1
|
Shinjo T, Nishimura F. The bidirectional association between diabetes and periodontitis, from basic to clinical. JAPANESE DENTAL SCIENCE REVIEW 2024; 60:15-21. [PMID: 38098853 PMCID: PMC10716706 DOI: 10.1016/j.jdsr.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/02/2023] [Accepted: 12/03/2023] [Indexed: 12/17/2023] Open
Abstract
The prevalence and severity of periodontitis are increased and advanced in diabetes. Severe periodontitis elicits adverse effects on diabetes by impairing insulin actions due to systemic microinflammation. Recent studies unveil the emerging findings and molecular basis of the bidirectional relationship between periodontitis and diabetes. In addition to conventional mechanisms such as hyperglycemia, hyperlipidemia, and chronic inflammation, deficient insulin action may play a pathogenic role in the progression of periodontitis under diabetes. Epidemiologically, from the viewpoint of the adverse effect of periodontitis on diabetes, recent studies have suggested that Asians including Japanese and Asian Americans with diabetes and mild obesity (BMI <25 kg/m2) should pay more attention to their increased risk for cardiovascular diseases. In this review, we summarize recent findings on the effect of diabetes on periodontitis from the viewpoint of abnormalities in metabolism and insulin resistance with novel mechanisms, and the influence of periodontitis on diabetes mainly focused on micro-inflammation related to mature adipose tissue and discuss future perspectives about novel approaches to interrupt the adverse interrelationship.
Collapse
Affiliation(s)
- Takanori Shinjo
- Section of Periodontology, Faculty of Dental Science, Kyushu University, Fukuoka, Fukuoka 812-8582, Japan
| | - Fusanori Nishimura
- Section of Periodontology, Faculty of Dental Science, Kyushu University, Fukuoka, Fukuoka 812-8582, Japan
| |
Collapse
|
2
|
Turner MB, Dalmasso C, Loria AS. The adipose tissue keeps the score: priming of the adrenal-adipose tissue axis by early life stress predisposes women to obesity and cardiometabolic risk. Front Endocrinol (Lausanne) 2024; 15:1481923. [PMID: 39493777 PMCID: PMC11527639 DOI: 10.3389/fendo.2024.1481923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/26/2024] [Indexed: 11/05/2024] Open
Abstract
Adverse Childhood Experiences (ACEs) refer to early life stress events, including abuse, neglect, and other psychosocial childhood traumas that can have long-lasting effects on a wide range of physiological functions. ACEs provoke sex-specific effects, whereas women have been shown to display a strong positive correlation with obesity and cardiometabolic disease. Notably, rodent models of chronic behavioral stress during postnatal life recapitulate several effects of ACEs in a sex-specific fashion. In this review, we will discuss the potential mechanisms uncovered by models of early life stress that may explain the greater susceptibility of females to obesity and metabolic risk compared with their male counterparts. We highlight the early life stress-induced neuroendocrine shaping of the adrenal-adipose tissue axis as a primary event conferring sex-dependent heightened sensitivity to obesity.
Collapse
Affiliation(s)
| | | | - Analia S. Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
3
|
Yang Z, Chen F, Zhang Y, Ou M, Tan P, Xu X, Li Q, Zhou S. Therapeutic targeting of white adipose tissue metabolic dysfunction in obesity: mechanisms and opportunities. MedComm (Beijing) 2024; 5:e560. [PMID: 38812572 PMCID: PMC11134193 DOI: 10.1002/mco2.560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 04/09/2024] [Accepted: 04/14/2024] [Indexed: 05/31/2024] Open
Abstract
White adipose tissue is not only a highly heterogeneous organ containing various cells, such as adipocytes, adipose stem and progenitor cells, and immune cells, but also an endocrine organ that is highly important for regulating metabolic and immune homeostasis. In individuals with obesity, dynamic cellular changes in adipose tissue result in phenotypic switching and adipose tissue dysfunction, including pathological expansion, WAT fibrosis, immune cell infiltration, endoplasmic reticulum stress, and ectopic lipid accumulation, ultimately leading to chronic low-grade inflammation and insulin resistance. Recently, many distinct subpopulations of adipose tissue have been identified, providing new insights into the potential mechanisms of adipose dysfunction in individuals with obesity. Therefore, targeting white adipose tissue as a therapeutic agent for treating obesity and obesity-related metabolic diseases is of great scientific interest. Here, we provide an overview of white adipose tissue remodeling in individuals with obesity including cellular changes and discuss the underlying regulatory mechanisms of white adipose tissue metabolic dysfunction. Currently, various studies have uncovered promising targets and strategies for obesity treatment. We also outline the potential therapeutic signaling pathways of targeting adipose tissue and summarize existing therapeutic strategies for antiobesity treatment including pharmacological approaches, lifestyle interventions, and novel therapies.
Collapse
Affiliation(s)
- Zi‐Han Yang
- Department of Plastic and Burn SurgeryWest China Hospital of Sichuan UniversityChengduChina
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fang‐Zhou Chen
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yi‐Xiang Zhang
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Min‐Yi Ou
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Poh‐Ching Tan
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xue‐Wen Xu
- Department of Plastic and Burn SurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Qing‐Feng Li
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shuang‐Bai Zhou
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
4
|
Sikder S, Pierce D, Sarkar ER, McHugh C, Quinlan KGR, Giacomin P, Loukas A. Regulation of host metabolic health by parasitic helminths. Trends Parasitol 2024; 40:386-400. [PMID: 38609741 DOI: 10.1016/j.pt.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 04/14/2024]
Abstract
Obesity is a worldwide pandemic and major risk factor for the development of metabolic syndrome (MetS) and type 2 diabetes (T2D). T2D requires lifelong medical support to limit complications and is defined by impaired glucose tolerance, insulin resistance (IR), and chronic low-level systemic inflammation initiating from adipose tissue. The current preventative strategies include a healthy diet, controlled physical activity, and medication targeting hyperglycemia, with underexplored underlying inflammation. Studies suggest a protective role for helminth infection in the prevention of T2D. The mechanisms may involve induction of modified type 2 and regulatory immune responses that suppress inflammation and promote insulin sensitivity. In this review, the roles of helminths in counteracting MetS, and prospects for harnessing these protective mechanisms for the development of novel anti-diabetes drugs are discussed.
Collapse
Affiliation(s)
- Suchandan Sikder
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia.
| | - Doris Pierce
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia
| | - Eti R Sarkar
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia; College of Public Health, Medical and Veterinary Sciences, James Cook University, Cairns, Queensland 4878, Australia
| | - Connor McHugh
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia; College of Public Health, Medical and Veterinary Sciences, James Cook University, Cairns, Queensland 4878, Australia
| | - Kate G R Quinlan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - Paul Giacomin
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia; Macrobiome Therapeutics Pty Ltd, Cairns, Queensland 4878, Australia
| | - Alex Loukas
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia; Macrobiome Therapeutics Pty Ltd, Cairns, Queensland 4878, Australia
| |
Collapse
|
5
|
Li N, Hu L, Li J, Ye Y, Bao Z, Xu Z, Chen D, Tang J, Gu Y. The Immunomodulatory effect of exosomes in diabetes: a novel and attractive therapeutic tool in diabetes therapy. Front Immunol 2024; 15:1357378. [PMID: 38720885 PMCID: PMC11076721 DOI: 10.3389/fimmu.2024.1357378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/03/2024] [Indexed: 05/12/2024] Open
Abstract
Exosomes carry proteins, metabolites, nucleic acids and lipids from their parent cell of origin. They are derived from cells through exocytosis, are ingested by target cells, and can transfer biological signals between local or distant cells. Therefore, exosomes are often modified in reaction to pathological processes, including infection, cancer, cardiovascular diseases and in response to metabolic perturbations such as obesity and diabetes, all of which involve a significant inflammatory aspect. Here, we discuss how immune cell-derived exosomes origin from neutrophils, T lymphocytes, macrophages impact on the immune reprogramming of diabetes and the associated complications. Besides, exosomes derived from stem cells and their immunomodulatory properties and anti-inflammation effect in diabetes are also reviewed. Moreover, As an important addition to previous reviews, we describes promising directions involving engineered exosomes as well as current challenges of clinical applications in diabetic therapy. Further research on exosomes will explore their potential in translational medicine and provide new avenues for the development of effective clinical diagnostics and therapeutic strategies for immunoregulation of diabetes.
Collapse
Affiliation(s)
- Na Li
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu, China
| | - Lingli Hu
- Graduate School of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jingyang Li
- Graduate School of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yang Ye
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu, China
| | - Zhengyang Bao
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu, China
| | - Zhice Xu
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu, China
| | - Daozhen Chen
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu, China
| | - Jiaqi Tang
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ying Gu
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Wuxi, Jiangsu, China
- Department of Obstetrics, Wuxi Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| |
Collapse
|
6
|
Su Z, Sun JY, Gao M, Sun W, Kong X. Molecular mechanisms and potential therapeutic targets in the pathogenesis of hypertension in visceral adipose tissue induced by a high-fat diet. Front Cardiovasc Med 2024; 11:1380906. [PMID: 38689862 PMCID: PMC11058983 DOI: 10.3389/fcvm.2024.1380906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/04/2024] [Indexed: 05/02/2024] Open
Abstract
Background Hypertension (HTN) presents a significant global public health challenge with diverse causative factors. The accumulation of visceral adipose tissue (VAT) due to a high-fat diet (HFD) is an independent risk factor for HTN. While various studies have explored pathogenic mechanisms, a comprehensive understanding of impact of VAT on blood pressure necessitates bioinformatics analysis. Methods Datasets GSE214618 and GSE188336 were acquired from the Gene Expression Omnibus and analyzed to identify shared differentially expressed genes between HFD-VAT and HTN-VAT. Gene Ontology enrichment and protein-protein interaction analyses were conducted, leading to the identification of hub genes. We performed molecular validation of hub genes using RT-qPCR, Western-blotting and immunofluorescence staining. Furthermore, immune infiltration analysis using CIBERSORTx was performed. Results This study indicated that the predominant characteristic of VAT in HTN was related to energy metabolism. The red functional module was enriched in pathways associated with mitochondrial oxidative respiration and ATP metabolism processes. Spp1, Postn, and Gpnmb in VAT were identified as hub genes on the pathogenic mechanism of HTN. Proteins encoded by these hub genes were closely associated with the target organs-specifically, the resistance artery, aorta, and heart tissue. After treatment with empagliflozin, there was a tendency for Spp1, Postn, and Gpnmb to decrease in VAT. Immune infiltration analysis confirmed that inflammation and immune response may not be the main mechanisms by which visceral adiposity contributes to HTN. Conclusions Our study pinpointed the crucial causative factor of HTN in VAT following HFD. Spp1, Postn, and Gpnmb in VAT acted as hub genes that promote elevated blood pressure and can be targets for HTN treatment. These findings contributed to therapeutic strategies and prognostic markers for HTN.
Collapse
Affiliation(s)
- Zhenyang Su
- School of Medicine, Southeast University, Nanjing, China
| | - Jin-Yu Sun
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Min Gao
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Wei Sun
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Xiangqing Kong
- School of Medicine, Southeast University, Nanjing, China
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| |
Collapse
|
7
|
Shrestha A, Zhu Y, Ali A. High-Fat Diet Influences Dendritic Cells and T-Cell Infiltration in Apical Periodontitis in Mice. J Endod 2024; 50:506-513.e2. [PMID: 38280515 DOI: 10.1016/j.joen.2024.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/29/2024]
Abstract
INTRODUCTION Diet-induced metabolic syndrome may influence the progression and healing of apical periodontitis (AP). The aim of this study was to evaluate the inflammatory immune response of dendritic cells (DCs) and T helper (Th) cells in normal versus obese mice with AP. METHODS Twenty male C57BL/6 mice were divided into 2 groups: normal chow (NC) and high-fat diet (HFD) for 12 wk. AP was induced in both groups by creating pulp exposure of the right first maxillary molar to the oral environment. Contralateral first molars from each mouse were used as a control. The animal's body mass was recorded on a weekly basis, and they were euthanized after 30 d. The maxillae were removed and processed for micro-computed tomography (micro-CT), histologic analysis, and immunofluorescence staining for DCs (CD11c), Th17 (IL-17A), and T regulatory cells (FOXP3 and IL-10). Different groups were analyzed by Mann-Whitney U test, Student t test, and ordinary 1-way analysis of variance followed by Tukey's multiple comparisons test. The level of significance (α) was set at 0.05. RESULTS The HFD group showed larger AP lesions than the NC group from micro-CT analysis. For the NC group, induction of AP significantly increased immune cell infiltration when compared with control. HFD showed increased DCs and Th17 infiltration in the control group without AP. In addition, there was no significant change in the amount of DCs and Th17 in the HFD-AP group when compared with the NC-AP and HFD-control groups. CONCLUSIONS HFD resulted in an increased immune cell infiltration in the periapical area without AP. Despite the larger AP lesion observed in HFD-AP than that of NC-AP, the amount of infiltrated inflammatory cells did not differ significantly. The results of this study suggest that the DCs and Th17 inflammatory pathways are affected by HFD in the periapical region, but their contribution toward AP complicated by metabolic syndrome requires further investigation.
Collapse
Affiliation(s)
- Annie Shrestha
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Dentistry, Mt. Sinai Hospital, Toronto, Toronto, Ontario, Canada.
| | - Yi Zhu
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Aiman Ali
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Soedono S, Sharlene S, Vo DHN, Averia M, Rosalie EE, Lee YK, Cho KW. Obese visceral adipose dendritic cells downregulate regulatory T cell development through IL-33. Front Immunol 2024; 15:1335651. [PMID: 38566998 PMCID: PMC10985834 DOI: 10.3389/fimmu.2024.1335651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 03/08/2024] [Indexed: 04/04/2024] Open
Abstract
Regulatory T cells (Tregs) residing in visceral adipose tissue (VAT) play a pivotal role in regulating tissue inflammation and metabolic dysfunction associated with obesity. However, the specific phenotypic and functional characteristics of Tregs in obese VAT, as well as the regulatory mechanisms shaping them, remain elusive. This study demonstrates that obesity selectively reduces Tregs in VAT, characterized by restrained proliferation, heightened PD-1 expression, and diminished ST2 expression. Additionally, obese VAT displays distinctive maturation of dendritic cells (DCs), marked by elevated expressions of MHC-II, CD86, and PD-L1, which are inversely correlated with VAT Tregs. In an in vitro co-culture experiment, only obese VAT DCs, not macrophages or DCs from subcutaneous adipose tissue (SAT) and spleen, result in decreased Treg differentiation and proliferation. Furthermore, Tregs differentiated by obese VAT DCs exhibit distinct characteristics resembling those of Tregs in obese VAT, such as reduced ST2 and IL-10 expression. Mechanistically, obesity lowers IL-33 production in VAT DCs, contributing to the diminished Treg differentiation. These findings collectively underscore the critical role of VAT DCs in modulating Treg generation and shaping Treg phenotype and function during obesity, potentially contributing to the regulation of VAT Treg populations.
Collapse
Affiliation(s)
- Shindy Soedono
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Republic of Korea
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Sharlene Sharlene
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Republic of Korea
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Dan Hoang Nguyet Vo
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Republic of Korea
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Maria Averia
- Magister of Biotechnology, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Eufrasia Elaine Rosalie
- Faculty of Biotechnology, Department of Food Technology, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Yun Kyung Lee
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Republic of Korea
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| | - Kae Won Cho
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Republic of Korea
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Republic of Korea
| |
Collapse
|
9
|
Satoh M, Iwabuchi K. Contribution of NKT cells and CD1d-expressing cells in obesity-associated adipose tissue inflammation. Front Immunol 2024; 15:1365843. [PMID: 38426085 PMCID: PMC10902011 DOI: 10.3389/fimmu.2024.1365843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 01/30/2024] [Indexed: 03/02/2024] Open
Abstract
Natural killer T (NKT) cell are members of the innate-like T lymphocytes and recognizes lipid antigens presented by CD1d-expressing cells. Obesity-associated inflammation in adipose tissue (AT) leads to metabolic dysfunction, including insulin resistance. When cellular communication is properly regulated among AT-residing immune cells and adipocytes during inflammation, a favorable balance of Th1 and Th2 immune responses is achieved. NKT cells play crucial roles in AT inflammation, influencing the development of diet-induced obesity and insulin resistance. NKT cells interact with CD1d-expressing cells in AT, such as adipocytes, macrophages, and dendritic cells, shaping pro-inflammatory or anti-inflammatory microenvironments with distinct characteristics depending on the antigen-presenting cells. Additionally, CD1d may be involved in the inflammatory process independently of NKT cells. In this mini-review, we provide a brief overview of the current understanding of the interaction between immune cells, focusing on NKT cells and CD1d signaling, which control AT inflammation both in the presence and absence of NKT cells. We aim to enhance our understanding of the mechanisms of obesity-associated diseases.
Collapse
Affiliation(s)
- Masashi Satoh
- Department of Immunology, Kitasato University School of Medicine, Sagamihara, Japan
| | | |
Collapse
|
10
|
Bradley D, Deng T, Shantaram D, Hsueh WA. Orchestration of the Adipose Tissue Immune Landscape by Adipocytes. Annu Rev Physiol 2024; 86:199-223. [PMID: 38345903 DOI: 10.1146/annurev-physiol-042222-024353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
Obesity is epidemic and of great concern because of its comorbid and costly inflammatory-driven complications. Extensive investigations in mice have elucidated highly coordinated, well-balanced interactions between adipocytes and immune cells in adipose tissue that maintain normal systemic metabolism in the lean state, while in obesity, proinflammatory changes occur in nearly all adipose tissue immune cells. Many of these changes are instigated by adipocytes. However, less is known about obesity-induced adipose-tissue immune cell alterations in humans. Upon high-fat diet feeding, the adipocyte changes its well-known function as a metabolic cell to assume the role of an immune cell, orchestrating proinflammatory changes that escalate inflammation and progress during obesity. This transformation is particularly prominent in humans. In this review, we (a) highlight a leading and early role for adipocytes in promulgating inflammation, (b) discuss immune cell changes and the time course of these changes (comparing humans and mice when possible), and (c) note how reversing proinflammatory changes in most types of immune cells, including adipocytes, rescues adipose tissue from inflammation and obese mice from insulin resistance.
Collapse
Affiliation(s)
- David Bradley
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA;
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Pennsylvania State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA;
| | - Tuo Deng
- Second Xiangya Hospital, Central South University, Changsha, China
| | - Dharti Shantaram
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA;
| | - Willa A Hsueh
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA;
| |
Collapse
|
11
|
Hildreth AD, Padilla ET, Gupta M, Wong YY, Sun R, Legala AR, O'Sullivan TE. Adipose cDC1s contribute to obesity-associated inflammation through STING-dependent IL-12 production. Nat Metab 2023; 5:2237-2252. [PMID: 37996702 DOI: 10.1038/s42255-023-00934-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 10/18/2023] [Indexed: 11/25/2023]
Abstract
Obesity is associated with chronic low-grade white adipose tissue (WAT) inflammation that can contribute to the development of insulin resistance in mammals. Previous studies have identified interleukin (IL)-12 as a critical upstream regulator of WAT inflammation and metabolic dysfunction during obesity. However, the cell types and mechanisms that initiate WAT IL-12 production remain unclear. Here we show that conventional type 1 dendritic cells (cDC1s) are the cellular source of WAT IL-12 during obesity through analysis of mouse and human WAT single-cell transcriptomic datasets, IL-12 reporter mice and IL-12p70 protein levels by enzyme-linked immunosorbent assay. We demonstrate that cDC1s contribute to obesity-associated inflammation by increasing group 1 innate lymphocyte interferon-γ production and inflammatory macrophage accumulation. Inducible depletion of cDC1s increased WAT insulin sensitivity and systemic glucose tolerance during diet-induced obesity. Mechanistically, endocytosis of apoptotic bodies containing self-DNA by WAT cDC1s drives stimulator of interferon genes (STING)-dependent IL-12 production. Together, these results suggest that WAT cDC1s act as critical regulators of adipose tissue inflammation and metabolic dysfunction during obesity.
Collapse
Affiliation(s)
- Andrew D Hildreth
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Eddie T Padilla
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Meha Gupta
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Yung Yu Wong
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ryan Sun
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Akshara R Legala
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Timothy E O'Sullivan
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
12
|
Guo C, Chi H. Immunometabolism of dendritic cells in health and disease. Adv Immunol 2023; 160:83-116. [PMID: 38042587 PMCID: PMC11086980 DOI: 10.1016/bs.ai.2023.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2023]
Abstract
Dendritic cells (DCs) are crucial mediators that bridge the innate and adaptive immune responses. Cellular rewiring of metabolism is an emerging regulator of the activation, migration, and functional specialization of DC subsets in specific microenvironments and immunological conditions. DCs undergo metabolic adaptation to exert immunogenic or tolerogenic effects in different contexts. Also, beyond their intracellular metabolic and signaling roles, metabolites and nutrients mediate the intercellular crosstalk between DCs and other cell types, and such crosstalk orchestrates DC function and immune responses. Here, we provide a comprehensive review of the metabolic regulation of DC biology in various contexts and summarize the current understanding of such regulation in directing immune homeostasis and inflammation, specifically with respect to infections, autoimmunity, tolerance, cancer, metabolic diseases, and crosstalk with gut microbes. Understanding context-specific metabolic alterations in DCs may identify mechanisms for physiological and pathological functions of DCs and yield potential opportunities for therapeutic targeting of DC metabolism in many diseases.
Collapse
Affiliation(s)
- Chuansheng Guo
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States.
| |
Collapse
|
13
|
Mukherjee S, Skrede S, Haugstøyl M, López M, Fernø J. Peripheral and central macrophages in obesity. Front Endocrinol (Lausanne) 2023; 14:1232171. [PMID: 37720534 PMCID: PMC10501731 DOI: 10.3389/fendo.2023.1232171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/28/2023] [Indexed: 09/19/2023] Open
Abstract
Obesity is associated with chronic, low-grade inflammation. Excessive nutrient intake causes adipose tissue expansion, which may in turn cause cellular stress that triggers infiltration of pro-inflammatory immune cells from the circulation as well as activation of cells that are residing in the adipose tissue. In particular, the adipose tissue macrophages (ATMs) are important in the pathogenesis of obesity. A pro-inflammatory activation is also found in other organs which are important for energy metabolism, such as the liver, muscle and the pancreas, which may stimulate the development of obesity-related co-morbidities, including insulin resistance, type 2 diabetes (T2D), cardiovascular disease (CVD) and non-alcoholic fatty liver disease (NAFLD). Interestingly, it is now clear that obesity-induced pro-inflammatory signaling also occurs in the central nervous system (CNS), and that pro-inflammatory activation of immune cells in the brain may be involved in appetite dysregulation and metabolic disturbances in obesity. More recently, it has become evident that microglia, the resident macrophages of the CNS that drive neuroinflammation, may also be activated in obesity and can be relevant for regulation of hypothalamic feeding circuits. In this review, we focus on the action of peripheral and central macrophages and their potential roles in metabolic disease, and how macrophages interact with other immune cells to promote inflammation during obesity.
Collapse
Affiliation(s)
- Sayani Mukherjee
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Silje Skrede
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Martha Haugstøyl
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Johan Fernø
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
14
|
Zhang Q, Jin Y, Xin X, An Z, Hu YY, Li Y, Feng Q. A high-trans fat, high-carbohydrate, high-cholesterol, high-cholate diet-induced nonalcoholic steatohepatitis mouse model and its hepatic immune response. Nutr Metab (Lond) 2023; 20:28. [PMID: 37244987 DOI: 10.1186/s12986-023-00749-w] [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: 10/20/2022] [Accepted: 05/08/2023] [Indexed: 05/29/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic progressive disease that can progress to non-alcoholic steatohepatitis (NASH). Animal models are important tools for basic NASH research. Immune activation plays a key role in liver inflammation in patients with NASH. We established a high-trans fat, high-carbohydrate, and high-cholesterol, high-cholate diet-induced (HFHCCC) mouse model. C57BL/6 mice were fed a normal or HFHCCC diet for 24 weeks, and the immune response characteristics of this model were evaluated. The proportion of immune cells in mouse liver tissues was detected by immunohistochemistry and flow cytometry, Multiplex bead immunoassay and Luminex technology was used to detecte the expression of cytokines in mouse liver tissues. The results showed that mice treated with HFHCCC diet exhibited remarkably increased hepatic triglycerides (TG) content, and the increase in plasma transaminases resulted in hepatocyte injury. Biochemical results showed that HFHCCC induced elevated hepatic lipids, blood glucose, insulin; marked hepatocyte steatosis, ballooning, inflammation, and fibrosis. The proportion of innate immunity-related cells, including Kupffer cells (KCs), neutrophils, dendritic cells (DCs), natural killer T cells (NKT), and adaptive immunity-related CD3+ T cells increased; interleukin-1α (IL-1α), IL-1β, IL-2, IL-6, IL-9, and chemokines, including CCL2, CCL3, and macrophage colony stimulating factor (G-CSF) increased. The constructed model closely approximated the characteristics of human NASH and evaluation of its immune response signature, showed that the innate immune response was more pronounced than adaptive immunity. Its use as an experimental tool for understanding innate immune responses in NASH is recommended.
Collapse
Affiliation(s)
- Qian Zhang
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong District, Shanghai, 201203, China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Pudong New Area, Shanghai, 201203, China
| | - Yue Jin
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong District, Shanghai, 201203, China
| | - Xin Xin
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong District, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, 201203, China
| | - Ziming An
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong District, Shanghai, 201203, China
| | - Yi-Yang Hu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong District, Shanghai, 201203, China
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Yajuan Li
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Pudong New Area, Shanghai, 201203, China.
| | - Qin Feng
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong District, Shanghai, 201203, China.
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, 201203, China.
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China.
| |
Collapse
|
15
|
Sullivan M, Fernandez-Aranda F, Camacho-Barcia L, Harkin A, Macrì S, Mora-Maltas B, Jiménez-Murcia S, O'Leary A, Ottomana AM, Presta M, Slattery D, Scholtz S, Glennon JC. Insulin and Disorders of Behavioural Flexibility. Neurosci Biobehav Rev 2023; 150:105169. [PMID: 37059405 DOI: 10.1016/j.neubiorev.2023.105169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/03/2023] [Accepted: 04/10/2023] [Indexed: 04/16/2023]
Abstract
Behavioural inflexibility is a symptom of neuropsychiatric and neurodegenerative disorders such as Obsessive-Compulsive Disorder, Autism Spectrum Disorder and Alzheimer's Disease, encompassing the maintenance of a behaviour even when no longer appropriate. Recent evidence suggests that insulin signalling has roles apart from its regulation of peripheral metabolism and mediates behaviourally-relevant central nervous system (CNS) functions including behavioural flexibility. Indeed, insulin resistance is reported to generate anxious, perseverative phenotypes in animal models, with the Type 2 diabetes medication metformin proving to be beneficial for disorders including Alzheimer's Disease. Structural and functional neuroimaging studies of Type 2 diabetes patients have highlighted aberrant connectivity in regions governing salience detection, attention, inhibition and memory. As currently available therapeutic strategies feature high rates of resistance, there is an urgent need to better understand the complex aetiology of behaviour and develop improved therapeutics. In this review, we explore the circuitry underlying behavioural flexibility, changes in Type 2 diabetes, the role of insulin in CNS outcomes and mechanisms of insulin involvement across disorders of behavioural inflexibility.
Collapse
Affiliation(s)
- Mairéad Sullivan
- Conway Institute of Biomedical and Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland.
| | - Fernando Fernandez-Aranda
- Department of Psychiatry, University Hospital of Bellvitge, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Barcelona, Spain; Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Lucía Camacho-Barcia
- Department of Psychiatry, University Hospital of Bellvitge, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Barcelona, Spain
| | - Andrew Harkin
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland
| | - Simone Macrì
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Bernat Mora-Maltas
- Department of Psychiatry, University Hospital of Bellvitge, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Susana Jiménez-Murcia
- Department of Psychiatry, University Hospital of Bellvitge, Barcelona, Spain; Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Barcelona, Spain; Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Aet O'Leary
- University Hospital Frankfurt, Frankfurt, Germany
| | - Angela Maria Ottomana
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; Neuroscience Unit, Department of Medicine, University of Parma, 43100 Parma, Italy
| | - Martina Presta
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
| | | | | | - Jeffrey C Glennon
- Conway Institute of Biomedical and Biomolecular Research, School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
16
|
Sun Y, Wang B, Hu Q, Zhang H, Lai X, Wang T, Zhao C, Wang J, Zhang X, Niu Q, He B, Jiang E, Shi M, Feng X, Luo Y. Loss of Lkb1 in CD11c + myeloid cells protects mice from diet-induced obesity while enhancing glucose intolerance and IL-17/IFN-γ imbalance. Cell Mol Life Sci 2023; 80:63. [PMID: 36781473 PMCID: PMC9925521 DOI: 10.1007/s00018-023-04707-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/04/2023] [Accepted: 01/22/2023] [Indexed: 02/15/2023]
Abstract
Adipose tissue CD11c+ myeloid cell is an independent risk factor associated with obesity and metabolic disorders. However, the underlying molecular basis remains elusive. Here, we demonstrated that liver kinase B1 (Lkb1), a key bioenergetic sensor, is involved in CD11c+ cell-mediated immune responses in diet-induced obesity. Loss of Lkb1 in CD11c+ cells results in obesity resistance but lower glucose tolerance, which accompanies tissue-specific immune abnormalities. The accumulation and CD80's expression of Lkb1 deficient adipose-tissue specific dendritic cells but not macrophages is restrained. Additionally, the balance of IL-17A and IFN-γ remarkably tips towards the latter in fat T cells and CD11c- macrophages. Mechanistically, IFN-γ promotes apoptosis of preadipocytes and inhibits their adipogenesis while IL-17A promotes the adipogenesis in vitro, which might account in part for the fat gain resistant phenotype. In summary, these findings reveal that Lkb1 is essential for fat CD11c+ dendritic cells responding to HFD exposure and provides new insights into the IL-17A/IFN-γ balance in HFD-induced obesity.
Collapse
Affiliation(s)
- Yunyan Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China.,Tianjin Institutes of Health Science, Tianjin, 301600, China.,Department of Hematology, Hematology Research Center of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, China.,Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Center, Kunming, China
| | - Bing Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China.,Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Qianwen Hu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China.,Tianjin Institutes of Health Science, Tianjin, 301600, China.,Department of Hematology, Hematology Research Center of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haixiao Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China.,Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Xun Lai
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Center, Kunming, China
| | - Tier Wang
- Department of Hematology, Hematology Research Center of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Chunxiao Zhao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China.,Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Jiali Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China.,Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Xi Zhang
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Center, Kunming, China
| | - Qing Niu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China.,Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Baolin He
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China.,Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Erlie Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China. .,Tianjin Institutes of Health Science, Tianjin, 301600, China.
| | - Mingxia Shi
- Department of Hematology, Hematology Research Center of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Xiaoming Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China. .,Tianjin Institutes of Health Science, Tianjin, 301600, China.
| | - Yuechen Luo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China. .,Tianjin Institutes of Health Science, Tianjin, 301600, China.
| |
Collapse
|
17
|
English J, Orofino J, Cederquist CT, Paul I, Li H, Auwerx J, Emili A, Belkina A, Cardamone D, Perissi V. GPS2-mediated regulation of the adipocyte secretome modulates adipose tissue remodeling at the onset of diet-induced obesity. Mol Metab 2023; 69:101682. [PMID: 36731652 PMCID: PMC9922684 DOI: 10.1016/j.molmet.2023.101682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 01/22/2023] [Indexed: 02/01/2023] Open
Abstract
OBJECTIVE Dysfunctional, unhealthy expansion of white adipose tissue due to excess dietary intake is a process at the root of obesity and Type 2 Diabetes development. The objective of this study is to contribute to a better understanding of the underlying mechanism(s) regulating the early stages of adipose tissue expansion and adaptation to dietary stress due to an acute, high-fat diet (HFD) challenge, with a focus on the communication between adipocytes and other stromal cells. METHODS We profiled the early response to high-fat diet exposure in wildtype and adipocyte-specific GPS2-KO (GPS2-AKO) mice at the cellular, tissue and organismal level. A multi-pronged approach was employed to disentangle the complex cellular interactions dictating tissue remodeling, via single-cell RNA sequencing and FACS profiling of the stromal fraction, and semi-quantitative proteomics of the adipocyte-derived exosomal cargo after 5 weeks of HFD feeding. RESULTS Our results indicate that loss of GPS2 in mature adipocytes leads to impaired adaptation to the metabolic stress imposed by HFD feeding. GPS2-AKO mice are significantly more inflamed, insulin resistant, and obese, compared to the WT counterparts. At the cellular level, lack of GPS2 in adipocytes impacts upon other stromal populations, with both the eWAT and scWAT depots exhibiting changes in the immune and non-immune compartments that contribute to an increase in inflammatory and anti-adipogenic cell types. Our studies also revealed that adipocyte to stromal cell communication is facilitated by exosomes, and that transcriptional rewiring of the exosomal cargo is crucial for tissue remodeling. Loss of GPS2 results in increased expression of secreted factors promoting a TGFβ-driven fibrotic microenvironment favoring unhealthy tissue remodeling and expansion. CONCLUSIONS Adipocytes serve as an intercellular signaling hub, communicating with the stromal compartment via paracrine signaling. Our study highlights the importance of proper regulation of the 'secretome' released by energetically stressed adipocytes at the onset of obesity. Altered transcriptional regulation of factors secreted via adipocyte-derived exosomes (AdExos), in the absence of GPS2, contributes to the establishment of an anti-adipogenic, pro-fibrotic adipose tissue environment, and to hastened progression towards a metabolically dysfunctional phenotype.
Collapse
Affiliation(s)
- Justin English
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA; Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| | - Joseph Orofino
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA.
| | - Carly T. Cederquist
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Indranil Paul
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA; Center for Network Systems Biology, Boston University, Boston, MA, USA.
| | - Hao Li
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland.
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland.
| | - Andrew Emili
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA; Center for Network Systems Biology, Boston University, Boston, MA, USA.
| | - Anna Belkina
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA.
| | - Dafne Cardamone
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA.
| | - Valentina Perissi
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA; School of Life Science, Northwestern Polytechnical University, Xi'an 710072, China.
| |
Collapse
|
18
|
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: 49] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [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
|
19
|
Essential Minerals and Metabolic Adaptation of Immune Cells. Nutrients 2022; 15:nu15010123. [PMID: 36615781 PMCID: PMC9824256 DOI: 10.3390/nu15010123] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/20/2022] [Accepted: 12/24/2022] [Indexed: 12/29/2022] Open
Abstract
Modern lifestyles deviated considerably from the ancestral routines towards major shifts in diets and increased sedentarism. The trace elements status of the human body is no longer adequately supported by micronutrient-inferior farmed meats and crop commodities produced by the existing agricultural food systems. This is particular evident in the increased obesogenic adipogenesis and low-grade inflammation that fails to resolve with time. The metabolically restrictive environment of the inflamed tissues drives activation and proliferation of transient and resident populations of immune cells in favor of pro-inflammatory phenotypes, as well as a part of the enhanced autoimmune response. As different stages of the immune activation and resolution depend on the availability of specific minerals to maintain the structural integrity of skin and mucus membranes, activation and migration of immune cells, activation of the complement system, and the release of pro-inflammatory cytokines and chemokines, this review discusses recent advances in our understanding of the contribution of select minerals in optimizing the responses of innate and adaptive immune outcomes. An abbreviated view on the absorption, transport, and delivery of minerals to the body tissues as related to metabolic adaptation is considered.
Collapse
|
20
|
Anti-Inflammatory Mechanisms of Dietary Flavones: Tapping into Nature to Control Chronic Inflammation in Obesity and Cancer. Int J Mol Sci 2022; 23:ijms232415753. [PMID: 36555392 PMCID: PMC9779861 DOI: 10.3390/ijms232415753] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Flavones are natural phytochemicals broadly distributed in our diet. Their anti-inflammatory properties provide unique opportunities to control the innate immune system and inflammation. Here, we review the role of flavones in chronic inflammation with an emphasis on their impact on the molecular mechanisms underlying inflammatory diseases including obesity and cancer. Flavones can influence the innate immune cell repertoire restoring the immune landscape. Flavones impinge on NF-κB, STAT, COX-2, or NLRP3 inflammasome pathways reestablishing immune homeostasis. Devoid of adverse side effects, flavones could present alternative opportunities for the treatment and prevention of chronic inflammation that contributes to obesity and cancer.
Collapse
|
21
|
Qian X, Meng X, Zhang S, Zeng W. Neuroimmune regulation of white adipose tissues. FEBS J 2022; 289:7830-7853. [PMID: 34564950 DOI: 10.1111/febs.16213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/21/2021] [Accepted: 09/24/2021] [Indexed: 01/14/2023]
Abstract
The white adipose tissues (WAT) are located in distinct depots throughout the body. They serve as an energy reserve, providing fatty acids for other tissues via lipolysis when needed, and function as an endocrine organ to regulate systemic metabolism. Their activities are coordinated through intercellular communications among adipocytes and other cell types such as residential and infiltrating immune cells, which are collectively under neuronal control. The adipocytes and immune subtypes including macrophages/monocytes, eosinophils, neutrophils, group 2 innate lymphoid cells (ILC2s), T and B cells, dendritic cells (DCs), and natural killer (NK) cells display cellular and functional diversity in response to the energy states and contribute to metabolic homeostasis and pathological conditions. Accumulating evidence reveals that neuronal innervations control lipid deposition and mobilization via regulating lipolysis, adipocyte size, and cellularity. Vice versa, the neuronal innervations and activity are influenced by cellular factors in the WAT. Though the literature describing adipose tissue cells is too extensive to cover in detail, we strive to highlight a selected list of neuronal and immune components in this review. The cell-to-cell communications and the perspective of neuroimmune regulation are emphasized to enlighten the potential therapeutic opportunities for treating metabolic disorders.
Collapse
Affiliation(s)
- Xinmin Qian
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Xia Meng
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Shan Zhang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Wenwen Zeng
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China.,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| |
Collapse
|
22
|
Li H, Meng Y, He S, Tan X, Zhang Y, Zhang X, Wang L, Zheng W. Macrophages, Chronic Inflammation, and Insulin Resistance. Cells 2022; 11:cells11193001. [PMID: 36230963 PMCID: PMC9562180 DOI: 10.3390/cells11193001] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/08/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
The prevalence of obesity has reached alarming levels, which is considered a major risk factor for several metabolic diseases, including type 2 diabetes (T2D), non-alcoholic fatty liver, atherosclerosis, and ischemic cardiovascular disease. Obesity-induced chronic, low-grade inflammation may lead to insulin resistance, and it is well-recognized that macrophages play a major role in such inflammation. In the current review, the molecular mechanisms underlying macrophages, low-grade tissue inflammation, insulin resistance, and T2D are described. Also, the role of macrophages in obesity-induced insulin resistance is presented, and therapeutic drugs and recent advances targeting macrophages for the treatment of T2D are introduced.
Collapse
Affiliation(s)
- He Li
- Beijing City Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ya Meng
- Beijing City Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Shuwang He
- Shandong DYNE Marine Biopharmaceutical Co., Ltd., Rongcheng 264300, China
| | - Xiaochuan Tan
- Beijing City Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yujia Zhang
- Beijing City Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xiuli Zhang
- Beijing City Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Lulu Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
- Correspondence: (L.W.); (W.Z.); Tel.: +86-010-63165233 (W.Z.)
| | - Wensheng Zheng
- Beijing City Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Correspondence: (L.W.); (W.Z.); Tel.: +86-010-63165233 (W.Z.)
| |
Collapse
|
23
|
Wang Y, Zhang Y, Li Y, Liu Y, Liu Y. Signature of gene expression profile of liver sinusoidal endothelial cells in nonalcoholic steatohepatitis. Front Cell Dev Biol 2022; 10:946566. [PMID: 36211451 PMCID: PMC9533023 DOI: 10.3389/fcell.2022.946566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/26/2022] [Indexed: 11/13/2022] Open
Abstract
Background: There has been emerging evidence that liver sinusoidal endothelial cells (LSECs) play an important role in the pathogenesis of nonalcoholic steatohepatitis (NASH). This study aims to figure out the signature of the gene expression profile of LSECs in NASH and to explore potential biomarkers related to damaged LSECs in NASH.Methods and materials: Animal experiments were performed to demonstrate the significant structural damage of LSECs in the NASH model. To further understand the functional changes of these damaged LSECs in NASH, we used the public GEO database that contained microarray data for the gene expression of LSECs in NASH and normal mouse liver. Differentially expressed genes (DEGs) were analyzed, and further Gene Ontology (GO) enrichment analysis was performed to understand the functional changes. The hub genes were then identified and validated via external GEO databases.Results: There was significant structural damage to LSECs in the NASH model, accompanied by remarkable functional changes of LSECs with 174 DEGs (156 upregulated and 18 downregulated genes). The functions of these DEGs were mainly enriched in the inflammatory reactions and immune responses. Nine specifically expressed hub genes were identified. Among them, CCL4 and ITGAX showed the most significant correlation with NASH, with AUROC of 0.77 and 0.86, respectively. The protein–protein interaction network, mRNA–miRNA interaction network, and ceRNA network were further predicted.Conclusion: LSECs show significant structural damage and functional changes in NASH. The LSEC-related DEGs, such as CCL4 and ITGAX, might be promising biomarkers as well as potential treatment targets for NASH.
Collapse
Affiliation(s)
- Yang Wang
- Department of Gastroenterology, Peking University People’s Hospital, Beijing, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing, China
- *Correspondence: Yang Wang, ; Yulan Liu,
| | - Yifan Zhang
- Department of Gastroenterology, Peking University People’s Hospital, Beijing, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing, China
| | - Yimin Li
- Department of Rheumatology and Immunology, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Peking University People’s Hospital, Beijing, China
| | - Yun Liu
- Department of Gastroenterology, Peking University People’s Hospital, Beijing, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing, China
| | - Yulan Liu
- Department of Gastroenterology, Peking University People’s Hospital, Beijing, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing, China
- *Correspondence: Yang Wang, ; Yulan Liu,
| |
Collapse
|
24
|
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: 13.0] [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
|
25
|
Barranco-Fragoso B, Pal SC, Díaz-Orozco LE, Dorantes-Heredia R, Qi X, Méndez-Sánchez N. Identification of Hepatic Dendritic Cells in Liver Biopsies Showing Steatosis in Patients with Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD) Associated with Obesity. MEDICAL SCIENCE MONITOR : INTERNATIONAL MEDICAL JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2022; 28:e937528. [PMID: 35934868 PMCID: PMC9373829 DOI: 10.12659/msm.937528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Background Metabolic dysfunction-associated fatty liver disease (MAFLD) is now the term used for hepatic steatosis in patients who are overweight or obese, have type 2 diabetes mellitus (T2DM), or evidence of metabolic dysregulation. The prevalence of MAFLD among morbidly obese subjects is 65–93%. Hepatic dendritic cells (hDCs) are antigen-presenting cells that induce T cell-mediated immunity. MAFLD pathogenesis involves numerous immune cell-mediated inflammatory processes, while the particular role of hDCs is yet to be well defined. This study aimed to identify hDCs in liver biopsies from 128 patients with MAFLD associated with obesity. Material/Methods In this cross-sectional study, 128 liver biopsies from 128 patients with MAFLD (diagnosed as presence of hepatic steatosis, plus T2DM, metabolic dysregulation or overweight/obesity) were collected and assessed for CD11c+ immunoreactivity degree (CD11c as dendritic cell biomarker), through antigen retrieval, reaction with CD11c antibodies (primary), and marking with diaminobenzidine chromogen. Results Among the 128 patients with MAFLD, 64 (50%) had MAFLD and fibrosis and 72 (56.2%) positively expressed hDCs (CD11c+). Among morbidly obese patients, 49 (64.5%) positively expressed hDCs (CD11c+) in liver tissue; from patients with obesity grade I- grade II (GI–II), 18 (54.5%) positively expressed hDCs (CD11c+) in liver tissue; and from non-obese patients with MAFLD, 5 (26.3%) positively expressed hDCs (CD11c+) in liver tissue. Conclusions hDC expression increases significantly in morbidly obese patients with MAFLD compared with non-obese patients, independent of the degree of fibrosis, suggesting the role of adaptive changes within hDCs in the perpetuation of inflammatory insults in chronic liver diseases.
Collapse
Affiliation(s)
- Beatriz Barranco-Fragoso
- Department of Gastroenterology, National Medical Center "20 de Noviembre", ISSSTE, Mexico City, Mexico.,University's Program of Research in Health (Programa Universitario de Investigación en Salud (PUIS), National Autonomous University of Mexico, Mexico City, Mexico
| | - Shreya C Pal
- Liver Research Unit, Medica Sur Clinic Foundation, Mexico City, Mexico.,Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Luis E Díaz-Orozco
- Liver Research Unit, Medica Sur Clinic Foundation, Mexico City, Mexico.,Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | | | - Xingshun Qi
- Department of Gastroenterology, General Hospital of Northern Theater Command (formerly General Hospital of Shenyang Military Area), Shenyang, Liaoning, China (mainland)
| | - Nahum Méndez-Sánchez
- Liver Research Unit, Medica Sur Clinic Foundation, Mexico City, Mexico.,Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| |
Collapse
|
26
|
Chen J, Wang W, Ni Q, Zhang L, Guo X. Interleukin 6-regulated macrophage polarization controls atherosclerosis-associated vascular intimal hyperplasia. Front Immunol 2022; 13:952164. [PMID: 35967343 PMCID: PMC9363591 DOI: 10.3389/fimmu.2022.952164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Vascular intimal hyperplasia (VIH) is an important stage of atherosclerosis (AS), in which macrophages not only play a critical role in local inflammation, but also transform into foam cells to participate into plaque formation, where they appear to be heterogeneous. Recently, it was shown that CD11c+ macrophages were more associated with active plaque progression. However, the molecular regulation of phenotypic changes of plaque macrophages during VIH has not been clarified and thus addressed in the current study. Since CD11c- cells were M2a-polarized anti-inflammatory macrophages, while CD11c+ cells were M1/M2b-polarized pro-inflammatory macrophages, we used bioinformatics tools to analyze the CD11c+ versus CD11c- plaque macrophages, aiming to detect the differential genes associated with M1/M2 macrophage polarization. We obtained 122 differential genes that were significantly altered in CD11c+ versus CD11c- plaque macrophages, regardless of CD11b expression. Next, hub genes were predicted in these 122 genes, from which we detected 3 candidates, interleukin 6 (Il6), Decorin (Dcn) and Tissue inhibitor matrix metalloproteinase 1 (Timp1). The effects of these 3 genes on CD11c expression as well as on the macrophage polarization were assessed in vitro, showing that only expression of Il6, but not expression of Dcn or Timp1, induced M1/M2b-like polarization in M2a macrophages. Moreover, only suppression of Il6, but not suppression of either of Dcn or Timp1, induced M2a-like polarization in M1/M2b macrophages. Furthermore, pharmaceutical suppression of Il6 attenuated VIH formation and progression of AS in a mouse model that co-applied apolipoprotein E-knockout and high-fat diet. Together, our data suggest that formation of VIH can be controlled through modulating macrophage polarization, as a promising therapeutic approach for prevent AS.
Collapse
|
27
|
Dai W, Liu X, Su H, Li X, Xu Y, Yu Y. Influence of adipose tissue immune dysfunction on childhood obesity. Cytokine Growth Factor Rev 2022; 65:27-38. [PMID: 35595599 DOI: 10.1016/j.cytogfr.2022.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 11/17/2022]
Abstract
In recent decades, a dramatic rise has been observed in the prevalence of obesity in childhood and adolescence, along with an increase in fetal microsomia rates. The increased risk of obesity during this key period in development negatively affects the health of the individual later in life. Immune cells residing and recruited to white adipose tissue have been highlighted as important factors contributing to the pathogenesis of childhood obesity. Immune dysfunction in the context of obesity begins early in childhood, which is different from the pathological characteristics and influencing factors of adipose immunity in adults. Here, we explore the current understanding of the roles of childhood and early life events that result in high risks for obesity by influencing adipose tissue immune dysfunction under the pathological condition of obesity. Such knowledge will help in determining the mechanisms of childhood and early life obesity in efforts to ameliorate chronic inflammation-related metabolic diseases.
Collapse
Affiliation(s)
- Wanlin Dai
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China; Innovation Institute, China Medical University, China Medical University, Shenyang 110122, Liaoning, China
| | - Xiyan Liu
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Han Su
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Xuan Li
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China; Innovation Institute, China Medical University, China Medical University, Shenyang 110122, Liaoning, China
| | - Yingxi Xu
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Yang Yu
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|
28
|
Kiran S, Rakib A, Kodidela S, Kumar S, Singh UP. High-Fat Diet-Induced Dysregulation of Immune Cells Correlates with Macrophage Phenotypes and Chronic Inflammation in Adipose Tissue. Cells 2022; 11:cells11081327. [PMID: 35456006 PMCID: PMC9031506 DOI: 10.3390/cells11081327] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 01/27/2023] Open
Abstract
Obesity is a complex disease associated with various metabolic abnormalities, cardiovascular diseases, and low-grade chronic inflammation. Inflammation associated with T helper 1 (Th1) immune cells is dominant in adipose tissue (AT) and exerts metabolically deleterious impacts. The precise mechanism of alteration in AT immune system and its effect on metabolic homeostasis remains unclear. In this study, we investigated how a high-fat diet (HFD) alters the AT immune response and influences inflammation during obesity. HFD consumption amends the metabolic parameters, including body weight, glucose, and insulin levels. We observed increased infiltration of Th17 cells, a subset of dendritic cells (CD103+), and M1 macrophages in AT of mice fed HFD compared to those fed a normal diet (ND). In mice that were fed HFD, we also observed a reduction in regulatory T cells (Tregs) relative to the numbers of these cells in mice fed ND. Corresponding with this, mice in the HFD group exhibited higher levels of proinflammatory cytokines and chemokines than those in the ND group. We also observed alterations in signaling pathways, including increased protein expression of IRF3, TGFβ1, and mRNA expression of IL-6, KLF4, and STAT3 in the AT of the mice fed HFD as compared to those fed ND. Further, HFD-fed mice exhibited decreased protein expression of peroxisome proliferator-activated receptor-gamma (PPAR-γ) compared to mice fed ND, suggesting that PPAR-γ functions as a negative regulator of Th17 cell differentiation. These results suggest that HFD induces increased levels of inflammatory cytokines and key immune cells, including Th17, M1 macrophages, and CD103+ dendritic cells, and reduces levels of PPAR-γ and Tregs to sustain AT inflammation. This study supports the notion that dysregulation of Th17/Tregs, which polarizes macrophages towards M1 phenotypes in part through TGFβ1-IRF3-STAT3 and negatively regulates PPAR-γ mediated pathways, results in AT inflammation during obesity.
Collapse
|
29
|
Stutte S, Ishikawa-Ankerhold H, Lynch L, Eickhoff S, Nasiscionyte S, Guo C, van den Heuvel D, Setzensack D, Colonna M, Maier-Begandt D, Weckbach L, Brocker T, Schulz C, Walzog B, von Andrian U. High-Fat Diet Rapidly Modifies Trafficking, Phenotype, and Function of Plasmacytoid Dendritic Cells in Adipose Tissue. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1445-1455. [PMID: 35181637 PMCID: PMC8919350 DOI: 10.4049/jimmunol.2100022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 01/05/2022] [Indexed: 06/14/2023]
Abstract
Plasmacytoid dendritic cells (pDCs) display an increased abundance in visceral adipose tissue (VAT) of humans with obesity. In the current study, we set out to decipher the molecular mechanisms of their recruitment to VAT and the functional relevance of this process. We observed increased pDC numbers in murine blood, liver, spleen, and VAT after feeding a high-fat diet (HFD) for 3 wk when compared with a standard diet. pDCs were enriched in fat-associated lymphoid clusters representing highly specific lymphoid regions within VAT. HFD led to an enlargement of fat-associated lymphoid clusters with an increased density and migratory speed of pDCs as shown by intravital multiphoton microscopy. For their recruitment into VAT, pDCs employed P-selectin with E-selectin and L-selectin being only critical in response to HFD, indicating that the molecular cues underlying pDC trafficking were dependent on the nutritional state. Subsequent recruitment steps required α4β1 and α4β7 integrins and engagement of CCR7. Application of fingolimod (FTY720) abrogated egress of pDCs from VAT, indicating the involvement of sphingosine-1-phosphate in this process. Furthermore, HFD altered pDC functions by promoting their activation and type 1 IFN expression. Blocking pDC infiltration into VAT prevented weight gain and improved glucose tolerance during HFD. In summary, a HFD fundamentally alters pDC biology by promoting their trafficking, retention, and activation in VAT, which in turn seems to regulate metabolism.
Collapse
Affiliation(s)
- Susanne Stutte
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany;
- Walter Brendel Center of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Institute for Immunology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA
| | - Hellen Ishikawa-Ankerhold
- Walter Brendel Center of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Internal Medicine I, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Lydia Lynch
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA
- Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Sarah Eickhoff
- Institute of Systems Immunology, University of Würzburg, Würzburg, Germany
| | - Simona Nasiscionyte
- Walter Brendel Center of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Chenglong Guo
- Walter Brendel Center of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Internal Medicine I, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Dominic van den Heuvel
- Walter Brendel Center of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Internal Medicine I, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Daniel Setzensack
- Walter Brendel Center of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Internal Medicine I, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Marco Colonna
- Washington University, School of Medicine, St. Louis, MO; and
| | - Daniela Maier-Begandt
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
- Walter Brendel Center of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ludwig Weckbach
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
- Walter Brendel Center of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Internal Medicine I, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Thomas Brocker
- Institute for Immunology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christian Schulz
- Walter Brendel Center of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Internal Medicine I, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Barbara Walzog
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
- Walter Brendel Center of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ulrich von Andrian
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA
| |
Collapse
|
30
|
Doukbi E, Soghomonian A, Sengenès C, Ahmed S, Ancel P, Dutour A, Gaborit B. Browning Epicardial Adipose Tissue: Friend or Foe? Cells 2022; 11:991. [PMID: 35326442 PMCID: PMC8947372 DOI: 10.3390/cells11060991] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 02/08/2023] Open
Abstract
The epicardial adipose tissue (EAT) is the visceral fat depot of the heart which is highly plastic and in direct contact with myocardium and coronary arteries. Because of its singular proximity with the myocardium, the adipokines and pro-inflammatory molecules secreted by this tissue may directly affect the metabolism of the heart and coronary arteries. Its accumulation, measured by recent new non-invasive imaging modalities, has been prospectively associated with the onset and progression of coronary artery disease (CAD) and atrial fibrillation in humans. Recent studies have shown that EAT exhibits beige fat-like features, and express uncoupling protein 1 (UCP-1) at both mRNA and protein levels. However, this thermogenic potential could be lost with age, obesity and CAD. Here we provide an overview of the physiological and pathophysiological relevance of EAT and further discuss whether its thermogenic properties may serve as a target for obesity therapeutic management with a specific focus on the role of immune cells in this beiging phenomenon.
Collapse
Affiliation(s)
- Elisa Doukbi
- INSERM, INRAE, C2VN, Aix-Marseille University, F-13005 Marseille, France; (E.D.); (A.S.); (S.A.); (P.A.); (A.D.)
| | - Astrid Soghomonian
- INSERM, INRAE, C2VN, Aix-Marseille University, F-13005 Marseille, France; (E.D.); (A.S.); (S.A.); (P.A.); (A.D.)
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, APHM, F-13005 Marseille, France
| | - Coralie Sengenès
- Stromalab, CNRS ERL5311, EFS, INP-ENVT, INSERM U1031, University of Toulouse, F-31100 Toulouse, France;
- Institut National de la Santé et de la Recherche Médicale, University Paul Sabatier, F-31100 Toulouse, France
| | - Shaista Ahmed
- INSERM, INRAE, C2VN, Aix-Marseille University, F-13005 Marseille, France; (E.D.); (A.S.); (S.A.); (P.A.); (A.D.)
| | - Patricia Ancel
- INSERM, INRAE, C2VN, Aix-Marseille University, F-13005 Marseille, France; (E.D.); (A.S.); (S.A.); (P.A.); (A.D.)
| | - Anne Dutour
- INSERM, INRAE, C2VN, Aix-Marseille University, F-13005 Marseille, France; (E.D.); (A.S.); (S.A.); (P.A.); (A.D.)
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, APHM, F-13005 Marseille, France
| | - Bénédicte Gaborit
- INSERM, INRAE, C2VN, Aix-Marseille University, F-13005 Marseille, France; (E.D.); (A.S.); (S.A.); (P.A.); (A.D.)
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, APHM, F-13005 Marseille, France
| |
Collapse
|
31
|
Schwartz C, Schmidt V, Deinzer A, Hawerkamp HC, Hams E, Bayerlein J, Röger O, Bailer M, Krautz C, El Gendy A, Elshafei M, Heneghan HM, Hogan AE, O'Shea D, Fallon PG. Innate PD-L1 limits T cell-mediated adipose tissue inflammation and ameliorates diet-induced obesity. Sci Transl Med 2022; 14:eabj6879. [PMID: 35263149 DOI: 10.1126/scitranslmed.abj6879] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Obesity has become a major health problem in the industrialized world. Immune regulation plays an important role in adipose tissue homeostasis; however, the initial events that shift the balance from a noninflammatory homeostatic environment toward inflammation leading to obesity are poorly understood. Here, we report a role for the costimulatory molecule programmed death-ligand 1 (PD-L1) in the limitation of diet-induced obesity. Functional ablation of PD-L1 on dendritic cells (DCs) using conditional knockout mice increased weight gain and metabolic syndrome during diet-induced obesity, whereas PD-L1 expression on type 2 innate lymphoid cells (ILC2s), T cells, and macrophages was dispensable for obesity control. Using in vitro cocultures, DCs interacted with T cells and ILC2s via the PD-L1:PD-1 axis to inhibit T helper type 1 proliferation and promote type 2 polarization, respectively. A role for PD-L1 in adipose tissue regulation was also shown in humans, with a positive correlation between PD-L1 expression in visceral fat of people with obesity and elevated body weight. Thus, we define a mechanism of adipose tissue homeostasis controlled by the expression of PD-L1 by DCs, which may be a clinically relevant finding with regard to immune-related adverse events during immune checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Christian Schwartz
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany.,Medical Immunology Campus Erlangen, FAU Erlangen-Nürnberg, D-91054 Erlangen, Germany.,Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, D02R590 Dublin 2, Ireland
| | - Viviane Schmidt
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Andrea Deinzer
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Heike C Hawerkamp
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, D02R590 Dublin 2, Ireland
| | - Emily Hams
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, D02R590 Dublin 2, Ireland
| | - Jasmin Bayerlein
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Ole Röger
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Moritz Bailer
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Christian Krautz
- Klinik für Allgemein- und Viszeralchirurgie, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Amr El Gendy
- Klinik für Allgemein- und Viszeralchirurgie, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Moustafa Elshafei
- Klinik für Allgemein- und Viszeralchirurgie, Universitätsklinikum Erlangen and Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Helen M Heneghan
- Department of Surgery, St. Vincent's University Hospital and University College Dublin, D04T6F4 Dublin 4, Ireland
| | - Andrew E Hogan
- Kathleen Lonsdale Human Health Institute, Maynooth University, W23F2H6 Maynooth, Co. Kildare, Ireland.,Obesity Immunology Research, St. Vincent's University Hospital and University College Dublin, D04T6F4 Dublin 4, Ireland
| | - Donal O'Shea
- Obesity Immunology Research, St. Vincent's University Hospital and University College Dublin, D04T6F4 Dublin 4, Ireland
| | - Padraic G Fallon
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, D02R590 Dublin 2, Ireland
| |
Collapse
|
32
|
Satoh M, Iizuka M, Majima M, Ohwa C, Hattori A, Van Kaer L, Iwabuchi K. Adipose invariant NKT cells interact with CD1d-expressing macrophages to regulate obesity-related inflammation. Immunology 2022; 165:414-427. [PMID: 35137411 DOI: 10.1111/imm.13447] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 01/06/2022] [Accepted: 01/11/2022] [Indexed: 11/28/2022] Open
Abstract
Obesity is accompanied by and accelerated with chronic inflammation in adipose tissue, especially visceral adipose tissue (VAT). This low-level inflammation predisposes the host to the development of metabolic disease, most notably type 2 diabetes. We have focused on the capacity of glycolipid-reactive, CD1d-restricted natural killer T (NKT) cells to modulate obesity and its associated metabolic sequelae. We previously reported that CD1d knockout (KO) mice are partially protected against the development of obesity-associated insulin-resistance, and these findings were recapitulated in mice with an adipocyte-specific CD1d deficiency, suggesting that NKT cell-adipocyte interactions play a critical role in exacerbating disease. However, many other CD1d-expressing cells contribute to the in vivo responses of NKT cells to lipid antigens. In the present study, we examined the role of CD1d expression by macrophages (Mϕ) to the development of obesity-associated metabolic inflammation using LysMcre-cd1d1f/f mice where the CD1d1 gene is disrupted in a Mϕ-specific manner. Unexpectedly, these animals contained a higher frequency of T-bet+ CD4+ T cells in VAT with increased production of Th1-cytokines that aggravated VAT inflammation. Mϕ from mutant mice displayed increased production of IL-12p40, suggesting M1 polarization. These findings indicate that interactions of CD1d on Mϕ with NKT cells play a beneficial role in obesity-associated VAT inflammation and insulin resistance with a sharp contrast to an aggravating role of CD1d on another type of antigen presenting cell, dendritic cells.
Collapse
Affiliation(s)
- Masashi Satoh
- Department of Immunology, Kitasato University School of Medicine.,Program in Cellular Immunology, Graduate School of Medical Sciences, Kitasato University
| | - Misao Iizuka
- Department of Immunology, Kitasato University School of Medicine
| | - Masataka Majima
- Department of Pharmacology, Kitasato University School of Medicine.,Program in Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan.,School of Health and Medical Sciences, Kanagawa Institute of Technology (KAIT), Atsugi, Japan
| | - Chizuru Ohwa
- Program in Cellular Immunology, Graduate School of Medical Sciences, Kitasato University
| | - Akito Hattori
- Program in Cellular Immunology, Graduate School of Medical Sciences, Kitasato University
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Kazuya Iwabuchi
- Department of Immunology, Kitasato University School of Medicine.,Program in Cellular Immunology, Graduate School of Medical Sciences, Kitasato University
| |
Collapse
|
33
|
Conventional type 1 dendritic cells protect against age-related adipose tissue dysfunction and obesity. Cell Mol Immunol 2022; 19:260-275. [PMID: 34983945 PMCID: PMC8803960 DOI: 10.1038/s41423-021-00812-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 02/08/2023] Open
Abstract
Conventional dendritic cells (cDCs) scan and integrate environmental cues in almost every tissue, including exogenous metabolic signals. While cDCs are critical in maintaining immune balance, their role in preserving energy homeostasis is unclear. Here, we showed that Batf3-deficient mice lacking conventional type 1 DCs (cDC1s) had increased body weight and adiposity during aging. This led to impaired energy expenditure and glucose tolerance, insulin resistance, dyslipidemia, and liver steatosis. cDC1 deficiency caused adipose tissue inflammation that was preceded by a paucity of NK1.1+ invariant NKT (iNKT) cells. Accordingly, among antigen-presenting cells, cDC1s exhibited notable induction of IFN-γ production by iNKT cells, which plays a metabolically protective role in lean adipose tissue. Flt3L treatment, which expands the dendritic cell (DC) compartment, mitigated diet-induced obesity and hyperlipidemia in a Batf3-dependent manner. This effect was partially mediated by NK1.1+ cells. These results reveal a new critical role for the cDC1-iNKT cell axis in the regulation of adipose tissue homeostasis.
Collapse
|
34
|
Resident and migratory adipose immune cells control systemic metabolism and thermogenesis. Cell Mol Immunol 2021; 19:421-431. [PMID: 34837070 PMCID: PMC8891307 DOI: 10.1038/s41423-021-00804-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/28/2021] [Indexed: 02/08/2023] Open
Abstract
Glucose is a vital source of energy for all mammals. The balance between glucose uptake, metabolism and storage determines the energy status of an individual, and perturbations in this balance can lead to metabolic diseases. The maintenance of organismal glucose metabolism is a complex process that involves multiple tissues, including adipose tissue, which is an endocrine and energy storage organ that is critical for the regulation of systemic metabolism. Adipose tissue consists of an array of different cell types, including specialized adipocytes and stromal and endothelial cells. In addition, adipose tissue harbors a wide range of immune cells that play vital roles in adipose tissue homeostasis and function. These cells contribute to the regulation of systemic metabolism by modulating the inflammatory tone of adipose tissue, which is directly linked to insulin sensitivity and signaling. Furthermore, these cells affect the control of thermogenesis. While lean adipose tissue is rich in type 2 and anti-inflammatory cytokines such as IL-10, obesity tips the balance in favor of a proinflammatory milieu, leading to the development of insulin resistance and the dysregulation of systemic metabolism. Notably, anti-inflammatory immune cells, including regulatory T cells and innate lymphocytes, protect against insulin resistance and have the characteristics of tissue-resident cells, while proinflammatory immune cells are recruited from the circulation to obese adipose tissue. Here, we review the key findings that have shaped our understanding of how immune cells regulate adipose tissue homeostasis to control organismal metabolism.
Collapse
|
35
|
Inflammatory Mediators and Gut Microbial Toxins Drive Colon Tumorigenesis by IL-23 Dependent Mechanism. Cancers (Basel) 2021; 13:cancers13205159. [PMID: 34680308 PMCID: PMC8533859 DOI: 10.3390/cancers13205159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/03/2021] [Accepted: 10/08/2021] [Indexed: 01/01/2023] Open
Abstract
Simple Summary Western-style diet, rich in high fat, is the major cause of obesity and enhanced risk of colon cancer in the USA and worldwide. The inflammatory molecules are a well-established link between obesity and the modulation of colon tumorigenesis. In particular, IL-23 plays an important role in the impact of a western-style diet on obesity, the gut microbiome, and colon tumorigenesis. However, the underlying mechanism of IL-23 production for colon tumor progression and whether IL-23 can be a potential target is not clear. Our findings signify the role of pro-tumorigenic innate immune cells, including dendritic cells and macrophages in IL-23 production by bacterial toxins and eicosanoids. IL-23 knockdown in the tumorigenic dendritic cells and macrophages inhibited the colon tumor cell and organoids growth. Taken together, targeting IL-23 may be a promising option for the prevention and treatment of high-fat/obesity-associated colon cancer in clinical trials. Abstract Obesity-associated chronic inflammation predisposes colon cancer risk development. Interleukin-23 (IL-23) is a potential inflammatory mediator linking obesity to chronic colonic inflammation, altered gut microbiome, and colon carcinogenesis. We aimed to elucidate the role of pro-inflammatory eicosanoids and gut bacterial toxins in priming dendritic cells and macrophages for IL-23 secretion to promote colon tumor progression. To investigate the association of IL-23 with obesity and colon tumorigenesis, we utilized TCGA data set and colonic tumors from humans and preclinical models. To understand IL-23 production by inflammatory mediators and gut microbial toxins, we performed several in vitro mechanistic studies to mimic the tumor microenvironment. Colonic tumors were utilized to perform the ex vivo experiments. Our findings showed that IL-23 is elevated in obese individuals, colonic tumors and correlated with reduced disease-free survival. In vitro studies showed that IL-23 treatment increased the colon tumor cell self-renewal, migration, and invasion while disrupting epithelial barrier permeability. Co-culture experiments of educated dendritic cells/macrophages with colon cancer cells significantly increased the tumor aggression by increasing the secretory levels of IL-23, and these observations are further supported by ex vivo rat colonic tumor organotypic experiments. Our results demonstrate gut microbe toxins and eicosanoids facilitate IL-23 production, which plays an important role in obesity-associated colonic tumor progression. This newly identified nexus represents a potential target for the prevention and treatment of obesity-associated colon cancer.
Collapse
|
36
|
The effects of dietary vitamin D supplementation and in vitro 1,25 dihydroxyvitamin D 3 treatment on autophagy in bone marrow-derived dendritic cells from high-fat diet-induced obese mice. J Nutr Biochem 2021; 100:108880. [PMID: 34655755 DOI: 10.1016/j.jnutbio.2021.108880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 06/18/2021] [Accepted: 09/22/2021] [Indexed: 12/11/2022]
Abstract
Obesity is associated with the dysregulation of vitamin D metabolism and altered immune responses in bone marrow-derived dendritic cells (BMDCs). Vitamin D can affect the differentiation, maturation, and activation of dendritic cells (DCs) and regulate autophagy via vitamin D receptor signaling. Autophagy was shown to be involved in the functions of DCs. We investigated the effects of dietary vitamin D supplementation and in vitro 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) treatment on autophagy in BMDCs from control diet (CON)-fed lean and high-fat diet (HFD)-induced obese mice. C57BL/6 male mice were fed CON or HFD with 10% or 45% kcal fat, respectively, supplemented with 1,000 or 10,000 IU vitamin D/kg diet (vDC or vDS) for 12 weeks. BMDCs were generated by culturing bone marrow cells from the mice with 20 ng/mL rmGM-CSF and treated with 1 nM 1,25(OH)2D3. Maturation of BMDCs was induced by lipopolysaccharide (50 ng/mL) stimulation. Treatment with 1,25(OH)2D3 inhibited the expression of phenotypes related to DC function (MHC class Ⅱ, CD86, CD80) and production of IL-12p70 by BMDCs from control and obese mice, regardless of dietary vitamin D supplementation. LC3Ⅱ/Ⅰ and VPS34 protein levels increased, and p62 expression decreased, after 1,25(OH)2D3 treatment of the BMDCs in CON-vDC only. Vdr mRNA levels decreased following 1,25(OH)2D3 treatment of BMDCs in the HFD-vDC. In conclusion, autophagy flux was increased by 1,25(OH)2D3 treatment of the BMDCs in CON-vDC but not in the HFD-vDC group. This suggests that the decreased expression of Vdr following 1,25(OH)2D3 treatment might have affected autophagy flux in BMDCs from obese mice.
Collapse
|
37
|
O'Brien CJO, Haberman ER, Domingos AI. A Tale of Three Systems: Toward a Neuroimmunoendocrine Model of Obesity. Annu Rev Cell Dev Biol 2021; 37:549-573. [PMID: 34613819 PMCID: PMC7614880 DOI: 10.1146/annurev-cellbio-120319-114106] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The prevalence of obesity is on the rise. What was once considered a simple disease of energy imbalance is now recognized as a complex condition perpetuated by neuro- and immunopathologies. In this review, we summarize the current knowledge of the neuroimmunoendocrine mechanisms underlying obesity. We examine the pleiotropic effects of leptin action in addition to its established role in the modulation of appetite, and we discuss the neural circuitry mediating leptin action and how this is altered with obesity, both centrally (leptin resistance) and in adipose tissues (sympathetic neuropathy). Finally, we dissect the numerous causal and consequential roles of adipose tissue macrophages in obesity and highlight recent key studies demonstrating their direct role in organismal energy homeostasis.
Collapse
Affiliation(s)
- Conan J O O'Brien
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom;
| | - Emma R Haberman
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom;
| | - Ana I Domingos
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom;
| |
Collapse
|
38
|
Butler MJ. The role of Western diets and obesity in peripheral immune cell recruitment and inflammation in the central nervous system. Brain Behav Immun Health 2021; 16:100298. [PMID: 34589790 PMCID: PMC8474237 DOI: 10.1016/j.bbih.2021.100298] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/09/2021] [Accepted: 07/10/2021] [Indexed: 12/25/2022] Open
Abstract
As the prevalence of obesity and chronic disease increases, the role of nutrition is taking center stage as a potential root cause of not just metabolic-related illnesses, but also of disorders of the central nervous system (CNS). Consumption of a modern, westernized diet, such as a high fat diet (HFD) that contains excess saturated fatty acids (SFAs), refined carbohydrates, and ultra-processed ingredients has been shown to induce neuroinflammation in multiple brain regions important for energy homeostasis, cognitive function, and mood regulation in rodents, non-human primates, and humans. This review article summarizes the literature showing Western diets, via SFA increases, can increase the reactivity and alter the function of multiple types of immune cells from both the innate and adaptive branches of the immune system, with a specific focus on microglia, macrophages, dendritic cells, and T-cells. These changes in immune and neuroimmune signaling have important implications for neuroinflammation and brain health and will be an important factor in future psychoneuroimmunology research.
Collapse
Affiliation(s)
- Michael J. Butler
- Institute for Behavioral Medicine Research, Ohio State University, Wexner Medical Center 460 Medical Center Drive, Columbus, OH, 43210, USA
| |
Collapse
|
39
|
Age and Sex: Impact on adipose tissue metabolism and inflammation. Mech Ageing Dev 2021; 199:111563. [PMID: 34474078 DOI: 10.1016/j.mad.2021.111563] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/19/2021] [Accepted: 08/26/2021] [Indexed: 02/08/2023]
Abstract
Age associated chronic inflammation is a major contributor to diseases with advancing age. Adipose tissue function is at the nexus of processes contributing to age-related metabolic disease and mediating longevity. Hormonal fluctuations in aging potentially regulate age-associated visceral adiposity and metabolic dysfunction. Visceral adiposity in aging is linked to aberrant adipogenesis, insulin resistance, lipotoxicity and altered adipokine secretion. Age-related inflammatory phenomena depict sex differences in macrophage polarization, changes in T and B cell numbers, and types of dendritic cells. Sex differences are also observed in adipose tissue remodeling and cellular senescence suggesting a role for sex steroid hormones in the regulation of the adipose tissue microenvironment. It is crucial to investigate sex differences in aging clinical outcomes to identify and better understand physiology in at-risk individuals. Early interventions aimed at targets involved in adipose tissue adipogenesis, remodeling and inflammation in aging could facilitate a profound impact on health span and overcome age-related functional decline.
Collapse
|
40
|
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: 16] [Impact Index Per Article: 5.3] [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
|
41
|
Li C, Wang G, Sivasami P, Ramirez RN, Zhang Y, Benoist C, Mathis D. Interferon-α-producing plasmacytoid dendritic cells drive the loss of adipose tissue regulatory T cells during obesity. Cell Metab 2021; 33:1610-1623.e5. [PMID: 34256015 PMCID: PMC8350961 DOI: 10.1016/j.cmet.2021.06.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/16/2021] [Accepted: 06/14/2021] [Indexed: 12/14/2022]
Abstract
The visceral adipose tissue (VAT) of lean mice hosts a unique population of regulatory T cells (Tregs) that have a distinct transcriptome and T cell receptor (TCR) repertoire and regulate local and systemic inflammation and metabolism. Perplexingly, this population disappears in obese mice, limiting the promise of Treg-based therapies for metabolic disorders. We exploited the power of a VAT-Treg TCR-transgenic mouse model to follow the dynamics of, and phenotypic changes in, the VAT-Treg population throughout the development of diet-induced obesity. Our results show that VAT-Tregs are lost under obesogenic conditions due to downregulation of their defining transcription factor, PPARγ, coupled with their strikingly enhanced responses to pro-inflammatory cytokines. In particular, the VAT from obese mice (and reportedly humans) was strongly enriched in plasmacytoid dendritic cells that actively express interferon-alpha. These cells were directly toxic to PPARγ+ VAT-Tregs. Blocking this pathway in obese mice by multiple approaches substantially restored the VAT-Treg population and enhanced insulin sensitivity.
Collapse
Affiliation(s)
- Chaoran Li
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Gang Wang
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Pulavendran Sivasami
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ricardo N Ramirez
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Yanbo Zhang
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Christophe Benoist
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
42
|
Inflammation in Metabolic and Cardiovascular Disorders-Role of Oxidative Stress. Life (Basel) 2021; 11:life11070672. [PMID: 34357044 PMCID: PMC8308054 DOI: 10.3390/life11070672] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/30/2021] [Accepted: 06/30/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases (CVD) constitute the main cause of death worldwide. Both inflammation and oxidative stress have been reported to be involved in the progress of CVD. It is well known that generation of oxidative stress during the course of CVD is involved in tissue damage and inflammation, causing deleterious effects such as hypertension, dysfunctional metabolism, endothelial dysfunction, stroke, and myocardial infarction. Remarkably, natural antioxidant strategies have been increasingly discovered and are subject to current scientific investigations. Here, we addressed the activation of immune cells in the context of ROS production, as well as how their interaction with other cellular players and further (immune) mediators contribute to metabolic and cardiovascular disorders. We also highlight how a dysregulated complement system contributes to immune imbalance and tissue damage in the context of increases oxidative stress. Additionally, modulation of hypothalamic oxidative stress is discussed, which may offer novel treatment strategies for type-2 diabetes and obesity. Together, we provide new perspectives on therapy strategies for CVD caused by oxidative stress, with a focus on oxidative stress.
Collapse
|
43
|
Meliț LE, Mărginean CO, Mărginean CD, Săsăran MO. The Peculiar Trialogue between Pediatric Obesity, Systemic Inflammatory Status, and Immunity. BIOLOGY 2021; 10:biology10060512. [PMID: 34207683 PMCID: PMC8229553 DOI: 10.3390/biology10060512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 12/24/2022]
Abstract
Pediatric obesity is not only an energetic imbalance, but also a chronic complex multisystem disorder that might impair both the life length and quality. Its pandemic status should increase worldwide awareness regarding the long-term life-threatening associated complications. Obesity related complications, such as cardiovascular, metabolic, or hepatic ones, affect both short and long-term wellbeing, and they do not spare pediatric subjects, defined as life-threatening consequences of the systemic inflammatory status triggered by the adipose tissue. The energetic imbalance of obesity clearly results in adipocytes hypertrophy and hyperplasia expressing different degrees of chronic inflammation. Adipose tissue might be considered an immune organ due to its rich content in a complex array of immune cells, among which the formerly mentioned macrophages, neutrophils, mast cells, but also eosinophils along with T and B cells, acting together to maintain the tissue homeostasis in normal weight individuals. Adipokines belong to the class of innate immunity humoral effectors, and they play a crucial role in amplifying the immune responses with a subsequent trigger effect on leukocyte activation. The usefulness of complete cellular blood count parameters, such as leukocytes, lymphocytes, neutrophils, erythrocytes, and platelets as predictors of obesity-triggered inflammation, was also proved in pediatric patients with overweight or obesity. The dogma that adipose tissue is a simple energy storage tissue is no longer accepted since it has been proved that it also has an incontestable multifunctional role acting like a true standalone organ resembling to endocrine or immune organs.
Collapse
Affiliation(s)
- Lorena Elena Meliț
- Department of Pediatrics I, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania; (L.E.M.); (C.D.M.)
| | - Cristina Oana Mărginean
- Department of Pediatrics I, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania; (L.E.M.); (C.D.M.)
- Correspondence: ; Tel.: +40-723-278543
| | - Cristian Dan Mărginean
- Department of Pediatrics I, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania; (L.E.M.); (C.D.M.)
| | - Maria Oana Săsăran
- Department of Pediatrics III, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania;
| |
Collapse
|
44
|
The Immunomodulating Effect of Baicalin on Inflammation and Insulin Resistance in High-Fat-Diet-Induced Obese Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5531367. [PMID: 34135978 PMCID: PMC8175130 DOI: 10.1155/2021/5531367] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/20/2021] [Accepted: 05/20/2021] [Indexed: 11/17/2022]
Abstract
Background Obesity is a chronic low-grade systemic inflammation state, which causes insulin resistance, diabetes, and other metabolic diseases. Baicalin is known to have anti-inflammatory and antiobesity effects. In this study, we investigated the cellular and molecular immunological effects of baicalin on obesity-induced inflammation. Methods Male C57BL/6 mice were assigned to four groups: the normal chow, high-fat diet (HFD), BC2 (HFD + baicalin 200 mg/kg), and BC4 (HFD + baicalin 400 mg/kg) group; the three groups except normal chow were fed with a high-fat diet for 8 weeks to induce obesity followed by baicalin treatment with two doses for 8 weeks. The body weight, epididymal fat weight, liver weight, food intake, oral glucose tolerance test (OGTT), oral fat tolerance test (OFTT), and serum lipids were measured. We evaluated insulin resistance by measuring the serum insulin level and homeostatic model assessment of insulin resistance (HOMA-IR). Also, the major obesity-associated immune cells including monocytes, macrophages, T lymphocytes, and dendritic cells in the blood, fat, and liver and the inflammatory and insulin signaling-related gene expressions in the fat and liver were evaluated. Results Baicalin significantly reduced the body weight and liver weight and improved serum fasting glucose, insulin, HOMA-IR, free fatty acid, HDL cholesterol, and the levels of glucose and triglyceride at each time point in the OGTT and OFTT. In the analysis of immune cells, baicalin significantly decreased inflammatory Ly6Chi monocytes, M1 adipose tissue macrophages (ATMs), and M1 Kupffer cells. On the contrary, baicalin increased anti-inflammatory M2 ATMs and liver CD4+ T cells and CD4/CD8 ratio. In the analysis of inflammatory and insulin signaling molecules, baicalin significantly downregulated the gene expression of tumor necrosis factor-α, F4/80, and C-C motif chemokine 2 while upregulated the insulin receptor mRNA expression. Conclusion From these results, baicalin can be a promising treatment option for obesity and its related metabolic diseases based on its anti-inflammatory property.
Collapse
|
45
|
Hayashi M, Iwashita M, Nishimura Y, Shinjo T, Sano T, Yamashita A, Fukuda T, Sanui T, Asano T, Nishimura F. Adipose-specific C-C motif chemokine ligand (CCL) 19 overexpression drives the mice to both insulin resistance and weight gain. BMJ Open Diabetes Res Care 2021; 9:9/1/e001871. [PMID: 34031140 PMCID: PMC8149363 DOI: 10.1136/bmjdrc-2020-001871] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 04/09/2021] [Accepted: 05/02/2021] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Enlarged adipose tissue is characterized by infiltration of activated immune cells and increased expression of chemokines recruiting these cells including C-C motif ligand 19 (CCL19), although the role of adipose CCL19 is still inconclusive. RESEARCH DESIGN AND METHODS Adipocyte-specific Ccl19 knock-in (KI) mice were generated, and the mice were fed either a normal diet or 40% or 60% fat diet (FD) to investigate the effects of CCL19 on the induction of inflammation and lipid metabolism. RESULTS Ccl19KI mice exhibited increased inflammatory signs in adipose tissue and enlarged subcutaneous white and brown adipose tissue than those of wild-type (WT) mice. The adipose tissue of Ccl19KI mice was characterized by increased extracellular signal-regulated kinase 1/2 and decreased AMP-activated protein kinase α phosphorylation. The protein expression of peroxisome proliferator-activated receptor γ coactivator 1α and uncoupling protein 1 was significantly reduced in brown adipose tissue of Ccl19KI mice compared with that in WT mice. The most remarkable changes between genotypes were observed in mice fed a 40% FD. CONCLUSION A 40% FD enhanced the effects of CCL19 overexpression, and these mice could be a suitable model to study metabolic disorders in overweight Asians.
Collapse
Affiliation(s)
- Masato Hayashi
- Section of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Misaki Iwashita
- Section of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yuki Nishimura
- Section of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Takanori Shinjo
- Section of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Tomomi Sano
- Section of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Akiko Yamashita
- Section of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Takao Fukuda
- Section of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Terukazu Sanui
- Section of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Tomoichiro Asano
- Department of Medical Science, Graduate School of Medicine, Hiroshima University, Hiroshima, Japan
| | - Fusanori Nishimura
- Section of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
46
|
Michailidou Z, Gomez-Salazar M, Alexaki VI. Innate Immune Cells in the Adipose Tissue in Health and Metabolic Disease. J Innate Immun 2021; 14:4-30. [PMID: 33849008 DOI: 10.1159/000515117] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/09/2021] [Indexed: 11/19/2022] Open
Abstract
Metabolic disorders, such as obesity, type 2 diabetes mellitus, and nonalcoholic fatty liver disease, are characterized by chronic low-grade tissue and systemic inflammation. During obesity, the adipose tissue undergoes immunometabolic and functional transformation. Adipose tissue inflammation is driven by innate and adaptive immune cells and instigates insulin resistance. Here, we discuss the role of innate immune cells, that is, macrophages, neutrophils, eosinophils, natural killer cells, innate lymphoid type 2 cells, dendritic cells, and mast cells, in the adipose tissue in the healthy (lean) and diseased (obese) state and describe how their function is shaped by the obesogenic microenvironment, and humoral, paracrine, and cellular interactions. Moreover, we particularly outline the role of hypoxia as a central regulator in adipose tissue inflammation. Finally, we discuss the long-lasting effects of adipose tissue inflammation and its potential reversibility through drugs, caloric restriction, or exercise training.
Collapse
Affiliation(s)
- Zoi Michailidou
- Centre for Cardiovascular Sciences, Edinburgh University, Edinburgh, United Kingdom
| | - Mario Gomez-Salazar
- Centre for Cardiovascular Sciences, Edinburgh University, Edinburgh, United Kingdom
| | - Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
47
|
Dwaib HS, AlZaim I, Eid AH, Obeid O, El-Yazbi AF. Modulatory Effect of Intermittent Fasting on Adipose Tissue Inflammation: Amelioration of Cardiovascular Dysfunction in Early Metabolic Impairment. Front Pharmacol 2021; 12:626313. [PMID: 33897419 PMCID: PMC8062864 DOI: 10.3389/fphar.2021.626313] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/18/2021] [Indexed: 12/15/2022] Open
Abstract
Cardiometabolic syndrome (CMS) is a cluster of maladaptive cardiovascular, renal, thrombotic, inflammatory, and metabolic disorders. It confers a high risk of cardiovascular mortality and morbidity. CMS is triggered by major shifts in lifestyle and dietary habits with increased consumption of refined, calorie-dense diets. Evidence indicates that diet-induced CMS is linked to Adipose tissue (AT) inflammation. This led to the proposal that adipose inflammation may be involved in metabolic derangements, such as insulin resistance and poor glycemic control, as well as the contribution to the inflammatory process predisposing patients to increased cardiovascular risk. Therefore, in the absence of direct pharmacological interventions for the subclinical phase of CMS, time restricted feeding regimens were anticipated to alleviate early metabolic damage and subsequent comorbidities. These regimens, referred to as intermittent fasting (IF), showed a strong positive impact on the metabolic state of obese and non-obese human subjects and animal models, positive AT remodeling in face of overnutrition and high fat diet (HFD) consumption, and improved CV outcomes. Here, we summarize the available evidence on the role of adipose inflammation in triggering cardiovascular impairment in the context of diet induced CMS with an emphasis on the involvement of perivascular adipose tissue. As well, we propose some possible molecular pathways linking intermittent fasting to the ameliorative effect on adipose inflammation and cardiovascular dysfunction under such circumstances. We highlight a number of targets, whose function changes in perivascular adipose tissue inflammation and could be modified by intermittent fasting acting as a novel approach to ameliorate the inflammatory status.
Collapse
Affiliation(s)
- Haneen S. Dwaib
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences, American University of Beirut, Beirut, Lebanon
| | - Ibrahim AlZaim
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Omar Obeid
- Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences, American University of Beirut, Beirut, Lebanon
| | - Ahmed F. El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Faculty of Pharmacy, Al-Alamein International University, Alamein, Egypt
| |
Collapse
|
48
|
Abstract
On this 100th anniversary of the discovery of insulin, we recognize the critical role that adipocytes, which are exquisitely responsive to insulin, have played in determining the mechanisms for insulin action at the cellular level. Our understanding of adipose tissue biology has evolved greatly, and it is now clear that adipocytes are far more complicated than simple storage depots for fat. A growing body of evidence documents how adipocytes, in response to insulin, contribute to the control of whole-body nutrient homeostasis. These advances highlight adipocyte plasticity, heterogeneity, and endocrine function, unique features that connect adipocyte metabolism to the regulation of other tissues important for metabolic homeostasis (e.g., liver, muscle, pancreas).
Collapse
Affiliation(s)
- Anna Santoro
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Timothy E McGraw
- Department of Biochemistry, Weill Medical College of Cornell University, New York, NY 10065, USA.
| | - Barbara B Kahn
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
49
|
The complex role of adipokines in obesity, inflammation, and autoimmunity. Clin Sci (Lond) 2021; 135:731-752. [PMID: 33729498 PMCID: PMC7969664 DOI: 10.1042/cs20200895] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/24/2021] [Accepted: 03/04/2021] [Indexed: 12/12/2022]
Abstract
The global obesity epidemic is a major contributor to chronic disease and disability in the world today. Since the discovery of leptin in 1994, a multitude of studies have characterized the pathological changes that occur within adipose tissue in the obese state. One significant change is the dysregulation of adipokine production. Adipokines are an indispensable link between metabolism and optimal immune system function; however, their dysregulation in obesity contributes to chronic low-grade inflammation and disease pathology. Herein, I will highlight current knowledge on adipokine structure and physiological function, and focus on the known roles of these factors in the modulation of the immune response. I will also discuss adipokines in rheumatic and autoimmune diseases.
Collapse
|
50
|
Kim J, Lee J. Role of obesity-induced inflammation in the development of insulin resistance and type 2 diabetes: history of the research and remaining questions. Ann Pediatr Endocrinol Metab 2021; 26:1-13. [PMID: 33819954 PMCID: PMC8026341 DOI: 10.6065/apem.2040188.094] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 01/12/2021] [Indexed: 12/13/2022] Open
Abstract
The prevalence of obesity has increased alarmingly both worldwide and in Korea. This has also dramatically increased the prevalence of chronic obesity-associated diseases, including type 2 diabetes (T2D). Extensive studies on the molecular etiology of T2D have revealed several potential mechanisms by which obesity induces the development of insulin resistance and T2D. One of these is low-grade chronic inflammation. Studies hinting at the existence of this phenomenon were first published about 30 years ago. Ten years later, several seminal papers confirmed its existence, which then led to a rapid and massive escalation of research in this field. Today, the notion that obesity-induced inflammation mediates T2D is now well-accepted. This paper will review the key developments in this field, including the discovery that obesity-induced inflammation and insulin resistance is mainly regulated by adipose tissue-resident immune cells, particularly those in visceral adipose tissue. This review further details the research areas, including (1) the obesity-related factors that induce adipose tissue macrophage (ATM) inflammation, (2) the precise effector functions by which adipose tissue immune cells promote insulin resistance, (3) whether there are early immunological events that have an outsize effect on later events and could be targeted to arrest the development of insulin resistance, (4) the roles played by nonimmunological functions of ATMs and other immune cells, and (5) whether there are noncanonical immune responses to obesity (i.e., immune responses that are unique to obesity and cannot be detected by following the discoveries in the classical immunity field).
Collapse
Affiliation(s)
- Jieun Kim
- Soonchunhyang Institute of MediBio Science (SIMS), Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Korea
| | - Jongsoon Lee
- Soonchunhyang Institute of MediBio Science (SIMS), Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Korea,Address for correspondence: Jongsoon Lee Soonchunhyang Institute of MediBio Science (SIMS), Department of Integrated Biomedical Science, Metabolic Disease Tissue Homeostasis Research Center, Soonchunhyang University, Cheonan 31151, Korea
| |
Collapse
|