151
|
Stafeev I, Podkuychenko N, Michurina S, Sklyanik I, Panevina A, Shestakova E, Yah'yaev K, Fedenko V, Ratner E, Vorotnikov A, Menshikov M, Yashkov Y, Parfyonova Y, Shestakova M. Low proliferative potential of adipose-derived stromal cells associates with hypertrophy and inflammation in subcutaneous and omental adipose tissue of patients with type 2 diabetes mellitus. J Diabetes Complications 2019; 33:148-159. [PMID: 30482492 DOI: 10.1016/j.jdiacomp.2018.10.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 09/25/2018] [Accepted: 10/16/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Obesity and type 2 diabetes mellitus (T2DM) are among the most important morbidity factors. In this study we tested the hypothesis that low proliferative potential of adipose derived stromal cells (ADSC) associates with reduced formation of new fat depots, excess accumulation of fat in the functional adipocytes and their hypertrophy, resulting in fat inflammation and insulin resistance. METHODS We screened two groups of obese patients with or without T2DM, matched for BMI, age, and duration of obesity to test the hypothesis that hypertrophy and decreased renewal of adipocytes may underlie transition from obesity to T2DM. All patients were matched for carbohydrate metabolism (fasting blood glucose level, glycated hemoglobin, HOMA-IR index and M-index). The subcutaneous and omental fat tissue biopsies were obtained during bariatric surgery from obese individuals with or without T2DM. The morphology and immunophenotype of subcutaneous and omental fat was assessed in frozen tissue sections. ADSC were isolated from both types of fat tissue biopsies and screened for morphology, proliferative potential and inflammatory status. RESULTS The non-diabetic patients had normal carbohydrate metabolism and moderate insulin resistance measured by HOMA-IR and hyperinsulinemic clamp (M-index), while T2DM patients were extremely insulin resistant by both indexes. The average size of diabetic adipocytes was higher than that of non-diabetic in both subcutaneous and omental fat tissues, indicating adipocyte hypertrophy in T2DM. Both these tissues contained higher level of macrophage infiltration and increased M1-like to M2-like ratio of macrophage subpopulations, suggesting increased fat inflammation in T2DM. This was confirmed by increased activatory phosphorylation of stress-induced JNK1/2 in diabetic ADSC. CONCLUSION These results suggest that blunted proliferation and increased hypertrophy of diabetic ADSC may lead to reduced insulin sensitivity via increased inflammation mediated by M1 macrophages and JNK1/2 pathway.
Collapse
Affiliation(s)
- I Stafeev
- National Medical Research Centre for Cardiology, Moscow, Russia; M.V. Lomonosov Moscow State University, Moscow, Russia; Endocrinology Research Centre, Moscow, Russia.
| | - N Podkuychenko
- National Medical Research Centre for Cardiology, Moscow, Russia; M.V. Lomonosov Moscow State University, Moscow, Russia; Endocrinology Research Centre, Moscow, Russia
| | - S Michurina
- National Medical Research Centre for Cardiology, Moscow, Russia; M.V. Lomonosov Moscow State University, Moscow, Russia
| | - I Sklyanik
- Endocrinology Research Centre, Moscow, Russia
| | - A Panevina
- Endocrinology Research Centre, Moscow, Russia
| | | | - K Yah'yaev
- Central Clinical Hospital #1 of LLC Russian Railways, Moscow, Russia
| | - V Fedenko
- V.I. Kulakov National Medical Research Centre for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - E Ratner
- National Medical Research Centre for Cardiology, Moscow, Russia; Endocrinology Research Centre, Moscow, Russia
| | - A Vorotnikov
- National Medical Research Centre for Cardiology, Moscow, Russia
| | - M Menshikov
- National Medical Research Centre for Cardiology, Moscow, Russia
| | - Y Yashkov
- V.I. Kulakov National Medical Research Centre for Obstetrics, Gynecology and Perinatology, Moscow, Russia
| | - Ye Parfyonova
- National Medical Research Centre for Cardiology, Moscow, Russia; M.V. Lomonosov Moscow State University, Moscow, Russia
| | | |
Collapse
|
152
|
Daryabor G, Kabelitz D, Kalantar K. An update on immune dysregulation in obesity-related insulin resistance. Scand J Immunol 2019; 89:e12747. [PMID: 30593678 DOI: 10.1111/sji.12747] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/22/2018] [Accepted: 12/25/2018] [Indexed: 12/29/2022]
Abstract
Obesity is associated with chronic low-grade inflammation of the adipose tissue (AT) that might develop into systemic inflammation, insulin resistance (IR) and an increased risk of type 2 diabetes mellitus (T2DM) in severe obese rodents and humans. In the lean state, small normal adipocytes and AT macrophages interact with each other to maintain metabolic homeostasis but during obesity, enlarged adipocytes secrete inflammatory mediators and express immune receptors to recruit immune cells and aggravate the inflammation. The better understanding of the obesity-related inflammatory milieu and the sequential events leading to IR could be helpful in designing new preventive and therapeutic strategies. The present review will discuss the cellular and molecular abnormalities participating in the pathogenesis of obesity in obese individuals as well as high-fat diet (HFD)-fed mice, a mouse model of obesity.
Collapse
Affiliation(s)
- Gholamreza Daryabor
- Department of Immunology, Shiraz Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Kurosh Kalantar
- Department of Immunology, Shiraz Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
153
|
Macdougall CE, Longhi MP. Adipose tissue dendritic cells in steady-state. Immunology 2019; 156:228-234. [PMID: 30552824 DOI: 10.1111/imm.13034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/21/2018] [Accepted: 11/28/2018] [Indexed: 12/17/2022] Open
Abstract
Healthy white adipose tissue (WAT) participates in regulating systemic metabolism, whereas dysfunctional WAT plays a prominent role in the development of obesity-associated co-morbidities. Tissue-resident immune cells are important for maintaining WAT homeostasis, including conventional dendritic cells (cDCs) which are critical in the initiation and regulation of adaptive immune responses. Due to phenotypic overlap with other myeloid cells, the distinct contribution of WAT cDCs has been poorly understood. This review will discuss the contribution of cDCs in the maintenance of WAT homeostasis. In particular, the review will focus on the metabolic cross-talk between cDCs and adipocytes that regulates local immune responses during physiological conditions.
Collapse
Affiliation(s)
- Claire E Macdougall
- William Harvey Research Institute, Barts and the London, Queen Mary University of London, London, UK
| | - M Paula Longhi
- William Harvey Research Institute, Barts and the London, Queen Mary University of London, London, UK
| |
Collapse
|
154
|
Wu D, Han JM, Yu X, Lam AJ, Hoeppli RE, Pesenacker AM, Huang Q, Chen V, Speake C, Yorke E, Nguyen N, Sampath S, Harris D, Levings MK. Characterization of regulatory T cells in obese omental adipose tissue in humans. Eur J Immunol 2019; 49:336-347. [DOI: 10.1002/eji.201847570] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 11/09/2018] [Accepted: 12/17/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Dan Wu
- Department of SurgeryUniversity of British Columbia Vancouver BC Canada
- BC Children's Hospital Research Institute Vancouver BC Canada
| | - Jonathan M. Han
- Department of SurgeryUniversity of British Columbia Vancouver BC Canada
- BC Children's Hospital Research Institute Vancouver BC Canada
| | - Xin Yu
- Prevention of Organ Failure (PROOF) Centre of Excellence Vancouver BC Canada
| | - Avery J. Lam
- Department of SurgeryUniversity of British Columbia Vancouver BC Canada
- BC Children's Hospital Research Institute Vancouver BC Canada
| | - Romy E. Hoeppli
- Department of SurgeryUniversity of British Columbia Vancouver BC Canada
- BC Children's Hospital Research Institute Vancouver BC Canada
| | - Anne M. Pesenacker
- Department of SurgeryUniversity of British Columbia Vancouver BC Canada
- BC Children's Hospital Research Institute Vancouver BC Canada
| | - Qing Huang
- Department of SurgeryUniversity of British Columbia Vancouver BC Canada
- BC Children's Hospital Research Institute Vancouver BC Canada
| | - Virginia Chen
- Prevention of Organ Failure (PROOF) Centre of Excellence Vancouver BC Canada
| | - Cate Speake
- Diabetes Clinical Research ProgramBenaroya Research Institute Seattle WA USA
| | - Ekua Yorke
- Department of SurgeryUniversity of British Columbia Vancouver BC Canada
- Richmond Metabolic and Bariatric Surgery ProgramRichmond Hospital Richmond BC Canada
| | - Nam Nguyen
- Department of SurgeryUniversity of British Columbia Vancouver BC Canada
- Richmond Metabolic and Bariatric Surgery ProgramRichmond Hospital Richmond BC Canada
| | - Sharadh Sampath
- Department of SurgeryUniversity of British Columbia Vancouver BC Canada
- Richmond Metabolic and Bariatric Surgery ProgramRichmond Hospital Richmond BC Canada
| | - David Harris
- Richmond Metabolic and Bariatric Surgery ProgramRichmond Hospital Richmond BC Canada
- Department of MedicineUniversity of British Columbia Vancouver BC Canada
| | - Megan K. Levings
- Department of SurgeryUniversity of British Columbia Vancouver BC Canada
- BC Children's Hospital Research Institute Vancouver BC Canada
| |
Collapse
|
155
|
Burhans MS, Hagman DK, Kuzma JN, Schmidt KA, Kratz M. Contribution of Adipose Tissue Inflammation to the Development of Type 2 Diabetes Mellitus. Compr Physiol 2018; 9:1-58. [PMID: 30549014 DOI: 10.1002/cphy.c170040] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The objective of this comprehensive review is to summarize and discuss the available evidence of how adipose tissue inflammation affects insulin sensitivity and glucose tolerance. Low-grade, chronic adipose tissue inflammation is characterized by infiltration of macrophages and other immune cell populations into adipose tissue, and a shift toward more proinflammatory subtypes of leukocytes. The infiltration of proinflammatory cells in adipose tissue is associated with an increased production of key chemokines such as C-C motif chemokine ligand 2, proinflammatory cytokines including tumor necrosis factor α and interleukins 1β and 6 as well as reduced expression of the key insulin-sensitizing adipokine, adiponectin. In both rodent models and humans, adipose tissue inflammation is consistently associated with excess fat mass and insulin resistance. In humans, associations with insulin resistance are stronger and more consistent for inflammation in visceral as opposed to subcutaneous fat. Further, genetic alterations in mouse models of obesity that reduce adipose tissue inflammation are-almost without exception-associated with improved insulin sensitivity. However, a dissociation between adipose tissue inflammation and insulin resistance can be observed in very few rodent models of obesity as well as in humans following bariatric surgery- or low-calorie-diet-induced weight loss, illustrating that the etiology of insulin resistance is multifactorial. Taken together, adipose tissue inflammation is a key factor in the development of insulin resistance and type 2 diabetes in obesity, along with other factors that likely include inflammation and fat accumulation in other metabolically active tissues. © 2019 American Physiological Society. Compr Physiol 9:1-58, 2019.
Collapse
Affiliation(s)
- Maggie S Burhans
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Derek K Hagman
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Jessica N Kuzma
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Kelsey A Schmidt
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Mario Kratz
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Epidemiology, University of Washington, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
156
|
Harmon DB, Wu C, Dedousis N, Sipula IJ, Stefanovic-Racic M, Schoiswohl G, O'Donnell CP, Alonso LC, Kershaw EE, Kelley EE, O'Doherty RM. Adipose tissue-derived free fatty acids initiate myeloid cell accumulation in mouse liver in states of lipid oversupply. Am J Physiol Endocrinol Metab 2018; 315:E758-E770. [PMID: 30086648 PMCID: PMC6293173 DOI: 10.1152/ajpendo.00172.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Accumulation of myeloid cells in the liver, notably dendritic cells (DCs) and monocytes/macrophages (MCs), is a major component of the metainflammation of obesity. However, the mechanism(s) stimulating hepatic DC/MC infiltration remain ill defined. Herein, we addressed the hypothesis that adipose tissue (AT) free fatty acids (FFAs) play a central role in the initiation of hepatic DC/MC accumulation, using a number of mouse models of altered FFA supply to the liver. In two models of acute FFA elevation (lipid infusion and fasting) hepatic DC/MC and triglycerides (TGs) but not AT DC/MC were increased without altering plasma cytokines (PCs; TNFα and monocyte chemoattractant protein 1) and with variable effects on oxidative stress (OxS) markers. However, fasting in mice with profoundly reduced AT lipolysis (AT-specific deletion of adipose TG lipase; AAKO) failed to elevate liver DC/MC, TG, or PC, but liver OxS increased. Livers of obese AAKO mice that are known to be resistant to steatosis were similarly protected from inflammation. In high-fat feeding studies of 1, 3, 6, or 20-wk duration, liver DC/MC accumulation dissociated from PC and OxS but tracked with liver TGs. Furthermore, decreasing OxS by ~80% in obese mice failed to decrease liver DC/MC. Therefore, FFA and more specifically AT-derived FFA stimulate hepatic DC/MC accumulation, thus recapitulating the pathology of the obese liver. In a number of cases the effects of FFA can be dissociated from OxS and PC but match well with liver TG, a marker of FFA oversupply.
Collapse
Affiliation(s)
- Daniel B Harmon
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh , Pittsburgh, Pennsylvania
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Chao Wu
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh , Pittsburgh, Pennsylvania
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
- Department of Metabolism & Endocrinology, The Second Xiangya Hospital, Central South University , Changsha , China
| | - Nikolaos Dedousis
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh , Pittsburgh, Pennsylvania
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Ian J Sipula
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh , Pittsburgh, Pennsylvania
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Maja Stefanovic-Racic
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh , Pittsburgh, Pennsylvania
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Gabriele Schoiswohl
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh , Pittsburgh, Pennsylvania
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Christopher P O'Donnell
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care Medicine University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Laura C Alonso
- Department of Medicine, Diabetes Division, University of Massachusetts , Worcester, Massachusetts
| | - Erin E Kershaw
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh , Pittsburgh, Pennsylvania
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Eric E Kelley
- Department of Physiology and Pharmacology, West Virginia University , Morgantown, West Virginia
| | - Robert M O'Doherty
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh , Pittsburgh, Pennsylvania
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
157
|
Wang Q, Wu H. T Cells in Adipose Tissue: Critical Players in Immunometabolism. Front Immunol 2018; 9:2509. [PMID: 30459770 PMCID: PMC6232870 DOI: 10.3389/fimmu.2018.02509] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/10/2018] [Indexed: 12/23/2022] Open
Abstract
Adipose tissue performs immunoregulatory functions in addition to fat storage. Various T cells in different fat depots either help maintain metabolic homeostasis under healthy conditions or contribute to metabolic disorders in pathological conditions such as obesity, diabetes, cardiovascular diseases, or even cancer. These T cells play critical roles in immunometabolism, which refers to the intersection of immunity and metabolism. Numerous studies have examined the presence and changes of different T cell subsets, including helper T cells, regulatory T cells, cytotoxic T cells, and natural killer T cells, in adipose depots in health and diseases. In this review, we will discuss the adipose tissue niches that influence the patterns and functions of T cell subsets and in turn the impact of these T cells on cell- or body-based immunometabolism accounting for health and obesity.
Collapse
Affiliation(s)
- Qun Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Huaizhu Wu
- Department of Medicine and Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
158
|
Russo L, Lumeng CN. Properties and functions of adipose tissue macrophages in obesity. Immunology 2018; 155:407-417. [PMID: 30229891 DOI: 10.1111/imm.13002] [Citation(s) in RCA: 439] [Impact Index Per Article: 62.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/06/2018] [Accepted: 09/07/2018] [Indexed: 12/12/2022] Open
Abstract
The expansion of adipose tissue (AT) in obesity is accompanied by the accumulation of immune cells that contribute to a state of low-grade, chronic inflammation and dysregulated metabolism. Adipose tissue macrophages (ATMs) represent the most abundant class of leukocytes in AT and are involved in the regulation of several regulatory physiological processes, such as tissue remodeling and insulin sensitivity. With progressive obesity, ATMs are key mediators of meta-inflammation, insulin resistance and impairment of adipocyte function. While macrophage recruitment from blood monocytes is a critical component of the generation of AT inflammation, new studies have revealed a role for ATM proliferation in the early stages of obesity and in sustaining AT inflammation. In addition, studies have revealed a more complex range of macrophage activation states than the previous M1/M2 model, and the existence of different macrophage profiles between human and animal models. This review will summarize the current understanding of the regulatory mechanisms of ATM function in relation to obesity, type 2 diabetes, depot of origin, and to other leukocytes such as AT dendritic cells, with hopes of emphasizing the regulatory nodes that can potentially be targeted to prevent and treat obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Lucia Russo
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Carey N Lumeng
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA.,Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
159
|
Gut adaptation after metabolic surgery and its influences on the brain, liver and cancer. Nat Rev Gastroenterol Hepatol 2018; 15:606-624. [PMID: 30181611 DOI: 10.1038/s41575-018-0057-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Metabolic surgery is the best treatment for long-term weight loss maintenance and comorbidity control. Metabolic operations were originally intended to change anatomy to alter behaviour, but we now understand that the anatomical changes can modulate physiology to change behaviour. They are no longer considered only mechanically restrictive and/or malabsorptive procedures; rather, they are considered metabolic procedures involving complex physiological changes, whereby gut adaptation influences signalling pathways in several other organs, including the liver and the brain, regulating hunger, satiation, satiety, body weight, glucose metabolism and immune functions. The integrative physiology of gut adaptation after these operations consists of a complex mechanistic web of communication between gut hormones, bile acids, gut microbiota, the brain and both enteric and central nervous systems. The understanding of nutrient sensing via enteroendocrine cells, the enteric nervous system, hypothalamic peptides and adipose tissue and of the role of inflammation has advanced our knowledge of this integrative physiology. In this Review, we focus on the adaptation of gut physiology to the anatomical alterations from Roux-en-Y gastric bypass and vertical sleeve gastrectomy and the influence of these procedures on food intake, weight loss, nonalcoholic fatty liver disease (NAFLD) and cancer. We also aim to demonstrate the underlying mechanisms that could explain how metabolic surgery could be used as a therapeutic option in NAFLD and certain obesity-related cancers.
Collapse
|
160
|
Abstract
During the last decades, research on adipose tissues has spread in parallel with the extension of obesity. Several observations converged on the idea that adipose tissues are organized in a large organ with endocrine and plastic properties. Two parenchymal components: white (WATs) and brown adipose tissues (BATs) are contained in subcutaneous and visceral compartments. Although both have endocrine properties, their function differs: WAT store lipids to allow intervals between meals, BAT burns lipids for thermogenesis. In spite of these opposite functions, they share the ability for reciprocal reversible transdifferentiation to tackle special physiologic needs. Thus, chronic need for thermogenesis induces browning and chronic positive energy balance induce whitening. Lineage tracing and data from explant studies strongly suggest other remodeling properties of this organ. During pregnancy and lactation breast WAT transdifferentiates into milk-secreting glands, composed by cells with abundant cytoplasmic lipids (pink adipocytes) and in the postlactation period pink adipocytes transdifferentiate back into WAT and BAT. The plastic properties of mature adipocytes are supported also by a liposecretion process in vitro where adult cell in culture transdifferentiate to differentiated fibroblast-like elements able to give rise to different phenotypes (rainbow adipocytes). In addition, the inflammasome system is activated in stressed adipocytes from obese adipose tissue. These adipocytes die and debris are reabsorbed by macrophages inducing a chronic low-grade inflammation, potentially contributing to insulin resistance and T2 diabetes. Thus, the plastic properties of this organ could open new therapeutic perspectives in the obesity-related metabolic disease and in breast pathologies. © 2018 American Physiological Society. Compr Physiol 8:1357-1431, 2018.
Collapse
Affiliation(s)
- Saverio Cinti
- Professor of Human Anatomy, Director, Center of Obesity, University of Ancona (Politecnica delle Marche), Ancona, Italy
| |
Collapse
|
161
|
Th17 and Treg lymphocytes in obesity and Type 2 diabetic patients. Clin Immunol 2018; 197:77-85. [PMID: 30218707 DOI: 10.1016/j.clim.2018.09.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/09/2018] [Accepted: 09/11/2018] [Indexed: 12/18/2022]
Abstract
Assumption that the pathogenesis of obesity-associated type 2 diabetes (T2DM) encompasses inflammation and autoimmune aspects is increasingly recognized. In the state of obesity and T2DM, the imbalance of T helper 17 (Th17) cells and regulatory T (Treg) cells are observed. These alterations reflect a loss of T cell homeostasis, which may contribute to tissue and systemic inflammation and immunity in T2DM. In this review we will discuss the accumulating data supporting the concept that Th17/Treg mediated immune responses are present in obesity-related T2DM pathogenesis, and provide evidences that restoration of Th17/Treg imbalance may be a possible therapeutic avenue for the prevention and treatment of T2DM and its complications.
Collapse
|
162
|
Unamuno X, Gómez-Ambrosi J, Rodríguez A, Becerril S, Frühbeck G, Catalán V. Adipokine dysregulation and adipose tissue inflammation in human obesity. Eur J Clin Invest 2018; 48:e12997. [PMID: 29995306 DOI: 10.1111/eci.12997] [Citation(s) in RCA: 411] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 07/10/2018] [Indexed: 12/13/2022]
Abstract
Obesity, a worldwide epidemic, confers increased risk for multiple serious conditions, including type 2 diabetes, cardiovascular diseases, nonalcoholic fatty liver disease and cancer. Adipose tissue is considered one of the largest endocrine organs in the body as well as an active tissue for cellular reactions and metabolic homeostasis rather than an inert tissue for energy storage. The functional pleiotropism of adipose tissue relies on its ability to synthesize and release a large number of hormones, cytokines, extracellular matrix proteins and growth and vasoactive factors, collectively termed adipokines that influence a variety of physiological and pathophysiological processes. In the obese state, excessive visceral fat accumulation causes adipose tissue dysfunctionality that strongly contributes to the onset of obesity-related comorbidities. The mechanisms underlying adipose tissue dysfunction include adipocyte hypertrophy and hyperplasia, increased inflammation, impaired extracellular matrix remodelling and fibrosis together with an altered secretion of adipokines. This review describes how adipose tissue becomes inflamed in obesity and summarizes key players and molecular mechanisms involved in adipose inflammation.
Collapse
Affiliation(s)
- Xabier Unamuno
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.,Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
163
|
Frydrych LM, Bian G, O'Lone DE, Ward PA, Delano MJ. Obesity and type 2 diabetes mellitus drive immune dysfunction, infection development, and sepsis mortality. J Leukoc Biol 2018; 104:525-534. [PMID: 30066958 DOI: 10.1002/jlb.5vmr0118-021rr] [Citation(s) in RCA: 178] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 06/18/2018] [Accepted: 06/19/2018] [Indexed: 12/14/2022] Open
Abstract
Obesity and type 2 diabetes mellitus (T2D) are global pandemics. Worldwide, the prevalence of obesity has nearly tripled since 1975 and the prevalence of T2D has almost doubled since 1980. Both obesity and T2D are indolent and chronic diseases that develop gradually, with cellular physiologic changes occurring before the clinical signs and symptoms of the diseases become apparent. Individuals with obesity and T2D are physiologically frail and have an increased risk of infections and mortality from sepsis. Improvement in the morbidity and mortality of these at-risk populations would provide a great societal benefit. We believe that the worsened outcomes observed in these patient populations is due to immune system dysfunction that is triggered by the chronic low-grade inflammation present in both diseases. As immune modulatory therapies have been utilized in other chronic inflammatory diseases, there is an emerging role for immune modulatory therapies that target the chronically affected immune pathways in obese and T2D patients. Additionally, bariatric surgery is currently the most successful treatment for obesity and is the only weight loss method that also causes a sustained, substantial improvement of T2D. Consequently, bariatric surgery may also have a role in improving immunity in these patient populations.
Collapse
Affiliation(s)
- Lynn M Frydrych
- Department of Surgery, Division of Acute Care Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Guowu Bian
- Department of Surgery, Division of Acute Care Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - David E O'Lone
- Department of Surgery, Division of Acute Care Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Peter A Ward
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew J Delano
- Department of Surgery, Division of Acute Care Surgery, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
164
|
Zhou H, Liu F. Regulation, Communication, and Functional Roles of Adipose Tissue-Resident CD4 + T Cells in the Control of Metabolic Homeostasis. Front Immunol 2018; 9:1961. [PMID: 30233575 PMCID: PMC6134258 DOI: 10.3389/fimmu.2018.01961] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/09/2018] [Indexed: 01/21/2023] Open
Abstract
Evidence accumulated over the past few years has documented a critical role for adipose tissue (AT)-resident immune cells in the regulation of local and systemic metabolic homeostasis. In the lean state, visceral adipose tissue (VAT) is predominated by anti-inflammatory T-helper 2 (Th2) and regulatory T (Treg) cell subsets. As obesity progresses, the population of Th2 and Treg cells decreases while that of the T-helper 1 (Th1) and T-helper 17 (Th17) cells increases, leading to augmented inflammation and insulin resistance. Notably, recent studies also suggest a potential role of CD4+ T cells in the control of thermogenesis and energy homeostasis. In this review, we have summarized recent advances in understanding the characteristics and functional roles of AT CD4+ T cell subsets during obesity and energy expenditure. We have also discussed new findings on the crosstalk between CD4+ T cells and local antigen-presenting cells (APCs) including adipocytes, macrophages, and dendritic cells (DCs) to regulate AT function and metabolic homeostasis. Finally, we have highlighted the therapeutic potential of targeting CD4+ T cells as an effective strategy for the treatment of obesity and its associated metabolic diseases.
Collapse
Affiliation(s)
- Haiyan Zhou
- Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center of Central South University, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feng Liu
- Department of Metabolism and Endocrinology, Metabolic Syndrome Research Center of Central South University, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
165
|
The asthma-obesity relationship: underlying mechanisms and treatment implications. Curr Opin Pulm Med 2018; 24:42-49. [PMID: 29176481 DOI: 10.1097/mcp.0000000000000446] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW Obesity is a worldwide epidemic with a prevalence that has tripled in the last two decades. Worldwide, more than 1.5 billion adults are overweight and more than 500 million obese. Obesity has been suggested to be a risk factor for the development of more difficult-to-control asthma. Although the mechanisms underlying the asthma-obesity relationship are not fully understood, several possible explanations have been put forward. These will be reviewed in this manuscript as well as the implications for the treatment of overweight and obese asthma patients. RECENT FINDINGS Insulin resistance is a possible factor contributing to the asthma-obesity relationship and the effect is independent of other components of the metabolic syndrome such as hypertriglyceridemia, hypertension, hyperglycemia, and systemic inflammation. Obesity has important effects on airway geometry, by especially reducing expiratory reserve volume causing obese asthmatics to breathe at low lung volumes. Furthermore, obesity affects the type of inflammation in asthma and is associated with reduced inhaled corticosteroids treatment responsiveness. SUMMARY Obesity induces the development of asthma with a difficult-to-control phenotype. Treatment targeting insulin resistance may be beneficial in obese asthma patients, especially when they have concomitant diabetes. Systemic corticosteroids should be avoided as much as possible as they are not very effective in obese asthma and associated with side-effects like diabetes, weight gain, and osteoporosis.
Collapse
|
166
|
Guzik TJ, Skiba DS, Touyz RM, Harrison DG. The role of infiltrating immune cells in dysfunctional adipose tissue. Cardiovasc Res 2018; 113:1009-1023. [PMID: 28838042 PMCID: PMC5852626 DOI: 10.1093/cvr/cvx108] [Citation(s) in RCA: 304] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 07/05/2017] [Indexed: 12/15/2022] Open
Abstract
Adipose tissue (AT) dysfunction, characterized by loss of its homeostatic functions, is a hallmark of non-communicable diseases. It is characterized by chronic low-grade inflammation and is observed in obesity, metabolic disorders such as insulin resistance and diabetes. While classically it has been identified by increased cytokine or chemokine expression, such as increased MCP-1, RANTES, IL-6, interferon (IFN) gamma or TNFα, mechanistically, immune cell infiltration is a prominent feature of the dysfunctional AT. These immune cells include M1 and M2 macrophages, effector and memory T cells, IL-10 producing FoxP3+ T regulatory cells, natural killer and NKT cells and granulocytes. Immune composition varies, depending on the stage and the type of pathology. Infiltrating immune cells not only produce cytokines but also metalloproteinases, reactive oxygen species, and chemokines that participate in tissue remodelling, cell signalling, and regulation of immunity. The presence of inflammatory cells in AT affects adjacent tissues and organs. In blood vessels, perivascular AT inflammation leads to vascular remodelling, superoxide production, endothelial dysfunction with loss of nitric oxide (NO) bioavailability, contributing to vascular disease, atherosclerosis, and plaque instability. Dysfunctional AT also releases adipokines such as leptin, resistin, and visfatin that promote metabolic dysfunction, alter systemic homeostasis, sympathetic outflow, glucose handling, and insulin sensitivity. Anti-inflammatory and protective adiponectin is reduced. AT may also serve as an important reservoir and possible site of activation in autoimmune-mediated and inflammatory diseases. Thus, reciprocal regulation between immune cell infiltration and AT dysfunction is a promising future therapeutic target.
Collapse
Affiliation(s)
- Tomasz J Guzik
- British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK.,Translational Medicine Laboratory, Department of Internal Medicine, Jagiellonian University, Collegium Medicum, Krakow, Poland
| | - Dominik S Skiba
- British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK.,Translational Medicine Laboratory, Department of Internal Medicine, Jagiellonian University, Collegium Medicum, Krakow, Poland
| | - Rhian M Touyz
- British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - David G Harrison
- British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK.,Department of Clinical Pharmacology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
167
|
Microenvironment of Immune Cells Within the Visceral Adipose Tissue Sensu Lato vs. Epicardial Adipose Tissue: What Do We Know? Inflammation 2018; 41:1142-1156. [PMID: 29846855 DOI: 10.1007/s10753-018-0798-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
The chronic low-grade inflammation of the visceral adipose tissue is now fully established as one of the main contributors to metabolic disorders such as insulin resistance, subsequently leading to metabolic syndrome and other associated cardiometabolic pathologies. The orchestration of immune response and the "ratio of responsibility" of different immune cell populations have been studied extensively over the last few years within the visceral adipose tissue in general sense (sensu lato). However, it is essential to clearly distinguish different types of visceral fat distribution. Visceral adipose tissue is not only the classical omental or epididymal depot, but includes also specific type of fat in the close vicinity to the myocardium-the epicardial adipose tissue. Disruption of this type of fat during obesity was found to have a unique and direct influence over the cardiovascular disease development. Therefore, epicardial adipose tissue and other types of visceral adipose tissue depots should be studied separately. The purpose of this review is to explore the present knowledge about the morphology and dynamics of individual populations of immune cells within the visceral adipose tissue sensu lato in comparison to the knowledge regarding the epicardial adipose tissue specifically.
Collapse
|
168
|
Sbierski-Kind J, Kath J, Brachs S, Streitz M, von Herrath MG, Kühl AA, Schmidt-Bleek K, Mai K, Spranger J, Volk HD. Distinct Housing Conditions Reveal a Major Impact of Adaptive Immunity on the Course of Obesity-Induced Type 2 Diabetes. Front Immunol 2018; 9:1069. [PMID: 29892281 PMCID: PMC5985496 DOI: 10.3389/fimmu.2018.01069] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 04/30/2018] [Indexed: 01/08/2023] Open
Abstract
Obesity is associated with adipose tissue inflammation, insulin resistance, and the development of type 2 diabetes (T2D). However, our knowledge is mostly based on conventional murine models and promising preclinical studies rarely translated into successful therapies. There is a growing awareness of the limitations of studies in laboratory mice, housed in abnormally hygienic specific pathogen-free (SPF) conditions, as relevant aspects of the human immune system remain unappreciated. Here, we assessed the impact of housing conditions on adaptive immunity and metabolic disease processes during high-fat diet (HFD). We therefore compared diet-induced obesity in SPF mice with those housed in non-SPF, so-called "antigen exposed" (AE) conditions. Surprisingly, AE mice fed a HFD maintained increased insulin levels to compensate for insulin resistance, which was reflected in islet hyperplasia and improved glucose tolerance compared to SPF mice. By contrast, we observed higher proportions of effector/memory T cell subsets in blood and liver of HFD AE mice accompanied by the development of non-alcoholic steatohepatitis-like liver pathology. Thus, our data demonstrate the impact of housing conditions on metabolic alterations. Studies in AE mice, in which physiological microbial exposure was restored, could provide a tool for revealing therapeutic targets for immune-based interventions for T2D patients.
Collapse
Affiliation(s)
- Julia Sbierski-Kind
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Partner Site Berlin, German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Jonas Kath
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sebastian Brachs
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Partner Site Berlin, German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Mathias Streitz
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Matthias G von Herrath
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Anja A Kühl
- Berlin Institute of Health (BIH), Berlin, Germany.,iPATH Berlin - Core Unit Immunopathology for Experimental Models, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Julius Wolff Institute (JWI), Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Knut Mai
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Partner Site Berlin, German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Joachim Spranger
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Partner Site Berlin, German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Hans-Dieter Volk
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
169
|
Kang M, Jeong J, Lee J, Park S, Sung Y, Choi M, Kwon W, Jang S, Choi KS, Choo YS, Yoon D, Kim MO, Ryoo ZY. Placental growth factor (PlGF) is linked to inflammation and metabolic disorders in mice with diet-induced obesity. Endocr J 2018; 65:437-447. [PMID: 29434073 DOI: 10.1507/endocrj.ej17-0363] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Placental growth factor (PlGF), a member of the vascular endothelial growth factor (VEGF) sub-family, plays a major role in angiogenesis and vasculogenesis. Previous study demonstrated that PlGF-overexpressing transgenic (Tg) mice had gestational loss. In addition, PlGF secretion was up-regulated in isolated T lymphocytes (T-cell) upon CD3/CD28 stimulation, suggesting that PlGF could be a regulator of T-cell differentiation and development. T-cells are well known to play a critical role in obesity-induced inflammation. Therefore, to verify the possible link of diet-induced obesity (DIO) with inflammation and related metabolic disorders, such as insulin resistance, we fed high-fat diet (HFD) to Tg mice for 16 weeks. Adiposity and glucose intolerance significantly increase in Tg mice fed a HFD (Tg HFD) compared to wild-type (WT) mice fed HFD (WT HFD). In addition, macrophage infiltrations were significantly higher in the epididymal white adipose tissue (EWAT), liver, and pancreatic islets of Tg HFD mice compared to WT HFD mice. In the in vitro study, we showed that isolated CD4+ T-cells from Tg mice further differentiate into type 1 (Th1) and type 17 (Th17) helper T-cells via CD3/CD28 stimulation. Furthermore, we observed that the pro-inflammatory cytokines IL-6, IL-17, and TNFα, are remarkably increased in Tg mice compared to WT mice. These findings demonstrate that PlGF overexpression in T-cells might lead to inflammatory T-cell differentiation and accumulation in adipose tissue (AT) or metabolism-related tissues, contributing to the development of systemic metabolic disorders. Thus, PlGF may provide an effective therapeutic target in the management of obesity-induced inflammation and related metabolic disorders.
Collapse
Affiliation(s)
- Mincheol Kang
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, 41566, Republic of Korea
| | - Jain Jeong
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, 41566, Republic of Korea
| | - Jinhee Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, 41566, Republic of Korea
| | - Song Park
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, 41566, Republic of Korea
| | - Yonghun Sung
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, 41566, Republic of Korea
| | - Minjee Choi
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, 41566, Republic of Korea
| | - Wookbong Kwon
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, 41566, Republic of Korea
| | - Soyoung Jang
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, 41566, Republic of Korea
| | - Kwang Shik Choi
- College of Natural Science, Kyungpook National University, 41566, Republic of Korea
| | - Yeon Sik Choo
- College of Natural Science, Kyungpook National University, 41566, Republic of Korea
| | - Duhak Yoon
- Department of Animal Science, Kyungpook National University, 37224, Republic of Korea
| | - Myoung Ok Kim
- School of Animal Biotechnology (BT) Science, Kyungpook National University, 37224, Republic of Korea
| | - Zae Young Ryoo
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, 41566, Republic of Korea
| |
Collapse
|
170
|
Del Cornò M, D'Archivio M, Conti L, Scazzocchio B, Varì R, Donninelli G, Varano B, Giammarioli S, De Meo S, Silecchia G, Pennestrì F, Persiani R, Masella R, Gessani S. Visceral fat adipocytes from obese and colorectal cancer subjects exhibit distinct secretory and ω6 polyunsaturated fatty acid profiles and deliver immunosuppressive signals to innate immunity cells. Oncotarget 2018; 7:63093-63105. [PMID: 27494857 PMCID: PMC5325349 DOI: 10.18632/oncotarget.10998] [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] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/22/2016] [Indexed: 01/16/2023] Open
Abstract
Obesity is a low-grade chronic inflammatory state representing an important risk factor for colorectal cancer (CRC). Adipocytes strongly contribute to inflammation by producing inflammatory mediators. In this study we investigated the role of human visceral fat adipocytes in regulating the functions of innate immunity cells. Adipocyte-conditioned media (ACM) from obese (n = 14) and CRC (lean, n = 14; obese, n = 13) subjects released higher levels of pro-inflammatory/immunoregulatory factors as compared to ACM from healthy lean subjects (n = 13). Dendritic cells (DC), differentiated in the presence of ACM from obese and CRC subjects, expressed elevated levels of the inhibitory molecules PD-L1 and PD-L2, and showed a reduced IL-12/IL-10 ratio in response to both TLR ligand- and γδ T lymphocyte-induced maturation. Furthermore, CRC patient-derived ACM inhibited DC-mediated γδ T cell activation. The immunosuppressive signals delivered by ACM from obese and CRC individuals were associated with a pro-inflammatory secretory and ω6 polyunsaturated fatty acid profile of adipocytes. Interestingly, STAT3 activation in adipocytes correlated with dihomo-γlinolenic acid content and was further induced by arachidonic acid, which conversely down-modulated PPARγ. These results provide novel evidence for a cross-talk between human adipocytes and innate immunity cells whose alteration in obesity and CRC may lead to immune dysfunctions, thus setting the basis for cancer development.
Collapse
Affiliation(s)
- Manuela Del Cornò
- Departments of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Massimo D'Archivio
- Departments of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità, Rome, Italy
| | - Lucia Conti
- Departments of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Beatrice Scazzocchio
- Departments of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità, Rome, Italy
| | - Rosaria Varì
- Departments of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità, Rome, Italy
| | - Gloria Donninelli
- Departments of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Barbara Varano
- Departments of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Stefania Giammarioli
- Departments of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità, Rome, Italy
| | - Simone De Meo
- Departments of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Gianfranco Silecchia
- Department of Medical-Surgical Sciences and Biotecnologies, Sapienza University of Rome, Rome, Italy
| | | | | | - Roberta Masella
- Departments of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità, Rome, Italy
| | - Sandra Gessani
- Departments of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
171
|
Repeated Binge-Like Alcohol Intoxication: Depot-Specific Adipose Tissue Immuno-Metabolic Dysregulation. Shock 2018; 48:243-250. [PMID: 28125531 DOI: 10.1097/shk.0000000000000843] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Repeated binge-like alcohol intoxication (RBAI) induces whole-body insulin resistance, which is predicted to increase the risk for metabolic syndrome and type 2 diabetes. Previously, we showed that acute alcohol intoxication increases mesenteric lymphatic permeability, perilymphatic adipose tissue (PLAT) inflammation, and circulating lipopolysaccharide levels in rats. We hypothesize that mesenteric lymphatic hyperpermeability, adipose tissue inflammation and associated dysregulated adipokine expression, and insulin signaling are central mechanisms underlying whole-body metabolic dysregulation resulting from RBAI. To test this hypothesis, male Sprague-Dawley rats surgically fitted with an intragastric catheter received a bolus of 2.5 g/kg/day of alcohol (12.5% alcohol w/v) or isocaloric dextrose in Vanilla Ensure (116 kcal/kg/day) for 3 days. Mesenteric lymphatic permeability, mesenteric (MFAT = PLAT) and subcutaneous (SFAT) adipose tissue inflammatory milieu, circulating adipokines, and markers of insulin responsiveness (pAKT and PTP1B protein expression) were determined following the last alcohol/dextrose administration. RBAI resulted in increased lymphatic permeability, MFAT-specific expression of inflammatory cytokines and markers of inflammatory cells (macrophages, dendritic, and T cells), decreased circulating adiponectin and visfatin levels, and MFAT-specific attenuation of insulin-stimulated protein kinase B phosphorylation (Ser) compared with dextrose-treated control animals. These results suggest that RBAI-induced mesenteric lymphatic hyperpermeability promotes inflammatory milieu, decreased insulin-sensitizing adipokines, and impaired insulin signaling in MFAT, which we propose may be an early event preceding systemic metabolic dysregulation. We speculate that RBAI-induced increase in gut-derived toxins, promoting lymphatic leak, and MFAT inflammatory milieu are mechanisms deserving further investigation to elucidate lymphatic-MFAT crosstalk events that precede and predispose for alcohol-induced insulin resistance.
Collapse
|
172
|
JAZF1 Inhibits Adipose Tissue Macrophages and Adipose Tissue Inflammation in Diet-Induced Diabetic Mice. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4507659. [PMID: 29765984 PMCID: PMC5885486 DOI: 10.1155/2018/4507659] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/19/2017] [Accepted: 12/28/2017] [Indexed: 12/26/2022]
Abstract
Background Juxtaposed with another zinc finger gene 1 (JAZF1) affects gluconeogenesis, insulin sensitivity, lipid metabolism, and inflammation, but its exact role in chronic inflammation remains unclear. This study aimed to examine JAZF1 overexpression in vivo on adipose tissue macrophages (ATMs). Methods Mouse models of high-fat diet- (HFD-) induced insulin resistance were induced using C57BL/6J and JAZF1-overexpressing (JAZF1-OX) mice. The mice were randomized (8–10/group) to C57BL/6J mice fed regular diet (RD) (NC group), C57BL/6J mice fed HFD (HF group), JAZF1-OX mice fed RD (NJ group), and JAZF1-OX mice fed HFD (HJ group). Adipose tissue was harvested 12 weeks later. ATMs were evaluated by flow cytometry. Inflammatory markers were evaluated by ELISA. Results JAZF1-OX mice had lower blood lipids, blood glucose, body weight, fat weight, and inflammatory markers compared with HF mice (all P < 0.05). JAZF1 overexpression decreased ATM number and secretion of proinflammatory cytokines. JAZF1 overexpression decreased total CD4+ T cells, active T cells, and memory T cells and increased Treg cells. JAZF1 overexpression downregulated IFN-γ and IL-17 levels and upregulated IL-4 levels. JAZF1 overexpression decreased MHCII, CD40, and CD86 in total ATM, CD11c+ ATM, and CD206+ ATM. Conclusions JAZF1 limits adipose tissue inflammation by limiting macrophage populations and restricting their antigen presentation function.
Collapse
|
173
|
Abstract
Obesity-induced adipose tissue inflammation is regulated by various immune cells for innate and adaptive immunity. Among adipose tissue immune cells, it has been proposed that invariant Natural Killer T (iNKT) cells play crucial roles in anti-inflammatory responses in obesity. iNKT cells recognize 'lipid' antigens loaded on CD1d of antigen presenting cells and modulate immune responses by secreting Th1 or Th2 type cytokines depending on species of lipid antigens, antigen presenting cell types, and environmental cytokine milieu. However, the regulatory mechanisms of antigen presenting cells for adipose iNKT cell stimulation have not been clearly elucidated. Recently, we have reported that CD1d expressing adipocytes could act as an antigen presenting cell for adipose iNKT cells by characterization of adipocyte-specific CD1d knockout (CD1dADKO) mice. Upon high-fat diet (HFD) feeding, CD1dADKO mice aggravated adipose tissue inflammation and insulin resistance compared with CD1df/f mice. In this commentary, we provide the additional data of adipocyte CD1d-dependent regulation of adipose iNKT cell responses as well as systemic insulin sensitivity. In addition, we discuss how the interaction between adipocytes and iNKT cells would be regulated with the progression of obesity.
Collapse
Affiliation(s)
- Jin Young Huh
- Department of Biological Science, Institute of Molecular Biology & Genetics, Seoul National University, Seoul, South Korea
- Department of Medicine, University of California San Diego, San Diego, California, USA
| | - Yoon Jeong Park
- Department of Biological Science, Institute of Molecular Biology & Genetics, Seoul National University, Seoul, South Korea
- Department of Biophysics and Chemical Biology, Seoul National University, Seoul, South Korea
| | - Jae Bum Kim
- Department of Biological Science, Institute of Molecular Biology & Genetics, Seoul National University, Seoul, South Korea
- Department of Biophysics and Chemical Biology, Seoul National University, Seoul, South Korea
| |
Collapse
|
174
|
Abstract
In this issue of Cell Metabolism, Macdougall et al. (2018) identify two subsets of conventional dendritic cells in visceral adipose tissue and demonstrate that these subsets engage distinct adipocyte-associated signaling pathways to drive their tolerogenic phenotypes in the lean state.
Collapse
Affiliation(s)
- Nelson M LaMarche
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lydia Lynch
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
175
|
Macdougall CE, Wood EG, Loschko J, Scagliotti V, Cassidy FC, Robinson ME, Feldhahn N, Castellano L, Voisin MB, Marelli-Berg F, Gaston-Massuet C, Charalambous M, Longhi MP. Visceral Adipose Tissue Immune Homeostasis Is Regulated by the Crosstalk between Adipocytes and Dendritic Cell Subsets. Cell Metab 2018; 27. [PMID: 29514067 PMCID: PMC5846800 DOI: 10.1016/j.cmet.2018.02.007] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Visceral adipose tissue (VAT) has multiple roles in orchestrating whole-body energy homeostasis. In addition, VAT is now considered an immune site harboring an array of innate and adaptive immune cells with a direct role in immune surveillance and host defense. We report that conventional dendritic cells (cDCs) in VAT acquire a tolerogenic phenotype through upregulation of pathways involved in adipocyte differentiation. While activation of the Wnt/β-catenin pathway in cDC1 DCs induces IL-10 production, upregulation of the PPARγ pathway in cDC2 DCs directly suppresses their activation. Combined, they promote an anti-inflammatory milieu in vivo delaying the onset of obesity-induced chronic inflammation and insulin resistance. Under long-term over-nutrition, changes in adipocyte biology curtail β-catenin and PPARγ activation, contributing to VAT inflammation.
Collapse
Affiliation(s)
- Claire E Macdougall
- William Harvey Research Institute, Barts, and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Elizabeth G Wood
- William Harvey Research Institute, Barts, and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Jakob Loschko
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Valeria Scagliotti
- William Harvey Research Institute, Barts, and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Féaron C Cassidy
- William Harvey Research Institute, Barts, and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Mark E Robinson
- Centre for Haematology, Department of Medicine, Imperial College London, W12 0NN London, UK; Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), Hammersmith Hospital, London W12 0NN, UK
| | - Niklas Feldhahn
- Centre for Haematology, Department of Medicine, Imperial College London, W12 0NN London, UK
| | - Leandro Castellano
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), Hammersmith Hospital, London W12 0NN, UK
| | - Mathieu-Benoit Voisin
- William Harvey Research Institute, Barts, and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Federica Marelli-Berg
- William Harvey Research Institute, Barts, and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Carles Gaston-Massuet
- William Harvey Research Institute, Barts, and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Marika Charalambous
- William Harvey Research Institute, Barts, and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - M Paula Longhi
- William Harvey Research Institute, Barts, and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK.
| |
Collapse
|
176
|
|
177
|
Sittipo P, Lobionda S, Lee YK, Maynard CL. Intestinal microbiota and the immune system in metabolic diseases. J Microbiol 2018; 56:154-162. [PMID: 29492872 DOI: 10.1007/s12275-018-7548-y] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/05/2018] [Accepted: 02/12/2018] [Indexed: 12/29/2022]
Abstract
The intestinal microbiota is comprised of millions of microorganisms that reside in the gastrointestinal tract and consistently interact with the host. Host factors such as diet and disease status affect the composition of the microbiota, while the microbiota itself produces metabolites that can further manipulate host physiology. Dysbiosis of the intestinal microbiota has been characterized in patients with certain metabolic diseases, some of which involve damage to the host intestinal epithelial barrier and alterations in the immune system. In this review, we will discuss the consequences of dietdependent bacterial dysbiosis in the gastrointestinal tract, and how the associated interaction with epithelial and immune cells impacts metabolic diseases.
Collapse
Affiliation(s)
- Panida Sittipo
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Stefani Lobionda
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Yun Kyung Lee
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea.
| | - Craig L Maynard
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
178
|
Abdel-Moneim A, Bakery HH, Allam G. The potential pathogenic role of IL-17/Th17 cells in both type 1 and type 2 diabetes mellitus. Biomed Pharmacother 2018; 101:287-292. [PMID: 29499402 DOI: 10.1016/j.biopha.2018.02.103] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/13/2018] [Accepted: 02/22/2018] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus (DM) is a serious medical problem affecting millions of peoples worldwide, and has a great socio-economic impacts. Cytokines possess a pivotal role in modulation of immune reactions and disease pathogenesis. T-helper type 17 (Th17) cells, an important proinflammatory CD4+ T cell subset secreting interleukin 17 (IL-17), has been embroiled in development of DM. There are recent evidences supporting a definitive role of Th17 cells in the etiology of type 1 diabetes (T1D). In addition, IL-17 has been shown to play a crucial role in inflammation, insulin resistance, and type 2 diabetes (T2D). Recently, small molecules which have been specified to block Th17 cells differentiation are considered as potential therapeutics for the disease. Anti-IL-17 neutralizing antibodies and/or antibodies targeting Th17 cells have been investigated to protect individuals at risk from disease development. In this review we aimed to shed light on the potential role of IL-17 and Th17 cells in both T1D and T2D pathogenesis and future therapeutic strategies.
Collapse
Affiliation(s)
- Adel Abdel-Moneim
- Physiology Division, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Heba H Bakery
- Physiology Division, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Gamal Allam
- Immunology Division, Department of Microbiology, College of Medicine, Taif University, Taif, Saudi Arabia; Immunology Division, Department of Zoology, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt.
| |
Collapse
|
179
|
Ivanov S, Merlin J, Lee MKS, Murphy AJ, Guinamard RR. Biology and function of adipose tissue macrophages, dendritic cells and B cells. Atherosclerosis 2018; 271:102-110. [PMID: 29482037 DOI: 10.1016/j.atherosclerosis.2018.01.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/22/2017] [Accepted: 01/12/2018] [Indexed: 12/20/2022]
Abstract
The increasing incidence of obesity and its socio-economical impact is a global health issue due to its associated co-morbidities, namely diabetes and cardiovascular disease [1-5]. Obesity is characterized by an increase in adipose tissue, which promotes the recruitment of immune cells resulting in low-grade inflammation and dysfunctional metabolism. Macrophages are the most abundant immune cells in the adipose tissue of mice and humans. The adipose tissue also contains other myeloid cells (dendritic cells (DC) and neutrophils) and to a lesser extent lymphocyte populations, including T cells, B cells, Natural Killer (NK) and Natural Killer T (NKT) cells. While the majority of studies have linked adipose tissue macrophages (ATM) to the development of low-grade inflammation and co-morbidities associated with obesity, emerging evidence suggests for a role of other immune cells within the adipose tissue that may act in part by supporting macrophage homeostasis. In this review, we summarize the current knowledge of the functions ATMs, DCs and B cells possess during steady-state and obesity.
Collapse
Affiliation(s)
- Stoyan Ivanov
- INSERM U1065, Mediterranean Center of Molecular Medicine, University of Nice Sophia-Antipolis, Faculty of Medicine, Nice, France.
| | - Johanna Merlin
- INSERM U1065, Mediterranean Center of Molecular Medicine, University of Nice Sophia-Antipolis, Faculty of Medicine, Nice, France
| | - Man Kit Sam Lee
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Andrew J Murphy
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Rodolphe R Guinamard
- INSERM U1065, Mediterranean Center of Molecular Medicine, University of Nice Sophia-Antipolis, Faculty of Medicine, Nice, France.
| |
Collapse
|
180
|
Trim W, Turner JE, Thompson D. Parallels in Immunometabolic Adipose Tissue Dysfunction with Ageing and Obesity. Front Immunol 2018; 9:169. [PMID: 29479350 PMCID: PMC5811473 DOI: 10.3389/fimmu.2018.00169] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/19/2018] [Indexed: 12/12/2022] Open
Abstract
Ageing, like obesity, is often associated with alterations in metabolic and inflammatory processes resulting in morbidity from diseases characterised by poor metabolic control, insulin insensitivity, and inflammation. Ageing populations also exhibit a decline in immune competence referred to as immunosenescence, which contributes to, or might be driven by chronic, low-grade inflammation termed "inflammageing". In recent years, animal and human studies have started to uncover a role for immune cells within the stromal fraction of adipose tissue in driving the health complications that come with obesity, but relatively little work has been conducted in the context of immunometabolic adipose function in ageing. It is now clear that aberrant immune function within adipose tissue in obesity-including an accumulation of pro-inflammatory immune cell populations-plays a major role in the development of systemic chronic, low-grade inflammation, and limiting the function of adipocytes leading to an impaired fat handling capacity. As a consequence, these changes increase the chance of multiorgan dysfunction and disease onset. Considering the important role of the immune system in obesity-associated metabolic and inflammatory diseases, it is critically important to further understand the interplay between immunological processes and adipose tissue function, establishing whether this interaction contributes to age-associated immunometabolic dysfunction and inflammation. Therefore, the aim of this article is to summarise how the interaction between adipose tissue and the immune system changes with ageing, likely contributing to the age-associated increase in inflammatory activity and loss of metabolic control. To understand the potential mechanisms involved, parallels will be drawn to the current knowledge derived from investigations in obesity. We also highlight gaps in research and propose potential future directions based on the current evidence.
Collapse
Affiliation(s)
- William Trim
- Department for Health, University of Bath, Bath, United Kingdom
| | - James E Turner
- Department for Health, University of Bath, Bath, United Kingdom
| | - Dylan Thompson
- Department for Health, University of Bath, Bath, United Kingdom
| |
Collapse
|
181
|
Tang CL, Liu ZM, Gao YR, Xiong F. Schistosoma Infection and Schistosoma-Derived Products Modulate the Immune Responses Associated with Protection against Type 2 Diabetes. Front Immunol 2018; 8:1990. [PMID: 29387059 PMCID: PMC5776330 DOI: 10.3389/fimmu.2017.01990] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 12/21/2017] [Indexed: 12/27/2022] Open
Abstract
Studies on parasite-induced immunoregulatory mechanisms could contribute to the development of new therapies for inflammatory diseases such as type 2 diabetes (T2D), which is a chronic inflammatory disease characterized by persistent elevated glucose levels due to insulin resistance. The association between previous Schistosoma infection and T2D has been confirmed—Schistosoma infection and Schistosoma-derived products modulate the immune system, including innate and acquired immune responses, contributing to T2D disease control. Schistosoma infections and Schistosoma-derived molecules affect the immune cell composition in adipose tissue, dampening inflammation and improving glucose tolerance. This protective role includes the polarization of immune cells to alternatively activated macrophages, dendritic cells, eosinophils, and group 2 innate lymphoid cells. Furthermore, Schistosoma infection and Schistosoma products are effective for the treatment of T2D, as they increase the number of type 2 helper T cells (Th2) and regulatory T cells (Tregs) and decrease type 1 helper T cells (Th1) and type 17 helper T cells (Th17) cells. Thus, our aim was to comprehensively review the mechanism through which Schistosoma infection and Schistosoma products modulate the immune response against T2D.
Collapse
Affiliation(s)
- Chun-Lian Tang
- Department of Science and Education, Wuchang Hospital, Wuhan, China
| | - Zhi-Ming Liu
- Department of Science and Education, Wuchang Hospital, Wuhan, China
| | - Yan Ru Gao
- Medical Department, City College, Wuhan University of Science and Technology, Wuhan, China
| | - Fei Xiong
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
182
|
Abstract
Adipose tissue represents a large volume of biologically active tissue that exerts substantial systemic effects in health and disease. Alcohol consumption can profoundly disturb the normal functions of adipose tissue by inducing adipocyte death and altering secretion of adipokines, pro-inflammatory mediators and free fatty acids from adipose tissue, which have important direct and indirect effects on the pathogenesis of alcoholic liver disease (ALD). Cessation of alcohol intake quickly reverses inflammatory changes in adipose tissue, and pharmacological treatment that normalizes adipose tissue function improves experimental ALD. Obesity exacerbates liver injury induced by chronic or binge alcohol consumption, and obesity and alcohol can synergize to increase risk of ALD and progression. Physicians who care for individuals with ALD should be aware of the effects of adipose tissue dysfunction on liver function, and consider strategies to manage obesity and insulin resistance. This Review examines the effect of alcohol on adiposity and adipose tissue and the relationship between alcohol, adipose tissue and the liver.
Collapse
|
183
|
Andreone L, Gimeno ML, Perone MJ. Interactions Between the Neuroendocrine System and T Lymphocytes in Diabetes. Front Endocrinol (Lausanne) 2018; 9:229. [PMID: 29867762 PMCID: PMC5966545 DOI: 10.3389/fendo.2018.00229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 04/20/2018] [Indexed: 12/16/2022] Open
Abstract
It is well established that there is a fine-tuned bidirectional communication between the immune and neuroendocrine tissues in maintaining homeostasis. Several types of immune cells, hormones, and neurotransmitters of different chemical nature are involved as communicators between organs. Apart of being key players of the adaptive arm of the immune system, it has been recently described that T lymphocytes are involved in the modulation of metabolism of several tissues in health and disease. Diabetes may result mainly from lack of insulin production (type 1 diabetes) or insufficient insulin and insulin resistance (type 2 diabetes), both influenced by genetic and environmental components. Herein, we discuss accumulating data regarding the role of the adaptive arm of the immune system in the pathogenesis of diabetes; including the action of several hormones and neurotransmitters influencing on central and peripheral T lymphocytes development and maturation, particularly under the metabolic burden triggered by diabetes. In addition, we comment on the role of T-effector lymphocytes in adipose and liver tissues during diabetes, which together enhances pancreatic β-cell stress aggravating the disease.
Collapse
|
184
|
Integrated Immunomodulatory Mechanisms through which Long-Chain n-3 Polyunsaturated Fatty Acids Attenuate Obese Adipose Tissue Dysfunction. Nutrients 2017; 9:nu9121289. [PMID: 29186929 PMCID: PMC5748740 DOI: 10.3390/nu9121289] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/14/2017] [Accepted: 11/16/2017] [Indexed: 12/13/2022] Open
Abstract
Obesity is a global health concern with rising prevalence that increases the risk of developing other chronic diseases. A causal link connecting overnutrition, the development of obesity and obesity-associated co-morbidities is visceral adipose tissue (AT) dysfunction, characterized by changes in the cellularity of various immune cell populations, altered production of inflammatory adipokines that sustain a chronic state of low-grade inflammation and, ultimately, dysregulated AT metabolic function. Therefore, dietary intervention strategies aimed to halt the progression of obese AT dysfunction through any of the aforementioned processes represent an important active area of research. In this connection, fish oil-derived dietary long-chain n-3 polyunsaturated fatty acids (PUFA) in the form of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have been demonstrated to attenuate obese AT dysfunction through multiple mechanisms, ultimately affecting AT immune cellularity and function, adipokine production, and metabolic signaling pathways, all of which will be discussed herein.
Collapse
|
185
|
Schindler TI, Wagner JJ, Goedicke-Fritz S, Rogosch T, Coccejus V, Laudenbach V, Nikolaizik W, Härtel C, Maier RF, Kerzel S, Zemlin M. T H17 Cell Frequency in Peripheral Blood Is Elevated in Overweight Children without Chronic Inflammatory Diseases. Front Immunol 2017; 8:1543. [PMID: 29201026 PMCID: PMC5696328 DOI: 10.3389/fimmu.2017.01543] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 10/30/2017] [Indexed: 12/23/2022] Open
Abstract
Background The prevalence of obesity has dramatically increased in children in the last few decades and is associated with chronic inflammatory diseases. Fat tissue produces IL-6 and TNF-α, which are stimuli for TH17 cell differentiation. These cells are characterized by expression of the transcription factor receptor-related orphan receptor C (RORC) and by IL-17A production. In murine models, obesity has been linked with elevated TH17 cell frequencies. The aim of this study was to explore whether being overweight was associated with an elevated frequency of circulating TH17 cells or elevated messenger RNA (mRNA)-levels of IL-17A and RORC in children without chronic inflammatory diseases. Methods We studied peripheral blood samples from 15 overweight and 50 non-overweight children without a history of autoimmune diseases, asthma, atopic dermatitis or allergic rhinoconjunctivitis. TH17 cells were quantified in Ionomycin stimulated peripheral blood mononuclear cells by flow cytometry using intracellular IL-17A staining. RORC- and IL-17A expressions were measured by real-time PCR. Results We found significantly elevated TH cell frequencies in overweight children compared then on-overweight controls with 34.7 ± 1.5% of CD3+CD4+ cells versus 25.4 ± 2.4% (mean ± SEM, p = 0.0023), respectively. Moreover, TH cell frequencies correlated positively with body mass index (r = 0.42, p = 0.0005, respectively). The relative mRNA expression of RORC (p = 0.013) and IL-17A (p = 0.014) were upregulated in overweight compared to non-overweight children. Conclusion Childhood obesity is an independent factor that is associated with an elevated frequency of circulating TH17 cells and higher expression of RORC- and IL-17A-mRNA after in vitro stimulation with Ionomycin. This might be due to the inflammatory activity of the fat tissue. Studies on TH17 immunity should not only be adjusted for acute and chronic inflammatory diseases but also for overweight.
Collapse
Affiliation(s)
| | - Johanna-Josophina Wagner
- Children's Hospital, Philipps University, Marburg, Germany.,Department of Pediatrics, Helios-Klinikum Buch, Berlin, Germany
| | - Sybelle Goedicke-Fritz
- Children's Hospital, Philipps University, Marburg, Germany.,Department of General Pediatrics and Neonatology, University Children's Hospital of Saarland, Homburg, Germany
| | - Tobias Rogosch
- Children's Hospital, Philipps University, Marburg, Germany
| | | | | | | | | | | | - Sebastian Kerzel
- Department of Pediatric Pneumology and Allergy, University Children's Hospital Regensburg, Regensburg, Germany
| | - Michael Zemlin
- Children's Hospital, Philipps University, Marburg, Germany.,Department of General Pediatrics and Neonatology, University Children's Hospital of Saarland, Homburg, Germany
| |
Collapse
|
186
|
Aravindhan V, Anand G. Cell Type-Specific Immunomodulation Induced by Helminthes: Effect on Metainflammation, Insulin Resistance and Type-2 Diabetes. Am J Trop Med Hyg 2017; 97:1650-1661. [PMID: 29141759 DOI: 10.4269/ajtmh.17-0236] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Recent epidemiological studies have documented an inverse relationship between the decreasing prevalence of helminth infections and the increasing prevalence of metabolic diseases ("metabolic hygiene hypothesis"). Chronic inflammation leading to insulin resistance (IR) has now been identified as a major etiological factor for a variety of metabolic diseases other than obesity and Type-2 diabetes (metainflammation). One way by which helminth infections such as filariasis can modulate IR is by inducing a chronic, nonspecific, low-grade, immune suppression mediated by modified T-helper 2 (Th2) response (induction of both Th2 and regulatory T cells) which can in turn suppress the proinflammatory responses and promote insulin sensitivity (IS). This article provides evidence on how the cross talk between the innate and adaptive arms of the immune responses can modulate IR/sensitivity. The cross talk between innate (macrophages, dendritic cells, natural killer cells, natural killer T cells, myeloid derived suppressor cells, innate lymphoid cells, basophils, eosinophils, and neutrophils) and adaptive (helper T [CD4+] cells, cytotoxic T [CD8+] cells and B cells) immune cells forms two opposing circuits, one associated with IR and the other associated with IS under the conditions of metabolic syndrome and helminth-mediated immunomodulation, respectively.
Collapse
|
187
|
Zhang J, Zhang L, Zhang S, Yu Q, Xiong F, Huang K, Wang CY, Yang P. HMGB1, an innate alarmin, plays a critical role in chronic inflammation of adipose tissue in obesity. Mol Cell Endocrinol 2017; 454:103-111. [PMID: 28619625 DOI: 10.1016/j.mce.2017.06.012] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/17/2017] [Accepted: 06/12/2017] [Indexed: 12/12/2022]
Abstract
Obesity has emerged as an imminent global public health concern over the past several decades. It has now become evident that obesity is characterized by the persistent and low-grade inflammation in the adipose tissue, and serves as an independent risk factor for many metabolic disorders such as diabetes and cardiovascular disease. Particularly, adipocytes originated from obese mice and humans likely predominate necrosis upon stressful insults, leading to passive release of cellular contents including the high mobility group box 1 (HMGB1) into the extracellular milieu. Extracellular HMGB1 acts as an innate alarmin to stimulate the activation of resident immune cells in the adipose tissue. Upon activation, those resident immune cells actively secrete additional HMGB1, which in turn activates/recruits additional immune cells, and induces adipocyte death. This review summarizes those novel discoveries in terms of HMGB1 in the initiation and maintenance of chronic inflammatory state in adipose tissue in obesity, and discusses its potential application in clinical settings.
Collapse
Affiliation(s)
- Jing Zhang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Lei Zhang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Shu Zhang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Qilin Yu
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Fei Xiong
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Kun Huang
- Tongji School of Pharmacy, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Cong-Yi Wang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China.
| | - Ping Yang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, 1095 Jiefang Ave., Wuhan, 430030, China.
| |
Collapse
|
188
|
Liu B, Yu H, Sun G, Sun X, Jin H, Zhang C, Shi W, Tian D, Liu K, Xu H, Li X, Yin J, Hong X, Zhang D. OX40 promotes obesity-induced adipose inflammation and insulin resistance. Cell Mol Life Sci 2017; 74:3827-3840. [PMID: 28612217 PMCID: PMC11107569 DOI: 10.1007/s00018-017-2552-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 05/28/2017] [Accepted: 05/30/2017] [Indexed: 12/14/2022]
Abstract
Adaptive immunity plays a critical role in IR and T2DM development; however, the biological mechanisms linking T cell costimulation and glucose metabolism have not been fully elucidated. In this study, we demonstrated that the costimulatory molecule OX40 controls T cell activation and IR development. Inflammatory cell accumulation and enhanced proinflammatory gene expression, as well as high OX40 expression levels on CD4+ T cells, were observed in the adipose tissues of mice with diet-induced obesity. OX40-KO mice exhibited significantly less weight gain and lower fasting glucose levels than those of WT mice, without obvious adipose tissue inflammation. The effects of OX40 on IR are mechanistically linked to the promotion of T cell activation, Th1 cell differentiation and proliferation-as well as the attenuation of Treg suppressive activity and the enhancement of proinflammatory cytokine production-in adipose tissues. Furthermore, OX40 expression on T cells was positively associated with obesity in humans, suggesting that our findings are clinically relevant. In summary, our study revealed that OX40 in CD4+ T cells is crucial for adipose tissue inflammation and IR development. Therefore, the OX40 signaling pathway may be a new target for preventing or treating obesity-related IR and T2DM.
Collapse
Affiliation(s)
- Bing Liu
- Endocrinology Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People's Republic of China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
| | - Hengchi Yu
- Endocrinology Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People's Republic of China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
| | - Guangyong Sun
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
- Beijing Clinical Research Institute, Beijing, 100050, People's Republic of China
| | - Xiaojing Sun
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
- Beijing Clinical Research Institute, Beijing, 100050, People's Republic of China
| | - Hua Jin
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
- Beijing Clinical Research Institute, Beijing, 100050, People's Republic of China
| | - Chunpan Zhang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
- Beijing Clinical Research Institute, Beijing, 100050, People's Republic of China
| | - Wen Shi
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
- Beijing Clinical Research Institute, Beijing, 100050, People's Republic of China
| | - Dan Tian
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
- Beijing Clinical Research Institute, Beijing, 100050, People's Republic of China
| | - Kai Liu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
- Beijing Clinical Research Institute, Beijing, 100050, People's Republic of China
| | - Hufeng Xu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
- Beijing Clinical Research Institute, Beijing, 100050, People's Republic of China
| | - Xinmin Li
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
- Beijing Clinical Research Institute, Beijing, 100050, People's Republic of China
| | - Jie Yin
- Endocrinology Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People's Republic of China
| | - Xu Hong
- Endocrinology Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People's Republic of China.
| | - Dong Zhang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China.
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China.
- Beijing Clinical Research Institute, Beijing, 100050, People's Republic of China.
| |
Collapse
|
189
|
Hannibal TD, Schmidt-Christensen A, Nilsson J, Fransén-Pettersson N, Hansen L, Holmberg D. Deficiency in plasmacytoid dendritic cells and type I interferon signalling prevents diet-induced obesity and insulin resistance in mice. Diabetologia 2017; 60:2033-2041. [PMID: 28660492 PMCID: PMC6448810 DOI: 10.1007/s00125-017-4341-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/19/2017] [Indexed: 12/30/2022]
Abstract
AIMS/HYPOTHESIS Obesity is associated with glucose intolerance and insulin resistance and is closely linked to the increasing prevalence of type 2 diabetes. In mouse models of diet-induced obesity (DIO) and type 2 diabetes, an increased fat intake results in adipose tissue expansion and the secretion of proinflammatory cytokines. The innate immune system not only plays a crucial role in obesity-associated chronic low-grade inflammation but it is also proposed to play a role in modulating energy metabolism. However, little is known about how the modulation of metabolism by the immune system may promote increased adiposity in the early stages of increased dietary intake. Here we aimed to define the role of type I IFNs in DIO and insulin resistance. METHODS Mice lacking the receptor for IFN-α (IFNAR-/-) and deficient in plasmacytoid dendritic cells (pDCs) (B6.E2-2 fl/fl .Itgax-cre) were fed a diet with a high fat content or normal chow. The mice were analysed in vivo and in vitro using cellular, biochemical and molecular approaches. RESULTS We found that the development of obesity was inhibited by an inability to respond to type I IFNs. Furthermore, the development of obesity and insulin resistance in this model was associated with pDC recruitment to the fatty tissues and liver of obese mice (a 4.3-fold and 2.7-fold increase, respectively). Finally, we demonstrated that the depletion of pDCs protects mice from DIO and from developing obesity-associated metabolic complications. CONCLUSIONS/INTERPRETATION Our results provide genetic evidence that pDCs, via type I IFNs, regulate energy metabolism and promote the development of obesity.
Collapse
Affiliation(s)
- Tine D Hannibal
- Department of Experimental Medical Sciences, Lund University, Biomedical Center, CRC, 205 02, Malmö, Sweden
- Department of Immunology & Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anja Schmidt-Christensen
- Department of Experimental Medical Sciences, Lund University, Biomedical Center, CRC, 205 02, Malmö, Sweden
| | - Julia Nilsson
- Department of Experimental Medical Sciences, Lund University, Biomedical Center, CRC, 205 02, Malmö, Sweden
| | - Nina Fransén-Pettersson
- Department of Experimental Medical Sciences, Lund University, Biomedical Center, CRC, 205 02, Malmö, Sweden
- Department of Immunology & Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lisbeth Hansen
- Department of Experimental Medical Sciences, Lund University, Biomedical Center, CRC, 205 02, Malmö, Sweden
- Department of Immunology & Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dan Holmberg
- Department of Experimental Medical Sciences, Lund University, Biomedical Center, CRC, 205 02, Malmö, Sweden.
- Department of Immunology & Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
190
|
Becker M, Levings MK, Daniel C. Adipose-tissue regulatory T cells: Critical players in adipose-immune crosstalk. Eur J Immunol 2017; 47:1867-1874. [PMID: 28849586 DOI: 10.1002/eji.201646739] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/13/2017] [Accepted: 08/24/2017] [Indexed: 12/19/2022]
Abstract
Obesity and type-2 diabetes (T2D) are associated with metabolic defects and inflammatory processes in fat depots. FoxP3+ regulatory T cells (Tregs) control immune tolerance, and have an important role in controlling tissue-specific inflammation. In this mini-review we will discuss current insights into how cross-talk between T cells and adipose tissue shapes the inflammatory environment in obesity-associated metabolic diseases, focusing on the role of CD4+ T cells and Tregs. We will also highlight potential opportunities for how the immunoregulatory properties of Tregs could be harnessed to control inflammation in obesity and T2D and emphasize the critical need for more research on humans to establish mechanisms that are conserved in both mice and humans.
Collapse
Affiliation(s)
- Maike Becker
- Institute for Diabetes Research, Research Group Immune Tolerance in Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, Munich, Germany.,Deutsches Zentrum für Diabetesforschung (DZD), Munich, Germany
| | - Megan K Levings
- Department of Surgery University of British Columbia and BC Children's Hospital Research Institute, Vancouver, B.C. Canada
| | - Carolin Daniel
- Institute for Diabetes Research, Research Group Immune Tolerance in Diabetes, Helmholtz Diabetes Center at Helmholtz Zentrum München, Munich, Germany.,Deutsches Zentrum für Diabetesforschung (DZD), Munich, Germany
| |
Collapse
|
191
|
Bharath LP, Ip BC, Nikolajczyk BS. Adaptive Immunity and Metabolic Health: Harmony Becomes Dissonant in Obesity and Aging. Compr Physiol 2017; 7:1307-1337. [PMID: 28915326 DOI: 10.1002/cphy.c160042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Adipose tissue (AT) is the primary energy reservoir organ, and thereby plays a critical role in energy homeostasis and regulation of metabolism. AT expands in response to chronic overnutrition or aging and becomes a major source of inflammation that has marked influence on systemic metabolism. The chronic, sterile inflammation that occurs in the AT during the development of obesity or in aging contributes to onset of devastating diseases such as insulin resistance, diabetes, and cardiovascular pathologies. Numerous studies have shown that inflammation in the visceral AT of humans and animals is a critical trigger for the development of metabolic syndrome. This work underscores the well-supported conclusion that the inflammatory immune response and metabolic pathways in the AT are tightly interwoven by multiple layers of relatively conserved mechanisms. During the development of diet-induced obesity or age-associated adiposity, cells of the innate and the adaptive immune systems infiltrate and proliferate in the AT. Macrophages, which dominate AT-associated immune cells in mouse models of obesity, but are less dominant in obese people, have been studied extensively. However, cells of the adaptive immune system, including T cells and B cells, contribute significantly to AT inflammation, perhaps more in humans than in mice. Lymphocytes regulate recruitment of innate immune cells into AT, and produce cytokines that influence the helpful-to-harmful inflammatory balance that, in turn, regulates organismal metabolism. This review describes inflammation, or more precisely, metabolic inflammation (metaflammation) with an eye toward the AT and the roles lymphocytes play in regulation of systemic metabolism during obesity and aging. © 2017 American Physiological Society. Compr Physiol 7:1307-1337, 2017.
Collapse
Affiliation(s)
- Leena P Bharath
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Blanche C Ip
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, USA.,Department of Molecular Pharmacology, Physiology and Biotechnology, Center of Biomedical Engineering, Brown University, Providence, Rhode Island, USA
| | | |
Collapse
|
192
|
Abstract
PURPOSE OF THE REVIEW Obesity and type 2 diabetes (T2D) are considered chronic inflammatory diseases. While early publications have reported the implication of innate immune cells such as macrophages to promote systemic inflammation and metabolic dysfunctions, recent publications underline the alterations of the T cell compartment in human obesity and type 2 diabetes. These recent findings are the focus of this review. RECENT FINDINGS In humans, obesity and T2D induce the expansion of proinflammatory T cells such as CD4 Th1, Th17, and CD8 populations, whereas innate T cells such as MAIT and iNKT cells are decreased. These alterations reflect a loss of total T cell homeostasis that may contribute to tissue and systemic inflammation. Whether these changes are adaptive to nutritional variations and/or contribute to the progression of metabolic diseases remains to be clarified. T cell phenotyping may improve obese and/or T2D patient stratification with therapeutic and prognostic implications.
Collapse
Affiliation(s)
- Sothea Touch
- INSERM, UMR_S 1166, Team 6 Nutriomics, 75013, Paris, France
- Sorbonne Universités, UPMC University Paris 06, UMR_S 1166, 75005, Paris, France
- ICAN, Pitié-Salpêtrière Hospital, Assistance Publique Hôpitaux de Paris, Institute of Cardiometabolism and Nutrition, 75013, Paris, France
| | - Karine Clément
- INSERM, UMR_S 1166, Team 6 Nutriomics, 75013, Paris, France
- Sorbonne Universités, UPMC University Paris 06, UMR_S 1166, 75005, Paris, France
- ICAN, Pitié-Salpêtrière Hospital, Assistance Publique Hôpitaux de Paris, Institute of Cardiometabolism and Nutrition, 75013, Paris, France
- Nutrition, Endocrinology and Cardiology Departments, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, 75013, Paris, France
| | - Sébastien André
- INSERM, UMR_S 1166, Team 6 Nutriomics, 75013, Paris, France.
- Sorbonne Universités, UPMC University Paris 06, UMR_S 1166, 75005, Paris, France.
- ICAN, Pitié-Salpêtrière Hospital, Assistance Publique Hôpitaux de Paris, Institute of Cardiometabolism and Nutrition, 75013, Paris, France.
| |
Collapse
|
193
|
Damouche A, Pourcher G, Pourcher V, Benoist S, Busson E, Lataillade JJ, Le Van M, Lazure T, Adam J, Favier B, Vaslin B, Müller-Trutwin M, Lambotte O, Bourgeois C. High proportion of PD-1-expressing CD4 + T cells in adipose tissue constitutes an immunomodulatory microenvironment that may support HIV persistence. Eur J Immunol 2017; 47:2113-2123. [PMID: 28762530 DOI: 10.1002/eji.201747060] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/14/2017] [Accepted: 07/26/2017] [Indexed: 11/11/2022]
Abstract
We and others have demonstrated that adipose tissue is a reservoir for HIV. Evaluation of the mechanisms responsible for viral persistence may lead to ways of reducing these reservoirs. Here, we evaluated the immune characteristics of adipose tissue in HIV-infected patients receiving antiretroviral therapy (ART) and in non-HIV-infected patients. We notably sought to determine whether adipose tissue's intrinsic properties and/or HIV induced alteration of the tissue environment may favour viral persistence. ART-controlled HIV infection was associated with a difference in the CD4/CD8 T-cell ratio and an elevated proportion of Treg cells in subcutaneous adipose tissue. No changes in Th1, Th2 and Th17 cell proportions or activation markers expression on T cell (Ki-67, HLA-DR) could be detected, and the percentage of CD69-expressing resident memory CD4+ T cells was not affected. Overall, our results indicate that adipose-tissue-resident CD4+ T cells are not extensively activated during HIV infection. PD-1 was expressed by a high proportion of tissue-resident memory CD4+ T cells in both HIV-infected patients and non-HIV-infected patients. Our findings suggest that adipose tissue's intrinsic immunomodulatory properties may limit immune activation and thus may strongly contribute to viral persistence.
Collapse
Affiliation(s)
- Abderaouf Damouche
- Univ Paris Sud, UMR INSERM 1184, Le Kremlin-Bicêtre, France.,CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
| | - Guillaume Pourcher
- Department of Digestive Diseases, Obesity center, Institut Mutualiste Montsouris, Paris-sud University, Paris, France
| | - Valérie Pourcher
- Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Service de Maladies Infectieuses et Tropicales, Sorbonne Universités, UPMC Université Paris 06, France
| | - Stéphane Benoist
- Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, Service de Chirurgie Digestive et Oncologique, Le Kremlin-Bicêtre, France
| | - Elodie Busson
- Hôpital d'Instruction des Armées Percy, Centre de Transfusion Sanguine des Armées, Clamart, France
| | - Jean-Jacques Lataillade
- Hôpital d'Instruction des Armées Percy, Centre de Transfusion Sanguine des Armées, Clamart, France
| | - Mélanie Le Van
- Univ Paris Sud, UMR INSERM 1184, Le Kremlin-Bicêtre, France.,CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
| | - Thierry Lazure
- Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, Service d'anatomo-pathologie, Le Kremlin-Bicêtre, France
| | - Julien Adam
- Institut Gustave Roussy, Plateforme d'évaluation préclinique, Villejuif, France
| | - Benoit Favier
- Univ Paris Sud, UMR INSERM 1184, Le Kremlin-Bicêtre, France.,CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
| | - Bruno Vaslin
- Univ Paris Sud, UMR INSERM 1184, Le Kremlin-Bicêtre, France.,CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
| | | | - Olivier Lambotte
- Univ Paris Sud, UMR INSERM 1184, Le Kremlin-Bicêtre, France.,CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France.,Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, Service de Médecine Interne et Immunologie clinique, Le Kremlin-Bicêtre, France
| | - Christine Bourgeois
- Univ Paris Sud, UMR INSERM 1184, Le Kremlin-Bicêtre, France.,CEA, DSV/iMETI, IDMIT, Fontenay-aux-Roses, France
| |
Collapse
|
194
|
Ganguly D. Do Type I Interferons Link Systemic Autoimmunities and Metabolic Syndrome in a Pathogenetic Continuum? Trends Immunol 2017; 39:28-43. [PMID: 28826817 DOI: 10.1016/j.it.2017.07.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/22/2017] [Accepted: 07/07/2017] [Indexed: 12/30/2022]
Abstract
The central pathogenetic role of type I interferons (IFNs) in several systemic autoimmune diseases is well established. Recent studies have also discovered a similar crucial role of type I IFNs in different components of metabolic disorders. Self nucleic acid-driven Toll-like receptor (TLR) activation in plasmacytoid dendritic cells (pDCs) and type I IFN induction appear to be the key initiating events shared by most of these autoimmune and metabolic diseases. Further strengthening this link, many patients with systemic autoimmunities also present with metabolic disorders. This concurrence of autoimmunities and metabolic disorders may be explained by a single pathogenetic continuum, and suggests shared targets for potential new therapies.
Collapse
Affiliation(s)
- Dipyaman Ganguly
- Dendritic Cell Biology Laboratory, CSIR-Indian Institute of Chemical Biology (IICB)-Translational Research Unit of Excellence, CN6 Sector V, Salt Lake, Kolkata, West Bengal, 700091, India; Division of Cancer Biology and Inflammatory Disorders, CSIR-Indian Institute of Chemical Biology (IICB), CN6 Sector V, Salt Lake, Kolkata, West Bengal, 700091, India.
| |
Collapse
|
195
|
Patouraux S, Rousseau D, Bonnafous S, Lebeaupin C, Luci C, Canivet CM, Schneck AS, Bertola A, Saint-Paul MC, Iannelli A, Gugenheim J, Anty R, Tran A, Bailly-Maitre B, Gual P. CD44 is a key player in non-alcoholic steatohepatitis. J Hepatol 2017; 67:328-338. [PMID: 28323124 DOI: 10.1016/j.jhep.2017.03.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 02/01/2017] [Accepted: 03/02/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Cluster of differentiation (CD)44 regulates adipose tissue inflammation in obesity and hepatic leukocyte recruitment in a lithogenic context. However, its role in hepatic inflammation in a mouse model of steatohepatitis and its relevance in humans have not yet been investigated. We aimed to evaluated the contribution of CD44 to non-alcoholic steatohepatitis (NASH) development and liver injury in mouse models and in patients at various stages of non-alcoholic fatty liver disease (NAFLD) progression. METHODS The role of CD44 was evaluated in CD44-/- mice and after injections of an αCD44 antibody in wild-type mice challenged with a methionine- and choline-deficient diet (MCDD). In obese patients, hepatic CD44 (n=30 and 5 NASH patients with a second liver biopsy after bariatric surgery) and serum sCD44 (n=64) were evaluated. RESULTS Liver inflammation (including inflammatory foci number, macrophage and neutrophil infiltration and CCL2/CCR2 levels), liver injury and fibrosis strongly decreased in CD44-/- mice compared to wild-type mice on MCDD. CD44 deficiency enhanced the M2 polarization and strongly decreased the activation of macrophages by lipopolysaccharide (LPS), hepatocyte damage-associated molecular patterns (DAMPs) and saturated fatty acids. Neutralization of CD44 in mice with steatohepatitis strongly decreased the macrophage infiltration and chemokine ligand (CCL)2 expression with a partial correction of liver inflammation and injury. In obese patients, hepatic CD44 was strongly upregulated in NASH patients (p=0.0008) and correlated with NAFLD activity score (NAS) (p=0.001), ballooning (p=0.003), alanine transaminase (p=0.005) and hepatic CCL2 (p<0.001) and macrophage marker CD68 (p<0.001) expression. Correction of NASH was associated with a strong decrease in liver CD44+ cells. Finally, the soluble form of CD44 increased with severe steatosis (p=0.0005) and NASH (p=0.007). CONCLUSION Human and experimental data suggest that CD44 is a marker and key player of hepatic inflammation and its targeting partially corrects NASH. LAY SUMMARY Human and experimental data suggest that CD44, a cellular protein mainly expressed in immune cells, is a marker and key player of non-alcoholic steatohepatitis (NASH). Indeed, CD44 enhances the non-alcoholic fatty liver (NAFL) (hepatic steatosis) to NASH progression by regulating hepatic macrophage polarization (pro-inflammatory phenotype) and infiltration (macrophage motility and the MCP1/CCL2/CCR2 system). Targeting CD44 partially corrects NASH, making it a potential therapeutic strategy.
Collapse
Affiliation(s)
- Stéphanie Patouraux
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France; CHU of Nice, Biological Center, Pasteur Hôpital, Nice, France
| | - Déborah Rousseau
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France
| | - Stéphanie Bonnafous
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France; CHU of Nice, Digestive Center, Nice, France
| | - Cynthia Lebeaupin
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France
| | - Carmelo Luci
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France
| | - Clémence M Canivet
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France; CHU of Nice, Digestive Center, Nice, France
| | - Anne-Sophie Schneck
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France; CHU of Nice, Digestive Center, Nice, France
| | - Adeline Bertola
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France
| | - Marie-Christine Saint-Paul
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France; CHU of Nice, Biological Center, Pasteur Hôpital, Nice, France
| | - Antonio Iannelli
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France; CHU of Nice, Digestive Center, Nice, France
| | - Jean Gugenheim
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France; CHU of Nice, Digestive Center, Nice, France
| | - Rodolphe Anty
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France; CHU of Nice, Digestive Center, Nice, France
| | - Albert Tran
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France; CHU of Nice, Digestive Center, Nice, France
| | - Béatrice Bailly-Maitre
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France
| | - Philippe Gual
- INSERM, U1065, C3M, Team 8 "Hepatic Complications in Obesity", Nice, France; Université Côte d'Azur, Nice, France.
| |
Collapse
|
196
|
Immune cell-derived cytokines contribute to obesity-related inflammation, fibrogenesis and metabolic deregulation in human adipose tissue. Sci Rep 2017; 7:3000. [PMID: 28592801 PMCID: PMC5462798 DOI: 10.1038/s41598-017-02660-w] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 04/18/2017] [Indexed: 12/03/2022] Open
Abstract
Adipose tissue contains a variety of immune cells, which vary in abundance and phenotype with obesity. The contribution of immune cell-derived factors to inflammatory, fibrotic and metabolic alterations in adipose tissue is not well established in human obesity. Human primary adipose tissue cells, including pre-adipocytes, endothelial cells and mature adipocytes, were used to investigate deregulation of cell- and pathway-specific gene profiles. Among factors known to alter adipose tissue biology, we focus on inflammatory (IL-1β and IL-17) and pro-fibrotic (TGF-β1) factors. rIL-1β and rIL-17 induced concordant pro-inflammatory transcriptional programs in pre-adipocytes and endothelial cells, with a markedly more potent effect of IL-1β than IL-17. None of these cytokines had significant effect on fibrogenesis-related gene expression, contrasting with rTGF-β1-induced up-regulation of extracellular matrix components and pro-fibrotic factors. In mature adipocytes, all three factors promoted down-regulation of genes functionally involved in lipid storage and release. IL-1β and IL-17 impacted adipocyte metabolic genes in relation with their respective pro-inflammatory capacity, while the effect of TGF-β1 occurred in face of an anti-inflammatory signature. These data revealed that IL-1β and IL-17 had virtually no effect on pro-fibrotic alterations but promote inflammation and metabolic dysfunction in human adipose tissue, with a prominent role for IL-1β.
Collapse
|
197
|
Role of immune cells in obesity induced low grade inflammation and insulin resistance. Cell Immunol 2017; 315:18-26. [DOI: 10.1016/j.cellimm.2017.03.001] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 02/09/2017] [Accepted: 03/05/2017] [Indexed: 12/11/2022]
|
198
|
Patel MS, Miranda-Nieves D, Chen J, Haller CA, Chaikof EL. Targeting P-selectin glycoprotein ligand-1/P-selectin interactions as a novel therapy for metabolic syndrome. Transl Res 2017; 183:1-13. [PMID: 28034759 PMCID: PMC5393932 DOI: 10.1016/j.trsl.2016.11.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 11/13/2016] [Indexed: 12/22/2022]
Abstract
Obesity-induced insulin resistance and metabolic syndrome continue to pose an important public health challenge worldwide as they significantly increase the risk of type 2 diabetes and atherosclerotic cardiovascular disease. Advances in the pathophysiologic understanding of this process has identified that chronic inflammation plays a pivotal role. In this regard, given that both animal models and human studies have demonstrated that the interaction of P-selectin glycoprotein ligand-1 (PSGL-1) with P-selectin is not only critical for normal immune response but also is upregulated in the setting of metabolic syndrome, PSGL-1/P-selectin interactions provide a novel target for preventing and treating resultant disease. Current approaches of interfering with PSGL-1/P-selectin interactions include targeted antibodies, recombinant immunoglobulins that competitively bind P-selectin, and synthetic molecular therapies. Experimental models as well as clinical trials assessing the role of these modalities in a variety of diseases have continued to contribute to the understanding of PSGL-1/P-selectin interactions and have demonstrated the difficulty in creating clinically relevant therapeutics. Most recently, however, computational simulations have further enhanced our understanding of the structural features of PSGL-1 and related glycomimetics, which are responsible for high-affinity selectin interactions. Leveraging these insights for the design of next generation agents has thus led to development of a promising synthetic method for generating PSGL-1 glycosulfopeptide mimetics for the treatment of metabolic syndrome.
Collapse
Affiliation(s)
- Madhukar S Patel
- Department of Surgery, Massachusetts General Hospital, Boston, Mass; Department of Surgery, Beth Israel Deaconess Medical Center, Boston, Mass; Harvard Medical School, Boston, Mass
| | - David Miranda-Nieves
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, Mass; Harvard Medical School, Boston, Mass; Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Mass
| | - Jiaxuan Chen
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Carolyn A Haller
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Elliot L Chaikof
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, Mass; Harvard Medical School, Boston, Mass.
| |
Collapse
|
199
|
Morin SO, Poggi M, Alessi MC, Landrier JF, Nunès JA. Modulation of T Cell Activation in Obesity. Antioxid Redox Signal 2017; 26:489-500. [PMID: 27225042 DOI: 10.1089/ars.2016.6746] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
SIGNIFICANCE Immune T cells are present in adipose tissues (AT), and the stoichiometry of the different T cell subsets is altered during diet-induced obesity (DIO). T cells contribute to the early steps of AT inflammation during DIO. Recent Advances: Many factors could potentially be responsible for this altered pro-inflammatory versus anti-inflammatory T cell balance. CRITICAL ISSUES T cells are potentially activated in AT, which vitamin D might contribute to, as will be discussed in this article. In addition, we will review the different possible contributors to T cell activation in AT, such as the CD28 and CD154 T cell costimulatory molecules in AT. FUTURE DIRECTIONS The potential antigen presentation capacities of adipocytes should be further investigated. Moreover, the properties of these AT resident (or migrating to AT) T cells must be further assessed. Antioxid. Redox Signal. 26, 489-500.
Collapse
Affiliation(s)
- Stéphanie O Morin
- 1 Inserm, U1068, Centre de Recherche en Cancérologie de Marseille , Marseille, France .,2 Institut Paoli-Calmettes , Marseille, France .,3 CNRS, UMR7258, Centre de Recherche en Cancérologie de Marseille , Marseille, France .,4 Aix-Marseille Université , UM105, Marseille, France
| | - Marjorie Poggi
- 5 Inserm U1062 , Marseille, France .,6 Inra , UMR1260, Marseille, France .,7 Aix-Marseille Université , Nutrition Obésité Risques Thrombotiques, Marseille, France
| | - Marie-Christine Alessi
- 5 Inserm U1062 , Marseille, France .,6 Inra , UMR1260, Marseille, France .,7 Aix-Marseille Université , Nutrition Obésité Risques Thrombotiques, Marseille, France
| | - Jean-François Landrier
- 5 Inserm U1062 , Marseille, France .,6 Inra , UMR1260, Marseille, France .,7 Aix-Marseille Université , Nutrition Obésité Risques Thrombotiques, Marseille, France
| | - Jacques A Nunès
- 1 Inserm, U1068, Centre de Recherche en Cancérologie de Marseille , Marseille, France .,2 Institut Paoli-Calmettes , Marseille, France .,3 CNRS, UMR7258, Centre de Recherche en Cancérologie de Marseille , Marseille, France .,4 Aix-Marseille Université , UM105, Marseille, France
| |
Collapse
|
200
|
Kępczyńska MA, Zaibi MS, Alomar SY, Trayhurn P. PCR arrays indicate that the expression of extracellular matrix and cell adhesion genes in human adipocytes is regulated by IL-1β (interleukin-1β). Arch Physiol Biochem 2017; 123:61-67. [PMID: 27855518 DOI: 10.1080/13813455.2016.1248979] [Citation(s) in RCA: 8] [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: 12/14/2022]
Abstract
The role of IL-1β in regulating the expression of extracellular matrix (ECM) and cell adhesion genes in human adipocytes has been examined. Adipocytes differentiated in culture were incubated with IL-1β for 4 or 24 h and RNA probed with PCR arrays for 84 ECM and cell adhesion genes. Treatment with IL-1β resulted in changes in the expression at one or both time points of ∼50% of the genes probed by the arrays, the majority being down-regulated. Genes whose expression was down-regulated by IL-1β included those encoding several collagen chains and integrin subunits. In contrast, IL-1β induced substantial increases (>10-fold) in the expression of ICAM1, VCAM1, MMP1 and MMP3; the secretion of the encoded proteins was also markedly stimulated. IL-1β has a pervasive effect on the expression of ECM and cell adhesion genes in human adipocytes, consistent with the derangement of tissue structure during inflammation in white fat.
Collapse
Affiliation(s)
| | - Mohamed S Zaibi
- a Clore Laboratory, University of Buckingham , Buckingham , United Kingdom
| | - Suliman Y Alomar
- b Zoology Department, College of Science, King Saud University , Riyadh , Saudi Arabia , and
| | - Paul Trayhurn
- a Clore Laboratory, University of Buckingham , Buckingham , United Kingdom
- b Zoology Department, College of Science, King Saud University , Riyadh , Saudi Arabia , and
- c Obesity Biology Unit, University of Liverpool , Liverpool , United Kingdom
| |
Collapse
|