1
|
Chew V. Why weight loss is only half the battle: The epigenetic memory of adipose tissue. J Hepatol 2025:S0168-8278(24)02765-X. [PMID: 39833020 DOI: 10.1016/j.jhep.2024.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 01/22/2025]
Affiliation(s)
- Valerie Chew
- Translational Immunology Institute (TII), SingHealth-DukeNUS Academic Medical Centre, Singapore 169856, Singapore.
| |
Collapse
|
2
|
Schuetz LT, Duran G, Baeten P, Lintsen D, Hermans D, Chenine S, Verreycken J, Vanmierlo T, Wouters K, Broux B. Sex differentially affects pro-inflammatory cell subsets in adipose tissue depots in a diet induced obesity model. Biol Sex Differ 2024; 15:105. [PMID: 39696610 DOI: 10.1186/s13293-024-00677-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024] Open
Abstract
Obesity is a growing pandemic that increases the risk for cardiovascular diseases, type 2 diabetes, and particularly in women also the risk of cancer and neurodegenerative disorders such as dementia and multiple sclerosis. Preclinical studies on obesity focus on male mice as they gain bodyweight faster and show a clear pro-inflammatory phenotype. Here, using male and female mice, we induced obesity by feeding a high fat diet (HFD), and compared adipose tissue (AT) inflammation at the same adiposity stage (% AT/bodyweight) between both sexes. Doing so, we identified that female mice show an increase in the number of pro-inflammatory immune cells in the visceral AT at a lower adiposity stage than male mice, but the effect of HFD is diminished with higher adiposity. Interestingly, only female mice showed an increase in immune cells in the subcutaneous AT after HFD feeding. Nonetheless, we found that pro-inflammatory cytokines in blood plasma mirror the inflammatory stage of the visceral AT in both male and female mice. Uniquely in male mice, myeloid cells in the visceral AT showed a higher inflammasome activation upon HFD. In summary, we showed that adiposity differentially affects immune cells in fat depots based on sex.
Collapse
Affiliation(s)
- Lisa T Schuetz
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- CARIM-School of Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
- Internal Medicine, Maastricht University, Maastricht, Netherlands
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
- MHeNs-Mental Health and Neuroscience Institute, Maastricht University, Maastricht, Netherlands
| | - Gayel Duran
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
| | - Paulien Baeten
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
| | - Daphne Lintsen
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
| | - Doryssa Hermans
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
| | - Sarah Chenine
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
- MHeNs-Mental Health and Neuroscience Institute, Maastricht University, Maastricht, Netherlands
| | - Janne Verreycken
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
| | - Tim Vanmierlo
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
- MHeNs-Mental Health and Neuroscience Institute, Maastricht University, Maastricht, Netherlands
| | - Kristiaan Wouters
- CARIM-School of Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands.
- Internal Medicine, Maastricht University, Maastricht, Netherlands.
| | - Bieke Broux
- Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center, Campus Diepenbeek, Diepenbeek, Belgium
| |
Collapse
|
3
|
Chen Y, Li X, Yang M, Jia C, He Z, Zhou S, Ruan P, Wang Y, Tang C, Pan W, Long H, Zhao M, Lu L, Peng W, Akbar A, Wu IX, Li S, Wu H, Lu Q. Time-restricted eating reveals a "younger" immune system and reshapes the intestinal microbiome in human. Redox Biol 2024; 78:103422. [PMID: 39561680 PMCID: PMC11616606 DOI: 10.1016/j.redox.2024.103422] [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: 10/10/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/21/2024] Open
Abstract
Time-restricted eating (TRE) has been shown to extent lifespans in drosophila and mouse models by affecting metabolic and anti-inflammatory activities. However, the effect of TRE on the human immune system, especially on immunosenescence, intestinal microbiome, and metabolism remains unclear. We conducted a 30-day 16:8 TRE single-arm clinical trial with 49 participants. Participants consumed daily meals from 9 a.m. to 5 p.m., provided by a nutrition canteen with a balanced, calorie-appropriate nutrition, which is designed by clinical nutritionists (ChiCTR2200058137). We monitored weight changes and weight-related parameters and focused on changes in the frequency of CD4+ senescent T cells, immune repertoire from peripheral blood, as well as serum metabolites and gut microbiota. We found that up to 95.9 % of subjects experienced sustained weight loss after TRE. The frequency of circulating senescent CD4+ T cells was decreased, while the frequency of Th1, Treg, Tfh-like, and B cells was increased. Regarding the immune repertoire, the proportions of T cell receptor alpha and beta chains were increased, whereas B cell receptor kappa and lambda chains were reduced. In addition, a reduced class switch recombination from immunoglobulin M (IgM) to immunoglobulin A (IgA) was observed. TRE upregulated the levels of anti-inflammatory and anti-aging serum metabolites named sphingosine-1-phosphate and prostaglandin-1. Additionally, several anti-inflammatory bacteria and probiotics were increased, such as Akkermansia and Rikenellaceae, and the composition of the gut microbiota tended to be "younger". Overall, TRE showed multiple anti-aging effects, which may help humans maintain a healthy lifestyle to stay "young". Clinical Trial Registration URL: https://www.chictr.org.cn/showproj.html?proj=159876.
Collapse
Affiliation(s)
- Yiran Chen
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, 210042, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, 210042, China; Research Unit of Key Technologies of Immune-related Skin Diseases Diagnosis and Treatment, Chinese Academy of Medical Sciences Institute of Dermatology, Nanjing, 210042, China
| | - Xi Li
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Ming Yang
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Chen Jia
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Zhenghao He
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Suqing Zhou
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Pinglang Ruan
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Yikun Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Congli Tang
- Nanjing ARP Biotechnology Co., Ltd, Nanjing, 210046, China
| | - Wenjing Pan
- Nanjing ARP Biotechnology Co., Ltd, Nanjing, 210046, China; Hunan Key Laboratory of Biomedical Nanomaterials and Devices, Hunan University of Technology, Zhuzhou, 412007, China
| | - Hai Long
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Ming Zhao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China; Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan, 410011, China
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, 518057, China
| | - Weijun Peng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Arne Akbar
- Associate of the Institute of Healthy Ageing, Division of Infection & Immunity, University College of London, London, WC1E 6BT, United Kingdom
| | - Irene Xy Wu
- Xiangya School of Public Health, Central South University, 4/F, Changsha, Hunan, 410006, China
| | - Song Li
- Key Laboratory of Rare Pediatric Diseases, Ministry of Education, Hengyang Medical School, University of South China, Hengyang, Hunan, China; National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China.
| | - Haijing Wu
- Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan, 410011, China.
| | - Qianjin Lu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, 210042, China; Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, 210042, China; Research Unit of Key Technologies of Immune-related Skin Diseases Diagnosis and Treatment, Chinese Academy of Medical Sciences Institute of Dermatology, Nanjing, 210042, China; Department of Dermatology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Medical Epigenomics, 139 Middle Renmin Road, Changsha, Hunan, 410011, China.
| |
Collapse
|
4
|
Capoccia D, Leonetti F, Natali A, Tricò D, Perrini S, Sbraccia P, Guglielmi V. Remission of type 2 diabetes: position statement of the Italian society of diabetes (SID). Acta Diabetol 2024; 61:1309-1326. [PMID: 38942960 PMCID: PMC11486812 DOI: 10.1007/s00592-024-02317-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 05/31/2024] [Indexed: 06/30/2024]
Abstract
The primary cause of the pandemic scale of type 2 diabetes (T2D) is the excessive and/or abnormal accumulation of adiposity resulting from a chronic positive energy balance. Any form of weight loss dramatically affects the natural history of T2D, favoring prevention, treatment, and even remission in the case of significant weight loss. However, weight regain, which is often accompanied by the recurrence or worsening of obesity complications such as T2D, is an inevitable biological phenomenon that is an integral part of the pathophysiology of obesity. This can occur not only after weight loss, but also during obesity treatment if it is not effective enough to counteract the physiological responses aimed at restoring adiposity to its pre-weight-loss equilibrium state. Over the past few years, many controlled and randomized studies have suggested a superior efficacy of bariatric surgery compared to conventional therapy in terms of weight loss, glycemic control, and rates of T2D remission. Recently, the therapeutic armamentarium in the field of diabetology has been enriched with new antihyperglycemic drugs with considerable efficacy in reducing body weight, which could play a pathogenetic role in the remission of T2D, not through the classical incretin effect, but by improving adipose tissue functions. All these concepts are discussed in this position statement, which aims to deepen the pathogenetic links between obesity and T2D, shift the paradigm from a "simple" interaction between insulin resistance and insulin deficiency, and evaluate the efficacy of different therapeutic interventions to improve T2D management and induce diabetes remission whenever still possible.
Collapse
Affiliation(s)
- Danila Capoccia
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Frida Leonetti
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy.
| | - Andrea Natali
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Domenico Tricò
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Sebastio Perrini
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Paolo Sbraccia
- Department of Systems Medicine, Unit of Internal Medicine - Obesity Center, Policlinico Tor Vergata, University of Rome Tor Vergata, Rome, Italy
| | - Valeria Guglielmi
- Department of Systems Medicine, Unit of Internal Medicine - Obesity Center, Policlinico Tor Vergata, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
5
|
Lima RS, Belchior-Bezerra M, Silva de Oliveira D, Rocha RDS, Medeiros NI, Mattos RT, Dos Reis IC, Marques AS, Rosário PW, Calsolari MR, Correa-Oliveira R, Dutra WO, Moreira PR, Gomes JA. Obesity Influences T CD4 Lymphocytes Subsets Profiles in Children and Adolescent's Immune Response. J Nutr 2024; 154:3133-3143. [PMID: 39019165 DOI: 10.1016/j.tjnut.2024.07.006] [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: 03/21/2024] [Revised: 06/14/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Evidence shows that CD4+ T cells are altered in obesity and play a significant role in the systemic inflammation in adults with the disease. OBJECTIVES Because the profile of these cells is poorly understood in the pediatric population, this study aims to investigate the profile of CD4+ T lymphocytes and the plasma levels of cytokines in this population. METHODS Using flow cytometry, we compared the expression profile of lymphocyte markers, master transcription factors, cytokines, and molecules involved in the regulation of the immune response in CD4+ T cells from children and adolescents with obesity (OB group, n = 20) with those with eutrophy group (EU group, n = 16). Plasma levels of cytokines in both groups were determined by cytometric bead array (CBA). RESULTS The OB group presents a lower frequency of CD3+ T cells, as well as a decreased frequency of CD4+ T cells expressing CD28, IL-4, and FOXP3, but an increased frequency of CD4+IL-17A+ cells compared with the EU group. The frequency of CD28 is increased in Th2 and Treg cells in the OB group, whereas CTLA-4 is decreased in all subpopulations compared with the EU group. Furthermore, Th2, Th17, and Treg profiles can differentiate the EU and OB groups. IL-10 plasma levels are reduced in the OB group and negatively correlated with adiposity and inflammatory parameters. CONCLUSIONS CD4+ T cells have an altered pattern of expression in children and adolescents with obesity, contributing to the inflammatory state and clinical characteristics of these patients.
Collapse
Affiliation(s)
- Rafael Silva Lima
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mayara Belchior-Bezerra
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Daniela Silva de Oliveira
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Nayara I Medeiros
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Imunologia Celular e Molecular, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ, Belo Horizonte, Brazil
| | - Rafael T Mattos
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabelle Camile Dos Reis
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Aiessa Santos Marques
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Pedro Ws Rosário
- Centro de Especialidades Médicas (CEM), Hospital Santa Casa, Belo Horizonte, Brazil
| | | | - Rodrigo Correa-Oliveira
- Imunologia Celular e Molecular, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ, Belo Horizonte, Brazil
| | - Walderez O Dutra
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Paula Rocha Moreira
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juliana As Gomes
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
6
|
Gallo G, Desideri G, Savoia C. Update on Obesity and Cardiovascular Risk: From Pathophysiology to Clinical Management. Nutrients 2024; 16:2781. [PMID: 39203917 PMCID: PMC11356794 DOI: 10.3390/nu16162781] [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: 07/25/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Obesity is an epidemic worldwide. Overweight and multiple obesity-related mechanisms, including dysmetabolic alterations, contribute to cardiovascular deleterious effects. Hence, overweight and obesity have been independently associated with increased cardiovascular risk, whose assessment is crucial for preserving life quality and reducing mortality, and to address appropriate therapeutic strategies in obese patients. Beyond the standard of care in managing overweight and obesity in adults (i.e., diet and physical exercise), several relevant pharmacotherapies have been approved, and several procedures and device types for weight loss have been recommended. In such a contest, medical weight management remains one option for treating excess weight. Most drugs used for obesity reduce appetite and increase satiety and, secondarily, slow gastric emptying to reduce body weight and, therefore, act also to improve metabolic parameters. In this contest, agonists of the glucagon-like peptide-1 receptor (GLP-1RAs) modulate different metabolic pathways associated with glucose metabolism, energy homeostasis, antioxidation, and inflammation. Moreover, this class of drugs has shown efficacy in improving glycemic control, reducing the incidence of cardiovascular events in type 2 diabetic patients, and reducing body weight independently of the presence of diabetes. Recently, in overweight or obese patients with pre-existing cardiovascular disease but without diabetes, the GLP-1RA semaglutide reduced the incidence of cardiovascular and cerebrovascular events and death from cardiovascular causes. Thus, semaglutide has been approved for secondary prevention in obese people with cardiovascular disease. Nevertheless, whether this class of drugs is equally effective for primary prevention in obese people has to be demonstrated. In this review, we will summarize updates on the pathophysiology of obesity, the effects of obesity on cardiovascular risk, the impact of different obesity phenotypes on cardiovascular diseases, and the novelties in the clinical management of obesity for cardiovascular prevention.
Collapse
Affiliation(s)
- Giovanna Gallo
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy;
| | - Giovambattista Desideri
- Department of Clinical, Internal Medicine, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy;
| | - Carmine Savoia
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy;
| |
Collapse
|
7
|
Valentine Y, Nikolajczyk BS. T cells in obesity-associated inflammation: The devil is in the details. Immunol Rev 2024; 324:25-41. [PMID: 38767210 PMCID: PMC11694249 DOI: 10.1111/imr.13354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Obesity presents a significant health challenge, affecting 41% of adults and 19.7% of children in the United States. One of the associated health challenges of obesity is chronic low-grade inflammation. In both mice and humans, T cells in circulation and in the adipose tissue play a pivotal role in obesity-associated inflammation. Changes in the numbers and frequency of specific CD4+ Th subsets and their contribution to inflammation through cytokine production indicate declining metabolic health, that is, insulin resistance and T2D. While some Th subset alterations are consistent between mice and humans with obesity, some changes mainly characterize male mice, whereas female mice often resist obesity and inflammation. However, protection from obesity and inflammation is not observed in human females, who can develop obesity-related T-cell inflammation akin to males. The decline in female sex hormones after menopause is also implicated in promoting obesity and inflammation. Age is a second underappreciated factor for defining and regulating obesity-associated inflammation toward translating basic science findings to the clinic. Weight loss in mice and humans, in parallel with these other factors, does not resolve obesity-associated inflammation. Instead, inflammation persists amid modest changes in CD4+ T cell frequencies, highlighting the need for further research into resolving changes in T-cell function after weight loss. How lingering inflammation after weight loss affecting the common struggle to maintain lower weight is unknown. Semaglutide, a newly popular pharmaceutical used for treating T2D and reversing obesity, holds promise for alleviating obesity-associated health complications, yet its impact on T-cell-mediated inflammation remains unexplored. Further work in this area could significantly contribute to the scientific understanding of the impacts of weight loss and sex/hormones in obesity and obesity-associated metabolic decline.
Collapse
Affiliation(s)
- Yolander Valentine
- Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, Kentucky, USA
| | - Barbara S. Nikolajczyk
- Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, Kentucky, USA
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
- Barnstable Brown Diabetes and Obesity Research Center, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
8
|
Wang H, He W, Yang G, Zhu L, Liu X. The Impact of Weight Cycling on Health and Obesity. Metabolites 2024; 14:344. [PMID: 38921478 PMCID: PMC11205792 DOI: 10.3390/metabo14060344] [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: 05/22/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024] Open
Abstract
Obesity is a systemic and chronic inflammation, which seriously endangers people's health. People tend to diet to control weight, and the short-term effect of dieting in losing weight is significant, but the prognosis is limited. With weight loss and recovery occurring frequently, people focus on weight cycling. The effect of weight cycling on a certain tissue of the body also has different conclusions. Therefore, this article systematically reviews the effects of body weight cycling on the body and finds that multiple weight cycling (1) increased fat deposition in central areas, lean mass decreased in weight loss period, and fat mass increased in weight recovery period, which harms body composition and skeletal muscle mass; (2) enhanced the inflammatory response of adipose tissue, macrophages infiltrated into adipose tissue, and increased the production of pro-inflammatory mediators in adipocytes; (3) blood glucose concentration mutation and hyperinsulinemia caused the increase or decrease in pancreatic β-cell population, which makes β-cell fatigue and leads to β-cell failure; (4) resulted in additional burden on the cardiovascular system because of cardiovascular rick escalation. Physical activity combined with calorie restriction can effectively reduce metabolic disease and chronic inflammation, alleviating the adverse effects of weight cycling on the body.
Collapse
Affiliation(s)
- Huan Wang
- Graduate School, Guangzhou Sport University, Guangzhou 510500, China; (H.W.); (W.H.); (G.Y.)
| | - Wenbi He
- Graduate School, Guangzhou Sport University, Guangzhou 510500, China; (H.W.); (W.H.); (G.Y.)
| | - Gaoyuan Yang
- Graduate School, Guangzhou Sport University, Guangzhou 510500, China; (H.W.); (W.H.); (G.Y.)
| | - Lin Zhu
- Graduate School, Guangzhou Sport University, Guangzhou 510500, China; (H.W.); (W.H.); (G.Y.)
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Guangzhou Sport University, Guangzhou 510500, China
| | - Xiaoguang Liu
- Graduate School, Guangzhou Sport University, Guangzhou 510500, China; (H.W.); (W.H.); (G.Y.)
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Guangzhou Sport University, Guangzhou 510500, China
| |
Collapse
|
9
|
Kühnen P, Argente J, Clément K, Dollfus H, Dubern B, Farooqi S, de Groot C, Grüters A, Holm JC, Hopkins M, Kleinendorst L, Körner A, Meeker D, Rydén M, von Schnurbein J, Tschöp M, Yeo GSH, Zorn S, Wabitsch M. IMPROVE 2022 International Meeting on Pathway-Related Obesity: Vision of Excellence. Clin Obes 2024; 14:e12659. [PMID: 38602039 DOI: 10.1111/cob.12659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 03/08/2024] [Indexed: 04/12/2024]
Abstract
Nearly 90 clinicians and researchers from around the world attended the first IMPROVE 2022 International Meeting on Pathway-Related Obesity. Delegates attended in person or online from across Europe, Argentina and Israel to hear the latest scientific and clinical developments in hyperphagia and severe, early-onset obesity, and set out a vision of excellence for the future for improving the diagnosis, treatment, and care of patients with melanocortin-4 receptor (MC4R) pathway-related obesity. The meeting co-chair Peter Kühnen, Charité Universitätsmedizin Berlin, Germany, indicated that change was needed with the rapidly increasing prevalence of obesity and the associated complications to improve the understanding of the underlying mechanisms and acknowledge that monogenic forms of obesity can play an important role, providing insights that can be applied to a wider group of patients with obesity. World-leading experts presented the latest research and led discussions on the underlying science of obesity, diagnosis (including clinical and genetic approaches such as the role of defective MC4R signalling), and emerging clinical data and research with targeted pharmacological approaches. The aim of the meeting was to agree on the questions that needed to be addressed in future research and to ensure that optimised diagnostic work-up was used with new genetic testing tools becoming available. This should aid the planning of new evidence-based treatment strategies for the future, as explained by co-chair Martin Wabitsch, Ulm University Medical Center, Germany.
Collapse
Affiliation(s)
- Peter Kühnen
- Department of Pediatric Endocrinology and Diabetology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Jesús Argente
- Departments of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Karine Clément
- Assistance Publique-Hôpitaux de Paris, Nutrition Department, Pitié-Salpêtrière Hospital, Paris, France
- INSERM, Nutrition and Obesity: Systemic Approaches, NutriOmics, Research Unit, Sorbonne Université, Paris, France
| | - Hélène Dollfus
- CARGO and Department of Medical Genetics, University of Strasbourg, Strasbourg, France
| | - Béatrice Dubern
- INSERM, Nutrition and Obesity: Systemic Approaches, NutriOmics, Research Unit, Sorbonne Université, Paris, France
- Sorbonne Université, Trousseau Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Sadaf Farooqi
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Corjan de Groot
- Sophia Children's Hospital, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Annette Grüters
- Department of Pediatric Endocrinology and Diabetes, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Jens-Christian Holm
- The Children's Obesity Clinic, accredited European Centre for Obesity Management, Department of Pediatrics, Copenhagen University Hospital Holbæk, Copenhagen, Denmark
| | - Mark Hopkins
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | - Lotte Kleinendorst
- Department of Clinical Genetics, Amsterdam UMC, Amsterdam, The Netherlands
| | - Antje Körner
- Center for Pediatric Research, Department of Pediatrics, LIFE Research Center for Civilization Diseases, University Hospital Leipzig, Leipzig, Germany
| | - David Meeker
- Rhythm Pharmaceuticals, Boston, Massachusetts, USA
| | - Mikael Rydén
- Department of Medicine H7, Karolinska Institute, Stockholm, Sweden
- Department of Endocrinology and Metabolism, Karolinska University Hospital, Stockholm, Sweden
| | - Julia von Schnurbein
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Matthias Tschöp
- Institute for Diabetes and Obesity, Helmholtz Zentrum, Munich, Germany
| | - Giles S H Yeo
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Stefanie Zorn
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Martin Wabitsch
- Division of Paediatric Endocrinology and Diabetes, Department of Paediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
10
|
Shimi G, Sohouli MH, Ghorbani A, Shakery A, Zand H. The interplay between obesity, immunosenescence, and insulin resistance. Immun Ageing 2024; 21:13. [PMID: 38317257 PMCID: PMC10840211 DOI: 10.1186/s12979-024-00414-7] [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: 12/12/2023] [Accepted: 01/22/2024] [Indexed: 02/07/2024]
Abstract
Obesity, which is the accumulation of fat in adipose tissue, has adverse impacts on human health. Obesity-related metabolic dysregulation has similarities to the metabolic alterations observed in aging. It has been shown that the adipocytes of obese individuals undergo cellular aging, known as senescence. Senescence can be transmitted to other normal cells through a series of chemical factors referred to as the senescence-associated secretory phenotype (SASP). Most of these factors are pro-inflammatory compounds. The immune system removes these senescent T-cells, but immunosenescence, which is the senescence of immune cells, disrupts the clearance of senescent T-cells. Immunosenescence occurs as a result of aging or indirectly through transmission from senescent tissues. The significant occurrence of senescence in obesity is expected to cause immunosenescence and impairs the immune response to resolve inflammation. The sustained and chronic inflammation disrupts insulin's metabolic actions in metabolic tissues. Therefore, this review focuses on the role of senescent adipocyte cells in obesity-associated immunosenescence and subsequent metabolic dysregulation. Moreover, the article suggests novel therapeutic approaches to improve metabolic syndrome by targeting senescent T-cells or using senotherapeutics.
Collapse
Affiliation(s)
- Ghazaleh Shimi
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, 1981619573, Iran
| | - Mohammad Hassan Sohouli
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, 1981619573, Iran
| | - Arman Ghorbani
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, 1981619573, Iran
| | - Azam Shakery
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, 1981619573, Iran
| | - Hamid Zand
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, 1981619573, Iran.
| |
Collapse
|
11
|
Gliniak CM, Pedersen L, Scherer PE. Adipose tissue fibrosis: the unwanted houseguest invited by obesity. J Endocrinol 2023; 259:e230180. [PMID: 37855264 PMCID: PMC11648981 DOI: 10.1530/joe-23-0180] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/27/2023] [Indexed: 09/28/2023]
Abstract
The prevalence of obesity is increasing exponentially across the globe. The lack of effective treatment options for long-term weight loss has magnified the enormity of this problem. Studies continue to demonstrate that adipose tissue holds a biological memory, one of the most important determinant of long-term weight maintenance. This phenomenon is consistent with the metabolically dynamic role of adipose tissue: it adapts and expands to store for excess energy and serves as an endocrine organ capable of synthesizing a number of biologically active molecules that regulate metabolic homeostasis. An important component of the plasticity of adipose tissue is the extracellular matrix, essential for structural support, mechanical stability, cell signaling and function. Chronic obesity upends a delicate balance of extracellular matrix synthesis and degradation, and the ECM accumulates in such a way that prevents the plasticity and function of the diverse cell types in adipose tissue. A series of maladaptive responses among the cells in adipose tissue leads to inflammation and fibrosis, major mechanisms that explain the link between obesity and insulin resistance, risk of type 2 diabetes, cardiovascular disease, and nonalcoholic fatty liver disease. Adipose tissue fibrosis persists after weight loss and further enhances adipose tissue dysfunction if weight is regained. Here, we highlight the current knowledge of the cellular events governing adipose tissue ECM remodeling during the development of obesity. Our goal is to delineate the relationship more clearly between adipose tissue ECM and metabolic disease, an important step toward better defining the pathophysiology of dysfunctional adipose tissue.
Collapse
Affiliation(s)
- Christy M Gliniak
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Line Pedersen
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Philipp E Scherer
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
12
|
Schleh MW, Caslin HL, Garcia JN, Mashayekhi M, Srivastava G, Bradley AB, Hasty AH. Metaflammation in obesity and its therapeutic targeting. Sci Transl Med 2023; 15:eadf9382. [PMID: 37992150 PMCID: PMC10847980 DOI: 10.1126/scitranslmed.adf9382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 08/29/2023] [Indexed: 11/24/2023]
Abstract
Obesity-associated inflammation is a systemic process that affects all metabolic organs. Prominent among these is adipose tissue, where cells of the innate and adaptive immune system are markedly changed in obesity, implicating these cells in a range of processes linking immune memory to metabolic regulation. Furthermore, weight loss and weight cycling have unexpected effects on adipose tissue immune populations. Here, we review the current literature on the roles of various immune cells in lean and obese adipose tissue. Within this context, we discuss pharmacological and nonpharmacological approaches to obesity treatment and their impact on systemic inflammation.
Collapse
Affiliation(s)
- Michael W. Schleh
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Heather L. Caslin
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Jamie N. Garcia
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Mona Mashayekhi
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Gitanjali Srivastava
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Weight Loss Center, Vanderbilt University Medical Center, Nashville, TN 37204 USA
| | - Anna B. Bradley
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Weight Loss Center, Vanderbilt University Medical Center, Nashville, TN 37204 USA
- VA Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| | - Alyssa H. Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- VA Tennessee Valley Healthcare System, Nashville, TN 37212, USA
| |
Collapse
|
13
|
Caslin HL, Cottam MA, Betjemann AM, Mashayekhi M, Silver HJ, Hasty AH. Single cell RNA-sequencing suggests a novel lipid associated mast cell population following weight cycling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.12.566786. [PMID: 38014269 PMCID: PMC10680619 DOI: 10.1101/2023.11.12.566786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Our recent study showed weight cycled mice have increased adipose mast cells compared to obese mice by single cell RNA-sequencing. Here, we aimed to confirm and elucidate these changes. Further analysis of our dataset showed that our initial mast cell cluster could subcluster into two unique populations: one with very high expression of classical mast cell markers and another with elevated lipid handling and antigen presentation genes. This new mast cell cluster accounted for most of the mast cells in the weight cycled group although it was not possible to detect the different populations by new studies with flow cytometry or Toluidine blue staining in mice, possibly due to a downregulation in classical mast cell genes. Interestingly, a pilot study in humans did suggest the existence of two mast cell populations in subcutaneous adipose tissue from obese women that appear similar to the murine populations detected by sequencing; one of which was significantly correlated with weight variance. Together, these data suggest that weight cycling may induce a unique population of mast cells similar to lipid associated macrophages. Future studies will focus on isolation of these cells to better determine their lineage, differentiation, and functional roles.
Collapse
|
14
|
Yan J, Hu C. Bone marrow immune cells stop weight regain. Cell Metab 2023; 35:1845-1846. [PMID: 37939653 DOI: 10.1016/j.cmet.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/03/2023] [Accepted: 10/07/2023] [Indexed: 11/10/2023]
Abstract
Weight regain is a major challenge in the long-term management of obesity; however, the underlying mechanisms remain unclear. Zhou et al. found that bone-marrow-derived CD7+ monocytes respond to fluctuating nutritional stress and suppress weight regain by promoting beige fat thermogenesis.
Collapse
Affiliation(s)
- Jing Yan
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cheng Hu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute for Metabolic Disease, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai, China.
| |
Collapse
|
15
|
Zhou HY, Feng X, Wang LW, Zhou R, Sun H, Chen X, Lu RB, Huang Y, Guo Q, Luo XH. Bone marrow immune cells respond to fluctuating nutritional stress to constrain weight regain. Cell Metab 2023; 35:1915-1930.e8. [PMID: 37703873 DOI: 10.1016/j.cmet.2023.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/19/2023] [Accepted: 08/18/2023] [Indexed: 09/15/2023]
Abstract
Weight regain after weight loss is a major challenge in the treatment of obesity. Immune cells adapt to fluctuating nutritional stress, but their roles in regulating weight regain remain unclear. Here, we identify a stem cell-like CD7+ monocyte subpopulation accumulating in the bone marrow (BM) of mice and humans that experienced dieting-induced weight loss. Adoptive transfer of CD7+ monocytes suppresses weight regain, whereas inducible depletion of CD7+ monocytes accelerates it. These cells, accumulating metabolic memories via epigenetic adaptations, preferentially migrate to the subcutaneous white adipose tissue (WAT), where they secrete fibrinogen-like protein 2 (FGL2) to activate the protein kinase A (PKA) signaling pathway and facilitate beige fat thermogenesis. Nevertheless, CD7+ monocytes gradually enter a quiescent state after weight loss, accompanied by increased susceptibility to weight regain. Notably, administration of FMS-like tyrosine kinase 3 ligand (FLT3L) remarkably rejuvenates CD7+ monocytes, thus ameliorating rapid weight regain. Together, our findings identify a unique bone marrow-derived metabolic-memory immune cell population that could be targeted to combat obesity.
Collapse
Affiliation(s)
- Hai-Yan Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Xu Feng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Li-Wen Wang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Rui Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Heng Sun
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Xin Chen
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Ren-Bin Lu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Yan Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Qi Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Xiang-Hang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan 410008, China; Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Hunan 410008, China.
| |
Collapse
|
16
|
van Baak MA, Mariman ECM. Obesity-induced and weight-loss-induced physiological factors affecting weight regain. Nat Rev Endocrinol 2023; 19:655-670. [PMID: 37696920 DOI: 10.1038/s41574-023-00887-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/27/2023] [Indexed: 09/13/2023]
Abstract
Weight regain after successful weight loss resulting from lifestyle interventions is a major challenge in the management of overweight and obesity. Knowledge of the causal mechanisms for weight regain can help researchers and clinicians to find effective strategies to tackle weight regain and reduce obesity-associated metabolic and cardiovascular complications. This Review summarizes the current understanding of a number of potential physiological mechanisms underlying weight regain after weight loss, including: the role of adipose tissue immune cells; hormonal and neuronal factors affecting hunger, satiety and reward; resting energy expenditure and adaptive thermogenesis; and lipid metabolism (lipolysis and lipid oxidation). We describe and discuss obesity-associated changes in these mechanisms, their persistence during weight loss and weight regain and their association with weight regain. Interventions to prevent or limit weight regain based on these factors, such as diet, exercise, pharmacotherapy and biomedical strategies, and current knowledge on the effectiveness of these interventions are also reviewed.
Collapse
Affiliation(s)
- Marleen A van Baak
- NUTRIM School for Nutrition and Translational Research in Metabolism, Department of Human Biology, Maastricht University, Maastricht, Netherlands.
| | - Edwin C M Mariman
- NUTRIM School for Nutrition and Translational Research in Metabolism, Department of Human Biology, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
17
|
Luo Q, Dwaraka VB, Chen Q, Tong H, Zhu T, Seale K, Raffaele JM, Zheng SC, Mendez TL, Chen Y, Carreras N, Begum S, Mendez K, Voisin S, Eynon N, Lasky-Su JA, Smith R, Teschendorff AE. A meta-analysis of immune-cell fractions at high resolution reveals novel associations with common phenotypes and health outcomes. Genome Med 2023; 15:59. [PMID: 37525279 PMCID: PMC10388560 DOI: 10.1186/s13073-023-01211-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/10/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND Changes in cell-type composition of tissues are associated with a wide range of diseases and environmental risk factors and may be causally implicated in disease development and progression. However, these shifts in cell-type fractions are often of a low magnitude, or involve similar cell subtypes, making their reliable identification challenging. DNA methylation profiling in a tissue like blood is a promising approach to discover shifts in cell-type abundance, yet studies have only been performed at a relatively low cellular resolution and in isolation, limiting their power to detect shifts in tissue composition. METHODS Here we derive a DNA methylation reference matrix for 12 immune-cell types in human blood and extensively validate it with flow-cytometric count data and in whole-genome bisulfite sequencing data of sorted cells. Using this reference matrix, we perform a directional Stouffer and fixed effects meta-analysis comprising 23,053 blood samples from 22 different cohorts, to comprehensively map associations between the 12 immune-cell fractions and common phenotypes. In a separate cohort of 4386 blood samples, we assess associations between immune-cell fractions and health outcomes. RESULTS Our meta-analysis reveals many associations of cell-type fractions with age, sex, smoking and obesity, many of which we validate with single-cell RNA sequencing. We discover that naïve and regulatory T-cell subsets are higher in women compared to men, while the reverse is true for monocyte, natural killer, basophil, and eosinophil fractions. Decreased natural killer counts associated with smoking, obesity, and stress levels, while an increased count correlates with exercise and sleep. Analysis of health outcomes revealed that increased naïve CD4 + T-cell and N-cell fractions associated with a reduced risk of all-cause mortality independently of all major epidemiological risk factors and baseline co-morbidity. A machine learning predictor built only with immune-cell fractions achieved a C-index value for all-cause mortality of 0.69 (95%CI 0.67-0.72), which increased to 0.83 (0.80-0.86) upon inclusion of epidemiological risk factors and baseline co-morbidity. CONCLUSIONS This work contributes an extensively validated high-resolution DNAm reference matrix for blood, which is made freely available, and uses it to generate a comprehensive map of associations between immune-cell fractions and common phenotypes, including health outcomes.
Collapse
Affiliation(s)
- Qi Luo
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Varun B Dwaraka
- TruDiagnostics, 881 Corporate Dr., Lexington, KY, 40503, USA
| | - Qingwen Chen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Huige Tong
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Tianyu Zhu
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Kirsten Seale
- Institute for Health and Sport (iHeS), Victoria University, Footscray, VIC, 3011, Australia
| | - Joseph M Raffaele
- PhysioAge LLC, 30 Central Park South / Suite 8A, New York, NY, 10019, USA
| | - Shijie C Zheng
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, USA
| | - Tavis L Mendez
- TruDiagnostics, 881 Corporate Dr., Lexington, KY, 40503, USA
| | - Yulu Chen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | | | - Sofina Begum
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Kevin Mendez
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Sarah Voisin
- Institute for Health and Sport (iHeS), Victoria University, Footscray, VIC, 3011, Australia
| | - Nir Eynon
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Jessica A Lasky-Su
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| | - Ryan Smith
- TruDiagnostics, 881 Corporate Dr., Lexington, KY, 40503, USA.
| | - Andrew E Teschendorff
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China.
| |
Collapse
|
18
|
Li W, Chen W. Weight cycling based on altered immune microenvironment as a result of metaflammation. Nutr Metab (Lond) 2023; 20:13. [PMID: 36814270 PMCID: PMC9945679 DOI: 10.1186/s12986-023-00731-6] [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] [Accepted: 02/07/2023] [Indexed: 02/24/2023] Open
Abstract
As a result of the obesity epidemic, more people are concerned about losing weight; however, weight regain is common, leading to repeated weight loss and weight cycling. The health benefits of early weight loss are nullified by weight regain after weight cycling, which has much more severe metabolic consequences. Weight cycling alters body composition, resulting in faster fat recovery and slower muscle reconstruction. This evident fat accumulation, muscle loss, and ectopic fat deposition destroy the intestinal barrier, increase the permeability of the small intestinal epithelium, and cause the lipotoxicity of lipid metabolites and toxins to leak into extraintestinal tissues and circulation. It causes oxidative stress and hypoxia in local tissues and immune cell infiltration in various tissues, all contributing to the adaptation to this metabolic change. Immune cells transmit inflammatory responses in adipose and skeletal muscle tissue by secreting cytokines and adipokines, which mediate immune cell pathways and cause metaflammation and inefficient metabolic degradation. In this review, we focus on the regulatory function of the immunological microenvironment in the final metabolic outcome, with a particular emphasis on the cellular and molecular processes of local and systemic metaflammation induced by weight cycling-induced changes in body composition. Metaflammation in adipose and muscle tissues that is difficult to relieve may cause weight cycling. As this chronic low-grade inflammation spreads throughout the body, metabolic complications associated with weight cycling are triggered. Inhibiting the onset and progression of metabolic inflammation and enhancing the immune microenvironment of adipose and muscle tissues may be the first step in addressing weight cycling.
Collapse
Affiliation(s)
- Wanyang Li
- grid.413106.10000 0000 9889 6335Department of Clinical Nutrition, Chinese Academy of Medical Sciences - Peking Union Medical College, Peking Union Medical College Hospital, No. 1 Shuaifuyuan, Dongcheng District, Beijing, 100730 China
| | - Wei Chen
- Department of Clinical Nutrition, Chinese Academy of Medical Sciences - Peking Union Medical College, Peking Union Medical College Hospital, No. 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
19
|
Shirakawa K, Sano M. Drastic transformation of visceral adipose tissue and peripheral CD4 T cells in obesity. Front Immunol 2023; 13:1044737. [PMID: 36685567 PMCID: PMC9846168 DOI: 10.3389/fimmu.2022.1044737] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Obesity has a pronounced effect on the immune response in systemic organs that results in not only insulin resistance but also altered immune responses to infectious diseases and malignant tumors. Obesity-associated microenvironmental changes alter transcriptional expression and metabolism in T cells, leading to alterations in T-cell differentiation, proliferation, function, and survival. Adipokines, cytokines, and lipids derived from obese visceral adipose tissue (VAT) may also contribute to the systemic T-cell phenotype, resulting in obesity-specific pathogenesis. VAT T cells, which have multiple roles in regulating homeostasis and energy utilization and defending against pathogens, are most susceptible to obesity. In particular, many studies have shown that CD4 T cells are deeply involved in the homeostasis of VAT endocrine and metabolic functions and in obesity-related chronic inflammation. In obesity, macrophages and adipocytes in VAT function as antigen-presenting cells and contribute to the obesity-specific CD4 T-cell response by inducing CD4 T-cell proliferation and differentiation into inflammatory effectors via interactions between major histocompatibility complex class II and T-cell receptors. When obesity persists, prolonged stimulation by leptin and circulating free fatty acids, repetitive antigen stimulation, activating stress responses, and hypoxia induce exhaustion of CD4 T cells in VAT. T-cell exhaustion is characterized by restricted effector function, persistent expression of inhibitory receptors, and a transcriptional state distinct from functional effector and memory T cells. Moreover, obesity causes thymic regression, which may result in homeostatic proliferation of obesity-specific T-cell subsets due to changes in T-cell metabolism and gene expression in VAT. In addition to causing T-cell exhaustion, obesity also accelerates cellular senescence of CD4 T cells. Senescent CD4 T cells secrete osteopontin, which causes further VAT inflammation. The obesity-associated transformation of CD4 T cells remains a negative legacy even after weight loss, causing treatment resistance of obesity-related conditions. This review discusses the marked transformation of CD4 T cells in VAT and systemic organs as a consequence of obesity-related microenvironmental changes.
Collapse
Affiliation(s)
| | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
20
|
Zhao J, Jiang Y, Qian J, Qian Z, Yang H, Shi W, Gong Y, Jiao Y, Tang L. A nomogram model based on the combination of the systemic immune-inflammation index and prognostic nutritional index predicts weight regain after laparoscopic sleeve gastrectomy. Surg Obes Relat Dis 2023; 19:50-58. [PMID: 36008279 DOI: 10.1016/j.soard.2022.07.014] [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: 02/04/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND The high rate of weight regain after laparoscopic sleeve gastrectomy is a great challenge. The systemic immune-inflammation index (SII; calculated by neutrophils, lymphocytes, and platelets) and prognostic nutritional index (PNI; calculated by albumin and lymphocytes) are widely used as prognostic factors in various diseases. OBJECTIVES The objective of this study was to investigate independent the independent risk factors associated with weight regain in patients after laparoscopic sleeve gastrectomy. SETTING A single-center retrospective study. METHODS Weight regain was defined as the percentage of increase in body weight ≥10% in comparison with the nadir weight postoperatively. Eligible patients admitted to the bariatric center of our hospital were consecutively enrolled and grouped according to the occurrence of weight regain within 5 postoperative years. Univariate and multivariate logistic regression analyses were performed to assess potential risk factors. A nomogram model containing the risk factors was then constructed and evaluated by R. RESULTS A total of 217 patients were enrolled, and 87 (40.1%) patients experienced weight regain. Univariate and logistic regression analyses indicated that depression (odds ratio [OR]: 2.51, 95% confidence interval [CI]: 1.20-5.22, P = .015), psychological counseling (OR: 2.27, 95% CI: 1.20-4.33, P = .017), preoperative C-reactive protein (OR: 2.20, 95% CI: 1.18-4.13, P = .012), and combination of SII-PNI scores (OR: .45, 95% CI: .31-.67, P < .001) were 4 independent risk factors for postoperative weight regain in laparoscopic sleeve gastrectomy patients. The area under the curve of the constructed nomogram model for predicting weight regain was .706. CONCLUSIONS This study concluded that the combination of the SII-PNI was an independent risk factor for weight regain and that the nomogram model based on the combination of the SII-PNI had a good predictive value.
Collapse
Affiliation(s)
- Jie Zhao
- Department of Gastrointestinal Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou City, Jiangsu Province, China
| | - Yicheng Jiang
- Department of Gastrointestinal Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou City, Jiangsu Province, China
| | - Jun Qian
- Department of Gastrointestinal Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou City, Jiangsu Province, China
| | - Zhifen Qian
- Department of Gastrointestinal Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou City, Jiangsu Province, China
| | - Haojun Yang
- Department of Gastrointestinal Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou City, Jiangsu Province, China
| | - Weihai Shi
- Department of Gastrointestinal Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou City, Jiangsu Province, China
| | - Yu Gong
- Department of Gastrointestinal Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou City, Jiangsu Province, China
| | - Yuwen Jiao
- Department of Gastrointestinal Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou City, Jiangsu Province, China.
| | - Liming Tang
- Department of Gastrointestinal Surgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou City, Jiangsu Province, China.
| |
Collapse
|
21
|
Chua D, Low ZS, Cheam GX, Ng AS, Tan NS. Utility of Human Relevant Preclinical Animal Models in Navigating NAFLD to MAFLD Paradigm. Int J Mol Sci 2022; 23:14762. [PMID: 36499091 PMCID: PMC9737809 DOI: 10.3390/ijms232314762] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/15/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Fatty liver disease is an emerging contributor to disease burden worldwide. The past decades of work established the heterogeneous nature of non-alcoholic fatty liver disease (NAFLD) etiology and systemic contributions to the pathogenesis of the disease. This called for the proposal of a redefinition in 2020 to that of metabolic dysfunction-associated fatty liver disease (MAFLD) to better reflect the current understanding of the disease. To date, several clinical cohort studies comparing NAFLD and MAFLD hint at the relevancy of the new nomenclature in enriching for patients with more severe hepatic injury and extrahepatic comorbidities. However, the underlying systemic pathogenesis is still not fully understood. Preclinical animal models have been imperative in elucidating key biological mechanisms in various contexts, including intrahepatic disease progression, interorgan crosstalk and systemic dysregulation. Furthermore, they are integral in developing novel therapeutics against MAFLD. However, substantial contextual variabilities exist across different models due to the lack of standardization in several aspects. As such, it is crucial to understand the strengths and weaknesses of existing models to better align them to the human condition. In this review, we consolidate the implications arising from the change in nomenclature and summarize MAFLD pathogenesis. Subsequently, we provide an updated evaluation of existing MAFLD preclinical models in alignment with the new definitions and perspectives to improve their translational relevance.
Collapse
Affiliation(s)
- Damien Chua
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore
| | - Zun Siong Low
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore
| | - Guo Xiang Cheam
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Aik Seng Ng
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore
| |
Collapse
|
22
|
Rogal J, Roosz J, Teufel C, Cipriano M, Xu R, Eisler W, Weiss M, Schenke‐Layland K, Loskill P. Autologous Human Immunocompetent White Adipose Tissue-on-Chip. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104451. [PMID: 35466539 PMCID: PMC9218765 DOI: 10.1002/advs.202104451] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 03/03/2022] [Indexed: 05/07/2023]
Abstract
Obesity and associated diseases, such as diabetes, have reached epidemic proportions globally. In this era of "diabesity", white adipose tissue (WAT) has become a target of high interest for therapeutic strategies. To gain insights into mechanisms of adipose (patho-)physiology, researchers traditionally relied on animal models. Leveraging Organ-on-Chip technology, a microphysiological in vitro model of human WAT is introduced: a tailored microfluidic platform featuring vasculature-like perfusion that integrates 3D tissues comprising all major WAT-associated cellular components (mature adipocytes, organotypic endothelial barriers, stromovascular cells including adipose tissue macrophages) in an autologous manner and recapitulates pivotal WAT functions, such as energy storage and mobilization as well as endocrine and immunomodulatory activities. A precisely controllable bottom-up approach enables the generation of a multitude of replicates per donor circumventing inter-donor variability issues and paving the way for personalized medicine. Moreover, it allows to adjust the model's degree of complexity via a flexible mix-and-match approach. This WAT-on-Chip system constitutes the first human-based, autologous, and immunocompetent in vitro adipose tissue model that recapitulates almost full tissue heterogeneity and can become a powerful tool for human-relevant research in the field of metabolism and its associated diseases as well as for compound testing and personalized- and precision medicine applications.
Collapse
Affiliation(s)
- Julia Rogal
- Department for Microphysiological Systems, Institute of Biomedical EngineeringEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGBNobelstr. 12Stuttgart70569Germany
| | - Julia Roosz
- NMI Natural and Medical Sciences Institute at the University of TübingenMarkwiesenstr. 55Reutlingen72770Germany
| | - Claudia Teufel
- Department for Microphysiological Systems, Institute of Biomedical EngineeringEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
| | - Madalena Cipriano
- Department for Microphysiological Systems, Institute of Biomedical EngineeringEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
- 3R‐Center for In vitro Models and Alternatives to Animal TestingEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
| | - Raylin Xu
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGBNobelstr. 12Stuttgart70569Germany
- Harvard Medical School (HMS)25 Shattuck StBostonMA02115USA
| | - Wiebke Eisler
- Clinic for PlasticReconstructiveHand and Burn SurgeryBG Trauma CenterEberhard Karls University TübingenSchnarrenbergstraße 95Tübingen72076Germany
| | - Martin Weiss
- NMI Natural and Medical Sciences Institute at the University of TübingenMarkwiesenstr. 55Reutlingen72770Germany
- Department of Women's HealthEberhard Karls University TübingenCalwerstrasse 7Tübingen72076Germany
| | - Katja Schenke‐Layland
- NMI Natural and Medical Sciences Institute at the University of TübingenMarkwiesenstr. 55Reutlingen72770Germany
- Department of Medicine/CardiologyCardiovascular Research LaboratoriesDavid Geffen School of Medicine at UCLA675 Charles E. Young Drive South, MRL 3645Los AngelesCA90095USA
- Cluster of Excellence iFIT (EXC2180) “Image‐Guided and Functionally Instructed Tumor Therapies”Eberhard Karls University TuebingenRöntgenweg 11Tuebingen72076Germany
- Department for Medical Technologies and Regenerative MedicineInstitute of Biomedical EngineeringEberhard Karls University TübingenSilcherstr. 7/1Tübingen72076Germany
| | - Peter Loskill
- Department for Microphysiological Systems, Institute of Biomedical EngineeringEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
- NMI Natural and Medical Sciences Institute at the University of TübingenMarkwiesenstr. 55Reutlingen72770Germany
- 3R‐Center for In vitro Models and Alternatives to Animal TestingEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
| |
Collapse
|
23
|
Cottam MA, Caslin HL, Winn NC, Hasty AH. Multiomics reveals persistence of obesity-associated immune cell phenotypes in adipose tissue during weight loss and weight regain in mice. Nat Commun 2022; 13:2950. [PMID: 35618862 PMCID: PMC9135744 DOI: 10.1038/s41467-022-30646-4] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 05/11/2022] [Indexed: 01/08/2023] Open
Abstract
Within adipose tissue (AT), immune cells and parenchymal cells closely interact creating a complex microenvironment. In obesity, immune cell derived inflammation contributes to insulin resistance and glucose intolerance. Diet-induced weight loss improves glucose tolerance; however, weight regain further exacerbates the impairment in glucose homeostasis observed with obesity. To interrogate the immunometabolic adaptations that occur in AT during murine weight loss and weight regain, we utilized cellular indexing of transcriptomes and epitopes by sequencing (CITEseq) in male mice. Obesity-induced imprinting of AT immune cells persisted through weight-loss and progressively worsened with weight regain, ultimately leading to impaired recovery of type 2 regulatory cells, activation of antigen presenting cells, T cell exhaustion, and enhanced lipid handling in macrophages in weight cycled mice. This work provides critical groundwork for understanding the immunological causes of weight cycling-accelerated metabolic disease. For further discovery, we provide an open-access web portal of diet-induced AT immune cell imprinting: https://hastylab.shinyapps.io/MAIseq .
Collapse
Affiliation(s)
- Matthew A Cottam
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Heather L Caslin
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Nathan C Winn
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA.
- VA Tennessee Valley Healthcare System, Nashville, TN, USA.
| |
Collapse
|
24
|
Thillainadesan S, Madsen S, James DE, Hocking SL. The impact of weight cycling on health outcomes in animal models: A systematic review and meta-analysis. Obes Rev 2022; 23:e13416. [PMID: 35075766 DOI: 10.1111/obr.13416] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 12/12/2022]
Abstract
The pattern of weight loss and regain, termed "weight cycling," is common in overweight individuals. It is unclear whether the well-established benefits of weight loss persist following weight regain or whether weight cycling is harmful. Human studies of weight cycling have conflicting results reflecting limitations of the observational designs of these studies. By controlling the macronutrient content of diets in animal studies, weight cycling can be studied in a highly controlled manner, thereby overcoming the limitations of human studies. We conducted a systematic review and meta-analysis of animal studies which assessed the health consequences of weight cycling. Studies were classified into those which compared weight cycling to lifelong obesity and those which compared weight cycling to later onset obesity. There were no differences in health outcomes between weight cycled animals and those with lifelong obesity, highlighting that weight regain reverses health benefits achieved by weight loss. In comparison with animals with later onset obesity, weight cycled animals had higher fasting glucose levels and more impaired glucose tolerance following weight regain. Our review of animal studies suggests that health benefits of diet-induced weight loss do not persist after weight regain and weight cycling results in adverse metabolic outcomes.
Collapse
Affiliation(s)
- Senthil Thillainadesan
- Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia.,Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia.,Department of Endocrinology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Søren Madsen
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia.,School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - David E James
- Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia.,Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia.,School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - Samantha L Hocking
- Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia.,Department of Endocrinology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| |
Collapse
|
25
|
Camell CD. Adipose tissue microenvironments during aging: Effects on stimulated lipolysis. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159118. [PMID: 35131468 PMCID: PMC8986088 DOI: 10.1016/j.bbalip.2022.159118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 10/17/2021] [Accepted: 01/20/2022] [Indexed: 12/15/2022]
Abstract
Adipose tissue is a critical organ for nutrient sensing, energy storage and maintaining metabolic health. The failure of adipose tissue homeostasis leads to metabolic disease that is seen during obesity or aging. Local metabolic processes are coordinated by interacting microenvironments that make up the complexity and heterogeneity of the adipose tissue. Catecholamine-induced lipolysis, a critical pathway in adipocytes that drives the release of stored triglyceride as free fatty acid after stimulation, is impaired during aging. The impairment of this pathway is associated with a failure to maintain a healthy body weight, core body-temperature during cold stress or mount an immune response. Along with impairments in aged adipocytes, aging is associated with an accumulation of inflammation, immune cell activation, and increased dysfunction in the nervous and lymphatic systems within the adipose tissue. Together these microenvironments support the initiation of stimulated lipolysis and the transport of free fatty acid under conditions of metabolic homeostasis. However, during aging, the defects in these cellular systems result in a reduction in ability to stimulate lipolysis. This review will focus on how the immune, nervous and lymphatic systems interact during tissue homeostasis, review areas that are impaired with aging and discuss areas of research that are currently unclear.
Collapse
Affiliation(s)
- Christina D Camell
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States of America.
| |
Collapse
|
26
|
Fuseini H, Smith R, Nochowicz CH, Simmons JD, Hannah L, Wanjalla CN, Gabriel CL, Mashayekhi M, Bailin SS, Castilho JL, Hasty AH, Koethe JR, Kalams SA. Leptin Promotes Greater Ki67 Expression in CD4 + T Cells From Obese Compared to Lean Persons Living With HIV. Front Immunol 2022; 12:796898. [PMID: 35111163 PMCID: PMC8801429 DOI: 10.3389/fimmu.2021.796898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/16/2021] [Indexed: 11/13/2022] Open
Abstract
While antiretroviral therapy (ART) has proven effective in suppressing viremia and disease progression among people living with human immunodeficiency virus (HIV; PLWH), suboptimal CD4+ T cell reconstitution remains a major obstacle in nearly 30% of ART-treated individuals. Epidemiological studies demonstrate that obesity, or a body mass index (BMI) ≥ 30 kg/m2, is positively correlated with greater CD4+ T cell recovery in PLWH on ART. Leptin is a known immunomodulator that is produced in proportion to fat mass and is increased in obese individuals, including PLWH. We hypothesized that CD4+ T cells from obese PLWH have increased cell proliferation and cytokine production compared to cells from lean PLWH, potentially modulated by differential effects of leptin signaling. To test this hypothesis, peripheral blood mononuclear cells from obese and lean PLWH with long-term virologic suppression on the same ART regimen were pretreated with recombinant leptin and then stimulated with anti-CD3/CD28 or PMA/ionomycin to measure Ki67 expression, leptin receptor (LepR) surface expression and cytokine production. In the absence of leptin, Ki67 expression and IL-17A production were significantly higher in CD4+ T cells from obese compared to lean PLWH. However, LepR expression was significantly lower on CD4+ T cells from obese compared to lean PLWH. After leptin treatment, Ki67 expression was significantly increased in CD4+ T cells from obese PLWH compared to the lean participants. Leptin also increased IL-17A production in CD4+ T cells from obese healthy controls. In contrast, leptin decreased IL-17A production in CD4+ T cells from both obese and lean PLWH. Combined, these results demonstrate that obesity is associated with greater CD4+ T cell proliferation among PLWH, and that higher circulating leptin levels in obesity may contribute to improved CD4+ T reconstitution in PLWH initiating ART.
Collapse
Affiliation(s)
- Hubaida Fuseini
- Divison of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Rita Smith
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Cindy H. Nochowicz
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Joshua D. Simmons
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, United States
| | - LaToya Hannah
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Celestine N. Wanjalla
- Divison of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Curtis L. Gabriel
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, United States
- Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Mona Mashayekhi
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Samuel S. Bailin
- Divison of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jessica L. Castilho
- Divison of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Alyssa H. Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, United States
- The Veterans Affairs Tennessee Healthcare System, Nashville, TN, United States
| | - John R. Koethe
- Divison of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, United States
- The Veterans Affairs Tennessee Healthcare System, Nashville, TN, United States
| | - Spyros A. Kalams
- Divison of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States
- Tennessee Center for AIDS Research, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
27
|
Cornejo MA, Ortiz RM. Body mass cycling and predictors of body mass regain and its impact on cardiometabolic health. Metabolism 2021; 125:154912. [PMID: 34648770 DOI: 10.1016/j.metabol.2021.154912] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/31/2021] [Accepted: 10/06/2021] [Indexed: 02/08/2023]
Abstract
Caloric restriction (CR) is the first line intervention to reduce adiposity and total body mass (BM) to improve insulin resistance and ameliorate metabolic derangements. However, the lost adipose mass is difficult to maintain reduced in the long term due to several factors including compensatory changes in orexigenic hormones, adipokine release, pro-inflammatory state, adipose tissue morphology, and resting metabolic rate as a consequence of the caloric deficit. Hence, most patients undergoing a BM reduction intervention ultimately regain the lost mass and too often additional adipose mass overtime, which is hypothesized to have increased deleterious effects chronically. In this mini-review we describe the effects of BM cycling (loss and regain) on insulin resistance and cardiometabolic health and factors that may predict BM regain in clinical studies. We also describe the factors that contribute to the chronic deleterious effects of BM cycling in rodent models of diet-induced obesity (DIO) and other metabolic defects. We conclude that most of the improvements in insulin resistance are observed after a profound loss in BM regardless of the diet and that BM cycling abrogates these beneficial effects. We also suggest that more BM cycling studies are needed in rodent models resembling the development of type 2 diabetes mellitus (T2DM) in humans.
Collapse
Affiliation(s)
- Manuel A Cornejo
- Department of Molecular & Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States of America.
| | - Rudy M Ortiz
- Department of Molecular & Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States of America
| |
Collapse
|
28
|
Diaz-Ruiz A, Rhinesmith T, Pomatto-Watson LCD, Price NL, Eshaghi F, Ehrlich MR, Moats JM, Carpenter M, Rudderow A, Brandhorst S, Mattison JA, Aon MA, Bernier M, Longo VD, de Cabo R. Diet composition influences the metabolic benefits of short cycles of very low caloric intake. Nat Commun 2021; 12:6463. [PMID: 34753921 PMCID: PMC8578605 DOI: 10.1038/s41467-021-26654-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 10/14/2021] [Indexed: 12/14/2022] Open
Abstract
Diet composition, calories, and fasting times contribute to the maintenance of health. However, the impact of very low-calorie intake (VLCI) achieved with either standard laboratory chow (SD) or a plant-based fasting mimicking diet (FMD) is not fully understood. Here, using middle-aged male mice we show that 5 months of short 4:10 VLCI cycles lead to decreases in both fat and lean mass, accompanied by improved physical performance and glucoregulation, and greater metabolic flexibility independent of diet composition. A long-lasting metabolomic reprograming in serum and liver is observed in mice on VLCI cycles with SD, but not FMD. Further, when challenged with an obesogenic diet, cycles of VLCI do not prevent diet-induced obesity nor do they elicit a long-lasting metabolic memory, despite achieving modest metabolic flexibility. Our results highlight the importance of diet composition in mediating the metabolic benefits of short cycles of VLCI.
Collapse
Affiliation(s)
- Alberto Diaz-Ruiz
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA.
- Nutritional Interventions Group, Precision Nutrition and Aging, Institute IMDEA Food, Crta. de Canto Blanco n° 8, E - 28049, Madrid, Spain.
| | - Tyler Rhinesmith
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Laura C D Pomatto-Watson
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Nathan L Price
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Farzin Eshaghi
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Margaux R Ehrlich
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Jacqueline M Moats
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Melissa Carpenter
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Annamaria Rudderow
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Sebastian Brandhorst
- Longevity Institute, School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA, 90089, USA
| | - Julie A Mattison
- Nonhuman Primate Core, Translational Gerontology Branch, National Institutes of Health, National Institute on Aging, Dickerson, MD, 20842, USA
| | - Miguel A Aon
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Michel Bernier
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Valter D Longo
- Longevity Institute, School of Gerontology, and Department of Biological Sciences, University of Southern California, Los Angeles, CA, 90089, USA
- IFOM, FIRC Institute of Molecular Oncology, 20139, Milano, Italy
| | - Rafael de Cabo
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA.
- Nutritional Interventions Group, Precision Nutrition and Aging, Institute IMDEA Food, Crta. de Canto Blanco n° 8, E - 28049, Madrid, Spain.
| |
Collapse
|
29
|
Le Garf S, Sibille B, Mothe-Satney I, Eininger C, Fauque P, Murdaca J, Chinetti G, Neels JG, Rousseau AS. Alpha-lipoic acid supplementation increases the efficacy of exercise- and diet-induced obesity treatment and induces immunometabolic changes in female mice and women. FASEB J 2021; 35:e21312. [PMID: 33742689 DOI: 10.1096/fj.202001817rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 01/29/2023]
Abstract
The decrease in the regulatory T cells (Tregs) population is highly involved in adipose tissue inflammation and insulin resistance in obesity. Tregs depend on fatty acids via β-oxidation for immunosuppressive function adapting their antioxidant systems to allow survival to oxidative stress. In this study, we have hypothesized that a dietary supplementation with alpha-lipoic acid (ALA), a powerful antioxidant, would improve immunometabolism when added to the classical strategy of obesity treatment. First, we showed by in vitro experiments that ALA favors the polarization of mice CD4 + T cells toward Tregs. Next, we have carried out a translational study where female obese mice and women were supplemented with ALA or vehicle/placebo (mice: 2.5 gALA /kgfood ; 6 weeks; women: 600 mgALA /day, 8 weeks) while following a protocol including regular exercise and a change in diet. Fatty acid oxidation potential and activity of nuclear erythroid-related factor 2 (NRF2) of mouse secondary lymphoid tissues were improved by ALA supplementation. ALA reduced visceral adipose tissue (VAT) mass and preserved Tregs in VAT in mice. In women, ALA supplementation induced significant metabolic changes of circulating CD4 + T cells including increased oxidative capacity and fatty acid oxidation, ameliorated their redox status, and improved the reduction of visceral fat mass. While appropriate biological markers are still required to be used in clinics to judge the effectiveness of long-term obesity treatment, further studies in female mice and women are needed to determine whether these immunometabolic changes would reduce VAT mass-associated risk for secondary health issues arising from obesity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jaap G Neels
- INSERM, C3M, Université Côte d'Azur, Nice, France
| | | |
Collapse
|
30
|
AlZaim I, Hammoud SH, Al-Koussa H, Ghazi A, Eid AH, El-Yazbi AF. Adipose Tissue Immunomodulation: A Novel Therapeutic Approach in Cardiovascular and Metabolic Diseases. Front Cardiovasc Med 2020; 7:602088. [PMID: 33282920 PMCID: PMC7705180 DOI: 10.3389/fcvm.2020.602088] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue is a critical regulator of systemic metabolism and bodily homeostasis as it secretes a myriad of adipokines, including inflammatory and anti-inflammatory cytokines. As the main storage pool of lipids, subcutaneous and visceral adipose tissues undergo marked hypertrophy and hyperplasia in response to nutritional excess leading to hypoxia, adipokine dysregulation, and subsequent low-grade inflammation that is characterized by increased infiltration and activation of innate and adaptive immune cells. The specific localization, physiology, susceptibility to inflammation and the heterogeneity of the inflammatory cell population of each adipose depot are unique and thus dictate the possible complications of adipose tissue chronic inflammation. Several lines of evidence link visceral and particularly perivascular, pericardial, and perirenal adipose tissue inflammation to the development of metabolic syndrome, insulin resistance, type 2 diabetes and cardiovascular diseases. In addition to the implication of the immune system in the regulation of adipose tissue function, adipose tissue immune components are pivotal in detrimental or otherwise favorable adipose tissue remodeling and thermogenesis. Adipose tissue resident and infiltrating immune cells undergo metabolic and morphological adaptation based on the systemic energy status and thus a better comprehension of the metabolic regulation of immune cells in adipose tissues is pivotal to address complications of chronic adipose tissue inflammation. In this review, we discuss the role of adipose innate and adaptive immune cells across various physiological and pathophysiological states that pertain to the development or progression of cardiovascular diseases associated with metabolic disorders. Understanding such mechanisms allows for the exploitation of the adipose tissue-immune system crosstalk, exploring how the adipose immune system might be targeted as a strategy to treat cardiovascular derangements associated with metabolic dysfunctions.
Collapse
Affiliation(s)
- Ibrahim AlZaim
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Safaa H. Hammoud
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon
| | - Houssam Al-Koussa
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
| | - Alaa Ghazi
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
| | - Ali H. Eid
- Department of Pharmacology and Therapeutics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Basic Medical Sciences, College of Medicine, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Ahmed F. El-Yazbi
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
31
|
Transcriptome Analysis of Testis from HFD-Induced Obese Rats ( Rattus norvigicus) Indicated Predisposition for Male Infertility. Int J Mol Sci 2020; 21:ijms21186493. [PMID: 32899471 PMCID: PMC7554891 DOI: 10.3390/ijms21186493] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/23/2020] [Accepted: 08/27/2020] [Indexed: 12/13/2022] Open
Abstract
Obesity is a worldwide life-threatening metabolic disorder, associated with various chronic diseases, including male infertility. Obesity was induced by high fat diet (HFD), and testis RNA was used for the transcriptome analysis using RNAseq via Illumina NovaSeq 6000 System and NovaSeq 6000 Kit. Gene expression level was estimated as FPKM (Fragments Per Kilobase of transcript per Million mapped reads). Differential expressed genes (DEGs) were annotated against gene ontology (GO) and KEGG databases. More than 63.66 million reads per sample were performed with 100 bp cutoff and 6 Gb sequencing depth. Results of this study revealed that 267 GO terms (245 biological processes (BP), 14 cellular components (CC), eight molecular functions (MF)), and 89 KEGG pathways were significantly enriched. Moreover, total numbers of 136 genes were differentially expressed (107 upregulated, 29 downregulated) with |FC| ≥ 2 and bh adjusted <0.05. Interesting DEGs were detected, including obesity and lipid metabolism-related genes, immune response-related genes, cytochrome P450 genes, including aromatase were upregulated, whereas genes related to male fertility and fertilization, cell adhesion, and olfactory receptors were downregulated. The combined expression pattern of the DEGs in obese animals indicated an increase in cholesterol metabolism. Furthermore, high aromatase activity enhances the testosterone turnover into estradiol and lowers the testosterone/estradiol (T/E) ratio, which ultimately reduces fertility. In addition, downregulation of cadherens junction components genes leads to the pre-mature release of sperm from Sertoli cells resulting in the reduction of fertility. Moreover, the downregulation of olfactory receptor genes reduces the chemotaxis capacity of sperms in tracking the oocyte for fertilization, which reduces male fertility. Furthermore, various obesity molecular markers were detected in our transcriptome. The results of this study will enhance our understanding of the molecular network of obesity development, development of obesity novel molecular diagnosis markers, molecular bases of obesity-induced infertility, and the development of anti-obesity drugs.
Collapse
|
32
|
Sbierski-Kind J, Mai K, Kath J, Jurisch A, Streitz M, Kuchenbecker L, Babel N, Nienen M, Jürchott K, Spranger L, Jumpertz von Schwartzenberg R, Decker AM, Krüger U, Volk HD, Spranger J. Association between Subcutaneous Adipose Tissue Inflammation, Insulin Resistance, and Calorie Restriction in Obese Females. THE JOURNAL OF IMMUNOLOGY 2020; 205:45-55. [PMID: 32482712 DOI: 10.4049/jimmunol.2000108] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/21/2020] [Indexed: 01/30/2023]
Abstract
The worldwide epidemic of overweight and obesity has led to an increase in associated metabolic comorbidities. Obesity induces chronic low-grade inflammation in white adipose tissue (WAT). However, the function and regulation of both innate and adaptive immune cells in human WAT under conditions of obesity and calorie restriction (CR) is not fully understood yet. Using a randomized interventional design, we investigated postmenopausal overweight or obese female subjects who either underwent CR for 3 mo followed by a 4-wk phase of weight maintenance or had to maintain a stable weight over the whole study period. A comprehensive immune phenotyping protocol was conducted using validated multiparameter flow cytometry analysis in blood and s.c. WAT (SAT). The TCR repertoire was analyzed by next-generation sequencing and cytokine levels were determined in SAT. Metabolic parameters were determined by hyperinsulinemic-euglycemic clamp. We found that insulin resistance correlates significantly with a shift toward the memory T cell compartment in SAT. TCR analysis revealed a diverse repertoire in SAT of overweight or obese individuals. Additionally, whereas weight loss improved systemic insulin sensitivity in the intervention group, SAT displayed no significant improvement of inflammatory parameters (cytokine levels and leukocyte subpopulations) compared with the control group. Our data demonstrate the accumulation of effector memory T cells in obese SAT and an association between systemic glucose homeostasis and inflammatory parameters in obese females. The long-standing effect of obesity-induced changes in SAT was demonstrated by preserved immune cell composition after short-term CR-induced weight loss.
Collapse
Affiliation(s)
- Julia Sbierski-Kind
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Department of Endocrinology and Metabolism, Berlin Institute of Health, 10178 Berlin, Germany; .,Berlin Institute of Health, 10178 Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité University Medicine Berlin, 10117 Berlin, Germany.,Institute for Medical Immunology, Charité University Medicine Berlin, 10117 Berlin, Germany
| | - Knut Mai
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Department of Endocrinology and Metabolism, Berlin Institute of Health, 10178 Berlin, Germany.,Berlin Institute of Health, 10178 Berlin, Germany.,Charité - Center for Cardiovascular Research, 10117 Berlin, Germany
| | - Jonas Kath
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Department of Endocrinology and Metabolism, Berlin Institute of Health, 10178 Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité University Medicine Berlin, 10117 Berlin, Germany
| | - Anke Jurisch
- Berlin Institute of Health Center for Regenerative Therapies, Charité University Medicine Berlin, 10117 Berlin, Germany.,Institute for Medical Immunology, Charité University Medicine Berlin, 10117 Berlin, Germany
| | - Mathias Streitz
- Berlin Institute of Health Center for Regenerative Therapies, Charité University Medicine Berlin, 10117 Berlin, Germany.,Institute for Medical Immunology, Charité University Medicine Berlin, 10117 Berlin, Germany
| | - Leon Kuchenbecker
- Berlin Institute of Health Center for Regenerative Therapies, Charité University Medicine Berlin, 10117 Berlin, Germany.,Institute for Medical Immunology, Charité University Medicine Berlin, 10117 Berlin, Germany
| | - Nina Babel
- Berlin Institute of Health Center for Regenerative Therapies, Charité University Medicine Berlin, 10117 Berlin, Germany.,Center for Translational Medicine, Department of Internal Medicine I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, 44625 Bochum, Germany
| | - Mikalai Nienen
- Berlin Institute of Health, 10178 Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité University Medicine Berlin, 10117 Berlin, Germany.,Center for Translational Medicine, Department of Internal Medicine I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, 44625 Bochum, Germany
| | - Karsten Jürchott
- Berlin Institute of Health Center for Regenerative Therapies, Charité University Medicine Berlin, 10117 Berlin, Germany.,Institute for Medical Immunology, Charité University Medicine Berlin, 10117 Berlin, Germany
| | - Leonard Spranger
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Department of Endocrinology and Metabolism, Berlin Institute of Health, 10178 Berlin, Germany.,Charité - Center for Cardiovascular Research, 10117 Berlin, Germany
| | - Reiner Jumpertz von Schwartzenberg
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Department of Endocrinology and Metabolism, Berlin Institute of Health, 10178 Berlin, Germany.,Berlin Institute of Health, 10178 Berlin, Germany.,Charité - Center for Cardiovascular Research, 10117 Berlin, Germany.,German Center for Cardiovascular Research, partner site Berlin, 13353 Berlin, Germany; and
| | - Anne-Marie Decker
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Department of Endocrinology and Metabolism, Berlin Institute of Health, 10178 Berlin, Germany.,Berlin Institute of Health, 10178 Berlin, Germany
| | - Ulrike Krüger
- Berlin Institute of Health Center for Regenerative Therapies, Charité University Medicine Berlin, 10117 Berlin, Germany.,Institute for Medical Immunology, Charité University Medicine Berlin, 10117 Berlin, Germany
| | - Hans-Dieter Volk
- Berlin Institute of Health Center for Regenerative Therapies, Charité University Medicine Berlin, 10117 Berlin, Germany.,Institute for Medical Immunology, Charité University Medicine Berlin, 10117 Berlin, Germany.,Berlin Center for Advanced Therapies, Charité University Medicine Berlin, 10117 Berlin, Germany
| | - Joachim Spranger
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Department of Endocrinology and Metabolism, Berlin Institute of Health, 10178 Berlin, Germany.,Berlin Institute of Health, 10178 Berlin, Germany.,Charité - Center for Cardiovascular Research, 10117 Berlin, Germany.,German Center for Cardiovascular Research, partner site Berlin, 13353 Berlin, Germany; and
| |
Collapse
|
33
|
Sudhakaran M, Doseff AI. The Targeted Impact of Flavones on Obesity-Induced Inflammation and the Potential Synergistic Role in Cancer and the Gut Microbiota. Molecules 2020; 25:E2477. [PMID: 32471061 PMCID: PMC7321129 DOI: 10.3390/molecules25112477] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/21/2020] [Accepted: 05/23/2020] [Indexed: 12/19/2022] Open
Abstract
Obesity is an inflammatory disease that is approaching pandemic levels, affecting nearly 30% of the world's total population. Obesity increases the risk of diabetes, cardiovascular disorders, and cancer, consequentially impacting the quality of life and imposing a serious socioeconomic burden. Hence, reducing obesity and related life-threatening conditions has become a paramount health challenge. The chronic systemic inflammation characteristic of obesity promotes adipose tissue remodeling and metabolic changes. Macrophages, the major culprits in obesity-induced inflammation, contribute to sustaining a dysregulated immune function, which creates a vicious adipocyte-macrophage crosstalk, leading to insulin resistance and metabolic disorders. Therefore, targeting regulatory inflammatory pathways has attracted great attention to overcome obesity and its related conditions. However, the lack of clinical efficacy and the undesirable side-effects of available therapeutic options for obesity provide compelling reasons for the need to identify additional approaches for the prevention and treatment of obesity-induced inflammation. Plant-based active metabolites or nutraceuticals and diets with an increased content of these compounds are emerging as subjects of intense scientific investigation, due to their ability to ameliorate inflammatory conditions and offer safe and cost-effective opportunities to improve health. Flavones are a class of flavonoids with anti-obesogenic, anti-inflammatory and anti-carcinogenic properties. Preclinical studies have laid foundations by establishing the potential role of flavones in suppressing adipogenesis, inducing browning, modulating immune responses in the adipose tissues, and hindering obesity-induced inflammation. Nonetheless, the understanding of the molecular mechanisms responsible for the anti-obesogenic activity of flavones remains scarce and requires further investigations. This review recapitulates the molecular aspects of obesity-induced inflammation and the crosstalk between adipocytes and macrophages, while focusing on the current evidence on the health benefits of flavones against obesity and chronic inflammation, which has been positively correlated with an enhanced cancer incidence. We conclude the review by highlighting the areas of research warranting a deeper investigation, with an emphasis on flavones and their potential impact on the crosstalk between adipocytes, the immune system, the gut microbiome, and their role in the regulation of obesity.
Collapse
Affiliation(s)
- Meenakshi Sudhakaran
- Physiology Graduate Program, Michigan State University, East Lansing, MI 48824, USA;
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Andrea I. Doseff
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
34
|
Caslin HL, Bhanot M, Bolus WR, Hasty AH. Adipose tissue macrophages: Unique polarization and bioenergetics in obesity. Immunol Rev 2020; 295:101-113. [PMID: 32237081 PMCID: PMC8015437 DOI: 10.1111/imr.12853] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/25/2020] [Accepted: 02/28/2020] [Indexed: 12/13/2022]
Abstract
Macrophages comprise a majority of the resident immune cells in adipose tissue (AT) and regulate both tissue homeostasis in the lean state and metabolic dysregulation in obesity. Since the AT environment rapidly changes based upon systemic energy status, AT macrophages (ATMs) must adapt phenotypically and metabolically. There is a distinct dichotomy in the polarization and bioenergetics of in vitro models, with M2 macrophages utilizing oxidative phosphorylation (OX PHOS) and M1 macrophages utilizing glycolysis. Early studies suggested differential polarization of ATMs, with M2-like macrophages predominant in lean AT and M1-like macrophages in obese AT. However, recent studies show that the phenotypic plasticity of ATMs is far more complicated, which is also reflected in their bioenergetics. Multiple ATM populations exist along the M2 to M1 continuum and appear to utilize both glycolysis and OX PHOS in obesity. The significance of the dual fuel bioenergetics is unclear and may be related to an intermediate polarization, their buffering capacity, or the result of a mixed population of distinct polarized ATMs. Recent evidence also suggests that ATMs of lean mice serve as a substrate buffer or reservoir to modulate lipid, catecholamine, and iron availability. Furthermore, recent models of weight loss and weight cycling reveal additional roles for ATMs in systemic metabolism. Evaluating ATM phenotype and intracellular metabolism together may more accurately illuminate the consequences of ATM accumulation in obese AT, lending further insight into obesity-related comorbidities in humans.
Collapse
Affiliation(s)
- Heather L Caslin
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Monica Bhanot
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt Medical Center, Nashville, TN, USA
| | - W Reid Bolus
- Diabetes Center, University of California San Francisco, San Francisco, CA, USA
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
- VA Tennessee Valley Healthcare System, Nashville, TN, USA
| |
Collapse
|
35
|
The role of immune cells in obesogenic memory. Cell Mol Immunol 2020; 17:884-886. [PMID: 32346101 DOI: 10.1038/s41423-020-0448-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 04/12/2020] [Indexed: 01/12/2023] Open
|
36
|
Surendar J, Karunakaran I, Frohberger SJ, Koschel M, Hoerauf A, Hübner MP. Macrophages Mediate Increased CD8 T Cell Inflammation During Weight Loss in Formerly Obese Mice. Front Endocrinol (Lausanne) 2020; 11:257. [PMID: 32411095 PMCID: PMC7198814 DOI: 10.3389/fendo.2020.00257] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 04/06/2020] [Indexed: 12/27/2022] Open
Abstract
Even after successful weight reduction, obese adults tend to quickly regain the lost weight. This raises the question of whether weight loss improves the underlying chronic adipose tissue inflammation characteristic of obesity. In order to improve our understanding of the mechanisms that reshape metabolic organs during weight loss, we investigated the macrophage and T cell function of the liver and adipose tissue on reversing high fat diet (HFD) mice to normal control diet (NCD). Obese mice that were switched to NCD showed an improvement in their metabolic profile that included enhanced glucose and insulin tolerance, decreased cholesterol, triglyceride, serum glutamic-oxaloacetic transaminase (SGOT), and serum glutamic pyruvic transaminase (SGPT) levels that were comparable to NCD controls. However, despite weight loss, increased frequencies, but not total numbers, of IL-17+ and IL-22+ CD4+ T cells, IFN-γ+ and TNF+ CD8+ T cells and IL-17+ and IL-22+ CD8+ T cells were observed in the adipose tissue of mice switched from HFD to NCD compared to NCD and even HFD fed mice. Further, in the liver, IFN-γ+ and TNF+ CD8+ T cell, IL-17+ and IL-22+ CD8+ T cell, macrophage frequencies and their expression of antigen presenting molecules were increased. To determine if macrophages are the major determinants of the sustained inflammation observed during weight loss, we depleted macrophages, which significantly reduced IFN-γ+, TNF+, IL-17+, and IL-22+ CD8+ T cell frequencies in the liver and the adipose tissue. In conclusion, we show that although weight loss improves the metabolic profile, there is an active and ongoing CD8+ T cell inflammation in liver and adipose tissue mediated by macrophages.
Collapse
Affiliation(s)
- Jayagopi Surendar
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Indulekha Karunakaran
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Stefan J. Frohberger
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Marianne Koschel
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Achim Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Marc P. Hübner
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- *Correspondence: Marc P. Hübner
| |
Collapse
|
37
|
Chen L, Shen Z. Tissue-resident memory T cells and their biological characteristics in the recurrence of inflammatory skin disorders. Cell Mol Immunol 2019; 17:64-75. [PMID: 31595056 DOI: 10.1038/s41423-019-0291-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 08/25/2019] [Indexed: 11/09/2022] Open
Abstract
The skin is the largest organ of the body. The establishment of immunological memory in the skin is a crucial component of the adaptive immune response. Once naive T cells are activated by antigen-presenting cells, a small fraction of them differentiate into precursor memory T cells. These precursor cells ultimately develop into several subsets of memory T cells, including central memory T (TCM) cells, effector memory T (TEM) cells, and tissue resident memory T (TRM) cells. TRM cells have a unique transcriptional profile, and their most striking characteristics are their long-term survival (longevity) and low migration in peripheral tissues, including the skin. Under physiological conditions, TRM cells that reside in the skin can respond rapidly to pathogenic challenges. However, there is emerging evidence to support the vital role of TRM cells in the recurrence of chronic inflammatory skin disorders, including psoriasis, vitiligo, and fixed drug eruption, under pathological or uncontrolled conditions. Clarifying and characterizing the mechanisms that are involved in skin TRM cells will help provide promising strategies for reducing the frequency and magnitude of skin inflammation recurrence. Here, we discuss recent insights into the generation, homing, retention, and survival of TRM cells and share our perspectives on the biological characteristics of TRM cells in the recurrence of inflammatory skin disorders.
Collapse
Affiliation(s)
- Ling Chen
- Department of Dermatology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Zhu Shen
- Department of Dermatology, Institute of Dermatology and Venereology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital; School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
38
|
Caslin HL, Hasty AH. Extrinsic and Intrinsic Immunometabolism Converge: Perspectives on Future Research and Therapeutic Development for Obesity. Curr Obes Rep 2019; 8:210-219. [PMID: 30919312 PMCID: PMC6661206 DOI: 10.1007/s13679-019-00344-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Research over the past decade has shown that immunologic and metabolic pathways are intricately linked. This burgeoning field of immunometabolism includes intrinsic and extrinsic pathways and is known to be associated with obesity-accelerated metabolic disease. Intrinsic immunometabolism includes the study of fuel utilization and bioenergetic pathways that influence immune cell function. Extrinsic immunometabolism includes the study of immune cells and products that influence systemic metabolism. RECENT FINDINGS Th2 immunity, macrophage iron handling, adaptive immune memory, and epigenetic regulation of immunity, which all require intrinsic metabolic changes, play a role in systemic metabolism and metabolic function, linking the two arms of immunometabolism. Together, this suggests that targeting intrinsic immunometabolism can directly affect immune function and ultimately systemic metabolism. We highlight important questions for future basic research that will help improve translational research and provide therapeutic targets to help establish new treatments for obesity and associated metabolic disorders.
Collapse
Affiliation(s)
- Heather L Caslin
- Molecular Physiology and Biophysics, Vanderbilt University, 813 Light Hall, 23rd Ave. South and Pierce, Nashville, TN, 37232, USA
| | - Alyssa H Hasty
- Molecular Physiology and Biophysics, Vanderbilt University, 813 Light Hall, 23rd Ave. South and Pierce, Nashville, TN, 37232, USA.
- VA Tennessee Valley Healthcare System, Nashville, TN, USA.
| |
Collapse
|
39
|
Kasher-Meron M, Youn DY, Zong H, Pessin JE. Lipolysis defect in white adipose tissue and rapid weight regain. Am J Physiol Endocrinol Metab 2019; 317:E185-E193. [PMID: 30964706 PMCID: PMC6732460 DOI: 10.1152/ajpendo.00542.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/07/2019] [Accepted: 04/03/2019] [Indexed: 11/22/2022]
Abstract
Weight regain after weight loss is a well-described phenomenon in both humans and animal models of obesity. Reduced energy expenditure and increased caloric intake are considered the main drivers of weight regain. We hypothesized that adipose tissue with obesity memory (OM) has a tissue-autonomous lipolytic defect, allowing for increased efficiency of lipid storage. We utilized a mouse model of diet-induced obesity, which was subjected to 60% caloric restriction to achieve lean body weight, followed by a short period of high-fat diet (HFD) rechallenge. Age-matched lean mice fed HFD for the first time were used as the control group. Upon rechallenge with HFD, mice with OM had higher respiratory exchange ratios than lean mice with no OM despite comparable body weight, suggesting higher utilization of glucose over fatty acid oxidation. White adipose tissue explants with OM had comparable lipolytic response after caloric restriction; however, reduced functional lipolytic response to norepinephrine was noted as early as 5 days after rechallenge with HFD and was accompanied by reduction in hormone-sensitive lipase serine phosphorylation. The relative lipolytic defect was associated with increased expression of inflammatory genes and a decrease in adrenergic receptor genes, most notably Adrb3. Taken together, white adipose tissue of lean mice with OM shows increased sensitization to HFD compared with white adipose tissue with no OM, rendering it resistant to catecholamine-induced lipolysis. This relative lipolytic defect is tissue-autonomous and could play a role in the rapid weight regain observed after weight loss.
Collapse
Affiliation(s)
- Michal Kasher-Meron
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Dou Y Youn
- Department of Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| | - Haihong Zong
- Department of Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| | - Jeffery E Pessin
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
- Department of Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
40
|
Abstract
One of the biggest challenges in the management of obesity is the prevention of weight regain after successful weight loss. Weight regain after weight loss has large interindividual variation. Although many factors probably contribute to this variation, we hypothesize that variability in biological responses associated with weight loss-induced shrinking of subcutaneous adipocytes has an important role. In this Review, we show that weight loss-induced variations in cellular stress, extracellular matrix remodelling, inflammatory responses, adipokine secretion and lipolysis seem to be associated with the amount of weight that is regained after successful weight loss. Weight regain could therefore, at least in part, depend on a combination of these factors. Further research on the causality of these associations could aid the development of effective strategies to prevent weight regain after successful weight loss.
Collapse
Affiliation(s)
- Marleen A van Baak
- NUTRIM School for Nutrition and Translational Research in Metabolism, Department of Human Biology, Maastricht University, Maastricht, Netherlands.
| | - Edwin C M Mariman
- NUTRIM School for Nutrition and Translational Research in Metabolism, Department of Human Biology, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
41
|
Cottam MA, Itani HA, Beasley AA, Hasty AH. Links between Immunologic Memory and Metabolic Cycling. THE JOURNAL OF IMMUNOLOGY 2019; 200:3681-3689. [PMID: 29784764 DOI: 10.4049/jimmunol.1701713] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/07/2018] [Indexed: 12/13/2022]
Abstract
Treatments for metabolic diseases, such as diet and therapeutics, often provide short-term therapy for metabolic stressors, but relapse is common. Repeated bouts of exposure to, and relief from, metabolic stimuli results in a phenomenon we call "metabolic cycling." Recent human and rodent data suggest metabolic cycling promotes an exaggerated response and ultimately worsened metabolic health. This is particularly evident with cycling of body weight and hypertension. The innate and adaptive immune systems have a profound impact on development of metabolic disease, and current data suggest that immunologic memory may partially explain this association, especially in the context of metabolic cycling. In this Brief Review, we highlight recent work in this field and discuss potential immunologic mechanisms for worsened disease prognosis in individuals who experience metabolic cycling.
Collapse
Affiliation(s)
- Matthew A Cottam
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, TN 37232
| | - Hana A Itani
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232.,Faculty of Medicine, American University of Beirut, Beirut, Lebanon; and
| | - Arch A Beasley
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, TN 37232
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, School of Medicine, Vanderbilt University, Nashville, TN 37232; .,Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN 37232
| |
Collapse
|
42
|
Spirulina supplementation in a mouse model of diet-induced liver fibrosis reduced the pro-inflammatory response of splenocytes. Br J Nutr 2019; 121:748-755. [PMID: 30806344 DOI: 10.1017/s0007114519000126] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Treatment of liver fibrosis is very limited as there is currently no effective anti-fibrotic therapy. Spirulina platensis (SP) is a blue-green alga that is widely supplemented in healthy foods. The objective of this study was to determine whether SP supplementation can prevent obesity-induced liver fibrosis in vivo. Male C57BL/6J mice were randomly assigned to a low-fat or a high-fat (HF)/high-sucrose/high-cholesterol diet or an HF diet supplemented with 2·5 % SP (w/w) (HF/SP) for 16 or 20 weeks. There were no significant differences in body weight, activity, energy expenditure, serum lipids or glucose tolerance between mice on HF and HF/SP diets. However, plasma alanine aminotransferase level was significantly reduced by SP at 16 weeks. Expression of fibrotic markers and trichrome stains showed no differences between HF and HF/SP. Splenocytes isolated from HF/SP fed mice had lower inflammatory gene expression and cytokine secretion compared with splenocytes from HF-fed mice. SP supplementation did not attenuate HF-induced liver fibrosis. However, the expression and secretion of inflammatory genes in splenocytes were significantly reduced by SP supplementation, demonstrating the anti-inflammatory effects of SP in vivo. Although SP did not show appreciable effect on the prevention of liver fibrosis in this mouse model, it may be beneficial for other inflammatory conditions.
Collapse
|
43
|
MicroRNA-99a mimics inhibit M1 macrophage phenotype and adipose tissue inflammation by targeting TNFα. Cell Mol Immunol 2018; 16:495-507. [PMID: 29849090 DOI: 10.1038/s41423-018-0038-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 04/21/2018] [Accepted: 04/21/2018] [Indexed: 01/19/2023] Open
Abstract
In human adipose tissue and obesity, miR-99a expression is negatively correlated with inflammation. Therefore, the present study investigated the role of miR-99a in macrophage phenotype activation and adipose tissue inflammation. M2 BMDMs showed a significant increase in miR-99a expression when compared to the M0 and M1 phenotypes. Phenotype-switching experiments established an association between upregulated miR-99a expression and the M2 phenotype. Overexpression of miR-99a prevented M1 phenotype activation and attenuated bactericidal activity. Likewise, knockdown of miR-99a abolished M2 phenotype activation. By means of in silico target prediction tools and a luciferase reporter assay, TNFα was identified as a direct target of miR-99a. Knockdown of TNFα recapitulated the effect of miR-99a overexpression in M1 BMDMs. In a db/db mice model, miR-99a expression was reduced in eWAT and F4/80+ ATMs. Systemic overexpression of miR-99a in db/db mice attenuated adipocyte hypertrophy with increased CD301 and reduced CD86 immunostaining. Flow cytometry analysis also showed an increased M2 and a reduced M1 macrophage population. Mimics of miR-99a also improved the diabetic dyslipidemia and insulin signaling in eWAT and liver, with an attenuated expression of gluconeogenesis and cholesterol metabolism genes in the liver. Furthermore, adoptive transfer of miR-99a-overexpressing macrophages in the db/db mice recapitulated in vivo miR-99a mimic effects with increased M2 and reduced M1 macrophage populations and improved systemic glucose, insulin sensitivity, and insulin signaling in the eWAT and liver. The present study demonstrates that miR-99a mimics can regulate macrophage M1 phenotype activation by targeting TNFα. miR-99a therapeutics in diabetic mice reduces the adipose tissue inflammation and improves insulin sensitivity.
Collapse
|