life and Environment Sciences, Wenzhou University, Wenzhou, Zhejiang, China
| | - Hong Wu
- Institute of Cardiovascular Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Grace K. Muller
- Department of Cell and Molecular Physiology, Loyola University, Chicago, IL, USA
| | - Yadan Qi
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Shuxia Wang
- Department of Cardiology, The General Hospital of Chinese PLA, Beijing, China
| | - Jinjie Xu
- Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Xiang Gao
- College of Life Sciences, Qingdao University, Qingdao, Shandong, China
| | - Kathleen E. Fifield
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - Lingyan Wang
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| | | | - Jacqueline L. Vanderluit
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - Suixin Liu
- Division of Cardiac Rehabilitation, Department of Physical Medicine & Rehabilitation, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Lin Leng
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Guang Sun
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| | - John McGuire
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Lawrence H. Young
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Dake Qi
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL, Canada
| |
Collapse
|
4
|
NGF and Its Role in Immunoendocrine Communication during Metabolic Syndrome. Int J Mol Sci 2023; 24:ijms24031957. [PMID: 36768281 PMCID: PMC9916855 DOI: 10.3390/ijms24031957] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 01/20/2023] Open
Abstract
Nerve growth factor (NGF) was the first neurotrophin described. This neurotrophin contributes to organogenesis by promoting sensory innervation and angiogenesis in the endocrine and immune systems. Neuronal and non-neuronal cells produce and secrete NGF, and several cell types throughout the body express the high-affinity neurotrophin receptor TrkA and the low-affinity receptor p75NTR. NGF is essential for glucose-stimulated insulin secretion and the complete development of pancreatic islets. Plus, this factor is involved in regulating lipolysis and thermogenesis in adipose tissue. Immune cells produce and respond to NGF, modulating their inflammatory phenotype and the secretion of cytokines, contributing to insulin resistance and metabolic homeostasis. This neurotrophin regulates the synthesis of gonadal steroid hormones, which ultimately participate in the metabolic homeostasis of other tissues. Therefore, we propose that this neurotrophin's imbalance in concentrations and signaling during metabolic syndrome contribute to its pathophysiology. In the present work, we describe the multiple roles of NGF in immunoendocrine organs that are important in metabolic homeostasis and related to the pathophysiology of metabolic syndrome.
Collapse
|
5
|
Somm E, Jornayvaz FR. Interleukin-18 in metabolism: From mice physiology to human diseases. Front Endocrinol (Lausanne) 2022; 13:971745. [PMID: 36313762 PMCID: PMC9596921 DOI: 10.3389/fendo.2022.971745] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
Interleukin-18 (IL-18) is a classical member of the IL-1 superfamily of cytokines. As IL-1β, IL-18 precursor is processed by inflammasome/caspase-1 into a mature and biologically active form. IL-18 binds to its specific receptor composed of two chains (IL-18Rα and IL-18Rβ) to trigger a similar intracellular signaling pathway as IL-1, ultimately leading to activation of NF-κB and inflammatory processes. Independently of this IL-1-like signaling, IL-18 also specifically induces IFN-γ production, driving the Th1 immune response. In circulation, IL-18 binds to the IL-18 binding protein (IL-18BP) with high affinity, letting only a small fraction of free IL-18 able to trigger receptor-mediated signaling. In contrast to other IL-1 family members, IL-18 is produced constitutively by different cell types, suggesting implications in normal physiology. If the roles of IL-18 in inflammatory processes and infectious diseases are well described, recent experimental studies in mice have highlighted the action of IL-18 signaling in the control of energy homeostasis, pancreatic islet immunity and liver integrity during nutritional stress. At the same time, clinical observations implicate IL-18 in various metabolic diseases including obesity, type 1 and 2 diabetes and nonalcoholic fatty liver disease (NAFLD)/nonalcoholic steatohepatitis (NASH). In the present review, we summarize and discuss both the physiological actions of IL-18 in metabolism and its potential roles in pathophysiological mechanisms leading to the most common human metabolic disorders, such as obesity, diabetes and NAFLD/NASH.
Collapse
Affiliation(s)
- Emmanuel Somm
- Service of Endocrinology, Diabetes, Nutrition and Therapeutic Patient Education, Department of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - François R. Jornayvaz
- Service of Endocrinology, Diabetes, Nutrition and Therapeutic Patient Education, Department of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
6
|
Gateva A, Kamenov Z, Karamfilova V, Assyov Y, Velikova T, El-Darawish Y, Okamura H. Higher levels of IL-18 in patients with prediabetes compared to obese normoglycaemic controls. Arch Physiol Biochem 2020; 126:449-452. [PMID: 30632794 DOI: 10.1080/13813455.2018.1555667] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Background: Overweight and obesity are linked to low-grade chronic inflammation that can impair normal insulin function and induce insulin resistance. The aim of this study was to compare IL-18 levels between patients with prediabetes and obese normoglycaemic controls.Patients and methods: In this study, we included 131 patients with mean age 54.9 ± 9.1 years, divided into two groups - group 1 with obesity without glycaemic disturbances (n = 66) and group 2 with prediabetes (n = 65). IL-18 was measured using enzyme-linked immunosorbent assay (ELISA) method.Results: Patients with prediabetes had significantly higher levels of IL-18 compared to obese controls (304.0 ± 220.4 vs. 233.6 ± 103.6 pg/l, p=.029). When patients with prediabetes were divided into IFG only, IGT only and IFG + IGT the highest levels of IL-18 were found in IGT only patients.Conclusions: Patients with prediabetes have higher levels of IL18 compared to obese normoglycemic controls.
Collapse
Affiliation(s)
- Antoaneta Gateva
- Department of Internal Medicine, Clinic of Endocrinology, University Hospital "Alexandrovska", Medical University - Sofia, Sofia, Bulgaria
| | - Zdravko Kamenov
- Department of Internal Medicine, Clinic of Endocrinology, University Hospital "Alexandrovska", Medical University - Sofia, Sofia, Bulgaria
| | - Vera Karamfilova
- Department of Internal Medicine, Clinic of Endocrinology, University Hospital "Alexandrovska", Medical University - Sofia, Sofia, Bulgaria
| | - Yavor Assyov
- Department of Internal Medicine, Clinic of Endocrinology, University Hospital "Alexandrovska", Medical University - Sofia, Sofia, Bulgaria
| | - Tsvetelina Velikova
- Department of Clinical Immunology, University Hospital Lozenetz, Sofia, Bulgaria
| | - Yosif El-Darawish
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, Tokyo, Japan
| | - Haruki Okamura
- Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, Tokyo, Japan
| |
Collapse
|
7
|
Lubow J, Collins KL. Vpr Is a VIP: HIV Vpr and Infected Macrophages Promote Viral Pathogenesis. Viruses 2020; 12:E809. [PMID: 32726944 PMCID: PMC7472745 DOI: 10.3390/v12080809] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/17/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023] Open
Abstract
HIV infects several cell types in the body, including CD4+ T cells and macrophages. Here we review the role of macrophages in HIV infection and describe complex interactions between viral proteins and host defenses in these cells. Macrophages exist in many forms throughout the body, where they play numerous roles in healthy and diseased states. They express pattern-recognition receptors (PRRs) that bind viral, bacterial, fungal, and parasitic pathogens, making them both a key player in innate immunity and a potential target of infection by pathogens, including HIV. Among these PRRs is mannose receptor, a macrophage-specific protein that binds oligosaccharides, restricts HIV replication, and is downregulated by the HIV accessory protein Vpr. Vpr significantly enhances infection in vivo, but the mechanism by which this occurs is controversial. It is well established that Vpr alters the expression of numerous host proteins by using its co-factor DCAF1, a component of the DCAF1-DDB1-CUL4 ubiquitin ligase complex. The host proteins targeted by Vpr and their role in viral replication are described in detail. We also discuss the structure and function of the viral protein Env, which is stabilized by Vpr in macrophages. Overall, this literature review provides an updated understanding of the contributions of macrophages and Vpr to HIV pathogenesis.
Collapse
Affiliation(s)
- Jay Lubow
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Kathleen L. Collins
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
8
|
Angelova P, Kamenov Z, Tsakova A, El-Darawish Y, Okamura H. Interleukin-18 and testosterone levels in men with metabolic syndrome. Aging Male 2018; 21:130-137. [PMID: 29168426 DOI: 10.1080/13685538.2017.1401993] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Interleukin 18 (IL-18) is an adipokine associated with obesity. Data about the relationship of IL-18 to the metabolic syndrome (MS) are still scarce. Low testosterone (T) levels are common in men with MS, but we did not find data about the levels of IL-18 in men with low T. The aim of this study was to determine the levels of IL-18 in men with MS with or without low T. PATIENTS AND METHODS A total of 251 men were included in the study. Of them 218 had MS (IDF 2005) and they were divided according to their morning total testosterone (TT) level (cutoff 10.4 nmol/l) into two groups: MS-low T (N = 84) and MS-normal T (N = 134). The control group consisted of 33 men without MS and low T. IL-18 was determined in serum using enzyme-linked immunosorbent assay. A small group of eight men with MS and low T levels received testosterone therapy for three months and physical and laboratory parameters were monitored at the end of that period. RESULTS MS men were at mean age (±SD) = 53.77 ± 9.59 years; body mass index (BMI) = 34.0 ± 6.3 kg/m2; and TT = 12.59 ± 5.66 nmol/l. The control group was at age = 52.12 ± 5.2 years (NS); BMI = 25.6 ± 2.4 kg/m2 (p < .001); and TT = 17.8 ± 5.68 nmol/l (p < .001), respectively. The levels of IL-18 were higher in the MS group - 345 pg/ml compared to the control one - 264 pg/ml (p < .01). There was no significant difference between MS-low T (330.6 pg/ml) and MS-normal T (350.2 pg/ml) subgroups. The MS-normal T differed more significantly from the control group (p < .001). Significant correlation of testosterone with IL-18 levels was not found. IL-18 correlated with parameters of obesity, lipids, fasting blood sugar (p < .05) and the number of criteria for MS (p < .001). Three months on T treatment showed improvement in obesity parameters and only in one patient IL-18 had clear reduction while the rest showed no change. CONCLUSIONS In this study, higher IL-18 levels were found in the presence of MS compared to healthy men, but they did not differ between men having MS with or without LOH.
Collapse
Affiliation(s)
- Petya Angelova
- a Clinic of Endocrinology , Alexandrovska University Hospital, Medical University-Sofia , Sofia , Bulgaria
| | - Zdravko Kamenov
- a Clinic of Endocrinology , Alexandrovska University Hospital, Medical University-Sofia , Sofia , Bulgaria
| | - Adelina Tsakova
- b Central Clinical Laboratory , Alexandrovska University Hospital, Medical University-Sofia , Sofia , Bulgaria
| | - Yosif El-Darawish
- c Laboratory of Tumor Immunology and Immunotherapy , Hyogo College of Medicine , Hyogo , Japan
| | - Haruki Okamura
- d Laboratory of Host Defense , Hyogo College of Medicine , Hyogo , Japan
| |
Collapse
|
9
|
Lindegaard B, Hvid T, Wolsk Mygind H, Hartvig-Mortensen O, Grøndal T, Abildgaard J, Gerstoft J, Pedersen BK, Baranowski M. Low expression of IL-18 and IL-18 receptor in human skeletal muscle is associated with systemic and intramuscular lipid metabolism-Role of HIV lipodystrophy. PLoS One 2018; 13:e0186755. [PMID: 29342149 PMCID: PMC5771554 DOI: 10.1371/journal.pone.0186755] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 10/07/2017] [Indexed: 01/22/2023] Open
Abstract
Introduction Interleukin (IL)-18 is involved in regulation of lipid and glucose metabolism. Mice lacking whole-body IL-18 signalling are prone to develop weight gain and insulin resistance, a phenotype which is associated with impaired fat oxidation and ectopic skeletal muscle lipid deposition. IL-18 mRNA is expressed in human skeletal muscle but a role for IL-18 in muscle has not been identified. Patients with HIV-infection and lipodystrophy (LD) are characterized by lipid and glucose disturbances and increased levels of circulating IL-18. We hypothesized that skeletal muscle IL-18 and IL-18 receptor (R) expression would be altered in patients with HIV-lipodystrophy. Design and methods Twenty-three HIV-infected patients with LD and 15 age-matched healthy controls were included in a cross-sectional study. Biopsies from the vastus lateralis muscle were obtained and IL-18 and IL-18R mRNA expression were measured by real-time PCR and sphingolipids (ceramides, sphingosine, sphingosine-1-Phosphate, sphinganine) were measured by HPLC. Insulin resistance was assessed by HOMA and the insulin response during an OGTT. Results Patients with HIV-LD had a 60% and 54% lower level of muscular IL-18 and IL-18R mRNA expression, respectively, compared to age-matched healthy controls. Patients with HIV-LD had a trend towards increased levels of ceramide (18.3±4.7 versus 14.8±3.0,p = 0.06) and sphingosine (0.41±0.13 versus 0.32±0.07, and lower level of sphinganine (p = 0.06). Low levels of muscle IL-18 mRNA correlated to high levels of ceramides (r = -0.31, p = 0.038) and sphingosine-1P (r = -0.29, p = 0.046) in skeletal muscle, whereas such a correlation was not found in healthy controls. Low expression of IL-18 mRNA in skeletal muscle correlated to elevated concentration of circulating triglycerides (Rp = -0.73, p<0.0001). Neither muscle expression of IL-18 mRNA or ceramide correlated to parameters of insulin resistance. Conclusion IL-18 (mRNA) in skeletal muscle appears to be involved in the regulation of intramuscular lipid metabolism and hypertriglyceridemia.
Collapse
Affiliation(s)
- Birgitte Lindegaard
- The Centre of Inflammation and Metabolism and The Centre of Physical Activity Research, Rigshospital, Copenhagen, Denmark
- The Department of Infectious Diseases, Rigshospitalet, Copenhagen, Denmark
- The Department of Lung- and Infectious Diseases, Nordsjællands Hospital, Hillerød, Denmark
- * E-mail:
| | - Thine Hvid
- The Centre of Inflammation and Metabolism and The Centre of Physical Activity Research, Rigshospital, Copenhagen, Denmark
| | - Helene Wolsk Mygind
- The Centre of Inflammation and Metabolism and The Centre of Physical Activity Research, Rigshospital, Copenhagen, Denmark
| | | | - Thomas Grøndal
- The Centre of Inflammation and Metabolism and The Centre of Physical Activity Research, Rigshospital, Copenhagen, Denmark
| | - Julie Abildgaard
- The Centre of Inflammation and Metabolism and The Centre of Physical Activity Research, Rigshospital, Copenhagen, Denmark
| | - Jan Gerstoft
- The Department of Infectious Diseases, Rigshospitalet, Copenhagen, Denmark
| | - Bente Klarlund Pedersen
- The Centre of Inflammation and Metabolism and The Centre of Physical Activity Research, Rigshospital, Copenhagen, Denmark
| | - Marcin Baranowski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
10
|
Zaharieva E, Kamenov Z, Velikova T, Tsakova A, El-Darawish Y, Okamura H. Interleukin-18 serum level is elevated in type 2 diabetes and latent autoimmune diabetes. Endocr Connect 2018; 7:179-185. [PMID: 29217651 PMCID: PMC5776671 DOI: 10.1530/ec-17-0273] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 12/07/2017] [Indexed: 02/05/2023]
Abstract
BACKGROUND Interleukin-18 (IL-18) is an inflammatory cytokine found to be elevated in obesity, metabolic syndrome and type 2 diabetes (T2D) as a part of the chronic low-grade inflammatory process in these states. The aim of the study was to evaluate the interleukin level in patients with latent autoimmune diabetes of the adults (LADA) in comparison to that in T2D subjects. MATERIALS AND METHODS IL-18 was analyzed through enzyme-linked immunosorbent assay in 76 participants with T2D and 24 with LADA and 14 control subjects. Evaluation was also carried out in body mass index (BMI)- and glycemic control-matched diabetic patients. RESULTS The serum concentration of IL-18 was higher in patients with T2D (389.04 ± 203.44 pg/mL) and LADA (327.04 ± 144.48 pg/mL) than that in control subjects (219.88 ± 91.03 pg/mL), P < 0.05. However, it was not significantly different between both diabetic groups (P = 0.255) despite higher IL-6 (4.78 ± 5.84 vs 1.79 ± 0.96 pg/mL, P < 0.001) and hs-CRP (2.60 ± 1.70 vs 1.29 ± 1.20 mg/L, P = 0.002) level in T2D patients. The results were persistent in BMI-matched subjects with diabetes (IL-18 = 403.48 ± 226.32 vs 329.30 ± 146.30 pg/mL, respectively for T2D and LADA, P = 0.391). The correlations in T2D group concerning HDL cholesterol (r = -0.377, P = 0.001), postprandial glucose (r = 0.244, P = 0.043), IL-6 (r = 0.398, P < 0.001) and hs-CRP (r = 0.427, P = 0.001) were not confirmed in LADA and control subjects. CONCLUSION The IL-18 serum level was higher in T2D and LADA than that in control subjects, but did not differ between both diabetic groups, even when they were BMI matched. Correlations with lipid, glycemic and inflammatory parameters were present in T2D only.
Collapse
Affiliation(s)
- Emanuela Zaharieva
- Department of Internal MedicineUniversity Hospital Alexandrovska, Clinic of Endocrinology, Faculty of Medicine, Medical University-Sofia, Sofia, Bulgaria
| | - Zdravko Kamenov
- Department of Internal MedicineUniversity Hospital Alexandrovska, Clinic of Endocrinology, Faculty of Medicine, Medical University-Sofia, Sofia, Bulgaria
| | - Tsvetelina Velikova
- Department of Clinical ImmunologyUniversity Hospital St. Ivan Rilski, Laboratory of Clinical Immunology, Faculty of Medicine, Medical University-Sofia, Sofia, Bulgaria
| | - Adelina Tsakova
- Department of Clinical LaboratoryUniversity Hospital Alexandrovska, Central Clinical Laboratory, Faculty of Medicine, Medical University-Sofia, Sofia, Bulgaria
| | - Yosif El-Darawish
- Laboratory of Tumor Immunology and Cell TherapyHyogo College of Medicine, Hyogo, Japan
| | - Haruki Okamura
- Laboratory of Tumor Immunology and Cell TherapyHyogo College of Medicine, Hyogo, Japan
| |
Collapse
|
11
|
Ahmad R, Thomas R, Kochumon S, Sindhu S. Increased adipose tissue expression of IL-18R and its ligand IL-18 associates with inflammation and insulin resistance in obesity. IMMUNITY INFLAMMATION AND DISEASE 2017; 5:318-335. [PMID: 28508444 PMCID: PMC5569378 DOI: 10.1002/iid3.170] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 03/23/2017] [Accepted: 04/18/2017] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The proinflammatory cytokine IL-18 is involved in the pathogenesis of metabolic syndrome. While the changes in IL-18 are known, IL-18R expression and relationship with IL-18 and other inflammatory markers in the adipose tissue in obesity/type-2 diabetes (T2D) remain unclear. METHODS We, therefore, determined the adipose tissue expression of IL-18R and IL-18 mRNA/protein in lean, overweight, and obese individuals with and without T2D, 15 each, using qRT-PCR, immunohistochemistry, and confocal microscopy. Data (mean ± SEM) were analyzed using unpaired t-test and Pearson's correlation (r); all P values ≤0.05 were considered statistically significant. RESULTS We found the upregulated gene/protein expression of IL-18R and IL-18 in non-diabetic obese/overweight as compared with lean individuals (P < 0.05). BMI correlated positively (P < 0.05) with the adipose tissue expression of IL-18R (mRNA: r = 0.90 protein: r = 0.84) and IL-18 (mRNA: r = 0.84 protein: r = 0.80). Similarly, in T2D individuals, gene and protein expression of IL-18R/IL-18 was significantly higher in obese as compared with overweight/lean individuals. The BMI was associated with the changes in both IL-18R (mRNA: r = 0.55 protein: r = 0.50) and IL-18 (mRNA: r = 0.53 protein: r = 0.57) expression. IL-18R/IL-18 gene expression in the adipose tissue was positively associated (P < 0.05) with local gene expression of other inflammatory markers including CD11c, CD86, CD68, CD163, TNF-α, and CCL5. Homeostatic model assessment of insulin resistance (HOMA-IR) was higher in diabetic/non-diabetic obese and it correlated with BMI (P < 0.05). IL-18R and IL-18 mRNA/protein expression in obesity was associated with HOMA-IR only in non-diabetics. CONCLUSIONS The adipose tissue IL-18R/IL-18 expression is enhanced in obesity which associates with proinflammatory gene signature and insulin resistance in these individuals.
Collapse
Affiliation(s)
- Rasheed Ahmad
- Immunology and Innovative Cell Therapy Unit, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Reeby Thomas
- Immunology and Innovative Cell Therapy Unit, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Shihab Kochumon
- Immunology and Innovative Cell Therapy Unit, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Sardar Sindhu
- Immunology and Innovative Cell Therapy Unit, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| |
Collapse
|
12
|
Li Z, Wang H, Chen Y, Niu J, Guo Q, Leng Q, Huang Z, Deng Z, Meng G. Interleukin-18 protects mice from Enterovirus 71 infection. Cytokine 2017; 96:132-137. [PMID: 28399485 DOI: 10.1016/j.cyto.2017.04.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 03/14/2017] [Accepted: 04/03/2017] [Indexed: 12/24/2022]
Abstract
Previous study has demonstrated that the NLRP3 inflammasome is essential for protecting murine host against Enterovirus 71 (EV71) infection. However, the underlying mechanism remained unknown. Here we discovered that the pleiotropic cytokine interleukin-18 (IL-18), an NLRP3 inflammasome-dependent effector protein, exhibits a protective capability against EV71 challenge. Deficiency of IL-18 in mice exacerbated EV71 infection, which was reflected by increased viral replication, elevated production of interferons (IFN-β, IFN-γ), proinflammatory cytokines (TNF-α, IL-6) and chemokine CCL2,as well as decreased survival of experimental animals. Conversely, administration of recombinant IL-18 considerably restrained EV71 infection in IL-18 deficient mice. Thus, our results revealed a protective role for IL-18 against EV71 challenge, and indicated a novel therapeutic application for IL-18 in EV71 associated hand, foot, and mouth disease (HFMD).
Collapse
Affiliation(s)
- Zheng Li
- School of Life Sciences, Shanghai University, Shanghai 200444, China; CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hongbin Wang
- CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yihui Chen
- CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Junling Niu
- CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiuhong Guo
- CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qibin Leng
- CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhong Huang
- CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhirui Deng
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Guangxun Meng
- CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
13
|
Kim BS, Tilstam PV, Hwang SS, Simons D, Schulte W, Leng L, Sauler M, Ganse B, Averdunk L, Kopp R, Stoppe C, Bernhagen J, Pallua N, Bucala R. D-dopachrome tautomerase in adipose tissue inflammation and wound repair. J Cell Mol Med 2016; 21:35-45. [PMID: 27605340 PMCID: PMC5192814 DOI: 10.1111/jcmm.12936] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 06/23/2016] [Indexed: 12/31/2022] Open
Abstract
D-dopachrome tautomerase (D-DT/MIF-2) is a member of the macrophage migration inhibitory factor (MIF) cytokine superfamily, and a close structural homolog of MIF. MIF and D-DT have been reported to be involved in obesity, but there is little known about the regulation of D-DT in adipose tissue inflammation and wound healing. Subcutaneous adipose tissue was collected from 54 healthy donors and 28 donors with acutely inflamed wounds undergoing wound debridement. In addition, epididymal fat pads of mice were injected with lipopolysaccharide to study receptor expression and cell migration in vivo. D-DT protein levels and mRNA expression were significantly decreased in subcutaneous adipose tissue adjacent to acutely inflamed wounds. D-DT improved fibroblast viability and increased proliferation in vitro. While D-DT alone did not have a significant effect on in vitro fibroblast wound healing, simultaneous addition of neutralizing MIF antibody resulted in a significant improvement of fibroblast wound healing. Interestingly, expression of the MIF and D-DT receptor CD74 was down-regulated while the MIF receptors CXCR2 and CXCR4 were up-regulated primarily on macrophages indicating that the MIF-CXCR2/4 axis may promote recruitment of inflammatory cells into adipose tissue. Our results describe a reciprocal role of D-DT to MIF in inflamed adipose tissue, and indicate that D-DT may be beneficial in wound repair by improving fibroblast survival and proliferation.
Collapse
Affiliation(s)
- Bong-Sung Kim
- Department of Plastic and Reconstructive Surgery, Hand Surgery - Burn Center, RWTH Aachen University, Aachen, Germany.,Section of Rheumatology, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Pathricia V Tilstam
- Section of Rheumatology, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Soo Seok Hwang
- Department of Immunology, Yale University School of Medicine, New Haven, CT, USA
| | - David Simons
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wibke Schulte
- Section of Rheumatology, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Lin Leng
- Section of Rheumatology, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Maor Sauler
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Bergita Ganse
- Department of Orthopedic Trauma Surgery, RWTH Aachen University, Aachen, Germany
| | - Luisa Averdunk
- Department of Intensive Care Medicine, RWTH Aachen University, Aachen, Germany
| | - Rüdger Kopp
- Department of Intensive Care Medicine, RWTH Aachen University, Aachen, Germany
| | - Christian Stoppe
- Department of Intensive Care Medicine, RWTH Aachen University, Aachen, Germany
| | - Jürgen Bernhagen
- Chair of Vascular Biology, Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Norbert Pallua
- Department of Plastic and Reconstructive Surgery, Hand Surgery - Burn Center, RWTH Aachen University, Aachen, Germany
| | - Richard Bucala
- Section of Rheumatology, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
14
|
Flynn MG, McFarlin BK, Markofski MM. The Anti-Inflammatory Actions of Exercise Training. Am J Lifestyle Med 2016; 1:220-235. [PMID: 25431545 DOI: 10.1177/1559827607300283] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The list of diseases with a known inflammatory etiology is growing. Cardiovascular disease, osteoporosis, diabetes, geriatric cachexia, and Alzheimer's disease have all been shown to be linked to or exacerbated by aberrantly regulated inflammatory processes. Nevertheless, there is mounting evidence that those who are physically active, or who become physically active, have a reduction in biomarkers associated with chronic inflammation. There was strong early consensus that exercise-induced reductions in inflammation were explained by body mass index or body fatness, but recent studies provide support for the contention that exercise has body fat-independent anti-inflammatory effects. With few exceptions, the anti-inflammatory effects of exercise appear to occur regardless of age or the presence of chronic diseases. What remains unclear are the mechanisms by which exercise training induces these anti-inflammatory effects, but there are several intriguing possibilities, including release of endogenous products, such as heat shock proteins; selective reduction of visceral adipose tissue mass or reducing infiltration of adipocytes by macrophages; shift in immune cell phenotype; cross-tolerizing effects; or exercise-induced shifts in accessory proteins of toll-like receptor signaling. However, future research endeavors are likely to uncover additional potential mechanisms, and it could be some time before functional mechanisms are made clear. In summary, the potential anti-inflammatory influences of exercise training may provide a low-cost, readily available, and effective treatment for low-grade systemic inflammation and could contribute significantly to the positive effects of exercise training on chronic disease.
Collapse
Affiliation(s)
- Michael G Flynn
- Wastl Human Performance Lab, Department of Health and Kinesiology, Purdue University West Lafayette, Indiana (MGF, MMM) and the Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, Houston, Texas (BKM)
| | - Brian K McFarlin
- Wastl Human Performance Lab, Department of Health and Kinesiology, Purdue University West Lafayette, Indiana (MGF, MMM) and the Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, Houston, Texas (BKM)
| | - Melissa M Markofski
- Wastl Human Performance Lab, Department of Health and Kinesiology, Purdue University West Lafayette, Indiana (MGF, MMM) and the Laboratory of Integrated Physiology, Department of Health and Human Performance, University of Houston, Houston, Texas (BKM)
| |
Collapse
|
15
|
Murphy AJ, Kraakman MJ, Kammoun HL, Dragoljevic D, Lee MKS, Lawlor KE, Wentworth JM, Vasanthakumar A, Gerlic M, Whitehead LW, DiRago L, Cengia L, Lane RM, Metcalf D, Vince JE, Harrison LC, Kallies A, Kile BT, Croker BA, Febbraio MA, Masters SL. IL-18 Production from the NLRP1 Inflammasome Prevents Obesity and Metabolic Syndrome. Cell Metab 2016; 23:155-64. [PMID: 26603191 DOI: 10.1016/j.cmet.2015.09.024] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 06/03/2015] [Accepted: 09/23/2015] [Indexed: 11/15/2022]
Abstract
Interleukin-18 (IL-18) is activated by Caspase-1 in inflammasome complexes and has anti-obesity effects; however, it is not known which inflammasome regulates this process. We found that mice lacking the NLRP1 inflammasome phenocopy mice lacking IL-18, with spontaneous obesity due to intrinsic lipid accumulation. This is exacerbated when the mice are fed a high-fat diet (HFD) or a high-protein diet, but not when mice are fed a HFD with low energy density (high fiber). Furthermore, mice with an activating mutation in NLRP1, and hence increased IL-18, have decreased adiposity and are resistant to diet-induced metabolic dysfunction. Feeding these mice a HFD further increased plasma IL-18 concentrations and strikingly resulted in loss of adipose tissue mass and fatal cachexia, which could be prevented by genetic deletion of IL-18. Thus, NLRP1 is an innate immune sensor that functions in the context of metabolic stress to produce IL-18, preventing obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Andrew J Murphy
- Haematopoiesis and Leukocyte Biology, Baker IDI Heart and Diabetes Institute, Melbourne 3004, Australia; Department of Immunology, Monash University, Melbourne 3004, Australia
| | - Michael J Kraakman
- Haematopoiesis and Leukocyte Biology, Baker IDI Heart and Diabetes Institute, Melbourne 3004, Australia
| | - Helene L Kammoun
- Haematopoiesis and Leukocyte Biology, Baker IDI Heart and Diabetes Institute, Melbourne 3004, Australia; Cellular and Molecular Metabolism Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne 3004, Australia
| | - Dragana Dragoljevic
- Haematopoiesis and Leukocyte Biology, Baker IDI Heart and Diabetes Institute, Melbourne 3004, Australia; Department of Immunology, Monash University, Melbourne 3004, Australia
| | - Man K S Lee
- Haematopoiesis and Leukocyte Biology, Baker IDI Heart and Diabetes Institute, Melbourne 3004, Australia; Department of Immunology, Monash University, Melbourne 3004, Australia
| | - Kate E Lawlor
- Division of Inflammation, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - John M Wentworth
- Division of Molecular Medicine, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Ajithkumar Vasanthakumar
- Division of Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Motti Gerlic
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Lachlan W Whitehead
- Division of Systems Biology and Personalised Medicine, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
| | - Ladina DiRago
- Division of Cancer and Hematology, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
| | - Louise Cengia
- Division of Cancer and Hematology, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
| | - Rachael M Lane
- Division of ACRF Chemical Biology, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
| | - Donald Metcalf
- Division of Cancer and Hematology, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - James E Vince
- Division of Inflammation, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Leonard C Harrison
- Division of Molecular Medicine, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Axel Kallies
- Division of Immunology, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Benjamin T Kile
- Division of ACRF Chemical Biology, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia
| | - Ben A Croker
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mark A Febbraio
- Cellular and Molecular Metabolism Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne 3004, Australia; Division of Diabetes and Metabolism, Garvan Institute of Medical Research, Darlinghurst 2010, Australia
| | - Seth L Masters
- Division of Inflammation, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville 3010, Australia.
| |
Collapse
|
16
|
Kim BS, Rongisch R, Hager S, Grieb G, Nourbakhsh M, Rennekampff HO, Bucala R, Bernhagen J, Pallua N. Macrophage Migration Inhibitory Factor in Acute Adipose Tissue Inflammation. PLoS One 2015; 10:e0137366. [PMID: 26348853 PMCID: PMC4562638 DOI: 10.1371/journal.pone.0137366] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 08/15/2015] [Indexed: 01/16/2023] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine and has been implicated in inflammatory diseases. However, little is known about the regulation of MIF in adipose tissue and its impact on wound healing. The aim of this study was to investigate MIF expression in inflamed adipose and determine its role in inflammatory cell recruitment and wound healing. Adipose tissue was harvested from subcutaneous adipose tissue layers of 24 healthy subjects and from adipose tissue adjacent to acutely inflamed wounds of 21 patients undergoing wound debridement. MIF protein and mRNA expression were measured by ELISA and RT-PCR. Cell-specific MIF expression was visualized by immunohistochemistry. The functional role of MIF in cell recruitment was investigated by a chemotaxis assay and by flow cytometry of labeled macrophages that were injected into Mif–/–and wildtype mice. Wound healing was evaluated by an in vitro scratch assay on human fibroblast monolayers. MIF protein levels of native adipose tissue and supernatants from acutely inflamed wounds were significantly elevated when compared to healthy controls. MIF mRNA expression was increased in acutely inflamed adipose tissue indicating the activation of MIF gene transcription in response to adipose tissue inflammation. MIF is expressed in mature adipocytes and in infiltrated macrophages. Peripheral blood mononuclear cell migration was significantly increased towards supernatants derived from inflamed adipose tissue. This effect was partially abrogated by MIF-neutralizing antibodies. Moreover, when compared to wildtype mice, Mif–/–mice showed reduced infiltration of labeled macrophages into LPS-stimulated epididymal fat pads in vivo. Finally, MIF antibodies partially neutralized the detrimental effect of MIF on fibroblast wound healing. Our results indicate that increased MIF expression and rapid activation of the MIF gene in fat tissue adjacent to acute wound healing disorders may play a role in cell recruitment to the site of inflammation and wound healing.
Collapse
Affiliation(s)
- Bong-Sung Kim
- Department of Plastic and Reconstructive Surgery, Hand Surgery—Burn Center, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Aachen, Germany
- * E-mail:
| | - Robert Rongisch
- Department of Plastic and Reconstructive Surgery, Hand Surgery—Burn Center, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Stephan Hager
- Department of Plastic and Reconstructive Surgery, Hand Surgery—Burn Center, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Gerrit Grieb
- Department of Plastic and Reconstructive Surgery, Hand Surgery—Burn Center, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Mahtab Nourbakhsh
- Department of Plastic and Reconstructive Surgery, Hand Surgery—Burn Center, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Hans-Oliver Rennekampff
- Department of Plastic and Reconstructive Surgery, Hand Surgery—Burn Center, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Richard Bucala
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Juergen Bernhagen
- Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Norbert Pallua
- Department of Plastic and Reconstructive Surgery, Hand Surgery—Burn Center, Medical Faculty, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
17
|
Kim BS, Pallua N, Bernhagen J, Bucala R. The macrophage migration inhibitory factor protein superfamily in obesity and wound repair. Exp Mol Med 2015; 47:e161. [PMID: 25930990 PMCID: PMC4454997 DOI: 10.1038/emm.2015.26] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 02/03/2015] [Indexed: 12/15/2022] Open
Abstract
The rising number of obese individuals has become a major burden to the healthcare systems worldwide. Obesity includes not only the increase of adipose tissue mass but importantly also the altered cellular functions that collectively lead to a chronic state of adipose tissue inflammation, insulin resistance and impaired wound healing. Adipose tissue undergoing chronic inflammation shows altered cytokine expression and an accumulation of adipose tissue macrophages (ATM). The macrophage migration inhibitory factor (MIF) superfamily consists of MIF and the recently identified homolog D-dopachrome tautomerase (D-DT or MIF-2). MIF and D-DT, which both bind to the CD74/CD44 receptor complex, are differentially expressed in adipose tissue and have distinct roles in adipogenesis. MIF positively correlates with obesity as well as insulin resistance and contributes to adipose tissue inflammation by modulating ATM functions. D-DT, however, is negatively correlated with obesity and reverses glucose intolerance. In this review, their respective roles in adipose tissue homeostasis, adipose tissue inflammation, insulin resistance and impaired wound healing will be reviewed.
Collapse
Affiliation(s)
- Bong-Sung Kim
- 1] Department of Medicine, Yale University School of Medicine, New Haven, CT, USA [2] Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Aachen, Germany [3] Department of Plastic and Reconstructive Surgery, Hand Surgery-Burn Center, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Norbert Pallua
- Department of Plastic and Reconstructive Surgery, Hand Surgery-Burn Center, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Jürgen Bernhagen
- Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Richard Bucala
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
18
|
Gao D, Madi M, Ding C, Fok M, Steele T, Ford C, Hunter L, Bing C. Interleukin-1β mediates macrophage-induced impairment of insulin signaling in human primary adipocytes. Am J Physiol Endocrinol Metab 2014; 307:E289-304. [PMID: 24918199 PMCID: PMC4121578 DOI: 10.1152/ajpendo.00430.2013] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 05/20/2014] [Indexed: 01/21/2023]
Abstract
Adipose tissue expansion during obesity is associated with increased macrophage infiltration. Macrophage-derived factors significantly alter adipocyte function, inducing inflammatory responses and decreasing insulin sensitivity. Identification of the major factors that mediate detrimental effects of macrophages on adipocytes may offer potential therapeutic targets. IL-1β, a proinflammatory cytokine, is suggested to be involved in the development of insulin resistance. This study investigated the role of IL-1β in macrophage-adipocyte cross-talk, which affects insulin signaling in human adipocytes. Using macrophage-conditioned (MC) medium and human primary adipocytes, we examined the effect of IL-1β antagonism on the insulin signaling pathway. Gene expression profile and protein abundance of insulin signaling molecules were determined, as was the production of proinflammatory cytokine/chemokines. We also examined whether IL-1β mediates MC medium-induced alteration in adipocyte lipid storage. MC medium and IL-1β significantly reduced gene expression and protein abundance of insulin signaling molecules, including insulin receptor substrate-1, phosphoinositide 3-kinase p85α, and glucose transporter 4 and phosphorylation of Akt. In contrast, the expression and release of the proinflammatory markers, including IL-6, IL-8, monocyte chemotactic protein-1, and chemokine (C-C motif) ligand 5 by adipocytes were markedly increased. These changes were significantly reduced by blocking IL-1β activity, its receptor binding, or its production by macrophages. MC medium-inhibited expression of the adipogenic factors and -stimulated lipolysis was also blunted with IL-1β neutralization. We conclude that IL-1β mediates, at least in part, the effect of macrophages on insulin signaling and proinflammatory response in human adipocytes. Blocking IL-1β could be beneficial for preventing obesity-associated insulin resistance and inflammation in human adipose tissue.
Collapse
MESH Headings
- Adipocytes, White/cytology
- Adipocytes, White/drug effects
- Adipocytes, White/immunology
- Adipocytes, White/metabolism
- Antibodies, Neutralizing/pharmacology
- Caspase 1/chemistry
- Caspase 1/metabolism
- Caspase Inhibitors/pharmacology
- Cell Communication
- Cell Line
- Cells, Cultured
- Culture Media, Conditioned/chemistry
- Culture Media, Conditioned/metabolism
- Gene Expression Regulation/drug effects
- Humans
- Hypoglycemic Agents/pharmacology
- Insulin/pharmacology
- Insulin Resistance
- Interleukin 1 Receptor Antagonist Protein/genetics
- Interleukin 1 Receptor Antagonist Protein/metabolism
- Interleukin-1beta/antagonists & inhibitors
- Interleukin-1beta/metabolism
- Lipolysis/drug effects
- Macrophages/cytology
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/metabolism
- Receptors, Interleukin-1/agonists
- Receptors, Interleukin-1/antagonists & inhibitors
- Receptors, Interleukin-1/metabolism
- Recombinant Proteins/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Dan Gao
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Mohamed Madi
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Cherlyn Ding
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Matthew Fok
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Thomas Steele
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Christopher Ford
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Leif Hunter
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Chen Bing
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
19
|
Bastiaansen-Jenniskens YM, Wei W, Feijt C, Waarsing JH, Verhaar JAN, Zuurmond AM, Hanemaaijer R, Stoop R, van Osch GJVM. Stimulation of fibrotic processes by the infrapatellar fat pad in cultured synoviocytes from patients with osteoarthritis: a possible role for prostaglandin f2α. ACTA ACUST UNITED AC 2013; 65:2070-80. [PMID: 23666869 DOI: 10.1002/art.37996] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 04/24/2013] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Stiffening of the joint is a feature of knee osteoarthritis (OA) that can be caused by fibrosis of the synovium. The infrapatellar fat pad (IPFP) present in the knee joint produces immune-modulatory and angiogenic factors. The goal of the present study was to investigate whether the IPFP can influence fibrotic processes in synovial fibroblasts, and to determine the role of transforming growth factor β (TGFβ) and prostaglandin F2α (PGF2α ) in these processes. METHODS Batches of fat-conditioned medium (FCM) were made by culturing pieces of IPFP obtained from the knees of 13 patients with OA. Human OA fibroblast-like synoviocytes (FLS) (from passage 3) were cultured in FCM with or without inhibitors of TGFβ/activin receptor-like kinase 5 or PGF2α for 4 days. The FLS were analyzed for production of collagen and expression of the gene for procollagen-lysine, 2-oxoglutarate 5-dioxygenase 2 (PLOD2; encoding lysyl hydroxylase 2b, an enzyme involved in collagen crosslinking) as well as the genes encoding α-smooth muscle actin and type I collagen α1 chain. In parallel, proliferation and migration of the synoviocytes were analyzed. RESULTS Collagen production and PLOD2 gene expression by the FLS were increased 1.8-fold (P < 0.05) and 6.0-fold (P < 0.01), respectively, in the presence of FCM, relative to control cultures without FCM. Moreover, the migration and proliferation of synoviocytes were stimulated by FCM. Collagen production was positively associated with PGF2α levels in the FCM (R = 0.89, P < 0.05), and inhibition of PGF2α levels reduced the extent of FCM-induced collagen production and PLOD2 expression. Inhibition of TGFβ signaling had no effect on the profibrotic changes. CONCLUSION These results indicate that the IPFP can contribute to the development of synovial fibrosis in the knee joint by increasing collagen production, PLOD2 expression, cell proliferation, and cell migration. In addition, whereas the findings showed that TGFβ is not involved, the more recently discovered profibrotic factor PGF2α appears to be partially involved in the regulation of profibrotic changes.
Collapse
|
20
|
Strength training and testosterone treatment have opposing effects on migration inhibitor factor levels in ageing men. Mediators Inflamm 2013; 2013:539156. [PMID: 24089589 PMCID: PMC3781991 DOI: 10.1155/2013/539156] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Revised: 07/24/2013] [Accepted: 08/02/2013] [Indexed: 12/24/2022] Open
Abstract
Background. The beneficial effects of testosterone treatment (TT) are debated. Methods. Double-blinded, placebo-controlled study of six months TT (gel) in 54 men aged 60–78 with bioavailable testosterone (BioT) <7.3 nmol/L and waist >94 cm randomized to TT (50–100 mg/day, n = 20), placebo (n = 18), or strength training (ST) (n = 16) for 24 weeks. Moreover, the ST group was randomized to TT (n = 7) or placebo (n = 9) after 12 weeks. Outcomes. Chemokines (MIF, MCP-1, and MIP-1α) and lean body mass (LBM), total, central, extremity, visceral, and subcutaneous (SAT) fat mass established by DXA and MRI. Results. From 0 to 24 weeks, MIF and SAT decreased during ST + placebo versus placebo, whereas BioT and LBM were unchanged. TT decreased fat mass (total, central, extremity, and SAT) and increased BioT and LBM versus placebo. MIF levels increased during TT versus ST + placebo. ST + TT decreased fat mass (total, central, and extremity) and increased BioT and LBM versus placebo. From 12 to 24 weeks, MCP-1 levels increased during TT versus placebo and MCP-1 levels decreased during ST + placebo versus placebo. Conclusion. ST + placebo was associated with decreased MIF levels suggesting decreased inflammatory activity. TT may be associated with increased inflammatory activity. This trial is registered with ClinicalTrials.gov NCT00700024.
Collapse
|
21
|
Dozio E, Dogliotti G, Malavazos AE, Bandera F, Cassetti G, Vianello E, Zelaschi R, Barassi A, Pellissero G, Solimene U, Morricone L, Sigruener A, Tarabin V, Schmitz G, Menicanti L, Corsi Romanelli MM. IL-18 level in patients undergoing coronary artery bypass grafting surgery or valve replacement: which link with epicardial fat depot? Int J Immunopathol Pharmacol 2013; 25:1011-20. [PMID: 23298491 DOI: 10.1177/039463201202500418] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Interleukin-18 (IL-18) is a member of the interleukin-1 family of cytokines produced constitutively by different cell types and by adipose tissue. Due to the link between obesity, inflammation and cardiovascular diseases, we aimed to measure IL-18 circulating level in patients undergoing open-heart surgery both for elective coronary artery bypass grafting (CABG) or for valve replacement (VR), and we also evaluated whether epicardial adipose tissue (EAT) depot may be a potential source of IL-18. Circulating IL-18 protein was quantified by enzyme-linked immunosorbent assay. IL-18, IL-18 receptor 1 (IL-18 R1) and IL-18 receptor accessory protein (IL-18-RAP) gene expression in EAT depot were evaluated by one colour microarray platform. EAT thickness was measured by echocardiography. In this study we found that all cardiovascular patients (CABG and VR) have increased circulating IL-18 level compared to healthy control subjects (p < 0.0001), but no statistical significant difference was observed between CABG and VR groups (p = 0.35). A great increase in the gene expression of IL-18 (p < 0.05), IL-18 R1 (p < 0.01) and IL-18 RAP (p < 0.001) was observed in EAT samples obtained from CABG vs VR patients. In conclusion, CABG and VR patients had similar increased level of circulating IL-18 protein, but in EAT depots isolated from CABG gene expression of IL-18, IL-18 R1 and IL-18-RAP resulted higher than in VR patients. Future investigation on local IL-18 protein production, its autocrine-paracrine effect and its correlation with plasmatic IL-18 level could give more information on the relationship between IL-18 and coronary artery disease.
Collapse
Affiliation(s)
- E Dozio
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Felipo V, Urios A, García-Torres ML, El Mlili N, del Olmo JA, Civera M, Ortega J, Ferrandez A, Martínez-Valls J, Cassinello N, Montoliu C. Alterations in adipocytokines and cGMP homeostasis in morbid obesity patients reverse after bariatric surgery. Obesity (Silver Spring) 2013; 21:229-37. [PMID: 23404955 DOI: 10.1002/oby.20008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 06/07/2012] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Obesity-associated nonalcoholic fatty liver disease (NAFLD), covering from simple steatosis to nonalcoholic steatohepatitis (NASH), is a common cause of chronic liver disease. Aberrant production of adipocytokines seems to play a main role in most obesity-associated disorders. Changes in adipocytokines in obesity could be mediated by alterations in cyclic GMP (cGMP) homeostasis. The aims of this work were: (1) to study the role of altered cGMP homeostasis in altered adipocytokines in morbid obesity, (2) to assess whether these alterations are different in simple steatosis or NASH, and (3) to assess whether these changes reverse in obese patients after bariatric surgery. DESIGN AND METHODS In 47 patients with morbid obesity and 45 control subjects, the levels in blood of adipocytokines, cGMP, nitric oxide (NO) metabolites, and atrial natriuretic peptide (ANP) were studied. Whether weight loss after a bariatric surgery reverses the changes in these parameters was evaluated. RESULTS NO metabolites and leptin increase (and adiponectin decreases) similarly in patients with steatosis or NASH, suggesting that these changes are due to morbid obesity and not to liver disease. Inflammation and cGMP homeostasis are affected both by morbid obesity and by liver disease. The increases in interleukin 6 (IL-6), interleukin 18 (IL-18), plasma cGMP, ANP, and the decrease in cGMP in lymphocytes are stronger in patients with NASH than with steatosis. All these changes reverse completely after bariatric surgery and weight loss, except IL-18. CONCLUSION Altered cGMP homeostasis seems to contribute more than inflammation to changes in leptin and adiponectin in morbid obesity.
Collapse
Affiliation(s)
- Vicente Felipo
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Negi SI, Pankow JS, Fernstrom K, Hoogeveen RC, Zhu N, Couper D, Schmidt MI, Duncan BB, Ballantyne CM. Racial differences in association of elevated interleukin-18 levels with type 2 diabetes: the Atherosclerosis Risk in Communities study. Diabetes Care 2012; 35:1513-8. [PMID: 22596175 PMCID: PMC3379601 DOI: 10.2337/dc11-1957] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Elevated plasma interleukin-18 (IL-18) has been linked to onset of diabetes mellitus (DM) and its complications. However, so far this association has been shown only in predominantly white populations. We examined IL-18 levels and their association with incident DM in a racially heterogeneous population. RESEARCH DESIGN AND METHODS In a nested case-cohort design representing a 9-year follow-up of 9,740 middle-aged, initially healthy, nondiabetic white and African American participants of the Atherosclerosis Risk in Communities Study, we selected and measured analytes on race-stratified (50% white, 50% African American) random samples of both cases of incident diabetes (n = 548) and eligible members of the full cohort (n = 536). RESULTS Baseline IL-18 levels were significantly higher in white participants compared with African American participants (P < 0.001). Although white participants in the fourth (versus first) quartile of IL-18 levels had a significant hazard ratio (HR) for developing DM (HR: 2.1, 95% CI: 1.3-3.4), after adjustment for age, sex, and study center, no difference was seen among African Americans (HR: 1.0, 95% CI: 0.6-1.7). Unlike those in African Americans, IL-18 levels in whites had a significant correlation with age (P < 0.01); anthropometric characteristics such as waist circumference (P < 0.001), height (P = 0.04), waist-to-hip ratio (P < 0.001), and BMI (P < 0.01); and total (P < 0.001) and high-molecular-weight (P < 0.001) adiponectin. CONCLUSIONS There are racial differences in levels of IL-18 and the association of IL-18 with risk factors and incident type 2 DM. In addition, there seems to be a complex interplay of inflammation and adiposity in the development of DM.
Collapse
Affiliation(s)
- Smita I Negi
- Section of Cardiovascular Research, Baylor College of Medicine, and Center for Cardiovascular Disease Prevention, Methodist DeBakey Heart Center, Houston, TX, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Bastiaansen-Jenniskens YM, Clockaerts S, Feijt C, Zuurmond AM, Stojanovic-Susulic V, Bridts C, de Clerck L, DeGroot J, Verhaar JAN, Kloppenburg M, van Osch GJVM. Infrapatellar fat pad of patients with end-stage osteoarthritis inhibits catabolic mediators in cartilage. Ann Rheum Dis 2012; 71:288-94. [PMID: 21998115 DOI: 10.1136/ard.2011.153858] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Adipose tissue is known to release inflammatory cytokines and growth factors. In this exploratory study, the authors examined whether the infrapatellar fat pad (IPFP) closely located to cartilage in the knee joint can affect cartilage metabolism. In addition, the authors analysed whether the macrophage types present in IPFP could explain the effect on cartilage. METHODS IPFP explants obtained during total knee replacement of 29 patients with osteoarthritis (OA) were used to make fat-conditioned medium (FCM). Explants of bovine cartilage were cultured with or without FCM. Nitric oxide (NO) and glycosaminoglycan release and gene expression of matrix-degrading enzymes in cartilage were analysed. To stimulate catabolic processes in the cartilage, the authors added interleukin 1β, and the effect of six FCMs was evaluated. The presence of different types of macrophages (CD68+, CD86+ and CD206+) in OA IPFPs was compared with subcutaneous adipose tissue samples and IPFP samples from patients with an anterior cruciate ligament rupture. RESULTS FCM alone reduced NO and glycosaminoglycan release and matrix metalloproteinase (MMP)1 gene expression by the cartilage. Moreover, when catabolic conditions were enhanced with interleukin 1β, FCM inhibited NO production as well as MMP1 and MMP3 gene expression and increased collagen type II gene expression. Significantly more CD206+ cells were present in OA IPFP samples than in subcutaneous fat or anterior cruciate ligament IPFP samples. CONCLUSION In contrast to the authors' expectations, medium conditioned by end-stage OA IPFP inhibited catabolic processes in cartilage. CD206+ cells present in the IPFPs used for making the FCM might have contributed to the inhibition of catabolic processes in the cartilage.
Collapse
MESH Headings
- Adipose Tissue/metabolism
- Adipose Tissue/pathology
- Adult
- Aged
- Aged, 80 and over
- Animals
- Arthroplasty, Replacement, Knee
- Cartilage, Articular/drug effects
- Cartilage, Articular/metabolism
- Cartilage, Articular/pathology
- Cattle
- Culture Media, Conditioned/pharmacology
- Glycosaminoglycans/metabolism
- Humans
- Interleukin-1beta/pharmacology
- Macrophages/pathology
- Matrix Metalloproteinases/metabolism
- Middle Aged
- Nitric Oxide/biosynthesis
- Osteoarthritis, Knee/metabolism
- Osteoarthritis, Knee/pathology
- Osteoarthritis, Knee/surgery
- Tissue Culture Techniques
- Young Adult
Collapse
|
25
|
Synergistic induction of interleukin-6 expression by endothelin-1 and cyclic AMP in adipocytes. Int J Obes (Lond) 2012; 37:197-203. [PMID: 22290536 DOI: 10.1038/ijo.2012.11] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND We have demonstrated previously that endothelin-1 (ET-1) may stimulate interleukin-6 (IL-6) release from 3T3-L1 adipocytes. In this study, we further examined the combined effect of ET-1 and cyclic adenosine monophosphate (cAMP) on IL-6 release. METHODS IL-6 release was measured by enzyme-linked immuosorbent assay. Reverse transcriptase-PCR and real-time PCR analyses were used to determine cellular mRNA levels. A luciferase reporter driven by promoter (-1310/+198) of mouse IL-6 gene was transfected into 3T3-L1 adipocytes to monitor IL-6 transcription. RESULTS ET-1 and cAMP induced IL-6 release in a synergistic manner that can be attributed to their synergistic induction of IL-6 gene expression, as evidenced by IL-6 mRNA analysis and the IL-6 promoter reporter assay. Both ET(A) and ET(B) receptors seem to be involved. In addition, enhanced IL-6 promoter activity can be similarly induced by ET-1 and catecholamines (epinephrine and norepinephrine). The cooperative interaction between ET-1 and cAMP on IL-6 expression seems distinctive, as no other proinflammatory cytokines, such as tumor necrosis factor-α (TNF-α) and IL-1β, are similarly affected. In fact, cAMP inhibited ET-1-stimulated TNF-α and IL-1β expressions in adipocytes. Furthermore, injection of mice with epinephrine and ET-1 induced a tremendously synergistic increase in serum IL-6 levels. Nevertheless, whereas cAMP induced IL-6 expression in RAW264.7 mouse macrophages, ET-1 had no effect on either the basal or the cAMP-induced IL-6 expression. CONCLUSION ET-1 and epinephrine may boost plasma IL-6 levels in mice in a synergistic manner, probably through their synergistic induction of IL-6 expression in adipocytes. SIGNIFICANCE This study should provide a new perspective for treating IL-6-related diseases, especially those accompanied with elevated ET-1 and catecholamine levels.
Collapse
|
26
|
Lindholm A, Blomquist C, Bixo M, Dahlbom I, Hansson T, Sundström Poromaa I, Burén J. No difference in markers of adipose tissue inflammation between overweight women with polycystic ovary syndrome and weight-matched controls. Hum Reprod 2011; 26:1478-85. [PMID: 21478181 PMCID: PMC3096560 DOI: 10.1093/humrep/der096] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Previous studies have indicated that peripheral circulating markers of inflammation are elevated in women with polycystic ovary syndrome (PCOS), but thus far no studies concerning markers of inflammation in adipose tissue have been published. The aim of the study was to investigate whether patients with PCOS display increased expression of inflammatory markers in adipose tissue. METHODS Twenty overweight patients with PCOS, 10 lean patients with PCOS and 20 overweight controls had subcutaneous fat biopsies and blood samples taken. Adipose tissue levels of mRNA of inflammatory markers were determined by use of real-time PCR. RESULTS Overweight patients with PCOS had higher relative adipose tissue chemokine ligand 2 (P < 0.01), and its cognate receptor (P < 0.05), tumour necrosis factor-α (P < 0.001), interleukin (IL)-10 (P < 0.001) and IL-18 (P < 0.001) and the monocyte/macrophage markers CD14 (P < 0.01) and CD163 (P < 0.01) mRNA levels compared with lean women with PCOS. There were no differences between overweight patients with PCOS and overweight control subjects in this respect. Within the PCOS group, markers of adipose tissue inflammation correlated significantly with obesity-related metabolic disturbances, but when data were adjusted for age and BMI, most correlations were lost. CONCLUSIONS Overweight, rather than the PCOS diagnosis per se, appears to be the main explanatory variable for elevated adipose tissue inflammation in patients with PCOS.
Collapse
Affiliation(s)
- Asa Lindholm
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden.
| | | | | | | | | | | | | |
Collapse
|
27
|
Sun L, Hu FB, Yu Z, Li H, Liu H, Wang X, Yu D, Wu H, Zhang G, Zong G, Liu Y, Lin X. Lean body mass, interleukin 18, and metabolic syndrome in apparently healthy Chinese. PLoS One 2011; 6:e18104. [PMID: 21437204 PMCID: PMC3060923 DOI: 10.1371/journal.pone.0018104] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Accepted: 02/21/2011] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE We aimed to investigate how lean body mass is related to circulating Interleukin 18 (IL-18) and its association with metabolic syndrome (MetS) among apparently healthy Chinese. METHODS A population-based sample of 1059 Chinese men and women aged 35-54 years was used to measure plasma IL-18, glucose, insulin, lipid profile, inflammatory markers and high-molecular-weight (HMW)-adiponectin. Fat mass index (FMI) and lean mass index (LMI) were measured by dual-energy X-ray absorptiometry. MetS was defined by the updated National Cholesterol Education Program Adult Treatment Panel III criteria for Asian-Americans. RESULTS Circulating IL-18 was positively correlated with LMI after adjustment for FMI (correlation coefficient = 0.11, P<0.001). The association with the MetS (odds ratio 3.43, 95% confidence interval 2.01-5.85) was substantially higher in the highest than the lowest quartile of IL-18 after multiple adjustments including body mass index. In the stratified multivariable regression analyses, the positive association between IL-18 and MetS was independent of tertiles of FMI, inflammatory markers and HMW-adiponectin, but significantly interacted with tertile of LMI (P for interaction = 0.010). CONCLUSION Elevated plasma IL-18 was associated with higher MetS prevalence in apparently healthy Chinese, independent of traditional risk factors, FMI, inflammatory markers and HMW-adiponectin. More studies are needed to clarify the role of lean mass in IL-18 secretion and its associated cardio-metabolic disorders.
Collapse
Affiliation(s)
- Liang Sun
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Frank B. Hu
- Departments of Nutrition and Epidemiology, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Zhijie Yu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Graduate School of the Chinese Academy of Sciences, Shanghai, China
- SIBS-Novo Nordisk Translational Research Centre for PreDiabetes, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Huaixing Li
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Huaiyu Liu
- Shanghai Luwan Center for Disease Control and Prevention, Shanghai, China
| | - Xiangdong Wang
- Shanghai Zhabei Center for Disease Control and Prevention, Shanghai, China
| | - Danxia Yu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Hongyu Wu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Geng Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Geng Zong
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Yong Liu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | - Xu Lin
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Graduate School of the Chinese Academy of Sciences, Shanghai, China
- * E-mail:
| |
Collapse
|
28
|
Circulating interleukin-18: A specific biomarker for atherosclerosis-prone patients with metabolic syndrome. Nutr Metab (Lond) 2011; 8:3. [PMID: 21251304 PMCID: PMC3038890 DOI: 10.1186/1743-7075-8-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2010] [Accepted: 01/20/2011] [Indexed: 12/22/2022] Open
Abstract
Background Metabolic syndrome (MetS) is associated with an increased risk of the development of atherosclerotic cardiovascular disease (CVD). Interleukin-18 (IL-18), which is a pleiotropic proinflammatory cytokine with important regulatory functions in the innate immune response system, plays a crucial role in vascular pathologies. IL-18 is also a predictor of cardiovascular death in patients with CVD and is involved in atherosclerotic plaque destabilization. Results In order to determine if circulating levels of IL-18 can serve as a specific biomarker for distinguishing MetS patients from pre-MetS subjects, we studied 78 patients with visceral fat deposition and 14 age-matched control subjects. Increased levels of IL-18 were observed more frequently in patients with MetS than in pre-MetS subjects and were positively associated with waist circumference. Serum levels of IL-18 were significantly reduced by a change in weight caused by lifestyle modifications. There was a significant interaction between waist circumference and serum IL-18 concentration. Weight loss of at least 5% of the body weight caused by lifestyle modification decreased IL-18 circulating levels relative to the reduction in waist circumference and blood pressure, suggesting that this degree of weight loss benefits the cardiovascular system. Conclusion IL-18 may be a useful biomarker of the clinical manifestations of MetS and for the management of the risk factors of CVD.
Collapse
|
29
|
Mathur SK, Jain P, Mathur P. Microarray evidences the role of pathologic adipose tissue in insulin resistance and their clinical implications. J Obes 2011; 2011:587495. [PMID: 21603273 PMCID: PMC3092611 DOI: 10.1155/2011/587495] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Accepted: 02/21/2011] [Indexed: 12/20/2022] Open
Abstract
Clustering of insulin resistance and dysmetabolism with obesity is attributed to pathologic adipose tissue. The morphologic hallmarks of this pathology are adipocye hypertrophy and heightened inflammation. However, it's underlying molecular mechanisms remains unknown. Study of gene function in metabolically active tissues like adipose tissue, skeletal muscle and liver is a promising strategy. Microarray is a powerful technique of assessment of gene function by measuring transcription of large number of genes in an array. This technique has several potential applications in understanding pathologic adipose tissue. They are: (1) transcriptomic differences between various depots of adipose tissue, adipose tissue from obese versus lean individuals, high insulin resistant versus low insulin resistance, brown versus white adipose tissue, (2) transcriptomic profiles of various stages of adipogenesis, (3) effect of diet, cytokines, adipokines, hormones, environmental toxins and drugs on transcriptomic profiles, (4) influence of adipokines on transcriptomic profiles in skeletal muscle, hepatocyte, adipose tissue etc., and (5) genetics of gene expression. The microarray evidences of molecular basis of obesity and insulin resistance are presented here. Despite the limitations, microarray has potential clinical applications in finding new molecular targets for treatment of insulin resistance and classification of adipose tissue based on future risk of insulin resistance syndrome.
Collapse
Affiliation(s)
- Sandeep Kumar Mathur
- Department of Endocrinology, S. M. S. Medical College, India
- *Sandeep Kumar Mathur:
| | - Priyanka Jain
- Institute of Genomics and Integrative Biology, Mall Road, New Delhi 110007, India
| | - Prashant Mathur
- Department of Pharmacology, S. M. S. Medical College, J. L. Marg, Jaipur 302004, India
| |
Collapse
|
30
|
Release of inflammatory mediators by human adipose tissue is enhanced in obesity and primarily by the nonfat cells: a review. Mediators Inflamm 2010; 2010:513948. [PMID: 20508843 PMCID: PMC2874930 DOI: 10.1155/2010/513948] [Citation(s) in RCA: 170] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 01/27/2010] [Accepted: 02/23/2010] [Indexed: 02/08/2023] Open
Abstract
This paper considers the role of putative adipokines that might be involved in the enhanced inflammatory response of human adipose tissue seen in obesity. Inflammatory adipokines [IL-6, IL-10, ACE, TGFbeta1, TNFalpha, IL-1beta, PAI-1, and IL-8] plus one anti-inflammatory [IL-10] adipokine were identified whose circulating levels as well as in vitro release by fat are enhanced in obesity and are primarily released by the nonfat cells of human adipose tissue. In contrast, the circulating levels of leptin and FABP-4 are also enhanced in obesity and they are primarily released by fat cells of human adipose tissue. The relative expression of adipokines and other proteins in human omental as compared to subcutaneous adipose tissue as well as their expression in the nonfat as compared to the fat cells of human omental adipose tissue is also reviewed. The conclusion is that the release of many inflammatory adipokines by adipose tissue is enhanced in obese humans.
Collapse
|
31
|
Fain JN, Tagele BM, Cheema P, Madan AK, Tichansky DS. Release of 12 adipokines by adipose tissue, nonfat cells, and fat cells from obese women. Obesity (Silver Spring) 2010; 18:890-6. [PMID: 19834460 DOI: 10.1038/oby.2009.335] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The relative release in vitro of endothelin-1, zinc-alpha2-glycoprotein (ZAG), lipocalin-2, CD14, RANTES (regulated on activation, normal T cell expressed and secreted protein), lipoprotein lipase (LPL), osteoprotegerin (OPG), fatty acid-binding protein 4 (FABP-4), visfatin/PBEF/Nampt, glutathione peroxidase-3 (GPX-3), intracellular cell adhesion molecule 1 (ICAM-1), and amyloid A was examined using explants of human adipose tissue as well as the nonfat cell fractions and adipocytes from obese women. Over a 48-h incubation the majority of the release of LPL was by fat cells whereas that of lipocalin-2, RANTES, and ICAM-1 was by the nonfat cells present in human adipose tissue. In contrast appreciable amounts of OPG, amyloid A, ZAG, FABP-4, GPX-3, CD14, and visfatin/PBEF/Nampt were released by both fat cells and nonfat cells. There was an excellent correlation (r = 0.75) between the ratios of adipokine release by fat cells to nonfat cells over 48 h and the ratio of their mRNAs in fat cells to nonfat cells at the start of the incubation. The total release of ZAG, OPG, RANTES, and amyloid A by incubated adipose tissue explants from women with a fat mass of 65 kg was not different from that by women with a fat mass of 29 kg. In contrast that of ICAM-1, FABP-4, GPX-3, visfatin/PBEF/Nampt, CD14, lipocalin-2, LP, and endothelin-1 was significantly greater in tissue from women with a total fat mass of 65 kg.
Collapse
Affiliation(s)
- John N Fain
- Department of Molecular Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA.
| | | | | | | | | |
Collapse
|
32
|
Trøseid M, Seljeflot I, Arnesen H. The role of interleukin-18 in the metabolic syndrome. Cardiovasc Diabetol 2010; 9:11. [PMID: 20331890 PMCID: PMC2858122 DOI: 10.1186/1475-2840-9-11] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2010] [Accepted: 03/23/2010] [Indexed: 12/18/2022] Open
Abstract
The metabolic syndrome is thought to be associated with a chronic low-grade inflammation, and a growing body of evidence suggests that interleukin-18 (IL-18) might be closely related to the metabolic syndrome and its consequences. Circulating levels of IL-18 have been reported to be elevated in subjects with the metabolic syndrome, to be closely associated with the components of the syndrome, to predict cardiovascular events and mortality in populations with the metabolic syndrome and to precede the development of type 2 diabetes. IL-18 is found in the unstable atherosclerotic plaque, in adipose tissue and in muscle tissue, and is subject to several regulatory steps including cleavage by caspase-1, inactivation by IL-18 binding protein and the influence of other cytokines in modulating its interaction with the IL-18 receptor. The purpose of this review is to outline the role of IL-18 in the metabolic syndrome, with particular emphasis on cardiovascular risk and the potential effect of life style interventions.
Collapse
Affiliation(s)
- Marius Trøseid
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Ullevål, Oslo, Norway.
| | | | | |
Collapse
|
33
|
Trøseid M, Lappegård KT, Mollnes TE, Arnesen H, Seljeflot I. The effect of exercise on serum levels of interleukin-18 and components of the metabolic syndrome. Metab Syndr Relat Disord 2010; 7:579-84. [PMID: 19900154 DOI: 10.1089/met.2009.0003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND There is increasing evidence that the metabolic syndrome is associated with a proinflammatory state. Interleukin-18 (IL-18) has been shown to be associated with multiple components of the syndrome and to predict the development of diabetes and cardiovascular events. The aim of the present work was to investigate if exercise could reduce serum levels of IL-18 in subjects with the metabolic syndrome, and if potential changes would be associated with changes in body composition and components of the syndrome. Pravastatin was used for comparison. METHODS We investigated the effect of 12 weeks of intervention with exercise, pravastatin, or both on serum levels of IL-18 in 34 subjects with the metabolic syndrome. RESULTS Levels of IL-18 were reduced by exercise (P = 0.036), pravastatin (P = 0.036), and the combination (P = 0.017) versus baseline, however without intergroup differences. Still, the reduction in the exercise group was not negligible (17.5%). Furthermore, reduced levels of IL-18 were significantly associated with improvement of an increasing number of components of the metabolic syndrome (P = 0.034). This effect is likely to be caused by exercise, because this intervention had a beneficial effect on components of the syndrome compared to the control group (P = 0.029). CONCLUSIONS Our findings suggest that the protective effect of exercise might be due in part to reduced inflammation associated with improvement of the metabolic syndrome and its components. Studies with larger sample sizes are warranted to confirm this hypothesis.
Collapse
Affiliation(s)
- Marius Trøseid
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Ullevål, Oslo, Norway.
| | | | | | | | | |
Collapse
|
34
|
Circulating levels of interleukin-1 family cytokines in overweight adolescents. Mediators Inflamm 2010; 2010:958403. [PMID: 20169140 PMCID: PMC2821754 DOI: 10.1155/2010/958403] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2009] [Accepted: 11/17/2009] [Indexed: 01/22/2023] Open
Abstract
Objectives.
Obesity and related diseases are dramatically increasing problems, particularly in children and adolescents. We determined circulating levels of different interleukin (IL)-1 family members in normal weight and overweight adolescents.
Methods.
Seventy male, Caucasian adolescents (13–17 years) were recruited. Thirty-five had a body-mass index (BMI) above the 90th age-specific percentile. IL-1α, IL-1β, IL-1 receptor antagonist (IL-1ra), and IL-18 were determined using multiplex-technology.
Results.
IL-18 concentrations were higher in the overweight group compared to normal weight (161.6 ± 40.7 pg/ml versus 134.7 ± 43.4 pg/ml, P = .009). Concentrations of circulating IL-1β levels were below the detection threshold. IL-18 (R2:0.355, P < .01) and IL-1ra (R2:0.287, P < .05) correlated with BMI, whereas IL-1α did not.
Conclusions.
Accumulating data indicate the importance of the endocrine function of adipose tissue for the pathophysiological consequences of obesity-related co-morbidities. Since IL-18 is involved in the pathogenesis of different cardiovascular diseases, we conclude that IL-18 may represent a link between obesity and related co-morbidities in children and adolescents.
Collapse
|
35
|
Aubin MC, Cardin S, Comtois P, Clément R, Gosselin H, Gillis MA, Le Quang K, Nattel S, Perrault LP, Calderone A. A high-fat diet increases risk of ventricular arrhythmia in female rats: enhanced arrhythmic risk in the absence of obesity or hyperlipidemia. J Appl Physiol (1985) 2010; 108:933-40. [PMID: 20133431 DOI: 10.1152/japplphysiol.01281.2009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Obesity increases the incidence of cardiac arrhythmias and impairs wound healing. However, it is presently unknown whether a high-fat diet affects arrhythmic risk or wound healing before the onset of overt obesity or hyperlipidemia. After 8 wk of feeding a high-fat diet to adult female rats, a nonsignificant increase in body weight was observed and associated with a normal plasma lipid profile. Following ischemia/reperfusion injury, scar length (standard diet 0.29 +/- 0.09 vs. high-fat 0.32 +/- 0.13 cm), thickness (standard diet 0.047 +/- 0.02 vs. high-fat 0.059 +/- 0.01 cm), and collagen alpha(1) type 1 content (standard diet 0.21 +/- 0.04 vs. high-fat 0.20 +/- 0.04 arbitrary units/mm(2)) of infarcted hearts were not altered by the high-fat diet. However, the mortality rate was greatly increased 24 h postinfarction (from 5% to 46%, P < 0.01 for ischemia/reperfusion rats; from 20% to 89%, P < 0.0001, in complete-occlusion rats) in high-fat fed rats, in association with a higher prevalence of ventricular arrhythmias. Ventricular arrhythmia inducibility was also significantly increased in noninfarcted rats fed a high-fat diet. In the hearts of rats fed a high-fat diet, connexin-40 expression was absent, connexin-43 was hypophosphorylated and lateralized, and neurofilament-M immunoreactive fiber density (standard diet 2,020 +/- 260 vs. high-fat diet 2,830 +/- 250 microm(2)/mm(2)) and tyrosine hydroxylase protein expression were increased (P < 0.05). Thus, in the absence of overt obesity and hyperlipidemia, sympathetic hyperinnervation and an aberrant pattern of gap junctional protein expression and regulation in the heart of female rats fed a high-fat diet may have contributed in part to the higher incidence of inducible cardiac arrhythmias.
Collapse
Affiliation(s)
- Marie-Claude Aubin
- Department of Pharmacology, Université de Montréal, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Glintborg D, Andersen M, Richelsen B, Bruun JM. Plasma monocyte chemoattractant protein-1 (MCP-1) and macrophage inflammatory protein-1alpha are increased in patients with polycystic ovary syndrome (PCOS) and associated with adiposity, but unaffected by pioglitazone treatment. Clin Endocrinol (Oxf) 2009; 71:652-8. [PMID: 19170716 DOI: 10.1111/j.1365-2265.2009.03523.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Hirsutism is most often caused by polycystic ovary syndrome (PCOS). PCOS patients are characterized by insulin resistance, abdominal obesity and low-grade inflammation. Insulin sensitizing treatment reduces the inflammatory state, but the effect on serum levels of migration inhibitor factor (MIF), monocyte chemoattractant protein (MCP)-1 and macrophage inflammatory protein (MIP)-1alpha have not been evaluated before in PCOS. RESEARCH DESIGN AND METHODS Plasma chemokine levels (MCP-1, MIP-1alpha and MIF) were measured in two study designs. (i) 51 hirsute patients and 63 matched controls and (ii) 30 PCOS patients before and after randomized treatment with 30 mg pioglitazone/placebo for 16 weeks. Clinical evaluations and whole body DXA-scans were performed in all participants. RESULTS Hirsute patients (n = 51) had significantly increased MCP-1 [121 (15-950) vs. 81 (18-365) pg/ml; P < 0.05] and MIP-1alpha[179 (8-4202) vs. 103 (4-1598) pg/ml; P < 0.05] than controls of matched body composition [geometric mean (-2SD to +2SD)]. In PCOS (n = 30), MCP-1, MIP-1alpha and MIF correlated positively with central fat mass. A BMI independent positive association was found between MIF and free testosterone (r = 0.49, P = 0.01) in PCOS. Pioglitazone treatment significantly improved insulin sensitivity without affecting testosterone, body composition, MCP-1, MIP-1alpha and MIF levels. CONCLUSIONS Chemokine levels were significantly increased and showed close associations with measures of adiposity in PCOS patients, but were unchanged during insulin sensitizing treatment with pioglitazone. Our data suggests a fat mass independent association between testosterone and MIF levels in PCOS and the effect of anti-androgen treatment on chemokine levels needs to be examined.
Collapse
Affiliation(s)
- Dorte Glintborg
- Department of Endocrinology and Metabolism, Odense University Hospital, Kløvervaenget 6, Odense C, Denmark.
| | | | | | | |
Collapse
|
37
|
Trøseid M, Arnesen H, Hjerkinn EM, Seljeflot I. Serum levels of interleukin-18 are reduced by diet and n-3 fatty acid intervention in elderly high-risk men. Metabolism 2009; 58:1543-9. [PMID: 19595382 DOI: 10.1016/j.metabol.2009.04.031] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Accepted: 04/24/2009] [Indexed: 11/16/2022]
Abstract
Inflammation plays a central role in the development and progression of atherosclerosis, and inflammatory markers have been reported to predict cardiovascular events. Mediterranean-like diet and very long chain omega-3 polyunsaturated fatty acid (n-3 PUFA) supplementation have been reported to reduce the risk of cardiovascular mortality and morbidity, but the mechanisms are not fully clarified. The aims of the present study were to investigate the effect of such interventions on serum levels of inflammatory markers, and potential associations with changes in serum fatty acids and anthropometric measures. This was a randomized 2 x 2 factorial-designed trial comparing the effect of 3 years of dietary counseling, n-3 PUFA supplementation (2.4 g/d), or both on different measures of atherosclerosis in elderly high-risk men (N = 563). Levels of interleukin-18 (IL-18) were decreased by diet (-10.5% vs baseline, P = .012 compared with no diet) and by n-3 PUFA supplementation (-9.9% vs baseline, P = .008 compared with placebo). Other measured inflammatory markers were not affected. Changes in IL-18 were significantly correlated to changes in triglycerides (r = 0.20, P < .001), eicosapentaenoic acid (r = -0.14, P = .030), docosahexaenoic acid (r = -0.14, P = .034), body mass index (r = 0.16, P < .001), and waist circumference (r = 0.12, P = .007). In conclusion, levels of IL-18 were significantly reduced by Mediterranean-like diet and n-3 PUFA supplementation. However, the changes correlated only weakly to changes in triglycerides, serum fatty acids, and anthropometric measures. The cardioprotective effects of both interventions might thus in part be explained by reduced levels of IL-18, but probably beyond changes in serum fatty acids and body composition.
Collapse
Affiliation(s)
- Marius Trøseid
- Center for Clinical Heart Research, Ullevål Department of Cardiology, Oslo University Hospital, University of Oslo, N-0407 Oslo, Norway
| | | | | | | |
Collapse
|
38
|
Hivert MF, Sun Q, Shrader P, Mantzoros CS, Meigs JB, Hu FB. Circulating IL-18 and the risk of type 2 diabetes in women. Diabetologia 2009; 52:2101-8. [PMID: 19669125 PMCID: PMC3758765 DOI: 10.1007/s00125-009-1455-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Accepted: 06/19/2009] [Indexed: 10/20/2022]
Abstract
AIMS/HYPOTHESIS The pathophysiology of type 2 diabetes involves pro-inflammatory pathways. We tested the hypothesis that IL-18 predicts future diabetes cases. METHODS We used a nested case-control design based in the Nurses' Health Study. Baseline blood samples were collected between 1989 and 1990. Questionnaires to assess body weight, lifestyle (physical activity, diet, smoking) and diabetes diagnosis were sent out and assessed biennially (follow-up until 2002). Cases (n = 1,012) were defined as women developing type 2 diabetes at least 1 year after blood sampling. Control women (n = 1,081) were matched to cases by age, date of blood draw, fasting status and race. We calculated the RR (95% CI) of type 2 diabetes in quintiles of IL-18 using conditional logistic regression with the first quintile as referent; adjustments included matching factors, diabetes risk factors, BMI, adipokine levels (adiponectin, resistin) and inflammatory proteins (C-reactive protein, tumour necrosis factor receptor 2 (TNFalpha-R2) and IL-6). RESULTS Higher IL-18 levels were associated with increased risk of developing diabetes, even after adjustment for matching factors and multiple diabetes risk factors: being in the highest quintile of IL-18 was associated with a RR of 1.75 (1.41-2.18) for diabetes relative to the first quintile (p < 0.0001 for trend). Significant trends in association were still observed after adjustment for BMI (RR 1.44 [1.15-1.80], p < 0.0001 for trend) and adiponectin levels (RR 1.28 [1.02-1.60], p = 0.006 for trend). Further adjustment for inflammatory markers in a sub-sample did not significantly change the results. CONCLUSIONS/INTERPRETATION Elevated IL-18 levels are associated with higher risk of diabetes. This association is independent of usual risk factors, including BMI and adipokine levels.
Collapse
Affiliation(s)
- M F Hivert
- Massachusetts General Hospital, Boston, USA
| | | | | | | | | | | |
Collapse
|
39
|
Madsen EL, Bruun JM, Skogstrand K, Hougaard DM, Christiansen T, Richelsen B. Long-term weight loss decreases the nontraditional cardiovascular risk factors interleukin-18 and matrix metalloproteinase-9 in obese subjects. Metabolism 2009; 58:946-53. [PMID: 19409578 DOI: 10.1016/j.metabol.2009.02.031] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Accepted: 02/13/2009] [Indexed: 01/04/2023]
Abstract
The objective of the study was to investigate the effect of long-term (3.2 years) weight loss on serum levels of the nontraditional cardiovascular risk factors interleukin (IL)-18 and matrix metalloproteinase (MMP)-9. Moreover, we wanted to assess the significance of the magnitude of the weight loss and evaluate the potential effects of 36 months of treatment with the lipase inhibitor orlistat on these parameters. Sixty-eight abdominally obese subjects completed 8 weeks of very low energy diet (600-800 kcal/d) followed by 36 months of randomized treatment with either orlistat or placebo together with lifestyle intervention. Serum levels of IL-18, MMP-9, and leptin were measured by flowmetric xMAP technology (Luminex, Austin, TX). Changes in the levels of IL-18, MMP-9, and leptin were similar in the orlistat and the placebo group during this study. Thus, the 2 groups were combined for further analysis. A weight loss of 8.4 +/- 8.8 kg from baseline to 3.2 years was associated with significant decreases in IL-18 (P < .001), MMP-9 (P < .01), and leptin (P < .001). Matrix metalloproteinase-9 was, however, significantly increased after 8 weeks of very low energy diet-induced weight loss (P < .05). The long-term changes in IL-18 were significantly associated with changes in body mass index independent of changes in blood pressure and lipids (P < .05). Levels and changes of IL-18 and MMP-9 were significantly positively associated at 3.2 years (P < .01). Long-term changes in leptin were significantly associated with changes in IL-18 (P < .01) at 3.2 years. Diet-induced long-term weight loss decreased IL-18 and MMP-9. The decrease in IL-18 was associated with changes in body mass index independent of changes in blood pressure and lipids, indicating that even a minor weight reduction (>5%) has beneficial effects on nontraditional cardiovascular risk markers. Orlistat treatment had no independent effects on IL-18, MMP-9, or leptin in the present study.
Collapse
Affiliation(s)
- Erik L Madsen
- Department of Endocrinology and Metabolism C, Aarhus University Hospital, Aarhus Sygehus, Aarhus C, Denmark
| | | | | | | | | | | |
Collapse
|
40
|
Swanson KS, Belsito KR, Vester BM, Schook LB. Adipose tissue gene expression profiles of healthy young adult and geriatric dogs. Arch Anim Nutr 2009; 63:160-71. [DOI: 10.1080/17450390902733934] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
41
|
Sherry CL, Kramer JM, York JM, Freund GG. Behavioral recovery from acute hypoxia is reliant on leptin. Brain Behav Immun 2009; 23:169-75. [PMID: 18854211 PMCID: PMC2652853 DOI: 10.1016/j.bbi.2008.09.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Revised: 09/19/2008] [Accepted: 09/20/2008] [Indexed: 01/20/2023] Open
Abstract
Individuals affected by hypoxia experience a variety of immune-associated sickness symptoms including malaise, fatigue, lethargy and loss of interest in the physical and social environment. Recently, we demonstrated that the interleukin (IL)-1beta arm of the neuroimmune system was critical to the sickness symptoms caused by hypoxia, and that IL-1 receptor antagonist (IL-1RA), IL-1beta's endogenous inhibitor, was critical to promoting sickness recovery. Here, we report that leptin is key to recovery from hypoxia because it dramatically augmented IL-1RA production in mice. We found that hypoxia increased leptin in white adipose tissue (WAT) which in turn, caused a marked rise in serum IL-1RA. Interestingly, in-vitro, leptin was a more potent inducer of IL-RA, in macrophages, than hypoxia. In leptin receptor defective (db/db) and leptin deficient (ob/ob) mice, sickness recovery from hypoxia was delayed 3-fold. Importantly, in ob/ob mice, leptin administration completely reversed this delayed recovery and induced a marked increase in serum IL-1RA. Finally, leptin administration to normal mice reduced hypoxia recovery time by 1/3 and dramatically increased WAT and serum IL-1RA. Leptin did not alter recovery from hypoxia in IL-1RA knock out mice. These results show that by enhancing IL-1RA production leptin promoted sickness recovery from hypoxia.
Collapse
Affiliation(s)
- Christina L. Sherry
- Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Jason M. Kramer
- Department of Pathology, University of Illinois, Urbana, IL 61801, USA
| | - Jason M. York
- Department of Animal Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Gregory G. Freund
- Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA, Department of Pathology, University of Illinois, Urbana, IL 61801, USA, Department of Animal Sciences, University of Illinois, Urbana, IL 61801, USA
| |
Collapse
|
42
|
Cartier A, Bergeron J, Poirier P, Alméras N, Tremblay A, Lemieux I, Després JP. Increased plasma interleukin-1 receptor antagonist levels in men with visceral obesity. Ann Med 2009; 41:471-8. [PMID: 19492200 DOI: 10.1080/07853890903022801] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
BACKGROUND Adipose tissue (AT) is an important source of interleukin-1 receptor antagonist (IL-1Ra). Its expression is markedly increased in obesity. AIMS AND METHODS To quantify the associations of IL-1Ra with body fat distribution as well as to examine the respective contributions of IL-1Ra and visceral adiposity to the variation in some cardiometabolic risk (CMR) markers. Plasma IL-1Ra levels were measured in a sample of 117 healthy non-diabetic men (age: 44.9+/-10.1 years; body mass index (BMI): 28.8+/-4.5 kg/m(2)). RESULTS Plasma IL-1Ra levels correlated positively with BMI, waist girth, and visceral and subcutaneous AT (0.39 < or = r<0.48; P < 0.0001). Multiple regression analyses revealed that visceral AT was the best independent predictor of IL-1Ra levels, explaining 22% (P < 0.0001) of its variance. IL-1Ra (P < 0.05) was an independent predictor of several CMR markers including triglyceride and high-density lipoprotein (HDL) cholesterol levels, cholesterol/HDL cholesterol ratio, glucose and insulin concentrations in response to a 75 g oral glucose load, and fasting insulin levels, in addition to the expected contribution of visceral AT (P < 0.05). CONCLUSIONS These results suggest that elevated IL-1Ra concentrations are influenced to a greater extent by visceral than subcutaneous adiposity and that IL-1Ra is independently related to some features of CMR beyond the known contribution of visceral adiposity.
Collapse
Affiliation(s)
- Amélie Cartier
- Centre de recherche de l'Institut Universitaire de cardiologie et de pneumologie de Québec, 2725 chemin Sainte-Foy, Québec, QC, Canada
| | | | | | | | | | | | | |
Collapse
|
43
|
Hausman GJ, Barb CR, Dean RG. Patterns of gene expression in pig adipose tissue: insulin-like growth factor system proteins, neuropeptide Y (NPY), NPY receptors, neurotrophic factors and other secreted factors. Domest Anim Endocrinol 2008; 35:24-34. [PMID: 18325722 DOI: 10.1016/j.domaniend.2008.01.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2007] [Revised: 01/08/2008] [Accepted: 01/09/2008] [Indexed: 11/25/2022]
Abstract
Although cDNA microarray studies have examined gene expression in human and rodent adipose tissue, only one microarray study of adipose tissue from growing pigs has been reported. Total RNA was collected at slaughter from outer subcutaneous adipose tissue (OSQ) and middle subcutaneous adipose tissue (MSQ) from gilts at 90, 150, and 210 d (n=5 age(-1)). Dye labeled cDNA probes were hybridized to custom porcine microarrays (70-mer oligonucleotides). Gene expression of insulin-like growth factor binding proteins (IGFBPs), hormones, growth factors, neuropeptide Y (NPY) receptors (NPYRs) and other receptors in OSQ and MSQ changed little with age in growing pigs. Distinct patterns of relative gene expression were evident within NPYR and IGFBP family members in adipose tissue from growing pigs. Relative gene expression levels of NPY2R, NPY4R and angiopoietin 2 (ANG-2) distinguished OSQ and MSQ depots in growing pigs. We demonstrated, for the first time, the expression of IGFBP-7, IGFBP-5, NPY1R, NPY2R, NPY, connective tissue growth factor (CTGF), brain-derived neurotrophic factor (BDNF) and ciliary neurotrophic factor (CNTF) genes in pig adipose tissue with microarray and RT-PCR assays. Furthermore, adipose tissue CTGF gene expression was upregulated while NPY and NPY2R gene expression were significantly down regulated by age. These studies demonstrate that expression of neuropeptides and neurotrophic factors in pig adipose tissue may be involved in regulation of leptin secretion. Many other regulatory factors were not influenced by age in growing pigs but may be influenced by location or depot.
Collapse
Affiliation(s)
- G J Hausman
- United States Department of Agriculture, Agricultural Research Service, Richard B. Russell Agricultural Research Center, 950 College Station Road, Athens, GA 30605, USA.
| | | | | |
Collapse
|
44
|
Fain JN, Buehrer B, Bahouth SW, Tichansky DS, Madan AK. Comparison of messenger RNA distribution for 60 proteins in fat cells vs the nonfat cells of human omental adipose tissue. Metabolism 2008; 57:1005-15. [PMID: 18555844 DOI: 10.1016/j.metabol.2008.02.019] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2007] [Accepted: 02/28/2008] [Indexed: 01/04/2023]
Abstract
The messenger RNA (mRNA) distribution of 60 proteins was examined in the 3 fractions obtained by collagenase digestion (fat cells and the nonfat cells comprising the tissue remaining after collagenase digestion [matrix] and the stromovascular cells) of omental adipose tissue obtained from morbidly obese women undergoing bariatric surgery. Fat cells were enriched by at least 3-fold as compared with nonfat cells in the mRNAs for retinol binding protein 4, angiotensinogen, adipsin, glutathione peroxidase 3, uncoupling protein 2, peroxisome proliferator-activated receptor gamma, cell death-inducing DFFA-like effector A, fat-specific protein 27, 11beta-hydroxysteroid dehydrogenase 1, glycerol channel aquaporin 7, NADPH:quinone oxidoreductase 1, cyclic adenosine monophosphate phosphodiesterase 3B, glyceraldehyde-3-phosphate dehydrogenase, insulin receptor, and amyloid A1. Fat cells were also enriched by at least 26-fold in the mRNAs for proteins involved in lipolysis such as hormone-sensitive lipase, lipoprotein lipase, adipose tissue triglyceride lipase, and FAT/CD36. The relative distribution of mRNAs in cultured preadipocytes was also compared with that of in vitro differentiated adipocytes derived from human omental adipose tissue. Cultured preadipocytes had far lower levels of the mRNAs for inflammatory proteins than the nonfat cells of omental adipose tissue. The nonfat cells were enriched by at least 5-fold in the mRNAs for proteins involved in the inflammatory response such as tumor necrosis factor alpha, interleukin lbeta, cyclooxygenase 2, interleukin 24, interleukin 6, and monocyte chemoattractant protein 1 plus the mRNAs for osteopontin, vaspin, endothelin, angiotensin II receptor 1, butyrylcholinesterase, lipocalin 2, and plasminogen activator inhibitor 1. The cells in the adipose tissue matrix were enriched at least 3-fold as compared with the isolated stromovascular cells in the mRNAs for proteins related to the inflammatory response, as well as osteopontin and endothelial nitric oxide synthase. We conclude that the mRNAs for inflammatory proteins are primarily present in the nonfat cells of human omental adipose tissue.
Collapse
Affiliation(s)
- John N Fain
- Department of Molecular Sciences, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | | | | | | | | |
Collapse
|
45
|
Wang P, Mariman E, Renes J, Keijer J. The secretory function of adipocytes in the physiology of white adipose tissue. J Cell Physiol 2008; 216:3-13. [PMID: 18264975 DOI: 10.1002/jcp.21386] [Citation(s) in RCA: 202] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
White adipose tissue, previously regarded as a passive lipid storage site, is now viewed as a dynamic tissue. It has the capacity to actively communicate by sending and receiving different types of signals. An overview of these signals, the external modulators that affect adipose tissue and the secreted signaling molecules, the adipokines, is presented. The secretory function is highlighted in relation to energy metabolism, inflammation and the extracellular matrix and placed in the context of adipose tissue biology. We observe that the endocrine function of adipocytes receives much attention, while its paracrine and autocrine functions are underestimated. Also, we provide examples that species specificity should not be neglected. We conclude that adipose tissue primarily is an energy storage organ, well supported by its secretory function.
Collapse
Affiliation(s)
- Ping Wang
- Functional Genomics Group, Department of Human Biology, The Nutrition and Toxicology Research Institute Maastricht (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | | | | | | |
Collapse
|
46
|
Bays HE, González-Campoy JM, Bray GA, Kitabchi AE, Bergman DA, Schorr AB, Rodbard HW, Henry RR. Pathogenic potential of adipose tissue and metabolic consequences of adipocyte hypertrophy and increased visceral adiposity. Expert Rev Cardiovasc Ther 2008; 6:343-68. [PMID: 18327995 DOI: 10.1586/14779072.6.3.343] [Citation(s) in RCA: 339] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
When caloric intake exceeds caloric expenditure, the positive caloric balance and storage of energy in adipose tissue often causes adipocyte hypertrophy and visceral adipose tissue accumulation. These pathogenic anatomic abnormalities may incite metabolic and immune responses that promote Type 2 diabetes mellitus, hypertension and dyslipidemia. These are the most common metabolic diseases managed by clinicians and are all major cardiovascular disease risk factors. 'Disease' is traditionally characterized as anatomic and physiologic abnormalities of an organ or organ system that contributes to adverse health consequences. Using this definition, pathogenic adipose tissue is no less a disease than diseases of other body organs. This review describes the consequences of pathogenic fat cell hypertrophy and visceral adiposity, emphasizing the mechanistic contributions of genetic and environmental predispositions, adipogenesis, fat storage, free fatty acid metabolism, adipocyte factors and inflammation. Appreciating the full pathogenic potential of adipose tissue requires an integrated perspective, recognizing the importance of 'cross-talk' and interactions between adipose tissue and other body systems. Thus, the adverse metabolic consequences that accompany fat cell hypertrophy and visceral adiposity are best viewed as a pathologic partnership between the pathogenic potential adipose tissue and the inherited or acquired limitations and/or impairments of other body organs. A better understanding of the physiological and pathological interplay of pathogenic adipose tissue with other organs and organ systems may assist in developing better strategies in treating metabolic disease and reducing cardiovascular disease risk.
Collapse
Affiliation(s)
- Harold E Bays
- L-MARC Research Center, 3288 Illinois Avenue, Louisville, KY 40213, USA.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Identification of omentin mRNA in human epicardial adipose tissue: comparison to omentin in subcutaneous, internal mammary artery periadventitial and visceral abdominal depots. Int J Obes (Lond) 2008; 32:810-5. [PMID: 18180782 DOI: 10.1038/sj.ijo.0803790] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The purpose of this study was to determine the relative distribution of omentin and visfatin mRNA in human epicardial, peri-internal mammary, upper thoracic, upper abdominal and leg vein subcutaneous adipose tissue as well as the distribution of omentin in the nonfat cells and adipocytes of human omental adipose tissue. BACKGROUND Omentin is found in human omentum but not subcutaneous fat. Omentin and visfatin are considered markers of visceral abdominal fat. RESEARCH DESIGN AND METHODS The mRNA content of omentin and visfatin was measured by qRT-PCR analysis of fat samples removed from humans undergoing cardiac or bariatric surgery. RESULTS Omentin mRNA in internal mammary fat was 3.5%, that in the upper thoracic subcutaneous fat was 4.7% while that in the other subcutaneous fat depots was less than 1% of omentin in epicardial fat. The distribution of visfatin mRNA did not vary between the five depots. Omentin mRNA was preferentially expressed in the nonfat cells of omental adipose tissue since the omentin mRNA content of isolated adipocytes was 9% of that in nonfat cells, and similar results were seen for visfatin. The amount of omentin mRNA in differentiated adipocytes was 0.3% and that of visfatin 4% of that in nonfat cells. The amount of omentin mRNA in preadipocytes was virtually undetectable while that of visfatin was 3% of that in freshly isolated nonfat cells from omental adipose tissue. CONCLUSION Omentin mRNA is predominantly found in epicardial and omental human fat whereas visfatin mRNA is found to the same extent in epicardial, subcutaneous and omental fat.
Collapse
|
48
|
Fain JN, Nesbit AS, Sudlow FF, Cheema P, Peeples JM, Madan AK, Tichansky DS. Release in vitro of adipsin, vascular cell adhesion molecule 1, angiotensin 1-converting enzyme, and soluble tumor necrosis factor receptor 2 by human omental adipose tissue as well as by the nonfat cells and adipocytes. Metabolism 2007; 56:1583-90. [PMID: 17950111 DOI: 10.1016/j.metabol.2007.06.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2007] [Accepted: 06/06/2007] [Indexed: 01/04/2023]
Abstract
The relative release in vitro of adipsin, vascular cell adhesion molecule (VCAM) 1, angiotensin 1-converting enzyme (ACE), and soluble tumor necrosis factor alpha receptor 2 (sTNFR2) by explants of human omental and subcutaneous adipose tissue as well as the nonfat cell fractions and adipocytes from morbidly obese gastric bypass women was compared with that by tissue from obese abdominoplasty patients. Release of VCAM-1 and ACE by omental adipose tissue explants was 220% and 80% greater, respectively, over 48 hours of incubation than that by subcutaneous adipose tissue explants. However, this difference was not seen when release by adipocytes derived from omental fat was compared with that by adipocytes from subcutaneous fat. The release of adipsin and sTNFR2 by omental adipose tissue explants did not differ from that by subcutaneous tissue adipose tissue. The release of adipsin by tissue explants over 48 hours was 100-fold greater than that of VCAM-1, ACE, or sTNFR2. Most of the release of all 4 adipokines was due to the nonfat cells because adipsin release by adipocytes was only 13% of that by the nonfat cells derived from the same amount of adipose tissue, whereas ACE release by adipocytes was 7% and release of VCAM-1 as well as sTNFR2 by adipocytes was 4% or less of that by nonfat cells. The total release in vitro of ACE and sTNFR2, but not that of adipsin or VCAM-1, was enhanced in adipose tissue explants from morbidly obese women as compared with those by explants derived from obese women. We conclude that although human adipose tissue explants release appreciable amounts of adipsin and far smaller amounts of VCAM-1, ACE, and sTNFR2 in vitro, more than 87% of the release is due to the nonfat cells present in adipose tissue. Furthermore, the enhanced release of VCAM-1 and ACE seen in omental adipose tissue explants as compared with explants of subcutaneous adipose tissue is due to release by nonfat cells.
Collapse
Affiliation(s)
- John N Fain
- Department of Molecular Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | | | | | | | | | | | | |
Collapse
|
49
|
Gomez-Merino D, Drogou C, Guezennec CY, Chennaoui M. Effects of chronic exercise on cytokine production in white adipose tissue and skeletal muscle of rats. Cytokine 2007; 40:23-9. [PMID: 17826174 DOI: 10.1016/j.cyto.2007.07.188] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Revised: 07/13/2007] [Accepted: 07/26/2007] [Indexed: 11/23/2022]
Abstract
White adipose tissue (WAT) is a major source of production of cytokines involved in chronic diseases such as obesity, type 2 diabetes, and atherosclerosis. Long-term exercise has been proposed as a therapy to reduce chronic inflammation. We investigated here the influence of an intense exercise training (over 7 weeks) on several cytokine concentrations including interleukin 1 receptor antagonist (IL-1ra), IL-1beta, and IL-12 in serum, WAT, and skeletal muscle (SM) from non-obese rats. Two groups of 10 rats were investigated: one group was progressively trained (the two last weeks: 120min per day, 25m/min, 7% grade, 5 days per week) and the other age-matched group was used as a sedentary control. Compared to sedentary rats, weight gain was lower in the trained rats (P<0.01). In WAT, concentrations of IL-1ra, IL-1beta, and IL-12 were lower (P<0.001 for IL-1ra and IL-12, P<0.05 for IL-1beta) while they were higher in SM (P<0.01 for IL-1ra, P<0.001 for IL-1beta, P<0.05 for IL-12), and similar in serum. Significant correlations were noted between (i) body weight and WAT concentrations of IL-1ra, IL-1beta, and IL-12 (0.595, 0.450, and 0.481, respectively), (ii) body weight and IL-1beta concentration in SM (-0.526). We also observed significant negative correlations between WAT and SM concentrations of the three cytokines. We show here for the first time that intense exercise training with weight loss reduced concentrations of IL-1ra, IL-1beta, and IL-12 in WAT, while it increased them in SM. These results suggest that exercise could help reduce inflammation in WAT through mobilization of immune cells producing pro- and anti-inflammatory cytokines in SM.
Collapse
Affiliation(s)
- D Gomez-Merino
- Department of Physiology, IMASSA, B.P. 73, 91223 Brétigny-sur-Orge Cedex, France.
| | | | | | | |
Collapse
|
50
|
Darimont C, Avanti O, Blancher F, Wagniere S, Mansourian R, Zbinden I, Leone-Vautravers P, Fuerholz A, Giusti V, Macé K. Contribution of mesothelial cells in the expression of inflammatory-related factors in omental adipose tissue of obese subjects. Int J Obes (Lond) 2007; 32:112-20. [PMID: 17637700 DOI: 10.1038/sj.ijo.0803688] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE The objective of this study was to determine the contribution of mesothelial cells, present in human omental adipose tissue (OAT) but not in the subcutaneous depot (SAT), on the expression of inflammation-related factors. DESIGN Comparison of the expression profiles of inflammation-related genes in mesothelial cells with those in the adipocyte-enriched (AEF) and stromal vascular fractions (SVF) and localization of interleukin-18 (IL-18) expression in adipose depots. SUBJECTS Eleven obese Caucasian female subjects undergoing gastric bypass surgery (body mass index: 43.6+/-1.3 kg/m(2); age: 41.6+/-2.3 years). MEASUREMENTS The expression profiles of cytokine and chemokine-related genes in mesothelial cells and in cell fractions prepared from OAT were assessed by the microarray technique. The differential expression of IL-18 was confirmed by real-time PCR and the protein was localized in adipose depots by immunohistochemistry. RESULTS Microarray data analysis demonstrated that, of the 16 cytokine and chemokine-related genes that were upregulated in mesothelial cells compared with the AEF, IL-18 was the cytokine with the highest differential expression. IL-18 expression was similar in mesothelial cells and the SVF. In both SAT and OAT, IL-18 was immunolocalized in neutrophils and mast cells, but not in macrophages nor adipocytes. This cytokine was also detected in mesothelial cells in OAT. This additional source of expression may explain the higher IL-18 expression levels in OAT than SAT (+5.9-fold). CONCLUSION By their capacity to express inflammatory-related factors, and in particular the proinflammatory cytokine IL-18 in OAT, mesothelial cells appear as a new player in the process of low-grade inflammation associated with obesity.
Collapse
Affiliation(s)
- C Darimont
- Nestlé Research Center, Department of Nutrition and Health, Lausanne, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|