1
|
Hemat Jouy S, Mohan S, Scichilone G, Mostafa A, Mahmoud AM. Adipokines in the Crosstalk between Adipose Tissues and Other Organs: Implications in Cardiometabolic Diseases. Biomedicines 2024; 12:2129. [PMID: 39335642 PMCID: PMC11428859 DOI: 10.3390/biomedicines12092129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Adipose tissue was previously regarded as a dormant organ for lipid storage until the identification of adiponectin and leptin in the early 1990s. This revelation unveiled the dynamic endocrine function of adipose tissue, which has expanded further. Adipose tissue has emerged in recent decades as a multifunctional organ that plays a significant role in energy metabolism and homeostasis. Currently, it is evident that adipose tissue primarily performs its function by secreting a diverse array of signaling molecules known as adipokines. Apart from their pivotal function in energy expenditure and metabolism regulation, these adipokines exert significant influence over a multitude of biological processes, including but not limited to inflammation, thermoregulation, immune response, vascular function, and insulin sensitivity. Adipokines are pivotal in regulating numerous biological processes within adipose tissue and facilitating communication between adipose tissue and various organs, including the brain, gut, pancreas, endothelial cells, liver, muscle, and more. Dysregulated adipokines have been implicated in several metabolic diseases, like obesity and diabetes, as well as cardiovascular diseases. In this article, we attempted to describe the significance of adipokines in developing metabolic and cardiovascular diseases and highlight their role in the crosstalk between adipose tissues and other tissues and organs.
Collapse
Affiliation(s)
- Shaghayegh Hemat Jouy
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Central Tehran Branch, Islamic Azad University, Tehran 14778-93855, Iran;
| | - Sukrutha Mohan
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (S.M.); (G.S.)
| | - Giorgia Scichilone
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (S.M.); (G.S.)
| | - Amro Mostafa
- Department of Pharmacology, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA;
| | - Abeer M. Mahmoud
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; (S.M.); (G.S.)
- Department of Kinesiology and Nutrition, College of Applied Health Sciences, University of Illinois Chicago, Chicago, IL 60612, USA
| |
Collapse
|
2
|
Zhuang Z, Zhou P, Wang J, Lu X, Chen Y. The Characteristics, Mechanisms and Therapeutics: Exploring the Role of Gut Microbiota in Obesity. Diabetes Metab Syndr Obes 2023; 16:3691-3705. [PMID: 38028999 PMCID: PMC10674108 DOI: 10.2147/dmso.s432344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/09/2023] [Indexed: 12/01/2023] Open
Abstract
Presently, obesity has emerged as a significant global public health concern due to its escalating prevalence and incidence rates. The gut microbiota, being a crucial environmental factor, has emerged as a key player in the etiology of obesity. Nevertheless, the intricate and specific interactions between obesity and gut microbiota, along with the underlying mechanisms, remain incompletely understood. This review comprehensively summarizes the gut microbiota characteristics in obesity, the mechanisms by which it induces obesity, and explores targeted therapies centered on gut microbiota restoration.
Collapse
Affiliation(s)
- Zequn Zhuang
- Department of General Surgery, the Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi, People’s Republic of China
| | - Peng Zhou
- Department of General Surgery, the Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi, People’s Republic of China
| | - Jing Wang
- Jiangnan University Medical Center, Wuxi, People’s Republic of China
| | - Xiaojing Lu
- Department of General Surgery, the Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi, People’s Republic of China
| | - Yigang Chen
- Department of General Surgery, the Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi, People’s Republic of China
- Jiangnan University Medical Center, Wuxi, People’s Republic of China
- Wuxi Clinical College, Nantong University, Wuxi, People’s Republic of China
| |
Collapse
|
3
|
Zhao X, An X, Yang C, Sun W, Ji H, Lian F. The crucial role and mechanism of insulin resistance in metabolic disease. Front Endocrinol (Lausanne) 2023; 14:1149239. [PMID: 37056675 PMCID: PMC10086443 DOI: 10.3389/fendo.2023.1149239] [Citation(s) in RCA: 146] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
Insulin resistance (IR) plays a crucial role in the development and progression of metabolism-related diseases such as diabetes, hypertension, tumors, and nonalcoholic fatty liver disease, and provides the basis for a common understanding of these chronic diseases. In this study, we provide a systematic review of the causes, mechanisms, and treatments of IR. The pathogenesis of IR depends on genetics, obesity, age, disease, and drug effects. Mechanistically, any factor leading to abnormalities in the insulin signaling pathway leads to the development of IR in the host, including insulin receptor abnormalities, disturbances in the internal environment (regarding inflammation, hypoxia, lipotoxicity, and immunity), metabolic function of the liver and organelles, and other abnormalities. The available therapeutic strategies for IR are mainly exercise and dietary habit improvement, and chemotherapy based on biguanides and glucagon-like peptide-1, and traditional Chinese medicine treatments (e.g., herbs and acupuncture) can also be helpful. Based on the current understanding of IR mechanisms, there are still some vacancies to follow up and consider, and there is also a need to define more precise biomarkers for different chronic diseases and lifestyle interventions, and to explore natural or synthetic drugs targeting IR treatment. This could enable the treatment of patients with multiple combined metabolic diseases, with the aim of treating the disease holistically to reduce healthcare expenditures and to improve the quality of life of patients to some extent.
Collapse
Affiliation(s)
| | | | | | | | - Hangyu Ji
- *Correspondence: Fengmei Lian, ; Hangyu Ji,
| | | |
Collapse
|
4
|
Tan T, Xiang Y, Deng C, Cao C, Ren Z, Huang G, Zhou Z. Variable frequencies of peripheral T-lymphocyte subsets in the diabetes spectrum from type 1 diabetes through latent autoimmune diabetes in adults (LADA) to type 2 diabetes. Front Immunol 2022; 13:974864. [PMID: 36091068 PMCID: PMC9449581 DOI: 10.3389/fimmu.2022.974864] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
T lymphocytes are key players in the pathogenesis of autoimmune diabetes. We recruited subjects with T1D (n=81), LADA (n=82), T2D (n=95) and NGT (n=218) and analyzed the percentages of T-lymphocyte subsets, including T helper 1 (Th1), T helper 2 (Th2), T helper 17 (Th17), T cytotoxic 1 (Tc1), regulatory T cells (Tregs), effector T (Teff), naïve T, central memory T (Tcm), and effector memory T (Tem) cells by flow cytometry. LADA patients possessed similar frequencies of IFN-γ+CD4+ T (Th1), IFN-γ+CD8+ T and CD4+ Teff cells compared with T1D patients, but much lower than those of NGT subjects. Like T2D patients, LADA patients had increased frequencies of CD4+ Tem and CD8+ Tem cells with respect to T1D and NGT subjects. In LADA patients, Th2 cells were decreased while CD4+ Tcm cells were increased compared with NGT subjects. Notably, we observed significant negative correlations between the CD4+ Tcm cell frequency and C-peptide in LADA subjects. These data demonstrates that LADA patients possess T-cell subset changes resembling both T1D and T2D and represent the middle of the diabetes spectrum between T1D and T2D. Based on these T-cell subset alterations, we speculate that autoimmunity-induced β-cell destruction and inflammation-induced insulin resistance might both be involved in the pathogenesis of LADA.
Collapse
|
5
|
Clement CC, Osan J, Buque A, Nanaware PP, Chang YC, Perino G, Shetty M, Yamazaki T, Tsai WL, Urbanska AM, Calvo-Calle JM, Ramsamooj S, Ramsamooj S, Vergani D, Mieli-Vergani G, Terziroli Beretta-Piccoli B, Gadina M, Montagna C, Goncalves MD, Sallusto F, Galluzzi L, Soni RK, Stern LJ, Santambrogio L. PDIA3 epitope-driven immune autoreactivity contributes to hepatic damage in type 2 diabetes. Sci Immunol 2022; 7:eabl3795. [PMID: 35984892 DOI: 10.1126/sciimmunol.abl3795] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A diet rich in saturated fat and carbohydrates causes low-grade chronic inflammation in several organs, including the liver, ultimately driving nonalcoholic steatohepatitis. In this setting, environment-driven lipotoxicity and glucotoxicity induce liver damage, which promotes dendritic cell activation and generates a major histocompatibility complex class II (MHC-II) immunopeptidome enriched with peptides derived from proteins involved in cellular metabolism, oxidative phosphorylation, and the stress responses. Here, we demonstrated that lipotoxicity and glucotoxicity, as driven by a high-fat and high-fructose (HFHF) diet, promoted MHC-II presentation of nested T and B cell epitopes from protein disulfide isomerase family A member 3 (PDIA3), which is involved in immunogenic cell death. Increased MHC-II presentation of PDIA3 peptides was associated with antigen-specific proliferation of hepatic CD4+ immune infiltrates and isotype switch of anti-PDIA3 antibodies from IgM to IgG3, indicative of cellular and humoral PDIA3 autoreactivity. Passive transfer of PDIA3-specific T cells or PDIA3-specific antibodies also exacerbated hepatocyte death, as determined by increased hepatic transaminases detected in the sera of mice subjected to an HFHF but not control diet. Increased humoral responses to PDIA3 were also observed in patients with chronic inflammatory liver conditions, including autoimmune hepatitis, primary biliary cholangitis, and type 2 diabetes. Together, our data indicated that metabolic insults caused by an HFHF diet elicited liver damage and promoted pathogenic immune autoreactivity driven by T and B cell PDIA3 epitopes.
Collapse
Affiliation(s)
- Cristina C Clement
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Jaspreet Osan
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Aitziber Buque
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Padma P Nanaware
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Yoke-Chen Chang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Giorgio Perino
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Madhur Shetty
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Wanxia Li Tsai
- Translational Immunology Section, National Institute of Arthritis Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 10916, USA
| | | | | | - Shakti Ramsamooj
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA.,Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Shakti Ramsamooj
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Diego Vergani
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana and Epatocentro Ticino, Lugano 6900, Switzerland.,King's College London Faculty of Life Sciences and Medicine, King's College Hospital, London WC2R 2LS, UK
| | - Giorgina Mieli-Vergani
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana and Epatocentro Ticino, Lugano 6900, Switzerland.,King's College London Faculty of Life Sciences and Medicine, King's College Hospital, London WC2R 2LS, UK
| | - Benedetta Terziroli Beretta-Piccoli
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana and Epatocentro Ticino, Lugano 6900, Switzerland.,King's College London Faculty of Life Sciences and Medicine, King's College Hospital, London WC2R 2LS, UK
| | - Massimo Gadina
- Translational Immunology Section, National Institute of Arthritis Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 10916, USA
| | - Cristina Montagna
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | | | - Federica Sallusto
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana and Epatocentro Ticino, Lugano 6900, Switzerland
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA.,Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA.,Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Rajesh K Soni
- Proteomics and Macromolecular Crystallography Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Lawrence J Stern
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01605, USA.,Immunology and Microbiology Program, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA.,Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA.,Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
6
|
Ding Q, Gao Z, Chen K, Zhang Q, Hu S, Zhao L. Inflammation-Related Epigenetic Modification: The Bridge Between Immune and Metabolism in Type 2 Diabetes. Front Immunol 2022; 13:883410. [PMID: 35603204 PMCID: PMC9120428 DOI: 10.3389/fimmu.2022.883410] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/11/2022] [Indexed: 12/11/2022] Open
Abstract
T2DM, as a typical metabolic inflammatory disease, is under the joint regulation of environmental factors and genetics, combining with a variety of epigenetic changes. Apart from epigenetic changes of islet β cells and glycometabolic tissues or organs, the inflammation-related epigenetics is also the core pathomechanism leading to β-cell dysfunction and insulin resistance. In this review, we focus on the epigenetic modification of immune cells’ proliferation, recruitment, differentiation and function, providing an overview of the key genes which regulated by DNA methylation, histone modifications, and non-coding RNA in the respect of T2DM. Meanwhile, we further summarize the present situation of T2DM epigenetic research and elucidate its prospect in T2DM clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Qiyou Ding
- Department of Endocrinology, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Metabolic Diseases, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Zezheng Gao
- Department of Endocrinology, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Metabolic Diseases, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Keyu Chen
- Department of Endocrinology, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Metabolic Diseases, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Qiqi Zhang
- Department of Endocrinology, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Metabolic Diseases, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shiwan Hu
- Department of Endocrinology, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Metabolic Diseases, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Linhua Zhao
- Institute of Metabolic Diseases, Guang’ anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Linhua Zhao,
| |
Collapse
|
7
|
Hod T, Ben-David A, Olmer L, Scott N, Ghinea R, Mor E, Levy I, Indenbaum V, Lustig Y, Grossman E, Rahav G. BNT162b2 Third Booster Dose Significantly Increases the Humoral Response Assessed by Both RBD IgG and Neutralizing Antibodies in Renal Transplant Recipients. Transpl Int 2022; 35:10239. [PMID: 35387393 PMCID: PMC8977405 DOI: 10.3389/ti.2022.10239] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/18/2022] [Indexed: 12/13/2022]
Abstract
Background: An impaired humoral response to full dose of BNT162b2 vaccine was observed in renal transplant recipients (RTR). Methods: To reveal predictors for humoral response to third vaccine, patients were stratified to positive (N = 85) and negative (N = 14) response groups based on receptor-binding domain (RBD) IgG ≥1.1 and neutralizing antibodies (NA) ≥ 16 dilution versus RBD IgG <1.1 or NA < 16, respectively. NA were detected using a SARS-CoV-2 pseudo-virus. Results: Response rate increased from 32.3% (32/99) before the third dose to 85.9% (85/99) post-third vaccine with a significant rise in geometric mean titers (GMTs) for RBD IgG and NA [0.79 (95% CI 0.65–0.96) vs. 3.08 (95% CI 2.76–3.45), p < 0.001 and 17.46 (95% CI 12.38–24.62) vs. 362.2 (95% CI 220.7–594.6), p < 0.001 respective. 80.6% (54/67) seroconverted and 96.9% (31/32) remained positive following the vaccine with a significant increase in GMTs for RBD IgG and NA. Age, ESRD secondary to diabetic nephropathy (DN) and renal allograft function were independent predictors for antibody response in RTR. Mycophenolic acid (MPA) use and dose had no impact on humoral response following the third booster. AEs were recorded for 70.1% of RTR population. Systemic AEs were more common in recipients with a positive humoral response as opposed to non-responders (45.2% versus 15.4% respectively, p = 0.04). Conclusion: 85.9% of RTR develop NA to BNT162b2 third vaccine, found effective in both negative and positive responders prior to the vaccine. Antigenic re-exposure overcame the suppressive effect of MPA on antibody response in RTR.
Collapse
Affiliation(s)
- Tammy Hod
- Renal Transplant Center, Sheba Medical Center, Tel Hashomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Aharon Ben-David
- Renal Transplant Center, Sheba Medical Center, Tel Hashomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Liraz Olmer
- Bio-statistical and Bio-mathematical Unit, The Gertner Institute of Epidemiology and Health Policy Research, Sheba Medical Center, Tel Hashomer, Israel
| | - Noa Scott
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ronen Ghinea
- Renal Transplant Center, Sheba Medical Center, Tel Hashomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Eytan Mor
- Renal Transplant Center, Sheba Medical Center, Tel Hashomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Itzchak Levy
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,The Infectious Diseases Unit, Sheba Medical Center, Tel Hashomer, Israel
| | - Victoria Indenbaum
- Central Virology Laboratory, Public Health Services, Ministry of Health and Sheba Medical Center, Tel-Hashomer, Israel
| | - Yaniv Lustig
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Central Virology Laboratory, Public Health Services, Ministry of Health and Sheba Medical Center, Tel-Hashomer, Israel
| | - Ehud Grossman
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Internal Medicine Wing, Sheba Medical Center, Tel Hashomer, Israel
| | - Galia Rahav
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,The Infectious Diseases Unit, Sheba Medical Center, Tel Hashomer, Israel
| |
Collapse
|
8
|
Ssekamatte P, Nakibuule M, Nabatanzi R, Egesa M, Musubika C, Bbuye M, Hepworth MR, Doherty DG, Cose S, Biraro IA. Type 2 Diabetes Mellitus and Latent Tuberculosis Infection Moderately Influence Innate Lymphoid Cell Immune Responses in Uganda. Front Immunol 2021; 12:716819. [PMID: 34512639 PMCID: PMC8432960 DOI: 10.3389/fimmu.2021.716819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 08/16/2021] [Indexed: 12/19/2022] Open
Abstract
Background Type 2 diabetes mellitus (T2DM) is a major risk factor for the acquisition of latent tuberculosis (TB) infection (LTBI) and development of active tuberculosis (ATB), although the immunological basis for this susceptibility remains poorly characterised. Innate lymphoid cells (ILCs) immune responses to TB infection in T2DM comorbidity is anticipated to be reduced. We compared ILC responses (frequency and cytokine production) among adult patients with LTBI and T2DM to patients (13) with LTBI only (14), T2DM only (10) and healthy controls (11). Methods Using flow cytometry, ILC phenotypes were categorised based on (Lin−CD127+CD161+) markers into three types: ILC1 (Lin−CD127+CD161+CRTH2-CD117−); ILC2 (Lin−CD127+CD161+CRTH2+) and ILC3 (Lin−CD127+CD161+CRTH2−NKp44+/−CD117+). ILC responses were determined using cytokine production by measuring percentage expression of interferon-gamma (IFN-γ) for ILC1, interleukin (IL)-13 for ILC2, and IL-22 for ILC3. Glycaemic control among T2DM patients was measured using glycated haemoglobin (HbA1c) levels. Data were analysed using FlowJo version 10.7.1, and GraphPad Prism version 8.3. Results Compared to healthy controls, patients with LTBI and T2DM had reduced frequencies of ILC2 and ILC3 respectively (median (IQR): 0.01 (0.005-0.04) and 0.002 (IQR; 0.002-0.007) and not ILC1 (0.04 (0.02-0.09) as expected. They also had increased production of IFN-γ [median (IQR): 17.1 (5.6-24.9)], but decreased production of IL-13 [19.6 (12.3-35.1)]. We however found that patients with T2DM had lower ILC cytokine responses in general but more marked for IL-22 production (median (IQR): IFN-γ 9.3 (4.8-22.6); IL-13 22.2 (14.7-39.7); IL-22 0.7 (IQR; 0.1-2.1) p-value 0.02), which highlights the immune suppression status of T2DM. We also found that poor glycaemic control altered ILC immune responses. Conclusion This study demonstrates that LTBI and T2DM, and T2DM were associated with slight alterations of ILC immune responses. Poor T2DM control also slightly altered these ILC immune responses. Further studies are required to assess if these responses recover after treatment of either TB or T2DM.
Collapse
Affiliation(s)
- Phillip Ssekamatte
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Marjorie Nakibuule
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute (MRC/UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda
| | - Rose Nabatanzi
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Moses Egesa
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute (MRC/UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda.,Department of Infection Biology, Faculty of Infectious and Tropical Diseases, LSHTM, London, United Kingdom
| | - Carol Musubika
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Mudarshiru Bbuye
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University College of Health Sciences, Kampala, Uganda
| | - Matthew R Hepworth
- Division of Infection, Immunity & Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Lydia Becker Institute of Immunology and Inflammation and Manchester Collaborative Centre for Inflammation Research (MCCIR), Manchester, United Kingdom
| | | | - Stephen Cose
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute (MRC/UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda
| | - Irene Andia Biraro
- Immunomodulation and Vaccines Programme, Medical Research Council/Uganda Virus Research Institute (MRC/UVRI) and London School of Hygiene and Tropical Medicine (LSHTM) Uganda Research Unit, Entebbe, Uganda.,Department of Internal Medicine, School of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| |
Collapse
|
9
|
Soedono S, Cho KW. Adipose Tissue Dendritic Cells: Critical Regulators of Obesity-Induced Inflammation and Insulin Resistance. Int J Mol Sci 2021; 22:ijms22168666. [PMID: 34445379 PMCID: PMC8395475 DOI: 10.3390/ijms22168666] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/31/2021] [Accepted: 08/09/2021] [Indexed: 12/22/2022] Open
Abstract
Chronic inflammation of the adipose tissue (AT) is a critical component of obesity-induced insulin resistance and type 2 diabetes. Adipose tissue immune cells, including AT macrophages (ATMs), AT dendritic cells (ATDCs), and T cells, are dynamically regulated by obesity and participate in obesity-induced inflammation. Among AT resident immune cells, ATDCs are master immune regulators and engage in crosstalk with various immune cells to initiate and regulate immune responses. However, due to confounding markers and lack of animal models, their exact role and contribution to the initiation and maintenance of AT inflammation and insulin resistance have not been clearly elucidated. This paper reviews the current understanding of ATDCs and their role in obesity-induced AT inflammation. We also provide the potential mechanisms by which ATDCs regulate AT inflammation and insulin resistance in obesity. Finally, this review offers perspectives on ways to better dissect the distinct functions and contributions of ATDCs to obesity.
Collapse
Affiliation(s)
- Shindy Soedono
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan 31151, Korea;
| | - Kae Won Cho
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan 31151, Korea;
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Korea
- Correspondence: ; Tel.: +82-41-413-5028
| |
Collapse
|
10
|
Brooks-Worrell BM, Tjaden AH, Edelstein SL, Palomino B, Utzschneider KM, Arslanian S, Mather KJ, Buchanan TA, Nadeau KJ, Atkinson K, Barengolts E, Kahn SE, Palmer JP. Islet Autoimmunity in Adults With Impaired Glucose Tolerance and Recently Diagnosed, Treatment Naïve Type 2 Diabetes in the Restoring Insulin SEcretion (RISE) Study. Front Immunol 2021; 12:640251. [PMID: 33981301 PMCID: PMC8108986 DOI: 10.3389/fimmu.2021.640251] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/24/2021] [Indexed: 12/26/2022] Open
Abstract
The presence of islet autoantibodies and islet reactive T cells (T+) in adults with established type 2 diabetes (T2D) have been shown to identify those patients with more severe β-cell dysfunction. However, at what stage in the progression toward clinical T2D does islet autoimmunity emerge as an important component influencing β-cell dysfunction? In this ancillary study to the Restoring Insulin SEcretion (RISE) Study, we investigated the prevalence of and association with β-cell dysfunction of T+ and autoantibodies to the 65 kDa glutamic acid decarboxylase antigen (GADA) in obese pre-diabetes adults with impaired glucose tolerance (IGT) and recently diagnosed treatment naïve (Ndx) T2D. We further investigated the effect of 12 months of RISE interventions (metformin or liraglutide plus metformin, or with 3 months of insulin glargine followed by 9 months of metformin or placebo) on islet autoimmune reactivity. We observed GADA(+) in 1.6% of NdxT2D and 4.6% of IGT at baseline, and in 1.6% of NdxT2D and 5.3% of IGT at 12 months, but no significant associations between GADA(+) and β-cell function. T(+) was observed in 50% of NdxT2D and 60.4% of IGT at baseline, and in 68.4% of NdxT2D and 83.9% of IGT at 12 months. T(+) NdxT2D were observed to have significantly higher fasting glucose (p = 0.004), and 2 h glucose (p = 0.0032), but significantly lower steady state C-peptide (sscpep, p = 0.007) compared to T(-) NdxT2D. T(+) IGT participants demonstrated lower but not significant (p = 0.025) acute (first phase) C-peptide response to glucose (ACPRg) compared to T(-) IGT. With metformin treatment, T(+) participants were observed to have a significantly lower Hemoglobin A1c (HbA1c, p = 0.002) and fasting C-peptide (p = 0.002) compared to T(-), whereas T(+) treated with liraglutide + metformin had significantly lower sscpep (p = 0.010) compared to T(-) participants. In the placebo group, T(+) participants demonstrated significantly lower ACPRg (p = 0.001) compared to T(-) participants. In summary, T(+) were found in a large percentage of obese pre-diabetes adults with IGT and in recently diagnosed T2D. Moreover, T(+) were significantly correlated with treatment effects and β-cell dysfunction. Our results demonstrate that T(+) are an important component in T2D.
Collapse
Affiliation(s)
- Barbara M Brooks-Worrell
- Department of Medicine, University of Washington, Seattle, WA, United States.,Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| | - Ashley H Tjaden
- Biostatistics Center, Milken School of Public Health, George Washington University Biostatistics Center, Rockville, MD, United States
| | - Sharon L Edelstein
- Biostatistics Center, Milken School of Public Health, George Washington University Biostatistics Center, Rockville, MD, United States
| | - Brenda Palomino
- Seattle Institute for Biochemical and Clinical Research, Seattle, WA, United States
| | - Kristina M Utzschneider
- Department of Medicine, University of Washington, Seattle, WA, United States.,Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| | - Silva Arslanian
- Department of Medicine, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Kieren J Mather
- Indiana University School of Medicine, Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, IN, United States
| | - Thomas A Buchanan
- University of Southern California Keck School of Medicine/Kaiser Permanente Southern California, Los Angeles, CA, United States
| | - Kristen J Nadeau
- University of Colorado Anschutz Medical Campus/Children's Hospital Colorado, Aurora, CO, United States
| | - Karen Atkinson
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| | - Elena Barengolts
- University of Chicago Clinical Research Center and Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States
| | - Steven E Kahn
- Department of Medicine, University of Washington, Seattle, WA, United States.,Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| | - Jerry P Palmer
- Department of Medicine, University of Washington, Seattle, WA, United States.,Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| | | |
Collapse
|
11
|
Mohammad S, Aziz R, Al Mahri S, Malik SS, Haji E, Khan AH, Khatlani TS, Bouchama A. Obesity and COVID-19: what makes obese host so vulnerable? IMMUNITY & AGEING 2021; 18:1. [PMID: 33390183 PMCID: PMC7779330 DOI: 10.1186/s12979-020-00212-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/03/2020] [Indexed: 12/13/2022]
Abstract
The disease (COVID-19) novel coronavirus pandemic has so far infected millions resulting in the death of over a million people as of Oct 2020. More than 90% of those infected with COVID-19 show mild or no symptoms but the rest of the infected cases show severe symptoms resulting in significant mortality. Age has emerged as a major factor to predict the severity of the disease and mortality rates are significantly higher in elderly patients. Besides, patients with underlying conditions like Type 2 diabetes, cardiovascular diseases, hypertension, and cancer have an increased risk of severe disease and death due to COVID-19 infection. Obesity has emerged as a novel risk factor for hospitalization and death due to COVID-19. Several independent studies have observed that people with obesity are at a greater risk of severe disease and death due to COVID-19. Here we review the published data related to obesity and overweight to assess the possible risk and outcome in Covid-19 patients based on their body weight. Besides, we explore how the obese host provides a unique microenvironment for disease pathogenesis, resulting in increased severity of the disease and poor outcome.
Collapse
Affiliation(s)
- Sameer Mohammad
- Experimental Medicine Department, King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences-MNGHA, Riyadh, 11426, Saudi Arabia.
| | - Rafia Aziz
- Government Medical College Baramulla, Baramulla, Kashmir, India
| | - Saeed Al Mahri
- Experimental Medicine Department, King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences-MNGHA, Riyadh, 11426, Saudi Arabia
| | - Shuja Shafi Malik
- Experimental Medicine Department, King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences-MNGHA, Riyadh, 11426, Saudi Arabia
| | - Esraa Haji
- Experimental Medicine Department, King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences-MNGHA, Riyadh, 11426, Saudi Arabia
| | - Altaf Husain Khan
- Biostatistics and Bioinformatics Department, King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences-MNGHA, Riyadh, 11426, Saudi Arabia
| | - Tanvir Saleem Khatlani
- Department of Cellular Therapy, Stem Cells Unit, King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences--MNGHA, Riyadh, 11426, Saudi Arabia
| | - Abderrezak Bouchama
- Experimental Medicine Department, King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences-MNGHA, Riyadh, 11426, Saudi Arabia
| |
Collapse
|
12
|
Song J, Deng T. The Adipocyte and Adaptive Immunity. Front Immunol 2020; 11:593058. [PMID: 33329579 PMCID: PMC7728694 DOI: 10.3389/fimmu.2020.593058] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 10/27/2020] [Indexed: 12/21/2022] Open
Abstract
Not only do Adipocytes have energy storage and endocrine functions, but they also play an immunological role. Adipocytes are involved in adaptive immunity to mediate the pathological processes of a variety of chronic inflammatory diseases and autoimmune syndromes. The adaptive immune response consists of T cell-mediated cellular immunity and B cell-mediated humoral immunity. Obese adipocytes overexpress MHC class II molecules and costimulators to act as antigen-presenting cells (APCs) and promote the activation of CD4+ T cells. In addition, various adipokines secreted by adipocytes regulate the proliferation and differentiation of T cells. Adipokines are also involved in B cell generation, development, activation, and antibody production. Therefore, adipocytes play an important role in B cell-mediated adaptive immunity. This review describes how adipocytes participate in adaptive immunity from the perspective of T cells and B cells, and discusses their role in the pathogenesis of various diseases.
Collapse
Affiliation(s)
- Jianfeng Song
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Tuo Deng
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Metabolic Syndrome Research Center, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.,Clinical Immunology Center, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
13
|
Elieh Ali Komi D, Shafaghat F, Christian M. Crosstalk Between Mast Cells and Adipocytes in Physiologic and Pathologic Conditions. Clin Rev Allergy Immunol 2020; 58:388-400. [PMID: 32215785 PMCID: PMC7244609 DOI: 10.1007/s12016-020-08785-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Excessive fatty acids and glucose uptake support the infiltration of adipose tissue (AT) by a variety of immune cells including neutrophils, pro-inflammatory M1 macrophages, and mast cells (MCs). These cells promote inflammation by releasing pro-inflammatory mediators. The involvement of MCs in AT biology is supported by their accumulation in the AT of obese individuals along with significantly higher serum levels of MC-derived tryptase. AT-resident MCs under the influence of locally derived adipokines such as leptin become activated and release pro-inflammatory cytokines including TNFα that worsens the inflammatory state. MCs support angiogenesis in AT by releasing chymase and inducing preadipocyte differentiation and also the proliferation of adipocytes through 15-deoxy-delta PGJ2/PPARγ interaction. Additionally, they contribute to the remodeling of the AT extracellular matrix (ECM) and play a role in the recruitment and activation of leukocytes. MC degranulation has been linked to brown adipocyte activation, and evidence indicates an important link between MCs and the appearance of BRITE/beige adipocytes in white AT. Cell crosstalk between MCs and AT-resident cells, mainly adipocytes and immune cells, shows that these cells play a critical role in the regulation of AT homeostasis and inflammation.
Collapse
Affiliation(s)
- Daniel Elieh Ali Komi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farzaneh Shafaghat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mark Christian
- School of Science and Technology, Nottingham, NG11 8NS, UK.
| |
Collapse
|
14
|
Tripathi D, Radhakrishnan RK, Sivangala Thandi R, Paidipally P, Devalraju KP, Neela VSK, McAllister MK, Samten B, Valluri VL, Vankayalapati R. IL-22 produced by type 3 innate lymphoid cells (ILC3s) reduces the mortality of type 2 diabetes mellitus (T2DM) mice infected with Mycobacterium tuberculosis. PLoS Pathog 2019; 15:e1008140. [PMID: 31809521 PMCID: PMC6919622 DOI: 10.1371/journal.ppat.1008140] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 12/18/2019] [Accepted: 10/09/2019] [Indexed: 12/14/2022] Open
Abstract
Previously, we found that pathological immune responses enhance the mortality rate of Mycobacterium tuberculosis (Mtb)-infected mice with type 2 diabetes mellitus (T2DM). In the current study, we evaluated the role of the cytokine IL-22 (known to play a protective role in bacterial infections) and type 3 innate lymphoid cells (ILC3s) in regulating inflammation and mortality in Mtb-infected T2DM mice. IL-22 levels were significantly lower in Mtb-infected T2DM mice than in nondiabetic Mtb-infected mice. Similarly, serum IL-22 levels were significantly lower in tuberculosis (TB) patients with T2DM than in TB patients without T2DM. ILC3s were an important source of IL-22 in mice infected with Mtb, and recombinant IL-22 treatment or adoptive transfer of ILC3s prolonged the survival of Mtb-infected T2DM mice. Recombinant IL-22 treatment reduced serum insulin levels and improved lipid metabolism. Recombinant IL-22 treatment or ILC3 transfer prevented neutrophil accumulation near alveoli, inhibited neutrophil elastase 2 (ELA2) production and prevented epithelial cell damage, identifying a novel mechanism for IL-22 and ILC3-mediated inhibition of inflammation in T2DM mice infected with an intracellular pathogen. Our findings suggest that the IL-22 pathway may be a novel target for therapeutic intervention in T2DM patients with active TB disease.
Collapse
Affiliation(s)
- Deepak Tripathi
- Department of Pulmonary Immunology, Center for Biomedical Research, The University of Texas Health Science Center, Tyler, Texas, TX, United States of America
| | - Rajesh Kumar Radhakrishnan
- Department of Pulmonary Immunology, Center for Biomedical Research, The University of Texas Health Science Center, Tyler, Texas, TX, United States of America
| | - Ramya Sivangala Thandi
- Department of Pulmonary Immunology, Center for Biomedical Research, The University of Texas Health Science Center, Tyler, Texas, TX, United States of America
| | - Padmaja Paidipally
- Department of Pulmonary Immunology, Center for Biomedical Research, The University of Texas Health Science Center, Tyler, Texas, TX, United States of America
| | - Kamakshi Prudhula Devalraju
- Immunology and Molecular Biology Department, Bhagwan Mahavir Medical Research Centre, Hyderabad, Telangana, India
| | - Venkata Sanjeev Kumar Neela
- Immunology and Molecular Biology Department, Bhagwan Mahavir Medical Research Centre, Hyderabad, Telangana, India
| | - Madeline Kay McAllister
- Department of Pulmonary Immunology, Center for Biomedical Research, The University of Texas Health Science Center, Tyler, Texas, TX, United States of America
| | - Buka Samten
- Department of Pulmonary Immunology, Center for Biomedical Research, The University of Texas Health Science Center, Tyler, Texas, TX, United States of America
| | - Vijaya Lakshmi Valluri
- Immunology and Molecular Biology Department, Bhagwan Mahavir Medical Research Centre, Hyderabad, Telangana, India
| | - Ramakrishna Vankayalapati
- Department of Pulmonary Immunology, Center for Biomedical Research, The University of Texas Health Science Center, Tyler, Texas, TX, United States of America
| |
Collapse
|
15
|
Van Herck MA, Weyler J, Kwanten WJ, Dirinck EL, De Winter BY, Francque SM, Vonghia L. The Differential Roles of T Cells in Non-alcoholic Fatty Liver Disease and Obesity. Front Immunol 2019; 10:82. [PMID: 30787925 PMCID: PMC6372559 DOI: 10.3389/fimmu.2019.00082] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 01/11/2019] [Indexed: 12/15/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) constitutes a spectrum of disease states characterized by hepatic steatosis and is closely associated to obesity and the metabolic syndrome. In non-alcoholic steatohepatitis (NASH), additionally, inflammatory changes and hepatocellular damage are present, representing a more severe condition, for which the treatment is an unmet medical need. Pathophysiologically, the immune system is one of the main drivers of NAFLD progression and other obesity-related comorbidities, and both the innate and adaptive immune system are involved. T cells form the cellular component of the adaptive immune system and consist of multiple differentially active subsets, i.e., T helper (Th) cells, regulatory T (Treg) cells, and cytotoxic T (Tc) cells, as well as several innate T-cell subsets. This review focuses on the role of these T-cell subsets in the pathogenesis of NAFLD, as well as the association with obesity and type 2 diabetes mellitus, reviewing the available evidence from both animal and human studies. Briefly, Th1, Th2, Th17, and Th22 cells seem to have an attenuating effect on adiposity. Th2, Th22, and Treg cells seem to decrease insulin resistance, whereas Th1, Th17, and Tc cells have an aggravating effect. Concerning NAFLD, both Th22 and Treg cells appear to have an overall tempering effect, whereas Th17 and Tc cells seem to induce more liver damage and fibrosis progression. The evidence regarding the role of the innate T-cell subsets is more controversial and warrants further exploration.
Collapse
Affiliation(s)
- Mikhaïl A Van Herck
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology and Hepatology, University of Antwerp, Antwerp, Belgium.,Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium
| | - Jonas Weyler
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology and Hepatology, University of Antwerp, Antwerp, Belgium.,Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium
| | - Wilhelmus J Kwanten
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology and Hepatology, University of Antwerp, Antwerp, Belgium.,Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium
| | - Eveline L Dirinck
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology and Hepatology, University of Antwerp, Antwerp, Belgium.,Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, Antwerp, Belgium
| | - Benedicte Y De Winter
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology and Hepatology, University of Antwerp, Antwerp, Belgium
| | - Sven M Francque
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology and Hepatology, University of Antwerp, Antwerp, Belgium.,Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium
| | - Luisa Vonghia
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology and Hepatology, University of Antwerp, Antwerp, Belgium.,Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium
| |
Collapse
|
16
|
Sun P, Jin Q, Nie S, Jia S, Li Y, Li X, Guo F. Unlike PD-L1, PD-1 Is Downregulated on Partial Immune Cells in Type 2 Diabetes. J Diabetes Res 2019; 2019:5035261. [PMID: 31008114 PMCID: PMC6441514 DOI: 10.1155/2019/5035261] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 09/21/2018] [Accepted: 01/10/2019] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Type 2 diabetes is a worldwide disease which is associated with chronic, low-grade inflammation. The PD-1/PD-L1 pathway has been reported to be a negative regulatory element in immune homeostasis and to be involved in many diseases. MATERIALS AND METHODS Peripheral blood mononuclear cells (PBMCs) were obtained from type 2 diabetes patients (n = 23) and healthy donors (n = 20). The PD-L1 and PD-1 expressions on corresponding immune cells were evaluated by flow cytometry. RESULTS The PD-L1 expression on corresponding immune cells has no significant difference between these two groups. We showed the downregulated PD-1 expression in type 2 diabetes patients. The correlation analysis indicated that the PD-1 on NK cells has a positive correlation with insulin and diabetes duration. And an inverse correlation has been shown between the PD-1 expression on monocytes and BMI (body mass index). CONCLUSIONS The results in this article suggest that PD-1, unlike PD-L1, might participate in the progression of type 2 diabetes. This investigation will provide evidence for the potential immune therapy for T2D.
Collapse
Affiliation(s)
- Peng Sun
- Department of Intervention Oncology, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Qingyan Jin
- Department of Intervention Oncology, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shengnan Nie
- Department of Intervention Oncology, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shijie Jia
- Department of Intervention Oncology, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yuanyuan Li
- Operation Room of Intervention Oncology, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiaoxue Li
- Department of Intervention Oncology, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Fang Guo
- Department of Intervention Oncology, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
17
|
Nyambuya TM, Dludla PV, Nkambule BB. T cell activation and cardiovascular risk in type 2 diabetes mellitus: a protocol for a systematic review and meta-analysis. Syst Rev 2018; 7:167. [PMID: 30342529 PMCID: PMC6195734 DOI: 10.1186/s13643-018-0835-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/03/2018] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION The burden of non-communicable diseases such as type 2 diabetes mellitus (T2DM) and cardiovascular diseases (CVDs) has drastically increased in developing countries over the years. Although recent evidence points to chronic immune activation to be a significant aspect in the pathogenesis and development of T2DM and CVDs, the exact role of T cells is not fully understood. Therefore, we aim to investigate T cell function and cardio vascular risk in T2DM. In addition, the therapeutic effect of blood glucose-lowering drugs to reverse hyperglycaemia induced T cell dysfunction and myocardial infarction will be reviewed. METHODS This will be a systematic review and meta-analysis of published studies assessing T cell activation and cardiovascular risk in adults with T2DM. The search strategy will include medical subject headings (MeSH) words for PubMed/MEDLINE database. The search terms will also be adapted to grey literature, Embase and Cochrane Central Register of Controlled Trials electronic databases. Studies will be independently screened by two reviewers using predefined criteria. Relevant eligible full texts will be screened and data will be extracted. Data extraction will be performed using a pre-piloted structured form. To assess the quality and strengths of evidence across selected studies, the Grading of Recommendations Assessment Development and Evaluation approach will be used. The Cochran's Q statistic and the I2 statistics will be used to analyse statistical heterogeneity between studies. If included studies show substantial level of statistical heterogeneity, a random-effects meta-analysis will be performed using R statistical software. DISCUSSIONS This review will not require ethical approval, and the findings will be disseminated through peer-reviewed publication and conferences. Although other previous studies have reported deregulated T cell function in hyperglycaemia, the underlying mechanisms remain controversial. However, evidence suggests that T cells may be a key component in the development of T2DM and CVDs as its complication. Furthermore, they are a potential diagnostic and therapeutic target in the management of the disease. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42018099745.
Collapse
Affiliation(s)
- Tawanda M. Nyambuya
- School of Laboratory Medicine and Medical Sciences (SLMMS), College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Phiwayinkosi V. Dludla
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
- Biomedical Research and Innovation Platform, Medical Research Council, Tygerberg, South Africa
| | - Bongani B. Nkambule
- School of Laboratory Medicine and Medical Sciences (SLMMS), College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
18
|
JAZF1 Inhibits Adipose Tissue Macrophages and Adipose Tissue Inflammation in Diet-Induced Diabetic Mice. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4507659. [PMID: 29765984 PMCID: PMC5885486 DOI: 10.1155/2018/4507659] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/19/2017] [Accepted: 12/28/2017] [Indexed: 12/26/2022]
Abstract
Background Juxtaposed with another zinc finger gene 1 (JAZF1) affects gluconeogenesis, insulin sensitivity, lipid metabolism, and inflammation, but its exact role in chronic inflammation remains unclear. This study aimed to examine JAZF1 overexpression in vivo on adipose tissue macrophages (ATMs). Methods Mouse models of high-fat diet- (HFD-) induced insulin resistance were induced using C57BL/6J and JAZF1-overexpressing (JAZF1-OX) mice. The mice were randomized (8–10/group) to C57BL/6J mice fed regular diet (RD) (NC group), C57BL/6J mice fed HFD (HF group), JAZF1-OX mice fed RD (NJ group), and JAZF1-OX mice fed HFD (HJ group). Adipose tissue was harvested 12 weeks later. ATMs were evaluated by flow cytometry. Inflammatory markers were evaluated by ELISA. Results JAZF1-OX mice had lower blood lipids, blood glucose, body weight, fat weight, and inflammatory markers compared with HF mice (all P < 0.05). JAZF1 overexpression decreased ATM number and secretion of proinflammatory cytokines. JAZF1 overexpression decreased total CD4+ T cells, active T cells, and memory T cells and increased Treg cells. JAZF1 overexpression downregulated IFN-γ and IL-17 levels and upregulated IL-4 levels. JAZF1 overexpression decreased MHCII, CD40, and CD86 in total ATM, CD11c+ ATM, and CD206+ ATM. Conclusions JAZF1 limits adipose tissue inflammation by limiting macrophage populations and restricting their antigen presentation function.
Collapse
|
19
|
Yang M, Su L, Tao Q, Zhang C, Wu Y, Liu J. Depletion of Regulatory T Cells in Visceral Adipose Tissues Contributes to Insulin Resistance in Hashimoto's Thyroiditis. Front Physiol 2018. [PMID: 29541033 PMCID: PMC5835527 DOI: 10.3389/fphys.2018.00136] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Hashimoto's Thyroiditis (HT) is a common organ-specific autoimmune disorder associated with a high incidence, and insulin resistance is highly related to autoimmune. Here, we examined the insulin sensitivity in HT patients and found decreased insulin sensitivity occurred in HT patients. To explore the relationship between impaired insulin sensitivity and immune status, we established HT model mice which showed similar pathological features and immune features to HT patients. In HT model mice, reinfusion of regulatory T cells (Tregs) from peripheral blood of normal mice could improve insulin sensitivity and decrease the inflammation. Anti-CD25 antibodies blocked beneficial effects from reinfusion of Tregs, but delayed administration of anti-CD25 antibodies could not abolished the effect from Tregs. Delayed administration of anti-CD25 antibodies abolished exogenous Tregs in peripheral blood, but there were increased exogenous Tregs located to visceral adipose tissues (VATs) which modulated the expression of cytokines in VATs. These findings suggest that insulin resistance exists in HT patients and it associates with the decreased Tregs and increased inflammation in the VATs.
Collapse
Affiliation(s)
- Min Yang
- Department of Endocrinology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Li Su
- Testing and Analysis Center, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Qin Tao
- Department of Endocrinology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Chenxi Zhang
- Testing and Analysis Center, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Yueyue Wu
- Department of Endocrinology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Jun Liu
- Department of Endocrinology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Huh JH, Kim HM, Lee ES, Kwon MH, Lee BR, Ko HJ, Chung CH. Dual CCR2/5 Antagonist Attenuates Obesity-Induced Insulin Resistance by Regulating Macrophage Recruitment and M1/M2 Status. Obesity (Silver Spring) 2018; 26:378-386. [PMID: 29280303 DOI: 10.1002/oby.22103] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 11/09/2017] [Accepted: 11/26/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Adipose tissue inflammation induced by macrophage infiltration through the C-C motif chemokine receptor (CCR) 2 or CCR5 pathway has a pivotal role in obesity-related disease and insulin resistance. Here, the effect of PF4178903, a dual CCR2/CCR5 antagonist, on obesity and insulin resistance was evaluated. METHODS Forty male C57BL/6J mice were divided into four groups as follows: (1) regular diet (RD), (2) RD with PF4178903, (3) high-fat diet (HFD), and (4) HFD with PF4178903. All mice were sacrificed 12 weeks after the beginning of the experiment. Biochemical analyses and adipose tissue examinations were performed. RESULTS After treatment with PF4178903, both body weight and adipocyte size in white adipose tissue were decreased in HFD-fed mice. Furthermore, PF4178903 treatment reduced adipose tissue macrophages (ATMs) and lowered serum proinflammatory cytokines in HFD-fed mice. PF4178903 treatment significantly improved HFD-induced insulin resistance and glucose intolerance. Fluorescence-activated cell sorter analysis revealed that PF4178903 treatment reduced the CD8 + T cell fraction in white adipose tissue of HFD-fed mice. PF4178903 treatment reduced M1-polarized macrophages while inducing an M2-dominant shift in macrophages within white adipose tissue in HFD-fed mice. CONCLUSIONS Dual CCR2/CCR5 antagonism ameliorates insulin resistance and inflammation in obesity by regulating ATM recruitment and polarization in white adipose tissue.
Collapse
Affiliation(s)
- Ji Hye Huh
- Department of Internal Medicine, Wonju College of Medicine, Yonsei University, Seoul, Korea
| | - Hong Min Kim
- Department of Internal Medicine, Wonju College of Medicine, Yonsei University, Seoul, Korea
| | - Eun Soo Lee
- Department of Internal Medicine, Wonju College of Medicine, Yonsei University, Seoul, Korea
| | - Mi Hye Kwon
- Department of Internal Medicine, Wonju College of Medicine, Yonsei University, Seoul, Korea
| | - Bo Ra Lee
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, Korea
| | - Hyun-Jeong Ko
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, Korea
| | - Choon Hee Chung
- Department of Internal Medicine, Wonju College of Medicine, Yonsei University, Seoul, Korea
| |
Collapse
|
21
|
Tang CL, Liu ZM, Gao YR, Xiong F. Schistosoma Infection and Schistosoma-Derived Products Modulate the Immune Responses Associated with Protection against Type 2 Diabetes. Front Immunol 2018; 8:1990. [PMID: 29387059 PMCID: PMC5776330 DOI: 10.3389/fimmu.2017.01990] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 12/21/2017] [Indexed: 12/27/2022] Open
Abstract
Studies on parasite-induced immunoregulatory mechanisms could contribute to the development of new therapies for inflammatory diseases such as type 2 diabetes (T2D), which is a chronic inflammatory disease characterized by persistent elevated glucose levels due to insulin resistance. The association between previous Schistosoma infection and T2D has been confirmed—Schistosoma infection and Schistosoma-derived products modulate the immune system, including innate and acquired immune responses, contributing to T2D disease control. Schistosoma infections and Schistosoma-derived molecules affect the immune cell composition in adipose tissue, dampening inflammation and improving glucose tolerance. This protective role includes the polarization of immune cells to alternatively activated macrophages, dendritic cells, eosinophils, and group 2 innate lymphoid cells. Furthermore, Schistosoma infection and Schistosoma products are effective for the treatment of T2D, as they increase the number of type 2 helper T cells (Th2) and regulatory T cells (Tregs) and decrease type 1 helper T cells (Th1) and type 17 helper T cells (Th17) cells. Thus, our aim was to comprehensively review the mechanism through which Schistosoma infection and Schistosoma products modulate the immune response against T2D.
Collapse
Affiliation(s)
- Chun-Lian Tang
- Department of Science and Education, Wuchang Hospital, Wuhan, China
| | - Zhi-Ming Liu
- Department of Science and Education, Wuchang Hospital, Wuhan, China
| | - Yan Ru Gao
- Medical Department, City College, Wuhan University of Science and Technology, Wuhan, China
| | - Fei Xiong
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Aleksandrova K, Mozaffarian D, Pischon T. Addressing the Perfect Storm: Biomarkers in Obesity and Pathophysiology of Cardiometabolic Risk. Clin Chem 2018; 64:142-153. [DOI: 10.1373/clinchem.2017.275172] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 10/25/2017] [Indexed: 02/06/2023]
Abstract
AbstractBACKGROUNDThe worldwide rise of obesity has provoked intensified research to better understand its pathophysiology as a means for disease prevention. Several biomarkers that may reflect various pathophysiological pathways that link obesity and cardiometabolic diseases have been identified over the past decades.CONTENTWe summarize research evidence regarding the role of established and novel obesity-related biomarkers, focusing on recent epidemiological evidence for detrimental associations with cardiometabolic diseases including obesity-related cancer. The reviewed biomarkers include biomarkers of glucose–insulin homeostasis (insulin, insulin-like growth factors, and C-peptide), adipose tissue biomarkers (adiponectin, omentin, apelin, leptin, resistin, and fatty-acid-binding protein-4), inflammatory biomarkers (C-reactive protein, interleukin 6, tumor necrosis factor α), and omics-based biomarkers (metabolites and microRNAs).SUMMARYAlthough the evidence for many classical obesity biomarkers, including adiponectin and C-reactive protein (CRP), in disease etiology has been initially promising, the evidence for a causal role in humans remains limited. Further, there has been little demonstrated ability to improve disease prediction beyond classical risk factors. In the era of “precision medicine,” there is an increasing interest in novel biomarkers, and the extended list of potentially promising biomarkers, such as adipokines, cytokines, metabolites, and microRNAs, implicated in obesity may bring new promise for improved, personalized prevention. To further evaluate the role of obesity-related biomarkers as etiological and early-disease-prediction targets, well-designed studies are needed to evaluate temporal associations, replicate findings, and test clinical utility of novel biomarkers. In particular, studies to determine the therapeutic implications of novel biomarkers beyond established metabolic risk factors are highly warranted.
Collapse
Affiliation(s)
- Krasimira Aleksandrova
- Nutrition, Immunity and Metabolism Start-up Lab, Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | | | - Tobias Pischon
- Molecular Epidemiology Research Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Berlin, Germany
- MDC/BIH Biobank, Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Berlin Institute of Health (BIH), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| |
Collapse
|
23
|
Liu B, Yu H, Sun G, Sun X, Jin H, Zhang C, Shi W, Tian D, Liu K, Xu H, Li X, Yin J, Hong X, Zhang D. OX40 promotes obesity-induced adipose inflammation and insulin resistance. Cell Mol Life Sci 2017; 74:3827-3840. [PMID: 28612217 PMCID: PMC11107569 DOI: 10.1007/s00018-017-2552-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 05/28/2017] [Accepted: 05/30/2017] [Indexed: 12/14/2022]
Abstract
Adaptive immunity plays a critical role in IR and T2DM development; however, the biological mechanisms linking T cell costimulation and glucose metabolism have not been fully elucidated. In this study, we demonstrated that the costimulatory molecule OX40 controls T cell activation and IR development. Inflammatory cell accumulation and enhanced proinflammatory gene expression, as well as high OX40 expression levels on CD4+ T cells, were observed in the adipose tissues of mice with diet-induced obesity. OX40-KO mice exhibited significantly less weight gain and lower fasting glucose levels than those of WT mice, without obvious adipose tissue inflammation. The effects of OX40 on IR are mechanistically linked to the promotion of T cell activation, Th1 cell differentiation and proliferation-as well as the attenuation of Treg suppressive activity and the enhancement of proinflammatory cytokine production-in adipose tissues. Furthermore, OX40 expression on T cells was positively associated with obesity in humans, suggesting that our findings are clinically relevant. In summary, our study revealed that OX40 in CD4+ T cells is crucial for adipose tissue inflammation and IR development. Therefore, the OX40 signaling pathway may be a new target for preventing or treating obesity-related IR and T2DM.
Collapse
Affiliation(s)
- Bing Liu
- Endocrinology Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People's Republic of China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
| | - Hengchi Yu
- Endocrinology Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People's Republic of China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
| | - Guangyong Sun
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
- Beijing Clinical Research Institute, Beijing, 100050, People's Republic of China
| | - Xiaojing Sun
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
- Beijing Clinical Research Institute, Beijing, 100050, People's Republic of China
| | - Hua Jin
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
- Beijing Clinical Research Institute, Beijing, 100050, People's Republic of China
| | - Chunpan Zhang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
- Beijing Clinical Research Institute, Beijing, 100050, People's Republic of China
| | - Wen Shi
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
- Beijing Clinical Research Institute, Beijing, 100050, People's Republic of China
| | - Dan Tian
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
- Beijing Clinical Research Institute, Beijing, 100050, People's Republic of China
| | - Kai Liu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
- Beijing Clinical Research Institute, Beijing, 100050, People's Republic of China
| | - Hufeng Xu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
- Beijing Clinical Research Institute, Beijing, 100050, People's Republic of China
| | - Xinmin Li
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China
- Beijing Clinical Research Institute, Beijing, 100050, People's Republic of China
| | - Jie Yin
- Endocrinology Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People's Republic of China
| | - Xu Hong
- Endocrinology Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People's Republic of China.
| | - Dong Zhang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-cheng District, Beijing, 100050, People's Republic of China.
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, People's Republic of China.
- Beijing Clinical Research Institute, Beijing, 100050, People's Republic of China.
| |
Collapse
|
24
|
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
|
25
|
Sarcopenic obesity or obese sarcopenia: A cross talk between age-associated adipose tissue and skeletal muscle inflammation as a main mechanism of the pathogenesis. Ageing Res Rev 2017; 35:200-221. [PMID: 27702700 DOI: 10.1016/j.arr.2016.09.008] [Citation(s) in RCA: 516] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/23/2016] [Accepted: 09/26/2016] [Indexed: 02/08/2023]
Abstract
Sarcopenia, an age-associated decline in skeletal muscle mass coupled with functional deterioration, may be exacerbated by obesity leading to higher disability, frailty, morbidity and mortality rates. In the combination of sarcopenia and obesity, the state called sarcopenic obesity (SOB), some key age- and obesity-mediated factors and pathways may aggravate sarcopenia. This review will analyze the mechanisms underlying the pathogenesis of SOB. In obese adipose tissue (AT), adipocytes undergo hypertrophy, hyperplasia and activation resulted in accumulation of pro-inflammatory macrophages and other immune cells as well as dysregulated production of various adipokines that together with senescent cells and the immune cell-released cytokines and chemokines create a local pro-inflammatory status. In addition, obese AT is characterized by excessive production and disturbed capacity to store lipids, which accumulate ectopically in skeletal muscle. These intramuscular lipids and their derivatives induce mitochondrial dysfunction characterized by impaired β-oxidation capacity and increased reactive oxygen species formation providing lipotoxic environment and insulin resistance as well as enhanced secretion of some pro-inflammatory myokines capable of inducing muscle dysfunction by auto/paracrine manner. In turn, by endocrine manner, these myokines may exacerbate AT inflammation and also support chronic low grade systemic inflammation (inflammaging), overall establishing a detrimental vicious circle maintaining AT and skeletal muscle inflammation, thus triggering and supporting SOB development. Under these circumstances, we believe that AT inflammation dominates over skeletal muscle inflammation. Thus, in essence, it redirects the vector of processes from "sarcopenia→obesity" to "obesity→sarcopenia". We therefore propose that this condition be defined as "obese sarcopenia", to reflect the direction of the pathological pathway.
Collapse
|
26
|
Liu MH, Li Y, Han L, Zhang YY, Wang D, Wang ZH, Zhou HM, Song M, Li YH, Tang MX, Zhang W, Zhong M. Adipose-derived stem cells were impaired in restricting CD4 +T cell proliferation and polarization in type 2 diabetic ApoE -/- mouse. Mol Immunol 2017; 87:152-160. [PMID: 28445787 DOI: 10.1016/j.molimm.2017.03.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/11/2017] [Accepted: 03/30/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND Atherosclerosis (AS) is the most common and serious complication of type 2 diabetes mellitus (T2DM) and is accelerated via chronic systemic inflammation rather than hyperglycemia. Adipose tissue is the major source of systemic inflammation in abnormal metabolic state. Pro-inflammatory CD4+T cells play pivotal role in promoting adipose inflammation. Adipose-derived stem cells (ADSCs) for fat regeneration have potent ability of immunosuppression and restricting CD4+T cells as well. Whether T2DM ADSCs are impaired in antagonizing CD4+T cell proliferation and polarization remains unclear. METHODS We constructed type 2 diabetic ApoE-/- mouse models and tested infiltration and subgroups of CD4+T cell in stromal-vascular fraction (SVF) in vivo. Normal/T2DM ADSCs and normal splenocytes with or without CD4 sorting were separated and co-cultured at different scales ex vivo. Immune phenotypes of pro- and anti-inflammation of ADSCs were also investigated. Flow cytometry (FCM) and ELISA were applied in the experiments above. RESULTS CD4+T cells performed a more pro-inflammatory phenotype in adipose tissue in T2DM ApoE-/- mice in vivo. Restriction to CD4+T cell proliferation and polarization was manifested obviously weakened after co-cultured with T2DM ADSCs ex vivo. No obvious distinctions were found in morphology and growth type of both ADSCs. However, T2DM ADSCs acquired a pro-inflammatory immune phenotype, with secreting less PGE2 and expressing higher MHC-II and co-stimulatory molecules (CD40, CD80). Normal ADSCs could also obtain the phenotypic change after cultured with T2DM SVF supernatant. CONCLUSION CD4+T cell infiltration and pro-inflammatory polarization exist in adipose tissue in type 2 diabetic ApoE-/- mice. T2DM ADSCs had impaired function in restricting CD4+T lymphocyte proliferation and pro-inflammatory polarization due to immune phenotypic changes.
Collapse
Affiliation(s)
- Ming-Hao Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ya Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Lu Han
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Department of General Practice, Qilu Hospital of Shandong University, Ji'nan, China
| | - Yao-Yuan Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Di Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zhi-Hao Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Department of Geriatrics, Qilu Hospital of Shandong University, Ji'nan, China
| | - Hui-Min Zhou
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ming Song
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yi-Hui Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Meng-Xiong Tang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Department of Emergency, Qilu Hospital of Shandong University, Ji'nan, China
| | - Wei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ming Zhong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health; The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
27
|
Chen S, Jiang H, Peng H, Wu X, Fang J. The Utility of Ovotransferrin and Ovotransferrin-Derived Peptides as Possible Candidates in the Clinical Treatment of Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:6504518. [PMID: 28386310 PMCID: PMC5366766 DOI: 10.1155/2017/6504518] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 03/08/2017] [Indexed: 12/21/2022]
Abstract
Several of the most prevalent etiological factors which contribute towards global death rates are associated with cardiovascular diseases (CVDs), which include a range of conditions such as angina, rheumatic heart disease, and venous thrombosis. Extensive research has been conducted into the role played by oxidative stress and inflammation in the functional transformations associated with the progression of CVDs, while the research findings from these investigations have been both fruitful and informative. In view of the adverse secondary effects that result from the clinical administration of many synthetic medications, research which explored the treatment of severe and long-lasting conditions, including CVDs, has primarily centered on the potential benefits displayed by natural agents, one of which is food protein-based bioactive peptides. Most importantly, previous research has revealed the possible benefits associated with these products' anti-inflammatory and antioxidant characteristics. In light of these considerations, this paper aims to review the degree to which ovotransferrin (otrf, also referred to as conalbumin) and otrf-derived peptides, including IRW, IQW, and KVREGT, are, by virtue of their anti-inflammatory and antioxidant characteristics, viable treatment agents for endothelial dysfunction and the prevention of CVD.
Collapse
Affiliation(s)
- Shuang Chen
- College of Bioscience and Biotechnology and College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Hongmei Jiang
- College of Bioscience and Biotechnology and College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Hanhui Peng
- College of Bioscience and Biotechnology and College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Xiaosong Wu
- College of Bioscience and Biotechnology and College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Jun Fang
- College of Bioscience and Biotechnology and College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan Province University Key Laboratory for Agricultural Biochemistry and Biotransformation, Hunan Agricultural University, Changsha 410128, China
- Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha 410128, China
| |
Collapse
|
28
|
|
29
|
Yan WJ, Sun P, Wei DD, Wang SX, Yang JJ, Li YH, Zhang C. T cell immunoglobulin and mucin domain-containing molecule 3 on CD14 + monocytes serves as a novel biological marker for diabetes duration in type 2 diabetes mellitus. J Diabetes Investig 2016; 7:867-873. [PMID: 27182056 PMCID: PMC5089949 DOI: 10.1111/jdi.12523] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 02/29/2016] [Accepted: 03/21/2016] [Indexed: 12/19/2022] Open
Abstract
Aims/Introduction Type 2 diabetes is a worldwide disease that is associated with increased rates of obesity and reduced physical activity. Obesity‐associated insulin resistance in type 2 diabetes is a disorder in the balance between pro‐inflammatory and anti‐inflammatory signals. T cell immunoglobulin and mucin domain‐containing molecule 3 (Tim‐3) has been reported as an important regulatory inflammation molecule, and plays a pivotal role in several inflammation‐related diseases. Materials and Methods Peripheral blood mononuclear cells were obtained from type 2 diabetes patients (n = 31) and healthy donors (n = 18), and Tim‐3 expression on peripheral blood mononuclear cells was evaluated by flow cytometry. Results We showed the downregulated expression of Tim‐3 on CD14+ monocytes from type 2 diabetes patients. In addition, the upregulated expression of Tim‐3 on peripheral CD4+ T cells and CD8+ T cells was observed in the present study. The correlation analysis between Tim‐3 expression on CD14+ monocytes and diabetes duration showed the longer diabetes duration time, the lower Tim‐3 expression on CD14 monocytes. Conclusions The present results suggest that Tim‐3 might participate in the progression of type 2 diabetes by its negative regulation on these immune cells, and Tim‐3 on CD14+ monocytes serves as a novel biological marker for diabetes duration in type 2 diabetes patients.
Collapse
Affiliation(s)
- Wen-Jiang Yan
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Peng Sun
- Department of Intervention Oncology, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Dan-Dan Wei
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Shuang-Xi Wang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jing-Jing Yang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yi-Hui Li
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Cheng Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
30
|
Dong Z, Luo Y, Cai H, Zhang Z, Peng Z, Jiang M, Li Y, Li C, Li ZP, Feng ST. Noninvasive fat quantification of the liver and pancreas may provide potential biomarkers of impaired glucose tolerance and type 2 diabetes. Medicine (Baltimore) 2016; 95:e3858. [PMID: 27281097 PMCID: PMC4907675 DOI: 10.1097/md.0000000000003858] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The aim of the study is to investigate if the fat content of the liver and pancreas may indicate impaired glucose tolerance (IGT) or type 2 diabetes mellitus (T2DM). A total of 83 subjects (34 men; aged 46.5 ± 13.5 years) were characterized as T2DM, IGT, or normal glucose tolerant (NGT). NGT individuals were stratified as <40 or ≥40 years. Standard laboratory tests were conducted for insulin resistance and β-cell dysfunction. The magnetic resonance imaging Dixon technique was used to determine fat distribution in the liver and pancreas. Correlations among liver and pancreatic fat volume fractions (LFVFs and PFVFs, respectively) and laboratory parameters were analyzed. Among the groups, fat distribution was consistent throughout sections of the liver and pancreas, and LFVFs closely correlated with PFVFs. LFVFs correlated more closely than PFVFs with insulin resistance and β-cell function. Both the LFVFs and PFVFs were the highest in the T2DM patients, less in the IGT, and least in the NGT; all differences were significant. The PFVFs of the NGT subjects ≥40 years were significantly higher than that of those <40 years. The fat content of the liver and pancreas, particularly the liver, may be a biomarker for IGT and T2DM.
Collapse
Affiliation(s)
- Zhi Dong
- Department of Radiology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China,
| | - Yanji Luo
- Department of Radiology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China,
| | - Huasong Cai
- Department of Radiology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China,
| | - Zhongwei Zhang
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX,
| | - Zhenpeng Peng
- Department of Radiology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China,
| | - Mengjie Jiang
- Department of Radiology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China,
| | - Yanbing Li
- Department of Endocrinology and Diabetes Center, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chang Li
- Department of Radiology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China,
| | - Zi-Ping Li
- Department of Radiology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China,
| | - Shi-Ting Feng
- Department of Radiology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China,
- ∗Correspondence: Shi-Ting Feng, Department of Radiology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China (e-mail: ); Zi-Ping Li, Department of Radiology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China (e-mail: )
| |
Collapse
|
31
|
The potential of antioxidative and anti-inflammatory peptides in reducing the risk of cardiovascular diseases. Curr Opin Food Sci 2016. [DOI: 10.1016/j.cofs.2016.01.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
32
|
Majdoubi A, Kishta OA, Thibodeau J. Role of antigen presentation in the production of pro-inflammatory cytokines in obese adipose tissue. Cytokine 2016; 82:112-21. [PMID: 26854212 DOI: 10.1016/j.cyto.2016.01.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 01/27/2016] [Accepted: 01/27/2016] [Indexed: 02/06/2023]
Abstract
Type II diabetes regroups different physiological anomalies that ultimately lead to low-grade chronic inflammation, insulin resistance and loss of pancreatic β-cells. Obesity is one of the best examples of such a condition that can develop into Metabolic Syndrome, causing serious health problems of great socio-economic consequences. The pathological outcome of obesity has a genetic basis and depends on the delicate balance between pro- and anti-inflammatory effectors of the immune system. The causal link between obesity and inflammation is well established. While innate immunity plays a key role in the development of a pro-inflammatory state in obese adipose tissues, it has now become clear that adaptive immune cells are also involved and participate in the cascade of events that lead to metabolic perturbations. The efficacy of some immunotherapeutic protocols in reducing the symptoms of obesity-driven metabolic syndrome in mice implicated all arms of the immune response. Recently, the production of pathogenic immunoglobulins and pro-inflammatory cytokines by B and T lymphocytes suggested an auto-immune basis for the establishment of a non-healthy obese state. Understanding the cellular landscape of obese adipose tissues and how immune cells sustain chronic inflammation holds the key to the development of targeted therapies. In this review, we emphasize the role of antigen-presenting cells and MHC molecules in obese adipose tissue and the general contribution of the adaptive arm of the immune system in inflammation-induced insulin resistance.
Collapse
Affiliation(s)
- Abdelilah Majdoubi
- Laboratoire d'Immunologie Moléculaire, Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Québec, Canada
| | - Osama A Kishta
- Laboratoire d'Immunologie Moléculaire, Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Québec, Canada
| | - Jacques Thibodeau
- Laboratoire d'Immunologie Moléculaire, Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Québec, Canada.
| |
Collapse
|