51
|
Abstract
Adipose tissue is a special tissue environment due to its high lipid content. Adipose tissue macrophages (ATMs) help maintain adipose tissue homeostasis in steady state by clearing dead adipocytes. However, adipose tissue changes drastically during obesity, resulting in a state of chronic low grade inflammation and a shift in the adipose immune landscape. In this review we will discuss how these changes influence the polarization of ATMs.
Collapse
Affiliation(s)
- Leen Catrysse
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium
| | - Geert van Loo
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium.
| |
Collapse
|
52
|
Chaves Filho AJM, Lima CNC, Vasconcelos SMM, de Lucena DF, Maes M, Macedo D. IDO chronic immune activation and tryptophan metabolic pathway: A potential pathophysiological link between depression and obesity. Prog Neuropsychopharmacol Biol Psychiatry 2018; 80:234-249. [PMID: 28595944 DOI: 10.1016/j.pnpbp.2017.04.035] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 04/03/2017] [Accepted: 04/10/2017] [Indexed: 12/12/2022]
Abstract
Obesity and depression are among the most pressing health problems in the contemporary world. Obesity and depression share a bidirectional relationship, whereby each condition increases the risk of the other. By inference, shared pathways may underpin the comorbidity between obesity and depression. Activation of cell-mediated immunity (CMI) is a key factor in the pathophysiology of depression. CMI cytokines, including IFN-γ, TNFα and IL-1β, induce the catabolism of tryptophan (TRY) by stimulating indoleamine 2,3-dioxygenase (IDO) resulting in the synthesis of kynurenine (KYN) and other tryptophan catabolites (TRYCATs). In the CNS, TRYCATs have been related to oxidative damage, inflammation, mitochondrial dysfunction, cytotoxicity, excitotoxicity, neurotoxicity and lowered neuroplasticity. The pathophysiology of obesity is also associated with a state of aberrant inflammation that activates aryl hydrocarbon receptor (AHR), a pathway involved in the detection of intracellular or environmental changes as well as with increases in the production of TRYCATs, being KYN an agonists of AHR. Both AHR and TRYCATS are involved in obesity and related metabolic disorders. These changes in the TRYCAT pathway may contribute to the onset of neuropsychiatric symptoms in obesity. This paper reviews the role of immune activation, IDO stimulation and increased TRYCAT production in the pathophysiology of depression and obesity. Here we suggest that increased synthesis of detrimental TRYCATs is implicated in comorbid obesity and depression and is a new drug target to treat both diseases.
Collapse
Affiliation(s)
- Adriano José Maia Chaves Filho
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Camila Nayane Carvalho Lima
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Silvânia Maria Mendes Vasconcelos
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - David Freitas de Lucena
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Michael Maes
- Impact Strategic Research Center, Deakin University, Geelong, Australia; Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Health Sciences Graduate Program, Health Sciences Center, State University of Londrina, Londrina, Brazil
| | - Danielle Macedo
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil.
| |
Collapse
|
53
|
Molloy CT, Adkins LJ, Griffin C, Singer K, Weinberg JB. Mouse adenovirus type 1 infection of adipose tissue. Virus Res 2017; 244:90-98. [PMID: 29141203 DOI: 10.1016/j.virusres.2017.11.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/10/2017] [Accepted: 11/10/2017] [Indexed: 01/13/2023]
Abstract
Human adenovirus (HAdV) type 36 seropositivity has been linked to obesity in humans. That link is supported by a small number of studies using HAdV-36 infection of animals that are not natural hosts for HAdVs. In this study, we infected mice with mouse adenovirus type 1 (MAV-1), a mouse pathogen, to determine whether MAV-1 infected adipose tissue and was associated with adipose tissue inflammation and obesity. We detected MAV-1 in adipose tissue during acute MAV-1 infection, but we did not detect virus-induced increases in adipose tissue cytokine expression or histological evidence of adipose tissue inflammation during acute infection. MAV-1 did not persist in adipose tissue at later times, and we did not detect long-term adipose inflammation, increased adipose tissue mass, or body weight in infected mice. Our data indicate that MAV-1 is not associated with obesity in infected mice.
Collapse
Affiliation(s)
- Caitlyn T Molloy
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, United States
| | - Laura J Adkins
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, United States
| | - Cameron Griffin
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, United States
| | - Kanakadurga Singer
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, United States
| | - Jason B Weinberg
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, United States; Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
54
|
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
|
55
|
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
|
56
|
Temporal and tissue-specific requirements for T-lymphocyte IL-6 signalling in obesity-associated inflammation and insulin resistance. Nat Commun 2017; 8:14803. [PMID: 28466852 PMCID: PMC5418621 DOI: 10.1038/ncomms14803] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 01/31/2017] [Indexed: 02/06/2023] Open
Abstract
Low-grade inflammation links obesity to insulin resistance through the activation of tissue-infiltrating immune cells. Interleukin-6 (IL-6) is a crucial regulator of T cells and is increased in obesity. Here we report that classical IL-6 signalling in T cells promotes inflammation and insulin resistance during the first 8 weeks on a high-fat diet (HFD), but becomes dispensable at later stages (after 16 weeks). Mice with T cell-specific deficiency of IL-6 receptor-α (IL-6RαT-KO) exposed to a HFD display improved glucose tolerance, insulin sensitivity and inflammation in liver and EWAT after 8 weeks. However, after 16 weeks, insulin resistance in IL-6RαT-KO epididymal white adipose tissue (EWAT) is comparable to that of controls, whereas the inflammatory profile is significantly worse. This coincided with a shift from classical T cell IL-6 signalling at 8 weeks, to enhanced IL-6 trans-signalling at 16 weeks. Collectively, our studies reveal that IL-6 action in T cells through classical IL-6 signalling promotes inflammation and insulin resistance early during obesity development, which can be compensated for by enhanced IL-6 trans-signalling at later stages. Interleukin-6 (IL-6) is increased in obesity and activates T cells to promote inflammation. Here, Xu et al. use mice that lack IL-6 receptors on T cells to uncover the temporal and tissue-specific effects of classic and trans IL-6 signalling on inflammation and insulin resistance on a high-fat diet.
Collapse
|
57
|
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]
|
58
|
Ghazarian M, Revelo XS, Nøhr MK, Luck H, Zeng K, Lei H, Tsai S, Schroer SA, Park YJ, Chng MHY, Shen L, D’Angelo JA, Horton P, Chapman WC, Brockmeier D, Woo M, Engleman EG, Adeyi O, Hirano N, Jin T, Gehring AJ, Winer S, Winer DA. Type I Interferon Responses Drive Intrahepatic T cells to Promote Metabolic Syndrome. Sci Immunol 2017; 2:eaai7616. [PMID: 28567448 PMCID: PMC5447456 DOI: 10.1126/sciimmunol.aai7616] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Obesity-related insulin resistance is driven by low-grade chronic inflammation of metabolic tissues. In the liver, non-alcoholic fatty liver disease (NAFLD) is associated with hepatic insulin resistance and systemic glucose dysregulation. However, the immunological factors supporting these processes are poorly understood. We found that the liver accumulates pathogenic CD8+ T cell subsets which control hepatic insulin sensitivity and gluconeogenesis during diet-induced obesity in mice. In a cohort of human patients, CD8+ T cells represent a dominant intrahepatic immune cell population which links to glucose dysregulation. Accumulation and activation of these cells are largely supported by type I interferon (IFN-I) responses in the liver. Livers from obese mice upregulate critical interferon regulatory factors (IRFs), interferon stimulatory genes (ISGs), and IFNα protein, while IFNαR1-/- mice, or CD8-specific IFNαR1-/- chimeric mice are protected from disease. IFNαR1 inhibitors improve metabolic parameters in mice, while CD8+ T cells and IFN-I responses correlate with NAFLD activity in human patients. Thus, IFN-I responses represent a central immunological axis that governs intrahepatic T cell pathogenicity during metabolic disease.
Collapse
Affiliation(s)
- Magar Ghazarian
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario M5G 1L7, Canada
- Department of Immunology, University of Toronto, 1 King’s College Circle, Toronto, Ontario M5S 3B3, Canada
| | - Xavier S. Revelo
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Mark K. Nøhr
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Helen Luck
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario M5G 1L7, Canada
- Department of Immunology, University of Toronto, 1 King’s College Circle, Toronto, Ontario M5S 3B3, Canada
| | - Kejing Zeng
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario M5G 1L7, Canada
- Department of Endocrinology and Metabolism, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Helena Lei
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Sue Tsai
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Stephanie A. Schroer
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Yoo Jin Park
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Melissa Hui Yen Chng
- Department of Pathology, Stanford University School of Medicine, Palo Alto, CA 94205, USA
| | - Lei Shen
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| | - June Ann D’Angelo
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Peter Horton
- Methodist University Hospital Transplant Institute, Memphis, TN 38104, USA
- Division of Abdominal Transplant, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - William C. Chapman
- Division of Abdominal Transplant, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Minna Woo
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Edgar G. Engleman
- Department of Pathology, Stanford University School of Medicine, Palo Alto, CA 94205, USA
| | - Oyedele Adeyi
- Department of Pathology, University Health Network, 200 Elizabeth Street, Toronto, Ontario M5G 2C4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Naoto Hirano
- Department of Immunology, University of Toronto, 1 King’s College Circle, Toronto, Ontario M5S 3B3, Canada
- Campbell Family Cancer Research Institute, University Health Network, Toronto, Ontario M5G 2M9, Canada
| | - Tianru Jin
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Adam J. Gehring
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
- Toronto Centre for Liver Disease, University Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Shawn Winer
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Department of Laboratory Medicine, St. Michael’s Hospital, Toronto, Ontario M5B 1W8, Canada
| | - Daniel A. Winer
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Research Institute, University Health Network, 101 College Street, Toronto, Ontario M5G 1L7, Canada
- Department of Immunology, University of Toronto, 1 King’s College Circle, Toronto, Ontario M5S 3B3, Canada
- Department of Pathology, University Health Network, 200 Elizabeth Street, Toronto, Ontario M5G 2C4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
59
|
HCV-induced regulatory alterations of IL-1β, IL-6, TNF-α, and IFN-ϒ operative, leading liver en-route to non-alcoholic steatohepatitis. Inflamm Res 2017; 66:477-486. [PMID: 28285394 DOI: 10.1007/s00011-017-1029-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/15/2017] [Accepted: 02/17/2017] [Indexed: 12/13/2022] Open
Abstract
Over the course of time, Hepatitis C has become a universal health menace. Its deleterious effects on human liver encompass a lot of physiological, genetic as well as epigenetic alterations. Fatty liver (Hepatic steatosis) is an inflammation having multifactorial ancestries; one of them is HCV (steatohepatitis). HCV boosts several cellular pathways involving up-regulation of a number of cytokines. Current study reviews the regulation of some selective key cytokines during HCV infection, to help generate an improved understanding of their role. These cytokines, IL-1β, IL-6, TNF-α, and IFN-ϒ, are inflammatory markers of the body. These particular markers along with others help hepatocytes against viral infestation. However, recently, their association has been found in degradation of liver on the trail heading to non-alcoholic steatohepatitis (NASH). Consequently, the disturbance in their equilibrium has been repeatedly reported during HCV infection. Quite a number of findings are affirming their up-regulation. Although these cell markers are stimulated by hepatocytes as their standard protection mechanism, but modern studies have testified the paradoxical nature of this defense line. Nevertheless, direct molecular or epigenetic research is needed to question the actual molecular progressions and directions commanding liver to steatosis, cirrhosis, or eventually HCC (Hepatocellular Carcinoma).
Collapse
|
60
|
Lorenz TK, Heiman JR, Demas GE. Testosterone and immune-reproductive tradeoffs in healthy women. Horm Behav 2017; 88:122-130. [PMID: 27865788 PMCID: PMC5303633 DOI: 10.1016/j.yhbeh.2016.11.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 10/14/2016] [Accepted: 11/04/2016] [Indexed: 11/24/2022]
Abstract
Although testosterone (T) has been characterized as universally immunosuppressive across species and sexes, recent ecoimmunology research suggests that T's immunomodulatory effects (enhancing/suppressing) depend on the organism's reproductive context. Very little is known about the immune effects of T in healthy females, and even less about how reproductive effort modulates the immune effects of T in humans. We investigated how the interaction between endogenous T and sexual activity predicted menstrual cycle-related changes in several measures of immunity: inflammation (indexed by interleukin-6, IL-6), adaptive immunity (indexed by immunoglobulin A, IgA), and functional immunity (indexed by bactericidal assay). Thirty-two healthy women (sexually abstinent, N=17; sexually active with one male partner, N=15) provided saliva samples at four points in the menstrual cycle: menses, follicular, ovulation, and luteal phases. Among sexually abstinent women, T was positively associated with IL-6 across the cycle; for sexually active women, however, T was positively associated with IL-6 in the luteal phase only, and negatively associated with IL-6 at ovulation. High T predicted higher IgA among women who reported infrequent intercourse, but lower IgA among women who reported very frequent intercourse. Finally, across groups, T was positively associated with greater bacterial killing at menses, but negatively associated in the luteal phase. Overall, rather than being universally immunosuppressive, T appeared to signal immunomodulation relevant to reproduction (e.g., lowering inflammation at ovulation, potentially preventing immune interference with conception). Our findings support the hypothesis that the immunomodulatory effects of endogenous T in healthy females depend on sexual and reproductive context.
Collapse
Affiliation(s)
- Tierney K Lorenz
- Center for Integrative Study for Animal Behavior, Indiana University, 409 N. Park Ave, Bloomington, IN 47405, United States; The Kinsey Institute, Indiana University, 1165 E 3rd St., Bloomington, IN 47405, United States; Department of Psychology, University of North Carolina at Charlotte, 9201 University City Blvd., Charlotte, NC 28223, United States.
| | - Julia R Heiman
- Center for Integrative Study for Animal Behavior, Indiana University, 409 N. Park Ave, Bloomington, IN 47405, United States; The Kinsey Institute, Indiana University, 1165 E 3rd St., Bloomington, IN 47405, United States; Department of Psychological and Brain Sciences, Indiana University, 1101 E. 10th St., Bloomington, IN 47405, United States.
| | - Gregory E Demas
- Center for Integrative Study for Animal Behavior, Indiana University, 409 N. Park Ave, Bloomington, IN 47405, United States; Department of Biology, Indiana University, 1001 E 3rd St., Bloomington, IN 47405, United States.
| |
Collapse
|
61
|
Garrison CB, Lastwika KJ, Zhang Y, Li CI, Lampe PD. Proteomic Analysis, Immune Dysregulation, and Pathway Interconnections with Obesity. J Proteome Res 2017; 16:274-287. [PMID: 27769113 PMCID: PMC5234688 DOI: 10.1021/acs.jproteome.6b00611] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Proteomic studies can offer information on hundreds to thousands of proteins and potentially provide researchers with a comprehensive understanding of signaling response during stress and disease. Large data sets, such as those obtained in high-dimensional proteomic studies, can be leveraged for pathway analysis to discover or describe the biological implications of clinical disease states. Obesity is a worldwide epidemic that is considered a risk factor for numerous other diseases. We performed analysis on plasma proteomic data from 3 separate sample sets of postmenopausal women to identify the pathways that are altered in subjects with a high body mass index (BMI) compared to normal BMI. We found many pathways consistently and significantly associated with inflammation dysregulated in plasma from obese/overweight subjects compared to plasma from normal BMI subjects. These pathways indicate alterations of soluble inflammatory regulators, cellular stress, and metabolic dysregulation. Our results highlight the importance of high-dimensional pathway analysis in complex diseases as well as provide information on the interconnections between pathways that are dysregulated with obesity. Specifically, overlap of obesity related pathways with those activated during cancer and infection could help describe why obesity is a risk factor for disease and help devise treatment options that mitigate its effect.
Collapse
Affiliation(s)
- Carly B. Garrison
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Kristin J. Lastwika
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Yuzheng Zhang
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Christopher I. Li
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Paul D. Lampe
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| |
Collapse
|
62
|
Engin AB, Engin A. The Interactions Between Kynurenine, Folate, Methionine and Pteridine Pathways in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:511-527. [PMID: 28585214 DOI: 10.1007/978-3-319-48382-5_22] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Obesity activates both innate and adaptive immune responses in adipose tissue. Elevated levels of eosinophils with depression of monocyte and neutrophil indicate the deficiencies in the immune system of morbidly obese individuals. Actually, adipose tissue macrophages are functional antigen-presenting cells that promote the proliferation of interferon-gamma (IFN-gamma)-producing CD4+ T cells in adipose tissue of obese subjects. Eventually, diet-induced obesity is associated with the loss of tissue homeostasis and development of type 1 inflammatory responses in visceral adipose tissue. Activity of inducible indoleamine 2,3-dioxygenase-1 (IDO-1) plays a major role under pro-inflammatory, IFN-gamma dominated settings. One of the two rate-limiting enzymes which can metabolize tryptophan to kynurenine is IDO-1. Tumor necrosis factor-alpha (TNF-alpha) correlates with IDO-1 in adipose compartments. Actually, IDO-1-mediated tryptophan catabolism due to chronic immune activation is the cause of reduced tryptophan plasma levels and be considered as the driving force for food intake in morbidly obese patients. Thus, decrease in plasma tryptophan levels and subsequent reduction in serotonin (5-HT) production provokes satiety dysregulation that leads to increased caloric uptake and obesity. However, after bariatric surgery, weight reduction does not lead to normalization of IDO-1 activity. Furthermore, there is a connection between arginine and tryptophan metabolic pathways in the generation of reactive nitrogen intermediates. Hence, abdominal obesity is associated with vascular endothelial dysfunction and reduced nitric oxide (NO) availability. IFN-gamma-induced activation of the inducible nitric oxide synthase (iNOS) and dissociation of endothelial adenosine monophosphate activated protein kinase (AMPK)- phosphoinositide 3-kinase (PI3K)-protein kinase B (Akt)- endothelial NO synthase (eNOS) pathway enhances oxidative stress production secondary to high-fat diet. Thus, reduced endothelial NO availability correlates with the increase in plasma non-esterified fatty acids and triglycerides levels. Additionally, in obese patients, folate-deficiency leads to hyperhomocysteinemia. Folic acid confers protection against hyperhomocysteinemia-induced oxidative stress.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- , Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
63
|
Influence of Antioxidants on Leptin Metabolism and its Role in the Pathogenesis of Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:399-413. [PMID: 28585209 DOI: 10.1007/978-3-319-48382-5_17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Obesity is associated with low-grade inflammation. Leptin, a hormone made by fat cells regulates appetite and hunger and thus food intake behavior. Interestingly, , food preservatives like sodium sulfite and sodium benzoate and also natural colorant and spice compounds such as curcumin were found to decrease the release of leptin in murine 3T3-L1 adipocytes, after co-incubation with LPS, which was added to mimic the pro-inflammatory status in obesity. Several of these compounds are well known food antioxidants.Whilst reducing oxidation events is beneficial in states of elevated oxidative stress, overexposure to food antioxidant can lead to adverse effects. There are hints from in vivo data, that antioxidant stress in younger age plays a role in the development of adiposity in later life. The insufficient exposure to oxidizing compounds like reactive oxygen species (ROS) cannot only cause an insufficient burning of calories but there is also a link to the regulation of food intake behavior. If the in vitro findings can be extrapolated to the in vivo situation, consumption of antioxidant supplemented food could lead to decreased leptin release and contribute to an obesogenic environment. This aspect sheds some new critical light on the potential role of an antioxidant-enriched nutrition in the obesity epidemic during the past few centuries. Doing sports could represent not only a proper strategy to initiate physiological ROS production and burning of calories, but also may shift the hormone milieu towards a reduction of hunger feelings and thus reduce appetite and food intake.
Collapse
|
64
|
Greer RL, Dong X, Moraes ACF, Zielke RA, Fernandes GR, Peremyslova E, Vasquez-Perez S, Schoenborn AA, Gomes EP, Pereira AC, Ferreira SRG, Yao M, Fuss IJ, Strober W, Sikora AE, Taylor GA, Gulati AS, Morgun A, Shulzhenko N. Akkermansia muciniphila mediates negative effects of IFNγ on glucose metabolism. Nat Commun 2016; 7:13329. [PMID: 27841267 PMCID: PMC5114536 DOI: 10.1038/ncomms13329] [Citation(s) in RCA: 209] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 09/23/2016] [Indexed: 12/19/2022] Open
Abstract
Cross-talk between the gut microbiota and the host immune system regulates host metabolism, and its dysregulation can cause metabolic disease. Here, we show that the gut microbe Akkermansia muciniphila can mediate negative effects of IFNγ on glucose tolerance. In IFNγ-deficient mice, A. muciniphila is significantly increased and restoration of IFNγ levels reduces A. muciniphila abundance. We further show that IFNγ-knockout mice whose microbiota does not contain A. muciniphila do not show improvement in glucose tolerance and adding back A. muciniphila promoted enhanced glucose tolerance. We go on to identify Irgm1 as an IFNγ-regulated gene in the mouse ileum that controls gut A. muciniphila levels. A. muciniphila is also linked to IFNγ-regulated gene expression in the intestine and glucose parameters in humans, suggesting that this trialogue between IFNγ, A. muciniphila and glucose tolerance might be an evolutionally conserved mechanism regulating metabolic health in mice and humans. Mice deficient in the pro-inflammatory cytokine IFNγ have improved glucose tolerance. Here, the authors show that this effect depends on the gut microbe Akkermansia muciniphila, whose abundance increases in the absence IFNγ, and which is known to have beneficial effects on host metabolism.
Collapse
Affiliation(s)
- Renee L Greer
- College of Veterinary Medicine, Oregon State University, 105 Dryden Hall, 450 SW 30th Street, Corvallis, Oregon 97331, USA
| | - Xiaoxi Dong
- College of Pharmacy, Oregon State University, 1601 SW Jefferson Way, Corvallis, Oregon 97331, USA
| | - Ana Carolina F Moraes
- Department of Epidemiology, School of Public Health, University of São Paulo, Av. Dr Arnaldo, 715, São Paulo, SP 01246-904, Brazil
| | - Ryszard A Zielke
- College of Pharmacy, Oregon State University, 1601 SW Jefferson Way, Corvallis, Oregon 97331, USA
| | - Gabriel R Fernandes
- Oswaldo Cruz Foundation, René Rachou Research Center, Av. Augusto de Lima, 1715, Belo Horizonte, MG 30190-002, Brazil
| | - Ekaterina Peremyslova
- College of Pharmacy, Oregon State University, 1601 SW Jefferson Way, Corvallis, Oregon 97331, USA
| | - Stephany Vasquez-Perez
- College of Veterinary Medicine, Oregon State University, 105 Dryden Hall, 450 SW 30th Street, Corvallis, Oregon 97331, USA
| | - Alexi A Schoenborn
- Division of Pediatric Gastroenterology, University of North Carolina at Chapel Hill, 260 MacNider Building, CB# 7220, Chapel Hill, North Carolina 27599, USA
| | - Everton P Gomes
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, Av. Dr Eneas de Carvalho Aguiar, 44, São Paulo, SP 05403-000, Brazil
| | - Alexandre C Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, Av. Dr Eneas de Carvalho Aguiar, 44, São Paulo, SP 05403-000, Brazil
| | - Sandra R G Ferreira
- Department of Epidemiology, School of Public Health, University of São Paulo, Av. Dr Arnaldo, 715, São Paulo, SP 01246-904, Brazil
| | - Michael Yao
- Mucosal Immunity Section, Laboratory of Immune Defenses, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland 20892, USA
| | - Ivan J Fuss
- Mucosal Immunity Section, Laboratory of Immune Defenses, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland 20892, USA
| | - Warren Strober
- Mucosal Immunity Section, Laboratory of Immune Defenses, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland 20892, USA
| | - Aleksandra E Sikora
- College of Pharmacy, Oregon State University, 1601 SW Jefferson Way, Corvallis, Oregon 97331, USA
| | - Gregory A Taylor
- Geriatric Research, Education and Clinical Center, VA Medical Center, Departments of Medicine, Molecular Genetics and Microbiology and Immunology, Division of Geriatrics and Center for the Study of Aging and Human Development, Duke Box 3003, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Ajay S Gulati
- Division of Pediatric Gastroenterology, University of North Carolina at Chapel Hill, 260 MacNider Building, CB# 7220, Chapel Hill, North Carolina 27599, USA
| | - Andrey Morgun
- College of Pharmacy, Oregon State University, 1601 SW Jefferson Way, Corvallis, Oregon 97331, USA
| | - Natalia Shulzhenko
- College of Veterinary Medicine, Oregon State University, 105 Dryden Hall, 450 SW 30th Street, Corvallis, Oregon 97331, USA
| |
Collapse
|
65
|
In utero exposure to gestational diabetes mellitus conditions TLR4 and TLR2 activated IL-1beta responses in spleen cells from rat offspring. Biochim Biophys Acta Mol Basis Dis 2016; 1862:2137-2146. [DOI: 10.1016/j.bbadis.2016.08.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 07/10/2016] [Accepted: 08/09/2016] [Indexed: 12/18/2022]
|
66
|
Azizian M, Mahdipour E, Mirhafez SR, Shoeibi S, Nematy M, Esmaily H, Ferns GA, Ghayour-Mobarhan M. Cytokine profiles in overweight and obese subjects and normal weight individuals matched for age and gender. Ann Clin Biochem 2016; 53:663-668. [PMID: 26787627 DOI: 10.1177/0004563216629997] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background Obesity is associated with a state of systemic inflammation, mediated by adipose tissue-derived cytokines that may also have metabolic effects, including an effect on insulin resistance. The aim of this study was to compare the serum profile of pro- and anti-inflammatory cytokines in obese and non-obese subjects. Methods A total of 242 subjects who were either overweight or obese (body mass index [BMI] ≥ 25 kg/m2) and non-obese subjects (body mass index <25 kg/m2), were recruited in Mashhad in northeastern Iran. The concentrations of serum interleukin-1α, -1β, -2, -4, -6, -8 and -10 (IL-1α, IL-1β, IL-2, IL-4, IL-6, IL-8 and IL-10), were measured in all subjects, together with serum vascular endothelial growth factor, interferon-γ, epidermal growth factor, monocyte chemoattractant protein-1 and tumour necrosis factor-α. Results The groups differed significantly with respect to measures of adiposity and fasted lipid profile. Serum pro-inflammatory cytokines interferon-γ and interleukin-1α, and anti-inflammatory cytokines, interleukin-10, and epidermal growth factor were significantly different between obese and non-obese individuals, as was serum high-sensitivity C-reactive protein. Multivariate regression showed that waist circumference was significantly and independently related to serum monocyte chemoattractant protein-1concentrations ( P = 0.001). Conclusion Despite significant differences in several cytokines between the groups, only monocyte chemoattractant protein-1appeared to be independently related to a measure of adiposity in this population sample from Iran.
Collapse
Affiliation(s)
- Maryam Azizian
- 1 Department of Nutrition, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elahe Mahdipour
- 2 Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Reza Mirhafez
- 3 Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Sara Shoeibi
- 2 Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Nematy
- 6 Biochemistry of Nutrition Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Habibollah Esmaily
- 5 Department of Biostatistics, Mashhad University of Medical Sciences, Mashhad, Iran.,6 Biochemistry of Nutrition Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon Aa Ferns
- 7 Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Majid Ghayour-Mobarhan
- 4 Cardiovascular Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,6 Biochemistry of Nutrition Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
67
|
Farhangi MA, Saboor-Yaraghi AA, Keshavarz SA. Vitamin A supplementation reduces the Th17-Treg - Related cytokines in obese and non-obese women. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2016; 60:29-35. [PMID: 26909479 PMCID: PMC10118919 DOI: 10.1590/2359-3997000000125] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 09/29/2015] [Indexed: 11/21/2022]
Abstract
OBJECTIVE The objective of the present study was to investigate the effect of vitamin A supplementation on serum Th17 (IL-6, IL-17, IFNγ) and Treg (TGF-β, IL-10) related cytokines in obese and non-obese women. SUBJECTS AND METHODS In a randomized double blind placebo controlled design, 56 obese women were randomly assigned to receive either an oral dose of 25,000 IU retinyl palmitate or placebo per day for 4 months. Twenty eight ages matched non-obese women were also received vitamin A. At the study entry, anthropometric variables were measured and serum Th17 and Treg related cytokine profile were determined at baseline and 4 months after intervention. RESULTS Significantly higher baseline concentrations of IL-6 were observed in obese compared with non-obese women (P < 0.05). However, the initial concentrations of other cytokines were not significantly different between groups. The mean concentrations of IL-17 and TGF-β were significantly decreased after vitamin A supplementation in non-obese and obese women respectively. Positive relationships between IL-17 and IL-10 (r = 0.42, P < 0.001), TGF-β and IL-17 (r = 0.35, P < 0.001) and between IL-10 and IFN-γ (r = 0.41, P = 0.002) in total participants were also observed. CONCLUSIONS The results of the present study showed for the first time that vitamin A supplementation reduces serum concentrations of IL-17 and TGF-β in reproductive age women. Further studies are needed to explore the possible underlying mechanisms.
Collapse
Affiliation(s)
| | - Ali Akbar Saboor-Yaraghi
- Department of Cellular and Molecular Nutrition, School of Nutrition and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyyed Ali Keshavarz
- Department of Clinical Nutrition, School of Nutrition and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
68
|
Yu HR, Tain YL, Sheen JM, Tiao MM, Chen CC, Kuo HC, Hung PL, Hsieh KS, Huang LT. Prenatal Dexamethasone and Postnatal High-Fat Diet Decrease Interferon Gamma Production through an Age-Dependent Histone Modification in Male Sprague-Dawley Rats. Int J Mol Sci 2016; 17:ijms17101610. [PMID: 27669212 PMCID: PMC5085643 DOI: 10.3390/ijms17101610] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/11/2016] [Accepted: 09/14/2016] [Indexed: 02/07/2023] Open
Abstract
Overexposure to prenatal glucocorticoid (GC) disturbs hypothalamic-pituitary-adrenocortical axis-associated neuroendocrine metabolism and susceptibility to metabolic syndrome. A high-fat (HF) diet is a major environmental factor that can cause metabolic syndrome. We aimed to investigate whether prenatal GC plus a postnatal HF diet could alter immune programming in rat offspring. Pregnant Sprague-Dawley rats were given intraperitoneal injections of dexamethasone or saline at 14-21 days of gestation. Male offspring were then divided into four groups: vehicle, prenatal dexamethasone exposure, postnatal HF diet (VHF), and prenatal dexamethasone exposure plus a postnatal HF diet (DHF). The rats were sacrificed and adaptive immune function was evaluated. Compared to the vehicle, the DHF group had lower interferon gamma (IFN-γ) production by splenocytes at postnatal day 120. Decreases in H3K9 acetylation and H3K36me3 levels at the IFN-γ promoter correlated with decreased IFN-γ production. The impaired IFN-γ production and aberrant site-specific histone modification at the IFN-γ promoter by prenatal dexamethasone treatment plus a postnatal HF diet resulted in resilience at postnatal day 180. Prenatal dexamethasone and a postnatal HF diet decreased IFN-γ production through a site-specific and an age-dependent histone modification. These findings suggest a mechanism by which prenatal exposure to GC and a postnatal environment exert effects on fetal immunity programming.
Collapse
Affiliation(s)
- Hong-Ren Yu
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - You-Lin Tain
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Jiunn-Ming Sheen
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Mao-Meng Tiao
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Chih-Cheng Chen
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Ho-Chang Kuo
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Pi-Lien Hung
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Kai-Sheng Hsieh
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Li-Tung Huang
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| |
Collapse
|
69
|
Yoshida K, Nakashima E, Kyoizumi S, Hakoda M, Hayashi T, Hida A, Ohishi W, Kusunoki Y. Metabolic Profile as a Potential Modifier of Long-Term Radiation Effects on Peripheral Lymphocyte Subsets in Atomic Bomb Survivors. Radiat Res 2016; 186:275-82. [PMID: 27541825 DOI: 10.1667/rr14336.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Immune system impairments reflected by the composition and function of circulating lymphocytes are still observed in atomic bomb survivors, and metabolic abnormalities including altered blood triglyceride and cholesterol levels have also been detected in such survivors. Based on closely related features of immune and metabolic profiles of individuals, we investigated the hypothesis that long-term effects of radiation exposure on lymphocyte subsets might be modified by metabolic profiles in 3,113 atomic bomb survivors who participated in health examinations at the Radiation Effect Research Foundation, Hiroshima and Nagasaki, in 2000-2002. The lymphocyte subsets analyzed involved T-, B- and NK-cell subsets, and their percentages in the lymphocyte fraction were assessed using flow cytometry. Health examinations included metabolic indicators, body mass index, serum levels of total cholesterol, high-density lipoprotein cholesterol, C-reactive protein and hemoglobin A1c, as well as diabetes and fatty liver diagnoses. Standard regression analyses indicated that several metabolic indicators of obesity/related disease, particularly high-density lipoprotein cholesterol levels, were positively associated with type-1 helper T- and B-cell percentages but were inversely associated with naïve CD4 T and NK cells. A regression analysis adjusted for high-density lipoprotein cholesterol revealed a radiation dose relationship with increasing NK-cell percentage. Additionally, an interaction effect was suggested between radiation dose and C-reactive protein on B-cell percentage with a negative coefficient of the interaction term. Collectively, these findings suggest that radiation exposure and subsequent metabolic profile changes, potentially in relationship to obesity-related inflammation, lead to such long-term alterations in lymphocyte subset composition. Because this study is based on cross-sectional and exploratory analyses, the implications regarding radiation exposure, metabolic profiles and circulating lymphocytes warrant future longitudinal and molecular mechanistic studies.
Collapse
Affiliation(s)
| | | | | | - Masayuki Hakoda
- e Department of Nutritional Sciences, Faculty of Human Ecology, Yasuda Women's University, Hiroshima, Japan
| | | | - Ayumi Hida
- d Department of Clinical Studies, Radiation Effects Research Foundation, Nagasaki, Japan; and
| | - Waka Ohishi
- c Clinical Studies, Radiation Effects Research Foundation, Hiroshima, Japan
| | | |
Collapse
|
70
|
High Fat Diet Inhibits Dendritic Cell and T Cell Response to Allergens but Does Not Impair Inhalational Respiratory Tolerance. PLoS One 2016; 11:e0160407. [PMID: 27483441 PMCID: PMC4970708 DOI: 10.1371/journal.pone.0160407] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 07/19/2016] [Indexed: 01/28/2023] Open
Abstract
The incidence of obesity has risen to epidemic proportions in recent decades, most commonly attributed to an increasingly sedentary lifestyle, and a ‘western’ diet high in fat and low in fibre. Although non-allergic asthma is a well-established co-morbidity of obesity, the influence of obesity on allergic asthma is still under debate. Allergic asthma is thought to result from impaired tolerance to airborne antigens, so-called respiratory tolerance. We sought to investigate whether a diet high in fats affects the development of respiratory tolerance. Mice fed a high fat diet (HFD) for 8 weeks showed weight gain, metabolic disease, and alteration in gut microbiota, metabolites and glucose metabolism compared to age-matched mice fed normal chow diet (ND). Respiratory tolerance was induced by repeated intranasal (i.n.) administration of ovalbumin (OVA), prior to induction of allergic airway inflammation (AAI) by sensitization with OVA in alum i.p. and subsequent i.n. OVA challenge. Surprisingly, respiratory tolerance was induced equally well in HFD and ND mice, as evidenced by decreased lung eosinophilia and serum OVA-specific IgE production. However, in a pilot study, HFD mice showed a tendency for impaired activation of airway dendritic cells and regulatory T cells compared with ND mice after induction of respiratory tolerance. Moreover, the capacity of lymph node cells to produce IL-5 and IL-13 after AAI was drastically diminished in HFD mice compared to ND mice. These results indicate that HFD does not affect the inflammatory or B cell response to an allergen, but inhibits priming of Th2 cells and possibly dendritic cell and regulatory T cell activation.
Collapse
|
71
|
Abstract
Adipose tissue has traditionally been defined as connective tissue that stores excess calories in the form of triacylglycerol. However, the physiologic functions attributed to adipose tissue are expanding, and it is now well established that adipose tissue is an endocrine gland. Among the endocrine factors elaborated by adipose tissue are the adipokines; hormones, similar in structure to cytokines, produced by adipose tissue in response to changes in adipocyte triacylglycerol storage and local and systemic inflammation. They inform the host regarding long-term energy storage and have a profound influence on reproductive function, blood pressure regulation, energy homeostasis, the immune response, and many other physiologic processes. The adipokines possess pro- and anti-inflammatory properties and play a critical role in integrating systemic metabolism with immune function. In calorie restriction and starvation, proinflammatory adipokines decline and anti-inflammatory adipokines increase, which informs the host of energy deficits and contributes to the suppression of immune function. In individuals with normal metabolic status, there is a balance of pro- and anti-inflammatory adipokines. This balance shifts to favor proinflammatory mediators as adipose tissue expands during the development of obesity. As a consequence, the proinflammatory status of adipose tissue contributes to a chronic low-grade state of inflammation and metabolic disorders associated with obesity. These disturbances are associated with an increased risk of metabolic disease, type 2 diabetes, cardiovascular disease, and many other pathological conditions. This review focuses on the impact of energy homeostasis on the adipokines in immune function.
Collapse
Affiliation(s)
- Peter Mancuso
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
72
|
Zarrati M, Salehi E, Razmpoosh E, Shoormasti RS, Hosseinzadeh-Attar MJ, Shidfar F. Relationship between leptin concentration and body fat with peripheral blood mononuclear cells cytokines among obese and overweight adults. Ir J Med Sci 2016; 186:133-142. [PMID: 27085343 DOI: 10.1007/s11845-016-1454-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 03/26/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Overweight and obesity has been suggested to be well correlated with altered levels of pro-inflammatory cytokines. AIM The purpose of this study is to assess the relationship of body fat mass (BFM), body fat percentage (BFP) and leptin levels with peripheral blood mononuclear cells (PBMCs) cytokines among obese and overweight adults. METHODS Eighty-two overweight and obese individuals were divided into two BMI-category groups (BMI <30 and BMI ≥30 kg/m2) in this study. Balanced blocked randomization was used based on their sex and BMI ranges. Fasting blood samples, PBMCs cytokines, leptin and anthropometric indices were measured and PBMCs were cultured. RESULTS Mean of leptin concentrations were 23.14 ± 4.07 and 28.25 ± 4.35 pg/ml among individuals with BMI <30 and BMI ≥30 kg/m2, respectively. The mean values of anthropometric measurements (all P < 0.001), the concentrations of TNF-α (P = 0.028) and IFN-γ (P = 0.029) were significantly higher among obese individuals. BFP had a significant positive correlation with leptin (P < 0.001, r = 0.445) and TGF-β (P = 0.03, r = 0.243). BFM has significant positive correlation with leptin (P < 0.001, r = 0.521). Leptin had a positive significant correlation with IFN-γ (p = 0.03, r = 0.251). CONCLUSIONS Regarding these results, we proved that BFP, BFM and leptin levels have significant correlations with some PBMC cytokines. Focusing on such strategies may lead to promises for alleviating obesity and its co-morbidities.
Collapse
Affiliation(s)
- M Zarrati
- School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - E Salehi
- Immunology Departments, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - E Razmpoosh
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Nutrition, Faculty of Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - R S Shoormasti
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - F Shidfar
- School of Public Health, Iran University of Medical Sciences, Tehran, Iran.
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
73
|
Aravindhan V, Madhumitha H. Metainflammation in Diabetic Coronary Artery Disease: Emerging Role of Innate and Adaptive Immune Responses. J Diabetes Res 2016; 2016:6264149. [PMID: 27610390 PMCID: PMC5004008 DOI: 10.1155/2016/6264149] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/19/2016] [Indexed: 02/06/2023] Open
Abstract
Globally, noncommunicable chronic diseases such as Type-2 Diabetes Mellitus (T2DM) and Coronary Artery Disease (CAD) are posing a major threat to the world. T2DM is known to potentiate CAD which had led to the coining of a new clinical entity named diabetic CAD (DM-CAD), leading to excessive morbidity and mortality. The synergistic interaction between these two comorbidities is through sterile inflammation which is now being addressed as metabolic inflammation or metainflammation, which plays a pivotal role during both early and late stages of T2DM and also serves as a link between T2DM and CAD. This review summarises the current concepts on the role played by both innate and adaptive immune responses in setting up metainflammation in DM-CAD. More specifically, the role played by innate pattern recognition receptors (PRRs) like Toll-like receptors (TLRs), NOD1-like receptors (NLRs), Rig-1-like receptors (RLRs), and C-type lectin like receptors (CLRs) and metabolic endotoxemia in fuelling metainflammation in DM-CAD would be discussed. Further, the role played by adaptive immune cells (Th1, Th2, Th17, and Th9 cells) in fuelling metainflammation in DM-CAD will also be discussed.
Collapse
Affiliation(s)
- Vivekanandhan Aravindhan
- Department of Genetics, Dr. ALM. PG. IBMS, University of Madras, Chennai 600113, India
- *Vivekanandhan Aravindhan:
| | - Haridoss Madhumitha
- AU-KBC Research Centre, MIT Campus of Anna University, Chennai 600044, India
| |
Collapse
|
74
|
Yamaguchi R, Yamamoto T, Sakamoto A, Ishimaru Y, Narahara S, Sugiuchi H, Yamaguchi Y. Chemokine profiles of human visceral adipocytes from cryopreserved preadipocytes: Neutrophil activation and induction of nuclear factor-kappa B repressing factor. Life Sci 2015; 143:225-30. [DOI: 10.1016/j.lfs.2015.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 07/18/2015] [Accepted: 11/11/2015] [Indexed: 12/31/2022]
|
75
|
Rau M, Schilling AK, Meertens J, Hering I, Weiss J, Jurowich C, Kudlich T, Hermanns HM, Bantel H, Beyersdorf N, Geier A. Progression from Nonalcoholic Fatty Liver to Nonalcoholic Steatohepatitis Is Marked by a Higher Frequency of Th17 Cells in the Liver and an Increased Th17/Resting Regulatory T Cell Ratio in Peripheral Blood and in the Liver. THE JOURNAL OF IMMUNOLOGY 2015; 196:97-105. [PMID: 26621860 DOI: 10.4049/jimmunol.1501175] [Citation(s) in RCA: 222] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 10/30/2015] [Indexed: 12/16/2022]
Abstract
Nonalcoholic fatty liver disease is increasing in prevalence. It can be subdivided into nonalcoholic fatty liver (NAFL) and nonalcoholic steatohepatitis (NASH). Five to twenty percent of cases progress from NAFL to NASH. Increased hepatic Th17 cells and IL-17 expression were observed in NASH mice and patients, respectively. We analyzed CD4(+) effector T cells and regulatory T cells (Tregs) from peripheral blood and livers of NAFL and NASH patients. A total of 51 NAFL patients, 30 NASH patients, 31 nonalcoholic fatty liver disease patients (without histology), and 43 healthy controls were included. FACS analysis was performed on PBMCs and intrahepatic lymphocytes. Compared with healthy controls, a lower frequency of resting Tregs (rTregs; CD4(+)CD45RA(+)CD25(++)) and higher frequencies of IFN-γ(+) and/or IL-4(+) cells were detected among CD4(+) T cells of peripheral blood in NASH, and to a lesser degree in NAFL. In hepatic tissue, NAFL to NASH progression was marked by an increase in IL-17(+) cells among intrahepatic CD4(+) T cells. To define immunological parameters in peripheral blood to distinguish NAFL from NASH, we calculated different ratios. Th17/rTreg and Th2/rTreg ratios were significantly increased in NASH versus NAFL. The relevance of our findings for NASH pathogenesis was highlighted by the normalization of all of the changes 1 y after bariatric surgery. In conclusion, our data indicate that NAFL patients show changes in their immune cell profile compared with healthy controls. NAFL to NASH progression is marked by an increased frequency of IL-17(+) cells among intrahepatic CD4(+) T cells and higher Th17/rTreg and Th2/rTreg ratios in peripheral blood.
Collapse
Affiliation(s)
- Monika Rau
- Division of Hepatology, Department of Internal Medicine II, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Anne-Kristin Schilling
- Division of Hepatology, Department of Internal Medicine II, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Jan Meertens
- Division of Hepatology, Department of Internal Medicine II, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Ilona Hering
- Division of Hepatology, Department of Internal Medicine II, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Johannes Weiss
- Division of Hepatology, Department of Internal Medicine II, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Christian Jurowich
- Department of General and Visceral Surgery, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Theodor Kudlich
- Division of Hepatology, Department of Internal Medicine II, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Heike M Hermanns
- Division of Hepatology, Department of Internal Medicine II, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Heike Bantel
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, 30625 Hannover, Germany; and
| | - Niklas Beyersdorf
- Institute for Virology and Immunobiology, University of Würzburg, 97080 Würzburg, Germany
| | - Andreas Geier
- Division of Hepatology, Department of Internal Medicine II, University Hospital Würzburg, 97080 Würzburg, Germany;
| |
Collapse
|
76
|
Sotoodeh Jahromi A, Sanie MS, Yusefi A, Zabetian H, Zareian P, Hakimelahi H, Madani A, Hojjat-Farsangi M. Association of Tumor Growth Factor-β and Interferon-γ Serum Levels With Insulin Resistance in Normal Pregnancy. Glob J Health Sci 2015; 8:25-32. [PMID: 26755467 PMCID: PMC4954908 DOI: 10.5539/gjhs.v8n6p25] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 08/19/2015] [Indexed: 11/28/2022] Open
Abstract
Pregnancy is related to change in glucose metabolism and insulin production. The aim of our study was to determine the association of serum IFN-γ and TGF-β levels with insulin resistance during normal pregnancy. This cross sectional study was carried out on 97 healthy pregnant (in different trimesters) and 28 healthy non-pregnant women. Serum TGF-β and IFN-γ level were measured by ELISA method. Pregnant women had high level TGF-β and low level IFN-γ as compared non-pregnant women. Maternal serum TGF-β concentration significantly increased in third trimester as compared first and second trimester of pregnancy. Maternal serum IFN-γ concentration significantly decreased in third trimester as compared first and second trimester of pregnancy. Pregnant women exhibited higher score of HOMA IR as compared non-pregnant women. There were association between gestational age with body mass index (r=0.28, P=0.005), TGF-β (r=0.45, P<0.001) and IFN-γ (r=-0.50, P<0.001). There was significant association between Insulin resistance and TGF-β (r=0.17, p=0.05). Our findings suggest that changes in maternal cytokine level in healthy pregnant women were anti-inflammatory. Furthermore, Tumor Growth Factor-β appears has a role in induction insulin resistance in healthy pregnant women. However, further studies needed to evaluate role of different cytokines on insulin resistance in normal pregnancy.
Collapse
|
77
|
Lorenz TK, Heiman JR, Demas GE. Sexual activity modulates shifts in TH1/TH2 cytokine profile across the menstrual cycle: an observational study. Fertil Steril 2015; 104:1513-21.e1-4. [PMID: 26385401 DOI: 10.1016/j.fertnstert.2015.09.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 09/01/2015] [Accepted: 09/01/2015] [Indexed: 01/31/2023]
Abstract
OBJECTIVE To investigate if sexual activity moderated menstrual cycle-related shifts in cytokines associated with T-helper type 1 (TH1) cells (e.g., interferon [IFN] γ) and T-helper type 2 (TH2) cells (e.g., interleukin [IL] 4). Immune activity shifts across the menstrual cycle, with higher follicular-phase TH1-cell activity but higher luteal-phase TH2-cell activity. Little is known about how social behaviors alter TH1-TH2 ratios, despite evidence that psychosocial factors can influence immunity. Of particular interest is how sexual activity influences immune responses that may support conception, such as the TH1-TH2 balance. DESIGN Participants provided saliva samples at four time points (menstrual, follicular, ovulatory, and luteal phases), which were assayed by means of ELISA. SETTING Academic laboratory. PARTICIPANT(S) Thirty healthy premenopausal women (16 sexually abstinent, 14 sexually active) not taking hormonal or immunoactive medications. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Salivary E2, P, IFN-γ, and IL-4. RESULT(S) Sexually active, but not abstinent, women were significantly more likely to express TH2-like cytokine ratios (IFN-γ < IL-4) in the luteal phase than in other phases. Similarly, sexually active women had significantly higher P, and higher P-E2 ratios, in the luteal phase than did abstinent women. The P-E2 ratio mediated menstrual variations in cytokine ratios in sexually active women. CONCLUSION(S) These results support the hypothesis that shifts in immune response across the menstrual cycle may reflect tradeoffs between reproduction and immunity. These findings point to the need for further research on the interaction between sexual behavior, the menstrual cycle, and immune response.
Collapse
Affiliation(s)
- Tierney K Lorenz
- Center for Integrative Study for Animal Behavior, Indiana University, Bloomington, Indiana; Kinsey Institute for Research on Sex, Gender, and Reproduction, Indiana University, Bloomington, Indiana.
| | - Julia R Heiman
- Center for Integrative Study for Animal Behavior, Indiana University, Bloomington, Indiana; Kinsey Institute for Research on Sex, Gender, and Reproduction, Indiana University, Bloomington, Indiana; Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana
| | - Gregory E Demas
- Center for Integrative Study for Animal Behavior, Indiana University, Bloomington, Indiana; Department of Biology, Indiana University, Bloomington, Indiana
| |
Collapse
|
78
|
Fang M, Li P, Wu X, Xu Y. Class II transactivator (CIITA) mediates transcriptional repression of pdk4 gene by interacting with hypermethylated in cancer 1 (HIC1). J Biomed Res 2015; 29:308-15. [PMID: 26243517 PMCID: PMC4547379 DOI: 10.7555/jbr.29.20150055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 05/09/2015] [Indexed: 12/30/2022] Open
Abstract
Increased accumulation and/or impaired utilization of fatty acid in extra-adipose tissues are implicated in the pathogenesis of insulin resistance and type 2 diabetes. Pyruvate dehydrogenase kinase 4 (Pdk4) is a key enzyme involved in fatty oxidation and energy expenditure, and its expression can be repressed by pro-inflammatory stimuli. Previously, we have shown that class II transactivator (CIITA) mediates the adverse effect of interferon gamma (IFN-γ) in skeletal muscle cells by cooperating with hypermethylated in cancer 1 (HIC1) to repress silent information regulator 1 (SIRT1) transcription. Building upon this finding, we report here that CIITA interacted with HIC1 via the GTP-binding domain (GBD) while HIC1 interacted with CIITA via the BTB/POZ domain. The GBD domain was required for CIITA to repress SIRT1 transcription probably acting as a bridge for CIITA to bind to HIC1 and consequently to bind to the SIRT1 promoter. IFN-γ stimulation, CIITA over-expression, or HIC1 over-expression repressed Pdk4 promoter activity while silencing either CIITA or HIC1 normalized Pdk4 expression in the presence of IFN-γ. An increase in SIRT1 expression or activity partially rescued Pdk4 expression in the presence of CIITA, but SIRT1 inhibition abrogated Pdk4 normalization even in the absence of CIITA. Taken together, our data have identified a HIC1-CIITA-SIRT1 axis that regulates Pdk4 transcription in response to IFN-γ stimulation.
Collapse
Affiliation(s)
- Mingming Fang
- Key Laboratory of Cardiovascular Disease, Department of Pathophysiology.,Department of Nursing, Jiangsu Jiankang Vocational University, Nanjing, Jiangsu 210029, China
| | - Ping Li
- Key Laboratory of Cardiovascular Disease, Department of Pathophysiology.,Department of Gastroenterology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, China
| | - Xiaoyan Wu
- Key Laboratory of Cardiovascular Disease, Department of Pathophysiology.,Laboratory Center for Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| | - Yong Xu
- Key Laboratory of Cardiovascular Disease, Department of Pathophysiology.,Laboratory Center for Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| |
Collapse
|
79
|
Adaptive Immunity and Antigen-Specific Activation in Obesity-Associated Insulin Resistance. Mediators Inflamm 2015; 2015:593075. [PMID: 26146464 PMCID: PMC4471324 DOI: 10.1155/2015/593075] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 05/11/2015] [Accepted: 05/13/2015] [Indexed: 02/07/2023] Open
Abstract
Type 2 diabetes mellitus (T2D) is a metabolic disease that is strongly tied to obesity and often preceded by insulin resistance (IR). It has been established that chronic inflammation of hypertrophic adipose tissue depots in obese individuals leads to obesity-associated IR and is mediated by cells of the innate immune system, particularly macrophages. More recently, cells of the adaptive immune system, B and T lymphocytes, have also emerged as important regulators of glucose homeostasis, raising the intriguing possibility that antigen-driven immune responses play a role in disease. In this review, we critically evaluate the roles that various B and T cell subsets play in IR, and then we examine the data suggesting that antigen-driven mechanisms, such as antigen presentation and costimulation, may drive the activity of these lymphocytes.
Collapse
|
80
|
Roy S, Chaudhuri TK. Assessment of Th1 and Th2 cytokine modulatory activity of an edible fern,Diplazium esculentum. FOOD AGR IMMUNOL 2015. [DOI: 10.1080/09540105.2015.1007449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
81
|
La leptine : un modulateur de l’activité des cellules Natural Killer ? NUTR CLIN METAB 2015. [DOI: 10.1016/j.nupar.2014.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
82
|
Ip BC, Hogan AE, Nikolajczyk BS. Lymphocyte roles in metabolic dysfunction: of men and mice. Trends Endocrinol Metab 2015; 26:91-100. [PMID: 25573740 PMCID: PMC4315738 DOI: 10.1016/j.tem.2014.12.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 11/21/2014] [Accepted: 12/05/2014] [Indexed: 02/07/2023]
Abstract
Type 2 diabetes (T2D) is a metabolic disease associated with obesity-related insulin resistance (IR) and chronic inflammation. Animal studies indicate that IR can be caused and/or exacerbated by systemic and/or tissue-specific alterations in lymphocyte differentiation and function. Human studies also indicate that obesity-associated inflammation promotes IR. Nevertheless, clinical trials with anti-inflammatory therapies have yielded modest impacts on established T2D. Unlike mouse models, where obesity is predominantly associated with IR, 20-25% of obese humans are metabolically healthy with high insulin sensitivity. The uncoupling of obesity from IR in humans but not in animal models advocates for a more comprehensive understanding of mediators and mechanisms of human obesity-promoted IR, and better integration of knowledge from human studies into animal experiments to efficiently pursue T2D prevention and treatment.
Collapse
Affiliation(s)
- Blanche C Ip
- Department of Microbiology, Boston University, Boston, MA, USA
| | - Andrew E Hogan
- Obesity Immunology Group, Education and Research Centre, St Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Barbara S Nikolajczyk
- Department of Microbiology, Boston University, Boston, MA, USA; Department of Medicine, Boston University, Boston, MA, USA.
| |
Collapse
|
83
|
3-Hydroxykynurenic Acid and Type 2 Diabetes: Implications for Aging, Obesity, Depression, Parkinson’s Disease, and Schizophrenia. TRYPTOPHAN METABOLISM: IMPLICATIONS FOR BIOLOGICAL PROCESSES, HEALTH AND DISEASE 2015. [DOI: 10.1007/978-3-319-15630-9_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
84
|
Inzaugarat ME, Billordo LA, Vodánovich F, Cervini GM, Casavalle PL, Vedire C, Cherñavsky AC. Alterations in innate and adaptive immune leukocytes are involved in paediatric obesity. Pediatr Obes 2014; 9:381-90. [PMID: 23852831 DOI: 10.1111/j.2047-6310.2013.00179.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 03/26/2013] [Accepted: 04/12/2013] [Indexed: 11/30/2022]
Abstract
BACKGROUND Adipose tissue is the main source of the cytokines and adipokines that are increased in the context of obesity. The production of reactive oxygen species (ROS) and cytokines by circulating immune cells can be regulated by these pro-inflammatory factors even before infiltration into adipose tissue. OBJECTIVE To investigate the alterations that can occur in circulating monocytes and lymphocytes in paediatric obesity. METHODS In this study, 54 paediatric obese patients and 30 age-matched metabolically healthy individuals were enrolled. Intracellular cytokines were analyzed after phorbol myristate acetate (PMA) or leptin plus PMA stimulation of lymphocytes and monocytes by flow cytometry. ROS generation was measured using dichlorofluorescein-diacetate. Both a 'stimulation index' and a 'fold of increase' were calculated for statistical purposes. RESULTS Both interferon gamma (IFN-γ) production by circulating CD4+ and CD8+ lymphocytes and ROS production by monocytes following PMA stimulation were increased in obese patients. Leptin induced an increased production of IFN-γ in both subsets of T cells and tumour necrosis factor alpha in monocytes, and linoleic acid induced a higher ROS production in monocytes. CONCLUSIONS The distinct functional responses of circulating cells suggest that alterations in both innate and adaptive immune cells are involved in the maintenance of low-grade inflammation in paediatric obesity.
Collapse
Affiliation(s)
- M E Inzaugarat
- Instituto de Inmunología, Genética y Metabolismo, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
85
|
Abstract
Adipose tissue (AT) lies at the crossroad of nutrition, metabolism, and immunity; AT inflammation was proposed as a central mechanism connecting obesity with its metabolic and vascular complications. Resident immune cells constitute the second largest AT cellular component after adipocytes and as such play important roles in the maintenance of AT homeostasis. Obesity-induced changes in their number and activity result in the activation of local and later systemic inflammatory response, marking the transition from simple adiposity to diseases such as type 2 diabetes mellitus, arterial hypertension, and ischemic heart disease. This review has focused on the various subsets of immune cells in AT and their role in the development of AT inflammation and obesity-induced insulin resistance.
Collapse
Affiliation(s)
- Milos Mraz
- Third Department of Medicine - Department of Endocrinology and MetabolismGeneral University Hospital, First Faculty of Medicine of Charles University in Prague, U nemocnice 1, 128 00 Prague 2, Czech Republic
| | - Martin Haluzik
- Third Department of Medicine - Department of Endocrinology and MetabolismGeneral University Hospital, First Faculty of Medicine of Charles University in Prague, U nemocnice 1, 128 00 Prague 2, Czech Republic
| |
Collapse
|
86
|
Suppressor of cytokine signalling (SOCS) proteins as guardians of inflammatory responses critical for regulating insulin sensitivity. Biochem J 2014; 461:177-88. [PMID: 24966052 DOI: 10.1042/bj20140143] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Overactivation of immune pathways in obesity is an important cause of insulin resistance and thus new approaches aimed to limit inflammation or its consequences may be effective for treating Type 2 diabetes. The SOCS (suppressors of cytokine signalling) are a family of proteins that play an essential role in mediating inflammatory responses in both immune cells and metabolic organs such as the liver, adipose tissue and skeletal muscle. In the present review we discuss the role of SOCS1 and SOCS3 in controlling immune cells such as macrophages and T-cells and the impact this can have on systemic inflammation and insulin resistance. We also dissect the mechanisms by which SOCS (1-7) regulate insulin signalling in different tissues including their impact on the insulin receptor and insulin receptor substrates. Lastly, we discuss the important findings from SOCS whole-body and tissue-specific null mice, which implicate an important role for these proteins in controlling insulin action and glucose homoeostasis in obesity.
Collapse
|
87
|
Manti S, Romano C, Chirico V, Filippelli M, Cuppari C, Loddo I, Salpietro C, Arrigo T. Nonalcoholic Fatty liver disease/non-alcoholic steatohepatitis in childhood: endocrine-metabolic "mal-programming". HEPATITIS MONTHLY 2014; 14:e17641. [PMID: 24829591 PMCID: PMC4013495 DOI: 10.5812/hepatmon.17641] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 02/13/2014] [Accepted: 02/19/2014] [Indexed: 02/08/2023]
Abstract
CONTEXT Nonalcoholic Fatty Liver Disease (NAFLD) is the major chronic liver disease in the pediatric population. NAFLD includes a broad spectrum of abnormalities (inflammation, fibrosis and cirrhosis), ranging from accumulation of fat (also known as steatosis) towards non-alcoholic steatohepatitis (NASH). The development of NAFLD in children is significantly increased. EVIDENCE ACQUISITION A literature search of electronic databases was undertaken for the major studies published from 1998 to today. The databases searched were: PubMed, EMBASE, Orphanet, Midline and Cochrane Library. We used the key words: "non-alcoholic fatty liver disease, children, non-alcoholic steatohepatitis and fatty liver". RESULTS NAFLD/NASH is probably promoted by "multiple parallel hits": environmental and genetic factors, systemic immunological disorders (oxidative stress, persistent-low grade of inflammation) as well as obesity and metabolic alterations (insulin resistance and metabolic syndrome). However its exact cause still underdiagnosed and unknown. CONCLUSIONS Pediatric NAFLD/NASH is emerging problem. Longitudinal follow-up studies, unfortunately still insufficient, are needed to better understand the natural history and outcome of NAFLD in children. This review focuses on the current knowledge regarding the epidemiology, pathogenesis, environmental, genetic and metabolic factors of disease. The review also highlights the importance of studying the underlying mechanisms of pediatric NAFLD and the need for complete and personalized approach in the management of NAFLD/NASH.
Collapse
Affiliation(s)
- Sara Manti
- Department of Pediatric Sciences, Genetics and Pediatric Immunology Unit, University of Messina, Messina, Italy
| | - Claudio Romano
- Department of Pediatric Sciences, Genetics and Pediatric Immunology Unit, University of Messina, Messina, Italy
| | - Valeria Chirico
- Department of Pediatric Sciences, Genetics and Pediatric Immunology Unit, University of Messina, Messina, Italy
| | - Martina Filippelli
- Department of Pediatric Sciences, Genetics and Pediatric Immunology Unit, University of Messina, Messina, Italy
| | - Caterina Cuppari
- Department of Pediatric Sciences, Genetics and Pediatric Immunology Unit, University of Messina, Messina, Italy
| | - Italia Loddo
- Department of Pediatric Sciences, Genetics and Pediatric Immunology Unit, University of Messina, Messina, Italy
| | - Carmelo Salpietro
- Department of Pediatric Sciences, Genetics and Pediatric Immunology Unit, University of Messina, Messina, Italy
| | - Teresa Arrigo
- Department of Pediatric Sciences, Genetics and Pediatric Immunology Unit, University of Messina, Messina, Italy
| |
Collapse
|
88
|
Pecht T, Gutman-Tirosh A, Bashan N, Rudich A. Peripheral blood leucocyte subclasses as potential biomarkers of adipose tissue inflammation and obesity subphenotypes in humans. Obes Rev 2014; 15:322-37. [PMID: 24251825 DOI: 10.1111/obr.12133] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Revised: 10/01/2013] [Accepted: 10/18/2013] [Indexed: 12/14/2022]
Abstract
While obesity is clearly accepted as a major risk factor for cardio-metabolic morbidity, it is also apparent that some obese patients largely escape this association, forming a unique obese subphenotype(s). Current approaches to define such subphenotypes include clinical biomarkers that largely reflect already manifested comorbidities, such as markers of dyslipidaemia, hyperglycaemia and impaired regulation of vascular tone, and anthropometric or imaging-based assessment of adipose tissue distribution. Low-grade inflammation, evident both systemically and within adipose tissue (particularly intra-abdominal fat depots), seems to characterize the more cardio-metabolically morbid forms of obesity. Indeed, several systemic inflammatory markers (C-reactive protein), adipokines (retinol-binding protein 4, adiponectin) and cytokines have been shown to correlate in humans with adipose tissue inflammation and with obesity-associated health risks. Circulating leucocytes constitute a diverse group of cells that form a major arm of the immune system. They are both major sources of cytokines and likely also of infiltrating adipose tissue immune cells in obesity. In the present review, we summarize currently available literature on 'classical' blood white cell classes and on more specific leucocyte subclasses present in the circulation in human obesity. We critically raise the possibility that leucocytes may constitute clinically available markers for the more morbidity-associated obesity subphenotype(s), and when available, for intra-abdominal adipose tissue inflammation.
Collapse
Affiliation(s)
- T Pecht
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel; The National Institute of Biotechnology (NIBN) in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | | |
Collapse
|
89
|
The Influence of an Obesogenic Diet on Oxysterol Metabolism in C57BL/6J Mice. CHOLESTEROL 2014; 2014:843468. [PMID: 24672716 PMCID: PMC3941159 DOI: 10.1155/2014/843468] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Revised: 12/18/2013] [Accepted: 12/21/2013] [Indexed: 11/18/2022]
Abstract
Our current understanding of oxysterol metabolism during different disease states such as obesity and dyslipidemia is limited. Therefore, the aim of this study was to determine the effect of diet-induced obesity on the tissue distribution of various oxysterols and the mRNA expression of key enzymes involved in oxysterol metabolism. To induce obesity, male C57BL/6J mice were fed a high fat-cholesterol diet for 24 weeks. Following diet-induced obesity, plasma levels of 4β-hydroxycholesterol, 5,6α-epoxycholesterol, 5,6β-epoxycholesterol, 7α-hydroxycholesterol, 7β-hydroxycholesterol, and 27-hydroxycholesterol were significantly (P < 0.05) increased. In the liver and adipose tissue of the obese mice, 4β-hydroxycholesterol was significantly (P < 0.05) increased, whereas 27-hydroxycholesterol was increased only in the adipose tissue. No significant changes in either hepatic or adipose tissue mRNA expression were observed for oxysterol synthesizing enzymes 4β-hydroxylase, 27-hydroxylase, or 7α-hydroxylase. Hepatic mRNA expression of SULT2B1b, a key enzyme involved in oxysterol detoxification, was significantly (P < 0.05) elevated in the obese mice. Interestingly, the appearance of the large HDL1 lipoprotein was observed with increased oxysterol synthesis during obesity. In diet-induced obese mice, dietary intake and endogenous enzymatic synthesis of oxysterols could not account for the increased oxysterol levels, suggesting that nonenzymatic cholesterol oxidation pathways may be responsible for the changes in oxysterol metabolism.
Collapse
|
90
|
Madhumitha H, Mohan V, Deepa M, Babu S, Aravindhan V. Increased Th1 and suppressed Th2 serum cytokine levels in subjects with diabetic coronary artery disease. Cardiovasc Diabetol 2014; 13:1. [PMID: 24383855 PMCID: PMC3893369 DOI: 10.1186/1475-2840-13-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 12/21/2013] [Indexed: 01/07/2023] Open
Abstract
Background The role played by T helper cytokines under chronic, low grade inflammation as seen in type-2 Diabetes Mellitus (T2DM) and Coronary Artery Disease (CAD) co-morbidity is less well studied. In the present study, we measured the serum levels of both Th1 and Th2 cytokines and correlated it with clinical risk factors for T2DM (Insulin Resistance (IR), Glycated haemoglobin (HbA1c)) and CAD (C-Reactive Protein (CRP), Intima Media Thickness (IMT) and Augmentation index (AGI)) in T2DM subjects with/without CAD. Methodology The study subjects were recruited from Chennai Urban Rural Epidemiology Study (CURES). Serum cytokine profile was determined by multiplex cytokine assay in Control (n = 61), T2DM (n = 60), CAD (n = 23) and T2DM-CAD (n = 21) subjects. Results T2DM subjects showed a mixed Th1-Th2 profile. CAD subjects presented a Th1 profile with modest Th2 suppression while T2DM-CAD subjects showed enhanced Th1 profile with strong suppression of Th2 cytokines. Both Th1 and Th2 cytokines showed a positive correlation with FPG, HbA1c, hsCRP, IMT and AGI. Logistic regression analysis revealed a significant association of IL-12 (OR = 9.3; 95% CI = 3.2-70.7; p = 0.016), IFN-γ (OR = 2.8; 95% CI = 2.7-2.9, p = 0.010), IL-4 (OR = 2.7; 95% CI 2.7-2.7, p = 0.010), IL-5 (OR = 1.1; 95% CI = 1.0-1.4; p = 0.003) and IL-13 (OR = 2; 95% CI = 1.7-2.6; p = 0.017) with T2DM-CAD. Conclusion In conclusion, from the present study it appears that transition from T2DM or CAD to T2DM-CAD co-morbidity is associated with strong down regulation of Th2 cytokines and enhancement of Th1 responses.
Collapse
|
91
|
CEACAM1 loss links inflammation to insulin resistance in obesity and non-alcoholic steatohepatitis (NASH). Semin Immunopathol 2013; 36:55-71. [PMID: 24258517 DOI: 10.1007/s00281-013-0407-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 10/13/2013] [Indexed: 02/06/2023]
Abstract
Mounting epidemiological evidence points to an association between metabolic syndrome and non-alcoholic steatohepatitis (NASH), an increasingly recognized new epidemic. NASH pathologies include hepatocellular ballooning, lobular inflammation, hepatocellular injury, apoptosis, and hepatic fibrosis. We will review the relationship between insulin resistance and inflammation in visceral obesity and NASH in an attempt to shed more light on the pathogenesis of these major metabolic diseases. Moreover, we will identify loss of the carcinoembryonic antigen-related cell adhesion molecule 1 as a unifying mechanism linking the immunological and metabolic abnormalities in NASH.
Collapse
|
92
|
Dao MC, Meydani SN. Iron biology, immunology, aging, and obesity: four fields connected by the small peptide hormone hepcidin. Adv Nutr 2013; 4:602-17. [PMID: 24228190 PMCID: PMC3823507 DOI: 10.3945/an.113.004424] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Iron status and immune response become impaired in situations that involve chronic inflammation, such as obesity or aging. Little is known, however, about the additional burden that obesity may place on the iron status and immune response in the elderly. This question is relevant given the rising numbers of elderly obese (BMI >30 kg/m(2)) individuals and the high prevalence of iron deficiency worldwide. Iron is necessary for proper function of both the innate and adaptive immune system. Hepcidin, a peptide hormone that regulates cellular iron export, is essential for the maintenance of iron homeostasis. Therefore, since immune cells require iron for proper function hepcidin may also play an important role in immune response. In this review, we summarize the evidence for hepcidin as a link between the fields of gerontology, obesity, iron biology, and immunology. We also identify several gaps in knowledge and unanswered questions pertaining to iron homeostasis and immunity in obese populations. Finally, we review studies that have shown the impact of weight loss, focusing on calorie restriction, iron homeostasis, and immunity. These studies are important both in elucidating mechanistic links between obesity and health impairments and identifying possible approaches to target immune impairment and iron deficiency as comorbidities of obesity.
Collapse
Affiliation(s)
- Maria Carlota Dao
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Nutrition Immunology Laboratory, Boston, MA
| | - Simin Nikbin Meydani
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Nutrition Immunology Laboratory, Boston, MA
| |
Collapse
|
93
|
Vonghia L, Michielsen P, Francque S. Immunological mechanisms in the pathophysiology of non-alcoholic steatohepatitis. Int J Mol Sci 2013; 14:19867-90. [PMID: 24084730 PMCID: PMC3821591 DOI: 10.3390/ijms141019867] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 09/11/2013] [Accepted: 09/22/2013] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is characterized by the presence of steatosis, inflammation and hepatocyte injury and constitutes hepatic manifestation of the metabolic syndrome. The pathogenesis of NASH is complex and implicates cross-talk between different metabolically active sites, such as liver and adipose tissue. Obesity is considered a chronic low-grade inflammatory state and the liver has been recognized as being an "immunological organ". The complex role of the immune system in the pathogenesis of NASH is currently raising great interest, also in view of the possible therapeutic potential of immunotherapy in NASH. This review focuses on the disturbances of the cells constituting the innate and adaptive immune system in the liver and in adipose tissue.
Collapse
Affiliation(s)
- Luisa Vonghia
- Department of Gastroenterology and Hepatology, University Hospital Antwerp, Wilrijkstraat 10, Edegem 2650, Belgium; E-Mails: (P.M.); (S.F.)
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, Policlinico, Piazza Giulio Cesare, Bari 70100, Italy
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +32-3821-3323; Fax: +32-3821-4478
| | - Peter Michielsen
- Department of Gastroenterology and Hepatology, University Hospital Antwerp, Wilrijkstraat 10, Edegem 2650, Belgium; E-Mails: (P.M.); (S.F.)
| | - Sven Francque
- Department of Gastroenterology and Hepatology, University Hospital Antwerp, Wilrijkstraat 10, Edegem 2650, Belgium; E-Mails: (P.M.); (S.F.)
| |
Collapse
|
94
|
Baumann S, Lorentz A. Obesity - a promoter of allergy? Int Arch Allergy Immunol 2013; 162:205-13. [PMID: 24021931 DOI: 10.1159/000353972] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The prevalence of both obesity and allergy has been increasing throughout the world, leading to the hypothesis that the two are linked to one another. This overview summarizes the results of 34 studies from 2002 to 2012 that investigated a possible contributing effect of increasing body mass on the development and prevalence of various atopic diseases. Obesity was found to clearly affect bronchial asthma. However, the correlation was stronger in the nonatopic asthma phenotype. Obesity was found to be associated with the development of atopic dermatitis in children only. No clear association was found between obesity and the prevalence of allergic rhinitis or allergic conjunctivitis or increased sensitization to food allergens. This review sums up our study results and discusses a possible role of obesity in the promotion of allergy and asthma.
Collapse
Affiliation(s)
- Susanne Baumann
- Department of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | | |
Collapse
|
95
|
Farhangi MA, Keshavarz SA, Eshraghian M, Ostadrahimi A, Saboor-Yaraghi AA. Vitamin A Supplementation and Serum Th1- and Th2-Associated Cytokine Response in Women. J Am Coll Nutr 2013; 32:280-5. [DOI: 10.1080/07315724.2013.816616] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
96
|
Cildir G, Akıncılar SC, Tergaonkar V. Chronic adipose tissue inflammation: all immune cells on the stage. Trends Mol Med 2013; 19:487-500. [PMID: 23746697 DOI: 10.1016/j.molmed.2013.05.001] [Citation(s) in RCA: 223] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 05/03/2013] [Accepted: 05/06/2013] [Indexed: 12/12/2022]
Abstract
Inflammation is indispensable for host homeostasis against invading pathogens and efficient wound healing upon tissue malfunction and has to be tightly controlled by various mechanisms to limit excess responses harmful to host tissues. A myriad of disease conditions ranging from type 2 diabetes (T2D) to neurodegenerative and cardiovascular disorders are now shown to progress due to persistent, unresolved inflammation in metabolic tissues such as adipose, liver, pancreas, muscle, and brain. However, their underlying mechanisms are incompletely understood. The actions of innate and adaptive immune cells in these ailments are increasingly appreciated so much so that a new research area called 'immunometabolism' has emerged. In this review, we will highlight the fundamental roles of various immune cells in adipose tissue during the initiation and progression of obesity-induced inflammation and discuss potential anti-inflammatory therapies from different mechanistic points of view.
Collapse
Affiliation(s)
- Gökhan Cildir
- Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology (IMCB), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | | | | |
Collapse
|
97
|
Class II major histocompatibility complex plays an essential role in obesity-induced adipose inflammation. Cell Metab 2013; 17:411-22. [PMID: 23473035 PMCID: PMC3619392 DOI: 10.1016/j.cmet.2013.02.009] [Citation(s) in RCA: 305] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 12/21/2012] [Accepted: 02/12/2013] [Indexed: 01/01/2023]
Abstract
Adipose-resident T cells (ARTs) regulate metabolic and inflammatory responses in obesity, but ART activation signals are poorly understood. Here, we describe class II major histocompatibility complex (MHCII) as an important component of high-fat-diet (HFD)-induced obesity. Microarray analysis of primary adipocytes revealed that multiple genes involved in MHCII antigen processing and presentation increased in obese women. In mice, adipocyte MHCII increased within 2 weeks on HFD, paralleling increases in proinflammatory ART markers and decreases in anti-inflammatory ART markers, and preceding adipose tissue macrophage (ATM) accumulation and proinflammatory M1 polarization. Mouse 3T3-L1 and primary adipocytes activated T cells in an antigen-specific, contact-dependent manner, indicating that adipocyte MHCII is functional. HFD-fed MHCII(-/-) mice developed less adipose inflammation and insulin resistance than did wild-type mice, despite developing similar adiposity. These investigations uncover a mechanism whereby a HFD-induced adipocyte/ART dialog involving MHCII instigates adipose inflammation and, together with ATM MHCII, escalates its progression.
Collapse
|
98
|
Ippoliti F, Canitano N, Businaro R. Stress and obesity as risk factors in cardiovascular diseases: a neuroimmune perspective. J Neuroimmune Pharmacol 2013; 8:212-26. [PMID: 23329173 DOI: 10.1007/s11481-012-9432-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 12/28/2012] [Indexed: 01/09/2023]
Abstract
Obesity is now growing at an alarming rate reaching epidemic proportions worldwide thus increasing morbidity and mortality rates for chronic disease. But although we have ample information on the complications associated with obesity, precisely what causes obesity remains poorly understood. Some evidence attributes a major role to a low-grade chronic inflammatory state (neurogenic inflammation) induced in obesity by inflammatory mediators produced and secreted within the expanded activated adipocyte pool. Adipose tissue is an endocrine organ that secretes numerous adipose tissue-specific or enriched hormones, known as adipokines, cytokine-like molecules thought to play a pathogenic role in cardiovascular diseases. The imbalance between increased inflammatory stimuli and decreased anti-inflammatory mechanisms may depend on chronic stress. Hence the positive correlation found between stress, obesity and cardiovascular diseases. The chronic inflammatory state associated with insulin resistance and endothelial dysfunction is highly deleterious for vascular function. This review focuses on the proposed neuroimmunodulatory mechanisms linking chronic (psychological) stress, obesity and cardiovascular diseases.
Collapse
Affiliation(s)
- Flora Ippoliti
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy.
| | | | | |
Collapse
|
99
|
Serum IL-12 is increased in Mexican obese subjects and associated with low-grade inflammation and obesity-related parameters. Mediators Inflamm 2013; 2013:967067. [PMID: 23533314 PMCID: PMC3590791 DOI: 10.1155/2013/967067] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 01/08/2013] [Accepted: 01/09/2013] [Indexed: 12/15/2022] Open
Abstract
Interleukin-(IL-) 12 has been recently suggested to participate during development of insulin resistance in obese mice. Nevertheless, serum IL-12 levels have not been accurately determined in overweight and obese humans. We thus studied serum concentrations of IL-12 in Mexican adult individuals, examining their relationship with low-grade inflammation and obesity-related parameters. A total of 147 healthy individuals, 43 normal weight, 61 overweight, and 43 obese subjects participated in the study. Circulating levels of IL-12, tumor necrosis factor-alpha (TNF-α), leptin, insulin, glucose, total cholesterol, and triglyceride were measured after overnight fasting in all of the study subjects. Waist circumference and body fat percentage were recorded for all the participants. Serum IL-12 was significantly higher in overweight and obese individuals than in normal weight controls. Besides being strongly related with body mass index (r = 0.5154), serum IL-12 exhibited a significant relationship with abdominal obesity (r = 0.4481), body fat percentage (r = 0.5625), serum glucose (r = 0.3158), triglyceride (r = 0.3714), and TNF-α (r = 0.4717). Thus, serum levels of IL-12 are increased in overweight and obese individuals and show a strong relationship with markers of low-grade inflammation and obesity in the Mexican adult population. Further research is needed to understand the role of IL-12 in developing obesity-associated alterations in humans.
Collapse
|
100
|
Nikolajczyk BS, Jagannathan-Bogdan M, Denis GV. The outliers become a stampede as immunometabolism reaches a tipping point. Immunol Rev 2013; 249:253-75. [PMID: 22889227 DOI: 10.1111/j.1600-065x.2012.01142.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Obesity and Type 2 diabetes mellitus (T2D) are characterized by pro-inflammatory alterations in the immune system including shifts in leukocyte subset differentiation and in cytokine/chemokine balance. The chronic, low-grade inflammation resulting largely from changes in T-cell, B-cell, and myeloid compartments promotes and/or exacerbates insulin resistance (IR) that, together with pancreatic islet failure, defines T2D. Animal model studies show that interruption of immune cell-mediated inflammation by any one of several methods almost invariably results in the prevention or delay of obesity and/or IR. However, anti-inflammatory therapies have had a modest impact on established T2D in clinical trials. These seemingly contradictory results indicate that a more comprehensive understanding of human IR/T2D-associated immune cell function is needed to leverage animal studies into clinical treatments. Important outstanding analyses include identifying potential immunological checkpoints in disease etiology, detailing immune cell/adipose tissue cross-talk, and defining strengths/weaknesses of model organism studies to determine whether we can harness the promising new field of immunometabolism to curb the global obesity and T2D epidemics.
Collapse
|