1
|
Kong Y, Yang H, Nie R, Zhang X, Zuo F, Zhang H, Nian X. Obesity: pathophysiology and therapeutic interventions. MOLECULAR BIOMEDICINE 2025; 6:25. [PMID: 40278960 PMCID: PMC12031720 DOI: 10.1186/s43556-025-00264-9] [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: 09/04/2024] [Revised: 03/15/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Over the past few decades, obesity has transitioned from a localized health concern to a pressing global public health crisis affecting over 650 million adults globally, as documented by WHO epidemiological surveys. As a chronic metabolic disorder characterized by pathological adipose tissue expansion, chronic inflammation, and neuroendocrine dysregulation that disrupts systemic homeostasis and impairs physiological functions, obesity is rarely an isolated condition; rather, it is frequently complicated by severe comorbidities that collectively elevate mortality risks. Despite advances in nutritional science and public health initiatives, sustained weight management success rates and prevention in obesity remain limited, underscoring its recognition as a multifactorial disease influenced by genetic, environmental, and behavioral determinants. Notably, the escalating prevalence of obesity and its earlier onset in younger populations have intensified the urgency to develop novel therapeutic agents that simultaneously ensure efficacy and safety. This review aims to elucidate the pathophysiological mechanisms underlying obesity, analyze its major complications-including type 2 diabetes mellitus (T2DM), cardiovascular diseases (CVD), non-alcoholic fatty liver disease (NAFLD), obesity-related respiratory disorders, obesity-related nephropathy (ORN), musculoskeletal impairments, malignancies, and psychological comorbidities-and critically evaluate current anti-obesity strategies. Particular emphasis is placed on emerging pharmacological interventions, exemplified by plant-derived natural compounds such as berberine (BBR), with a focus on their molecular mechanisms, clinical efficacy, and therapeutic advantages. By integrating mechanistic insights with clinical evidence, this review seeks to provide innovative perspectives for developing safe, accessible, and effective obesity treatments.
Collapse
Affiliation(s)
- Yue Kong
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | | | - Rong Nie
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xuxiang Zhang
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fan Zuo
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | | | - Xin Nian
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
2
|
Li D, Zhu J, Zhang M, Shi Q, Guo R, Zhang D, Zheng P, Zhang H, Li G, Wu J, Sun G, Wen Q, Tan J, Liu Z, Liu X, Yang H, Lu H, Cao G, Yin Z, Wang Q. SOSTDC1 downregulation in CD4 + T cells confers protection against obesity-induced insulin resistance. Cell Rep 2025; 44:115496. [PMID: 40173040 DOI: 10.1016/j.celrep.2025.115496] [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: 01/30/2024] [Revised: 01/28/2025] [Accepted: 03/10/2025] [Indexed: 04/04/2025] Open
Abstract
Adipose-resident T cells play a crucial role in the development of obesity-induced insulin resistance. However, the specific mechanisms, particularly those involving non-immune cytokines, remain unclear. Here, we report significantly elevated levels of sclerostin domain-containing protein 1 (SOSTDC1) in individuals with type 2 diabetes (T2D), showing positive correlations with fasting glucose and HbA1c. T cell-specific Sostdc1-deficient mice exhibit resistance to age-induced adipose lipid accumulation and glucose dysregulation at 12 months and protect against obesity-induced insulin resistance without affecting proinflammatory macrophage infiltration or adipose inflammation. Mechanistically, SOSTDC1 disrupts the lipid balance in adipocytes by promoting lipogenesis and inhibiting lipolysis through the LRP5/6-β-catenin pathway. Furthermore, T cell receptor (TCR) signaling significantly amplifies SOSTDC1 secretion in CD4+ T cells. In summary, our study uncovers an additional mechanism by which T cells contribute to obesity and insulin resistance, suggesting that inhibiting SOSTDC1 could be a promising immunotherapeutic strategy for metabolic disorders.
Collapse
Affiliation(s)
- Dehai Li
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai 519000, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, China; Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jing Zhu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai 519000, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
| | - Mingyue Zhang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai 519000, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
| | - Qiping Shi
- Department of Endocrinology, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - Rong Guo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, China
| | - Daming Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, China
| | - Pei Zheng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, China
| | - Hua Zhang
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Guangqiang Li
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai 519000, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
| | - Jie Wu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai 519000, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
| | - Guodong Sun
- Department of Orthopedics, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China; Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan 517000, China
| | - Qiong Wen
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai 519000, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
| | - Jingyi Tan
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai 519000, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
| | - Zonghua Liu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai 519000, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
| | - Xindong Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hengwen Yang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai 519000, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China.
| | - Hongyun Lu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai 519000, China.
| | - Guangchao Cao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai 519000, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China.
| | - Zhinan Yin
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai 519000, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China.
| | - Qian Wang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai 519000, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China.
| |
Collapse
|
3
|
Yeom E, Mun H, Lim J, Chun YL, Min KW, Lambert J, Cowart LA, Pierce JS, Ogretmen B, Cho JH, Chang JH, Buchan JR, Pitt J, Kaeberlein M, Kang SU, Kwon ES, Ko S, Choi KM, Lee YS, Ha YS, Kim SJ, Lee KP, Kim HS, Yang SY, Shin CH, Yoon JH, Lee KS. Phosphorylation of an RNA-Binding Protein Rck/Me31b by Hippo Is Essential for Adipose Tissue Aging. Aging Cell 2025:e70022. [PMID: 40070010 DOI: 10.1111/acel.70022] [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: 09/12/2024] [Revised: 01/20/2025] [Accepted: 02/10/2025] [Indexed: 05/15/2025] Open
Abstract
The metazoan lifespan is determined in part by a complex signaling network that regulates energy metabolism and stress responses. Key signaling hubs in this network include insulin/IGF-1, AMPK, mTOR, and sirtuins. The Hippo/Mammalian Ste20-like Kinase1 (MST1) pathway has been reported to maintain lifespan in Caenorhabditis elegans, but its role has not been studied in higher metazoans. In this study, we report that overexpression of Hpo, the MST1 homolog in Drosophila melanogaster, decreased lifespan with concomitant changes in lipid metabolism and aging-associated gene expression, while RNAi Hpo depletion increased lifespan. These effects were mediated primarily by Hpo-induced transcriptional activation of the RNA-binding protein maternal expression at 31B (Me31b)/RCK, resulting in stabilization of mRNA-encoding a lipolytic hormone, Akh. In mouse adipocytes, Hpo/Mst1 mediated adipocyte differentiation, phosphorylation of RNA-binding proteins such as Rck, decapping MRNA 2 (Dcp2), enhancer Of MRNA decapping 3 (Edc3), nucleolin (NCL), and glucagon mRNA stability by interacting with Rck. Decreased lifespan in Hpo-overexpressing Drosophila lines required expression of Me31b, but not DCP2, which was potentially mediated by recovering expression of lipid metabolic genes and formation of lipid droplets. Taken together, our findings suggest that Hpo/Mst1 plays a conserved role in longevity by regulating adipogenesis and fatty acid metabolism.
Collapse
Affiliation(s)
- Eunbyul Yeom
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea
- KNU G-LAMP Project Group, KNU Institute of Basic Sciences, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu, Korea
- Neurophysiology and Metabolism Research Group, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Hyejin Mun
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Oncology Science, University of Oklahoma, Oklahoma City, Oklahoma, USA
| | - Jinhwan Lim
- Department of Environmental and Occupational Heatlh, University of California, Irvine, California, USA
- Translational Gerontology Branch, National Institute of Aging Intramural Research Program, Baltimore, Maryland, USA
| | - Yoo Lim Chun
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kyung-Won Min
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Biology, College of Natural Sciences, Gangneung-Wonju National University, Gangneung, South Korea
| | - Johana Lambert
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
- Hunter Holmes McGuire Veteran's Affairs Medical Center, Richmond, Virginia, USA
| | - L Ashley Cowart
- Department of Biochemistry and Molecular Biology and the Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
- Hunter Holmes McGuire Veteran's Affairs Medical Center, Richmond, Virginia, USA
| | - Jason S Pierce
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Besim Ogretmen
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jung-Hyun Cho
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jeong Ho Chang
- Department of Biology Education, Kyungpook National University, Daegu, Republic of Korea
| | - J Ross Buchan
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona, USA
| | - Jason Pitt
- Department of Laboratory Medicine and Pathology, University of Washington, Washington, DC, USA
| | - Matt Kaeberlein
- Department of Laboratory Medicine and Pathology, University of Washington, Washington, DC, USA
| | - Sung-Ung Kang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eun-Soo Kwon
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Seungbeom Ko
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kyoung-Min Choi
- Department of Oncology Science, University of Oklahoma, Oklahoma City, Oklahoma, USA
| | - Yong Sun Lee
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Yoon-Su Ha
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Republic of Korea
| | - Seung-Jin Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Republic of Korea
| | - Kwang-Pyo Lee
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - Hyo-Sung Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea
- KNU G-LAMP Project Group, KNU Institute of Basic Sciences, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu, Korea
| | - Seo Young Yang
- Department of Biology Education, Kyungpook National University, Daegu, Republic of Korea
| | - Chang Hoon Shin
- Department of Oncology Science, University of Oklahoma, Oklahoma City, Oklahoma, USA
| | - Je-Hyun Yoon
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Oncology Science, University of Oklahoma, Oklahoma City, Oklahoma, USA
- Department of Pathology, University of Oklahoma, Oklahoma City, Oklahoma, USA
| | - Kyu-Sun Lee
- Neurophysiology and Metabolism Research Group, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
4
|
Heifner JJ, Pannu TS, Gomez O, Sakalian PA, Corces A. Body Mass Index is Not an Appropriate Proxy for the Condition of Peri-Incisional Adiposity in Primary Total Joint Arthroplasty Patients. J Arthroplasty 2025; 40:346-351. [PMID: 39178972 DOI: 10.1016/j.arth.2024.08.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/11/2024] [Accepted: 08/14/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND The American Academy of Orthopaedic Surgeons defined the acceptable threshold for elective safe surgery as a body mass index (BMI) under 40 due to the increased risk of complications. A consequence of this recommendation has been a hard cutoff based on BMI, which restricts access to care for an increasingly large and diverse population. There is an improved understanding that excess adipose tissue confers additional risk for postoperative complications, including infection, through mechanical and physiologic mechanisms. But, it is unclear if BMI is an accurate indicator of adiposity in total joint arthroplasty (TJA) patients and, thus, whether BMI is capturing clinically relevant information in obese patients. Our objective was to determine the relationship between peri-incisional adiposity (PIA) and BMI in a consecutive series of diverse primary TJA patients. METHODS A consecutive series of patients indicated for primary TJA were preoperatively evaluated. For each patient, the following variables were collected: BMI and measures of PIA on radiographs and ultrasounds. RESULTS In THA patients (N = 99), Pearson's correlation coefficient (r) = 0.436, which indicates a moderate correlation between BMI and adiposity. In total knee arthroplasty patients (N = 271), r = 0.395 for femoral PIA (FPIA) and r = 0.249 for tibial PIA, which indicates a weak correlation between BMI and adiposity measured on radiography. In total knee arthroplasty patients, r = 0.560 for femoral PIA and r = 0.544 for tibial PIA, which indicates a moderate correlation between BMI and adiposity measured on ultrasound. CONCLUSIONS Quantification of obesity has become a common practice in the preoperative evaluation of primary TJA patients. The intent is to determine the magnitude of adipose tissue, which is one of the main drivers of postoperative complications in obesity. The BMI is ubiquitously used as a proxy for obesity due to its simplicity of attainment and calculation. We report that BMI has a weak-to-moderate association with PIA in this population. These findings indicate that BMI may not accurately represent the condition of peri-incisional adipose tissue and, thus, is not capturing the relevant obesity data for preoperative risk stratification in primary TJA patients. LEVEL OF EVIDENCE III.
Collapse
Affiliation(s)
| | - Tejbir S Pannu
- Larkin Hospital Department of Orthopedic Surgery, Miami, Florida
| | - Osmanny Gomez
- Larkin Hospital Department of Orthopedic Surgery, Miami, Florida
| | | | - Arturo Corces
- Larkin Hospital Department of Orthopedic Surgery, Miami, Florida
| |
Collapse
|
5
|
Bakondi E, Jung T, Marg S, Schnell V, Weber D, Schulz TJ, Grune T, Höhn A. Palmitic acid and eicosapentaenoic acid supplementation in 3T3 adipocytes: impact on lipid storage and oxidative stress. Redox Rep 2024; 29:2430882. [PMID: 39607809 DOI: 10.1080/13510002.2024.2430882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024] Open
Abstract
OBJECTIVES Obesity is a worldwide public health problem, predisposing individuals to serious cardiovascular and metabolic complications such as type 2 diabetes mellitus. White adipose tissue serves as an important regulator of energy balance, and its expansion in obesity can trigger inflammatory reactions and oxidative stress, which can also lead to insulin resistance. Adipocytes, with a key role in regulating metabolic homeostasis, respond to increased calorie intake and altered fatty acid composition with hypertrophy or hyperplasia. Of particular interest are saturated fatty acids such as palmitic acid and omega-3 polyunsaturated fatty acids such as eicosapentaenoic acid (EPA), which have differential effects on adipocyte function and inflammation. METHODS Using 3T3-L1 cells as a model for adipocytes, we evaluated the effects of PA and EPA on lipid accumulation, droplet size, and oxidative stress markers. RESULTS We were able to show that EPA supplementation in 3T3 adipocytes does not lead to excessive lipid accumulation, but rather reduces the size of lipid droplets and also induces redox changes due to the unsaturated nature of EPA. DISCUSSION These results emphasize the contrasting roles of PA and EPA and the importance of fatty acid composition in the regulation of adipocyte function.
Collapse
Affiliation(s)
- Edina Bakondi
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Tobias Jung
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Susanna Marg
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Vanessa Schnell
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Daniela Weber
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Tim J Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Annika Höhn
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
6
|
Toğuç H, Yılmaz HÖ, Yaprak B. The effect of diets with different inflammatory scores on inflammation and sleep in obese subjects: a randomized controlled trial. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2024; 70:e20240588. [PMID: 39356960 PMCID: PMC11444204 DOI: 10.1590/1806-9282.20240588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVE The aim of this study was to investigate the effect of different dietary inflammatory index diets on inflammatory markers, anthropometric measurements, and sleep quality in obese subjects. METHODS This study was conducted in a public hospital in Turkey between November 2021 and May 2022. Participants with pro-inflammatory dietary habits were included in the study. Randomly divided into two groups of 33 participants, they were subjected to an anti-inflammatory diet or a control diet for 8 weeks. The study evaluated the anthropometric parameters, inflammatory biomarkers, and sleep quality indices of the diet groups. RESULTS Significant reductions in body mass index were observed in both groups, more marked in the anti-inflammatory diet cohort. C-reactive protein levels, indicative of inflammation, also decreased substantially in both groups, with a more marked reduction in the anti-inflammatory diet cohort. Despite the improvement in sleep quality in both groups, the variation was not statistically significant. CONCLUSION This study demonstrates the importance of anti-inflammatory diets in nutritional strategies for obesity by reducing body mass index and inflammation.
Collapse
Affiliation(s)
- Hakan Toğuç
- Inonu University, Faculty of Health Sciences, Department of Nutrition and Dietetics – Malatya, Turkey
| | - Hande Öngün Yılmaz
- Bandırma Onyedi Eylül University, Faculty of Health Sciences, Department of Nutrition and Dietetics – Balıkesir, Turkey
| | - Bülent Yaprak
- Malatya Training and Research Hospital, Department of Internal Medicine – Malatya, Turkey
| |
Collapse
|
7
|
Das S, Mukhuty A, Mullen GP, Rudolph MC. Adipocyte Mitochondria: Deciphering Energetic Functions across Fat Depots in Obesity and Type 2 Diabetes. Int J Mol Sci 2024; 25:6681. [PMID: 38928386 PMCID: PMC11203708 DOI: 10.3390/ijms25126681] [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: 04/25/2024] [Revised: 06/13/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
Adipose tissue, a central player in energy balance, exhibits significant metabolic flexibility that is often compromised in obesity and type 2 diabetes (T2D). Mitochondrial dysfunction within adipocytes leads to inefficient lipid handling and increased oxidative stress, which together promote systemic metabolic disruptions central to obesity and its complications. This review explores the pivotal role that mitochondria play in altering the metabolic functions of the primary adipocyte types, white, brown, and beige, within the context of obesity and T2D. Specifically, in white adipocytes, these dysfunctions contribute to impaired lipid processing and an increased burden of oxidative stress, worsening metabolic disturbances. Conversely, compromised mitochondrial function undermines their thermogenic capabilities, reducing the capacity for optimal energy expenditure in brown adipocytes. Beige adipocytes uniquely combine the functional properties of white and brown adipocytes, maintaining morphological similarities to white adipocytes while possessing the capability to transform into mitochondria-rich, energy-burning cells under appropriate stimuli. Each type of adipocyte displays unique metabolic characteristics, governed by the mitochondrial dynamics specific to each cell type. These distinct mitochondrial metabolic phenotypes are regulated by specialized networks comprising transcription factors, co-activators, and enzymes, which together ensure the precise control of cellular energy processes. Strong evidence has shown impaired adipocyte mitochondrial metabolism and faulty upstream regulators in a causal relationship with obesity-induced T2D. Targeted interventions aimed at improving mitochondrial function in adipocytes offer a promising therapeutic avenue for enhancing systemic macronutrient oxidation, thereby potentially mitigating obesity. Advances in understanding mitochondrial function within adipocytes underscore a pivotal shift in approach to combating obesity and associated comorbidities. Reigniting the burning of calories in adipose tissues, and other important metabolic organs such as the muscle and liver, is crucial given the extensive role of adipose tissue in energy storage and release.
Collapse
Affiliation(s)
- Snehasis Das
- Harold Hamm Diabetes Center, Department of Biochemistry and Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Alpana Mukhuty
- Department of Zoology, Rampurhat College, Rampurhat 731224, India
| | - Gregory P. Mullen
- Harold Hamm Diabetes Center, Department of Biochemistry and Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Michael C. Rudolph
- Harold Hamm Diabetes Center, Department of Biochemistry and Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
8
|
Hager M, Chang P, Lee M, Burns CM, Endicott SJ, Miller RA, Li X. Recapitulation of anti-aging phenotypes by global overexpression of PTEN in mice. GeroScience 2024; 46:2653-2670. [PMID: 38114855 PMCID: PMC10828233 DOI: 10.1007/s11357-023-01025-8] [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] [Received: 09/25/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
The PTEN gene negatively regulates the oncogenic PI3K-AKT pathway by encoding a lipid and protein phosphatase that dephosphorylates lipid phosphatidylinositol-3,4,5-triphosphate (PIP3) resulting in the inhibition of PI3K and downstream inhibition of AKT. Overexpression of PTEN in mice leads to a longer lifespan compared to control littermates, although the mechanism is unknown. Here, we provide evidence that young adult PTENOE mice exhibit many characteristics shared by other slow-aging mouse models, including those with mutations that affect GH/IGF1 pathways, calorie-restricted mice, and mice treated with anti-aging drugs. PTENOE white adipose tissue (WAT) has increased UCP1, a protein linked to increased thermogenesis. WAT of PTENOE mice also shows a change in polarization of fat-associated macrophages, with elevated levels of arginase 1 (Arg1, characteristic of M2 macrophages) and decreased production of inducible nitric oxide synthase (iNOS, characteristic of M1 macrophages). Muscle and hippocampus showed increased expression of the myokine FNDC5, and higher levels of its cleavage product irisin in plasma, which has been linked to increased conversion of WAT to more thermogenic beige/brown adipose tissue. PTENOE mice also have an increase, in plasma and liver, of GPLD1, which is known to improve cognition in mice. Hippocampus of the PTENOE mice has elevation of both BDNF and DCX, indices of brain resilience and neurogenesis. These changes in fat, macrophages, liver, muscle, hippocampus, and plasma may be considered "aging rate indicators" in that they seem to be consistently changed across many of the long-lived mouse models and may help to extend lifespan by delaying many forms of late-life illness. Our new findings show that PTENOE mice can be added to the group of long-lived mice that share this multi-tissue suite of biochemical characteristics.
Collapse
Affiliation(s)
- Mary Hager
- College of Literature, Sciences, & the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Peter Chang
- College of Literature, Sciences, & the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Michael Lee
- College of Literature, Sciences, & the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Calvin M Burns
- Department of Pathology, University of Michigan School of Medicine, Room 3160, BSRB ,109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - S Joseph Endicott
- Department of Pathology, University of Michigan School of Medicine, Room 3160, BSRB ,109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
- University of Michigan Geriatrics Center, Ann Arbor, MI, 48109, USA
| | - Richard A Miller
- Department of Pathology, University of Michigan School of Medicine, Room 3160, BSRB ,109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
- University of Michigan Geriatrics Center, Ann Arbor, MI, 48109, USA
| | - Xinna Li
- Department of Pathology, University of Michigan School of Medicine, Room 3160, BSRB ,109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
- University of Michigan Geriatrics Center, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
9
|
Reyes-Farias M, Fernández-García P, Corrales P, González L, Soria-Gondek A, Martínez E, Pellitero S, Tarascó J, Moreno P, Sumoy L, Medina-Gómez G, Sánchez-Infantes D, Herrero L. Interleukin-16 is increased in obesity and alters adipogenesis and inflammation in vitro. Front Endocrinol (Lausanne) 2024; 15:1346317. [PMID: 38544694 PMCID: PMC10965774 DOI: 10.3389/fendo.2024.1346317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/23/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction Obesity is a chronic condition associated with low-grade inflammation mainly due to immune cell infiltration of white adipose tissue (WAT). WAT is distributed into two main depots: subcutaneous WAT (sWAT) and visceral WAT (vWAT), each with different biochemical features and metabolic roles. Proinflammatory cytokines including interleukin (IL)-16 are secreted by both adipocytes and infiltrated immune cells to upregulate inflammation. IL-16 has been widely studied in the peripheral proinflammatory immune response; however, little is known about its role in adipocytes in the context of obesity. Aim & Methods We aimed to study the levels of IL-16 in WAT derived from sWAT and vWAT depots of humans with obesity and the role of this cytokine in palmitate-exposed 3T3-L1 adipocytes. Results The results demonstrated that IL-16 expression was higher in vWAT compared with sWAT in individuals with obesity. In addition, IL-16 serum levels were higher in patients with obesity compared with normal-weight individuals, increased at 6 months after bariatric surgery, and at 12 months after surgery decreased to levels similar to before the intervention. Our in vitro models showed that IL-16 could modulate markers of adipogenesis (Pref1), lipid metabolism (Plin1, Cd36, and Glut4), fibrosis (Hif1a, Col4a, Col6a, and Vegf), and inflammatory signaling (IL6) during adipogenesis and in mature adipocytes. In addition, lipid accumulation and glycerol release assays suggested lipolysis alteration. Discussion Our results suggest a potential role of IL-16 in adipogenesis, lipid and glucose homeostasis, fibrosis, and inflammation in an obesity context.
Collapse
Affiliation(s)
- Marjorie Reyes-Farias
- Endocrinology department, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institute of Biomedicine of the University of Barcelona (IBUB), Universitat de Barcelona (UB), Barcelona, Spain
| | | | - Patricia Corrales
- Department of Basic Health Sciences, University Rey Juan Carlos (URJC), Madrid, Spain
| | - Lorena González
- Endocrinology department, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | - Andrea Soria-Gondek
- Pediatric Surgery Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Ester Martínez
- Department of Basic Health Sciences, University Rey Juan Carlos (URJC), Madrid, Spain
| | - Silvia Pellitero
- Endocrinology and Nutrition Department, Institute Research and Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jordi Tarascó
- General Surgery Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Pau Moreno
- General Surgery Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Lauro Sumoy
- Endocrinology department, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | - Gema Medina-Gómez
- Department of Basic Health Sciences, University Rey Juan Carlos (URJC), Madrid, Spain
| | - David Sánchez-Infantes
- Department of Basic Health Sciences, University Rey Juan Carlos (URJC), Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institute of Biomedicine of the University of Barcelona (IBUB), Universitat de Barcelona (UB), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
10
|
Fernández-García P, Taxerås SD, Reyes-Farias M, González L, Soria-Gondek A, Pellitero S, Tarascó J, Moreno P, Sumoy L, Stephens JM, Yoo LG, Galán M, Izquierdo A, Medina-Gómez G, Herrero L, Corrales P, Villarroya F, Cereijo R, Sánchez-Infantes D. Claudin-1 as a novel target gene induced in obesity and associated to inflammation, fibrosis, and cell differentiation. Eur J Endocrinol 2024; 190:201-210. [PMID: 38375549 DOI: 10.1093/ejendo/lvae018] [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: 09/07/2023] [Revised: 12/13/2023] [Accepted: 02/06/2024] [Indexed: 02/21/2024]
Abstract
OBJECTIVE T lymphocytes from visceral and subcutaneous white adipose tissues (vWAT and sWAT, respectively) can have opposing roles in the systemic metabolic changes associated with obesity. However, few studies have focused on this subject. Claudin-1 (CLDN1) is a protein involved canonically in tight junctions and tissue paracellular permeability. We evaluated T-lymphocyte gene expression in vWAT and sWAT and in the whole adipose depots in human samples. METHODS A Clariom D-based transcriptomic analysis was performed on T lymphocytes magnetically separated from vWAT and sWAT from patients with obesity (Cohort 1; N = 11). Expression of candidate genes resulting from that analysis was determined in whole WAT from individuals with and without obesity (Cohort 2; patients with obesity: N = 13; patients without obesity: N = 14). RESULTS We observed transcriptional differences between T lymphocytes from sWAT compared with vWAT. Specifically, CLDN1 expression was found to be dramatically induced in vWAT T cells relative to those isolated from sWAT in patients with obesity. CLDN1 was also induced in obesity in vWAT and its expression correlates with genes involved in inflammation, fibrosis, and adipogenesis. CONCLUSION These results suggest that CLDN1 is a novel marker induced in obesity and differentially expressed in T lymphocytes infiltrated in human vWAT as compared with sWAT. This protein may have a crucial role in the crosstalk between T lymphocytes and other adipose tissue cells and may contribute to inflammation, fibrosis, and alter homeostasis and promote metabolic disease in obesity.
Collapse
Affiliation(s)
- Pablo Fernández-García
- Department of Basic Health Sciences, Campus Alcorcón, University Rey Juan Carlos (URJC), Madrid E-28922, Spain
| | - Siri D Taxerås
- Endocrinology Department, Fundació Institut Germans Trias i Pujol, Barcelona 08916, Spain
| | - Marjorie Reyes-Farias
- Endocrinology Department, Fundació Institut Germans Trias i Pujol, Barcelona 08916, Spain
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona 08028, Spain
| | - Lorena González
- Endocrinology Department, Fundació Institut Germans Trias i Pujol, Barcelona 08916, Spain
| | - Andrea Soria-Gondek
- Pediatric Surgery Department, Hospital Universitari Germans Trias i Pujol, Badalona 08916, Spain
| | - Silvia Pellitero
- Endocrinology Department, Hospital Universitari Germans Trias i Pujol, Badalona 08916, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Jordi Tarascó
- General Surgery Department, Hospital Universitari Germans Trias i Pujol, Badalona 08916, Spain
| | - Pau Moreno
- General Surgery Department, Hospital Universitari Germans Trias i Pujol, Badalona 08916, Spain
| | - Lauro Sumoy
- Endocrinology Department, Fundació Institut Germans Trias i Pujol, Barcelona 08916, Spain
| | - Jacqueline M Stephens
- Adipocyte Biology Department, Pennington Biomedical Research Center (PBRC), Louisiana State University, Baton Rouge, LA 70808, United States
| | - Lindsey G Yoo
- Adipocyte Biology Department, Pennington Biomedical Research Center (PBRC), Louisiana State University, Baton Rouge, LA 70808, United States
| | - María Galán
- Department of Basic Health Sciences, Campus Alcorcón, University Rey Juan Carlos (URJC), Madrid E-28922, Spain
| | - Adriana Izquierdo
- Department of Basic Health Sciences, Campus Alcorcón, University Rey Juan Carlos (URJC), Madrid E-28922, Spain
| | - Gema Medina-Gómez
- Department of Basic Health Sciences, Campus Alcorcón, University Rey Juan Carlos (URJC), Madrid E-28922, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona 08028, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Patricia Corrales
- Department of Basic Health Sciences, Campus Alcorcón, University Rey Juan Carlos (URJC), Madrid E-28922, Spain
| | - Francesc Villarroya
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine, University of Barcelona, Barcelona 08028, Spain
| | - Rubén Cereijo
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine, University of Barcelona, Barcelona 08028, Spain
- Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau (IIB-Sant Pau), Infectious Diseases Unit, Barcelona 08041, Spain
| | - David Sánchez-Infantes
- Department of Basic Health Sciences, Campus Alcorcón, University Rey Juan Carlos (URJC), Madrid E-28922, Spain
- Endocrinology Department, Fundació Institut Germans Trias i Pujol, Barcelona 08916, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| |
Collapse
|
11
|
Naik N, Patel M, Sen R. Developmental Impacts of Epigenetics and Metabolism in COVID-19. J Dev Biol 2024; 12:9. [PMID: 38390960 PMCID: PMC10885083 DOI: 10.3390/jdb12010009] [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: 12/31/2023] [Revised: 02/04/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
Developmental biology is intricately regulated by epigenetics and metabolism but the mechanisms are not completely understood. The situation becomes even more complicated during diseases where all three phenomena are dysregulated. A salient example is COVID-19, where the death toll exceeded 6.96 million in 4 years, while the virus continues to mutate into different variants and infect people. Early evidence during the pandemic showed that the host's immune and inflammatory responses to COVID-19 (like the cytokine storm) impacted the host's metabolism, causing damage to the host's organs and overall physiology. The involvement of angiotensin-converting enzyme 2 (ACE2), the pivotal host receptor for the SARS-CoV-2 virus, was identified and linked to epigenetic abnormalities along with other contributing factors. Recently, studies have revealed stronger connections between epigenetics and metabolism in COVID-19 that impact development and accelerate aging. Patients manifest systemic toxicity, immune dysfunction and multi-organ failure. Single-cell multiomics and other state-of-the-art high-throughput studies are only just beginning to demonstrate the extent of dysregulation and damage. As epigenetics and metabolism directly impact development, there is a crucial need for research implementing cutting-edge technology, next-generation sequencing, bioinformatics analysis, the identification of biomarkers and clinical trials to help with prevention and therapeutic interventions against similar threats in the future.
Collapse
Affiliation(s)
- Noopur Naik
- Department of Molecular, Cellular & Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Mansi Patel
- Institute of Genomics and Integrative Biology, Delhi 110007, India
| | - Rwik Sen
- Active Motif, Inc., Carlsbad, CA 92008, USA
| |
Collapse
|
12
|
Kang H, Lee J. Adipose tissue macrophage heterogeneity in the single-cell genomics era. Mol Cells 2024; 47:100031. [PMID: 38354858 PMCID: PMC10960114 DOI: 10.1016/j.mocell.2024.100031] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/07/2024] [Accepted: 02/07/2024] [Indexed: 02/16/2024] Open
Abstract
It is now well-accepted that obesity-induced inflammation plays an important role in the development of insulin resistance and type 2 diabetes. A key source of the inflammation is the murine epididymal and human visceral adipose tissue. The current paradigm is that obesity activates multiple proinflammatory immune cell types in adipose tissue, including adipose-tissue macrophages (ATMs), T Helper 1 (Th1) T cells, and natural killer (NK) cells, while concomitantly suppressing anti-inflammatory immune cells such as T Helper 2 (Th2) T cells and regulatory T cells (Tregs). A key feature of the current paradigm is that obesity induces the anti-inflammatory M2 ATMs in lean adipose tissue to polarize into proinflammatory M1 ATMs. However, recent single-cell transcriptomics studies suggest that the story is much more complex. Here we describe the single-cell genomics technologies that have been developed recently and the emerging results from studies using these technologies. While further studies are needed, it is clear that ATMs are highly heterogeneous. Moreover, while a variety of ATM clusters with quite distinct features have been found to be expanded by obesity, none truly resemble classical M1 ATMs. It is likely that single-cell transcriptomics technology will further revolutionize the field, thereby promoting our understanding of ATMs, adipose-tissue inflammation, and insulin resistance and accelerating the development of therapies for type 2 diabetes.
Collapse
Affiliation(s)
- Haneul Kang
- Soonchunhyang Institute of Medi-Bio Science (SIMS) and Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-si, South Korea
| | - Jongsoon Lee
- Soonchunhyang Institute of Medi-Bio Science (SIMS) and Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-si, South Korea.
| |
Collapse
|
13
|
Liu Y, Hu G, Jia Y, Qin L, Xu L, Chang Y, Li B, Li H. Wnt10b knockdown regulates the relative balance of adipose tissue-resident T cells and inhibits white fat deposition. Mol Biol Rep 2024; 51:272. [PMID: 38302806 DOI: 10.1007/s11033-024-09249-3] [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] [Received: 09/21/2023] [Accepted: 01/12/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND Wnt10b is one of critical Wnt family members that being involved in networks controlling stemness, pluripotency and cell fate decisions. However, its role in adipose-resident T lymphocytes and further in fat metabolism yet remains largely unknown. METHODS AND RESULTS In the present study, we demonstrated a distinctive effect for Wnt10b on the relative balance of T lymphocytes in adipose tissue by using a Wnt10b knockdown mouse model. Wnt10b knockdown led to a reduction of adipose-resident CD4+ T cells and an elevation of Foxp3+/CD4+ Treg cells. Wnt10b-knockdown mice fed with standard diet showed less white fat deposition owing to the suppressed adipogenic process. Moreover, under high fat diet conditions, Wnt10b knockdown resulted in an alleviated obesity symptoms, as well as an improvement of glucose homeostasis and hepatic steatosis. CONCLUSIONS Collectively, we reveal an unexpected and novel function for Wnt10b in mediating the frequency of adipose-resident T cell subsets, that when knockdown skewing toward a Treg-dominated phenotype and further improving fat metabolism.
Collapse
Affiliation(s)
- Yan Liu
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Geng Hu
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, China
| | - Yanxin Jia
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, China
| | - Lining Qin
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Longfei Xu
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Yaxin Chang
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Bin Li
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Haifang Li
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China.
| |
Collapse
|
14
|
Avtanski D, Stojchevski R. Significance of Adipose Tissue as an Endocrine Organ. CONTEMPORARY ENDOCRINOLOGY 2024:1-46. [DOI: 10.1007/978-3-031-72570-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
15
|
Pérez-Gómez JM, Montero-Hidalgo AJ, Fuentes-Fayos AC, Sarmento-Cabral A, Guzmán-Ruiz R, Malagón MM, Herrera-Martínez AD, Gahete MD, Luque RM. Exploring the role of the inflammasomes on prostate cancer: Interplay with obesity. Rev Endocr Metab Disord 2023; 24:1165-1187. [PMID: 37819510 PMCID: PMC10697898 DOI: 10.1007/s11154-023-09838-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/21/2023] [Indexed: 10/13/2023]
Abstract
Obesity is a weight-related disorder characterized by excessive adipose tissue growth and dysfunction which leads to the onset of a systemic chronic low-grade inflammatory state. Likewise, inflammation is considered a classic cancer hallmark affecting several steps of carcinogenesis and tumor progression. In this regard, novel molecular complexes termed inflammasomes have been identified which are able to react to a wide spectrum of insults, impacting several metabolic-related disorders, but their contribution to cancer biology remains unclear. In this context, prostate cancer (PCa) has a markedly inflammatory component, and patients frequently are elderly individuals who exhibit weight-related disorders, being obesity the most prevalent condition. Therefore, inflammation, and specifically, inflammasome complexes, could be crucial players in the interplay between PCa and metabolic disorders. In this review, we will: 1) discuss the potential role of each inflammasome component (sensor, molecular adaptor, and targets) in PCa pathophysiology, placing special emphasis on IL-1β/NF-kB pathway and ROS and hypoxia influence; 2) explore the association between inflammasomes and obesity, and how these molecular complexes could act as the cornerstone between the obesity and PCa; and, 3) compile current clinical trials regarding inflammasome targeting, providing some insights about their potential use in the clinical practice.
Collapse
Affiliation(s)
- Jesús M Pérez-Gómez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Antonio J Montero-Hidalgo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Antonio C Fuentes-Fayos
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - André Sarmento-Cabral
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Rocio Guzmán-Ruiz
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - María M Malagón
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Aura D Herrera-Martínez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Endocrinology and Nutrition Service, HURS/IMIBIC, Córdoba, Spain
| | - Manuel D Gahete
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain
| | - Raúl M Luque
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), IMIBIC Building, Av. Menéndez Pidal s/n, 14004, Córdoba, Spain.
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Cordoba, Spain.
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, (CIBERobn), Cordoba, Spain.
| |
Collapse
|
16
|
Ishikawa H, Nagashima R, Kuno Y, Sasaki H, Kohda C, Iyoda M. Effects of NKT Cells on Metabolic Disorders Caused by High-Fat Diet Using CD1d-Knockout Mice. Diabetes Metab Syndr Obes 2023; 16:2855-2864. [PMID: 37744699 PMCID: PMC10517681 DOI: 10.2147/dmso.s428190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/14/2023] [Indexed: 09/26/2023] Open
Abstract
Purpose The purpose of this study was to investigate whether NKT cells play an important role in preventing or exacerbating diseases caused by high-fat diet (HFD) using CD1d-knockout (KO) mice which lack NKT cells. Methods Five-week-old male Balb/c (wild-type; WT) or CD1dKO mice were fed with control-diet (CTD) or HFD for 16 weeks. Results The present study revealed four main findings. First, CD1dKO mice were susceptible to obesity caused by HFD in comparison to WT mice. Second, clinical conditions of fatty liver caused by HFD were comparable between CD1dKO mice and WT mice. Third, HFD-fed WT mice showed high levels of serum biochemical markers, involved in lipid metabolisms, in comparison to WT mice fed a CTD. Notably, the serum concentrations of ALT, T-CHO, TG and HDL-C in CD1dKO mice fed a HFD were almost comparable to those of CD1dKO mice fed a CTD. Fourth, the expression of peroxisome proliferator-activated receptor (PPAR) γ, low-density lipoprotein receptor (LDLR), CD36 of epididymal adipose tissue enhanced and proprotein convertase subtilisin/kexin type (PCSK) 9 in serum decreased. Conclusion NKT cells were responsible for protection against HFD-induced obesity. However, CD1dKO mice were resistant to serum biochemical marker abnormalities after HFD feeding. One possible explanation is that the epididymal adipose tissue of CD1dKO mice could take up greater amounts of excess lipids in serum in comparison to WT mice.
Collapse
Affiliation(s)
- Hiroki Ishikawa
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Ryuichi Nagashima
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Yoshihiro Kuno
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, 142-8555, Japan
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, 142-8666, Japan
| | - Hiraku Sasaki
- Department of Health Science, Faculty of Health and Sports Science, Juntendo University, Inzai, Chiba, 270-1695, Japan
| | - Chikara Kohda
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Masayuki Iyoda
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, 142-8555, Japan
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, 142-8666, Japan
| |
Collapse
|
17
|
Sharifi-Zahabi E, Rezvani N, Hajizadeh-Sharafabad F, Hosseini-Baharanchi FS, Shidfar F, Rahimi M. Theobromine supplementation in combination with a low-calorie diet improves cardiovascular risk factors in overweight and obese subjects with metabolic syndrome: a randomized controlled trial. Food Funct 2023; 14:8431-8441. [PMID: 37615657 DOI: 10.1039/d3fo00555k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Background & aims: The beneficial effects of theobromine (TB) on obesity and features of metabolic syndrome (MetS) have been reported in several studies. However, the findings are equivocal. The present study aimed to investigate the effects of 12 week pure TB supplementation (450 mg day-1) combined with a low-calorie diet on the anthropometric and metabolic syndrome indices in overweight and obese adults with MetS. Methods: In a randomized double-blind parallel controlled trial, 80 participants aged 40-55 years were randomly assigned to take 450 mg day-1 TB or placebo along with a low-calorie diet for 12 weeks. Dietary intake, anthropometric indices, blood pressure, lipid profile and glycemic indices were assessed at the start and end of the intervention. Results: Seventy-two participants completed the study. After 12 weeks, TB supplementation significantly decreased the waist circumference (WC) (-0.86 cm; P = 0.045), LDL-c/HDL-c (-0.26; P = 0.008), TG/HDL-c (-0.41; P = 0.001), TC/HDL-c (-0.38; P = 0.006) and increased HDL-c (1.72 mg dl-1; P = 0.036) compared to the placebo group. There were no significant differences regarding body weight, BMI, hip circumference (HC), hip-to-waist circumference ratio (WHR), systolic and diastolic blood pressure, fasting levels of total cholesterol (TC), triacylglycerol (TAG), low-density lipoprotein cholesterol (LDL-c), fasting blood glucose, insulin, homoeostatic model assessment for insulin resistance (HOMA-IR) and homeostasis model assessment of β-cell function (HOMA-β) between the two groups (p > 0.05). Conclusion: The results of the current study revealed that TB supplementation along with a low-calorie diet had favorable effects on WC, LDL-c/HDL-c, TG/HDL-c, TC/HDL-c, and serum level of HDL-c in overweight and obese subjects with MetS. Trial registration number: IRCT20091114002709N59. Registration date: 5 March 2022.
Collapse
Affiliation(s)
- Elham Sharifi-Zahabi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Nayebali Rezvani
- Department of Clinical Biochemistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Fatemeh Sadat Hosseini-Baharanchi
- Minimally Invasive Surgery Research Center, & Department of Biostatistics, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Shidfar
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran.
| | - Mehrali Rahimi
- School of Medicine, Diabetic Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
18
|
Sharifi-Zahabi E, Hajizadeh-Sharafabad F, Nachvak SM, Mirzaian S, Darbandi S, Shidfar F. A comprehensive insight into the molecular effect of theobromine on cardiovascular-related risk factors: A systematic review of in vitro and in vivo studies. Phytother Res 2023; 37:3765-3779. [PMID: 37309834 DOI: 10.1002/ptr.7916] [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: 10/16/2022] [Revised: 05/11/2023] [Accepted: 05/27/2023] [Indexed: 06/14/2023]
Abstract
Theobromine may have beneficial effects on cardiovascular risk factors. This study aimed to find molecular effects of theobromine on lipid profile, glycemic status, inflammatory factors, and vascular function through a comprehensive assessment of all in vitro and in vivo studies. The search process was started at 18 July 2022. Databases including PubMed, Scopus, and Web of Science were searched to find all articles published up to 18 July 2022. Nineteen studies were included in this study. In vitro studies showed the improving effects of theobromine on inflammatory markers. Of four animal studies assessing the effect of theobromine on inflammatory markers, two reported favorable effects. Among five animal studies assessing the effects of theobromine on lipid profile, three reported improving effects on either triglyceride, total cholesterol, low- or high-density lipoprotein cholesterol. Of the three human studies, two revealed that theobromine had improving effects on lipid profile. A favorable effect of theobromine on augmentation index was also reported in two RCTs. The results for other outcomes were inconclusive. Theobromine may have favorable effects on inflammatory factors, lipid profile, and vascular function markers. However, studies with a longer duration and lower, dietary-relevant doses are required for future confirmation.
Collapse
Affiliation(s)
| | | | - Seyed Mostafa Nachvak
- School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Soheila Mirzaian
- Department of Food and Human Nutrition Sciences, University of Manitoba, Winnipeg, Canada
| | - Sahar Darbandi
- Imam Khomeini comprehensive health center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farzad Shidfar
- Department of nutrition, School of public health, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Gonçalves CCRA, Feitosa BM, Cavalcante BV, Lima ALGDSB, de Souza CM, Joventino LB, Cavalcante MB. Obesity and recurrent miscarriage: The interconnections between adipose tissue and the immune system. Am J Reprod Immunol 2023; 90:e13757. [PMID: 37641378 DOI: 10.1111/aji.13757] [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: 05/08/2023] [Revised: 07/16/2023] [Accepted: 07/17/2023] [Indexed: 08/31/2023] Open
Abstract
Currently, obesity is considered a global public health problem. It is the main risk factor for noncommunicable diseases and reproductive complications, such as recurrent miscarriage (RM). RM affects approximately 1% of couples of reproductive age, and recent studies suggest that its prevalence is increasing. Immunological abnormalities may be responsible for a significant number of cases of unexplained RM. Obesity is recognized as a chronic low-grade inflammatory condition. The accumulation of fat in obese adipose tissue promotes changes in the local and systemic immune response. Adipokines, exosomes, micro-RNAs, lipids, and other factors released or secreted by adipose tissue are responsible for the interconnection between obesity and the immune system. Obesity-induced dysregulation of the innate and acquired immune response is also involved in the immunopathology of pregnancy loss in patients with unexplained RM. Therefore, understanding the communication pathways between maternal adipose tissue and the immune response in women living with obesity and RM is an important objective. Thus, diagnostic tools and new immunomodulatory therapies may be proposed for the management of patients with concurrent obesity and RM.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Marcelo Borges Cavalcante
- Medical School, Universidade de Fortaleza (UNIFOR), Fortaleza, CE, Brazil
- Postgraduate Program in Medical Sciences, Universidade de Fortaleza (UNIFOR), Fortaleza, CE, Brazil
- CONCEPTUS - Reproductive Medicine, Fortaleza, Brazil
| |
Collapse
|
20
|
Szukiewicz D. Molecular Mechanisms for the Vicious Cycle between Insulin Resistance and the Inflammatory Response in Obesity. Int J Mol Sci 2023; 24:9818. [PMID: 37372966 DOI: 10.3390/ijms24129818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 05/31/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
The comprehensive anabolic effects of insulin throughout the body, in addition to the control of glycemia, include ensuring lipid homeostasis and anti-inflammatory modulation, especially in adipose tissue (AT). The prevalence of obesity, defined as a body mass index (BMI) ≥ 30 kg/m2, has been increasing worldwide on a pandemic scale with accompanying syndemic health problems, including glucose intolerance, insulin resistance (IR), and diabetes. Impaired tissue sensitivity to insulin or IR paradoxically leads to diseases with an inflammatory component despite hyperinsulinemia. Therefore, an excess of visceral AT in obesity initiates chronic low-grade inflammatory conditions that interfere with insulin signaling via insulin receptors (INSRs). Moreover, in response to IR, hyperglycemia itself stimulates a primarily defensive inflammatory response associated with the subsequent release of numerous inflammatory cytokines and a real threat of organ function deterioration. In this review, all components of this vicious cycle are characterized with particular emphasis on the interplay between insulin signaling and both the innate and adaptive immune responses related to obesity. Increased visceral AT accumulation in obesity should be considered the main environmental factor responsible for the disruption in the epigenetic regulatory mechanisms in the immune system, resulting in autoimmunity and inflammation.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
21
|
Agnese M, Toia P, Sollami G, Militello C, Rundo L, Vitabile S, Maffei E, Agnello F, Gagliardo C, Grassedonio E, Galia M, Cademartiri F, Midiri M, La Grutta L. Epicardial and thoracic subcutaneous fat texture analysis in patients undergoing cardiac CT. Heliyon 2023; 9:e15984. [PMID: 37215845 PMCID: PMC10196784 DOI: 10.1016/j.heliyon.2023.e15984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/09/2023] [Accepted: 04/28/2023] [Indexed: 05/24/2023] Open
Abstract
Introduction The aim of our study was to evaluate the feasibility of texture analysis of epicardial fat (EF) and thoracic subcutaneous fat (TSF) in patients undergoing cardiac CT (CCT). Materials and methods We compared a consecutive population of 30 patients with BMI ≤25 kg/m2 (Group A, 60.6 ± 13.7 years) with a control population of 30 patients with BMI >25 kg/m2 (Group B, 63.3 ± 11 years). A dedicated computer application for quantification of EF and a texture analysis application for the study of EF and TSF were employed. Results The volume of EF was higher in group B (mean 116.1 cm3 vs. 86.3 cm3, p = 0.014), despite no differences were found neither in terms of mean density (-69.5 ± 5 HU vs. -68 ± 5 HU, p = 0.28), nor in terms of quartiles distribution (Q1, p = 0.83; Q2, p = 0.22, Q3, p = 0.83, Q4, p = 0.34). The discriminating parameters of the histogram class were mean (p = 0.02), 0,1st (p = 0.001), 10th (p = 0.002), and 50th percentiles (p = 0.02). DifVarnc was the discriminating parameter of the co-occurrence matrix class (p = 0.007).The TSF thickness was 15 ± 6 mm in group A and 19.5 ± 5 mm in group B (p = 0.003). The TSF had a mean density of -97 ± 19 HU in group A and -95.8 ± 19 HU in group B (p = 0.75). The discriminating parameters of texture analysis were 10th (p = 0.03), 50th (p = 0.01), 90th percentiles (p = 0.04), S(0,1)SumAverg (p = 0.02), S(1,-1)SumOfSqs (p = 0.02), S(3,0)Contrast (p = 0.03), S(3,0)SumAverg (p = 0.02), S(4,0)SumAverg (p = 0.04), Horzl_RLNonUni (p = 0.02), and Vertl_LngREmph (p = 0.0005). Conclusions Texture analysis provides distinctive radiomic parameters of EF and TSF. EF and TSF had different radiomic features as the BMI varies.
Collapse
Affiliation(s)
- Manfredi Agnese
- Department of Biomedicine, Neurosciences and Advanced Diagnostics - BIND, University of Palermo, Via del Vespro 127, 90100, Palermo, Italy
| | - Patrizia Toia
- Department of Biomedicine, Neurosciences and Advanced Diagnostics - BIND, University of Palermo, Via del Vespro 127, 90100, Palermo, Italy
| | - Giulia Sollami
- Department of Biomedicine, Neurosciences and Advanced Diagnostics - BIND, University of Palermo, Via del Vespro 127, 90100, Palermo, Italy
| | - Carmelo Militello
- Institute for High-Performance Computing and Networking, National Research Council (ICAR-CNR), Palermo, Italy
| | - Leonardo Rundo
- Department of Information and Electrical Engineering and Applied Mathematics (DIEM), University of Salerno, Salerno, Italy
| | - Salvatore Vitabile
- Department of Biomedicine, Neurosciences and Advanced Diagnostics - BIND, University of Palermo, Via del Vespro 127, 90100, Palermo, Italy
| | - Erica Maffei
- Department of Radiology, Fondazione Monasterio, Pisa, Italy
| | - Francesco Agnello
- Department of Biomedicine, Neurosciences and Advanced Diagnostics - BIND, University of Palermo, Via del Vespro 127, 90100, Palermo, Italy
| | - Cesare Gagliardo
- Department of Biomedicine, Neurosciences and Advanced Diagnostics - BIND, University of Palermo, Via del Vespro 127, 90100, Palermo, Italy
| | - Emanuele Grassedonio
- Department of Biomedicine, Neurosciences and Advanced Diagnostics - BIND, University of Palermo, Via del Vespro 127, 90100, Palermo, Italy
| | - Massimo Galia
- Department of Biomedicine, Neurosciences and Advanced Diagnostics - BIND, University of Palermo, Via del Vespro 127, 90100, Palermo, Italy
| | | | - Massimo Midiri
- Department of Biomedicine, Neurosciences and Advanced Diagnostics - BIND, University of Palermo, Via del Vespro 127, 90100, Palermo, Italy
| | - Ludovico La Grutta
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties - ProMISE, University of Palermo, Palermo, Italy
| |
Collapse
|
22
|
Pezhman L, Hopkin SJ, Begum J, Heising S, Nasteska D, Wahid M, Ed Rainger G, Hodson DJ, Iqbal AJ, Chimen M, McGettrick HM. PEPITEM modulates leukocyte trafficking to reduce obesity-induced inflammation. Clin Exp Immunol 2023; 212:1-10. [PMID: 36891817 PMCID: PMC10081110 DOI: 10.1093/cei/uxad022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/10/2023] [Accepted: 02/13/2023] [Indexed: 03/10/2023] Open
Abstract
Dysregulation of leukocyte trafficking, lipid metabolism, and other metabolic processes are the hallmarks that underpin and drive pathology in obesity. Current clinical management targets alternations in lifestyle choices (e.g. exercise, weight loss) to limit the impact of the disease. Crucially, re-gaining control over the pathogenic cellular and molecular processes may offer an alternative, complementary strategy for obese patients. Here we investigate the impact of the immunopeptide, PEPITEM, on pancreas homeostasis and leukocyte trafficking in mice on high-fed obesogenic diet (HFD). Both prophylactic and therapeutic treatment with PEPITEM alleviated the effects of HFD on the pancreas, reducing pancreatic beta cell size. Moreover, PEPITEM treatment also limited T-cell trafficking (CD4+ T-cells and KLRG1+ CD3+ T-cells) to obese visceral, but not subcutaneous, adipose tissue. Similarly, PEPITEM treatment reduced macrophage numbers within the peritoneal cavity of mice on HFD diet at both 6 and 12 weeks. By contrast, PEPITEM therapy elevated numbers of T and B cells were observed in the secondary lymphoid tissues (e.g. spleen and inguinal lymph node) when compared to the untreated HFD controls. Collectively our data highlights the potential for PEPITEM as a novel therapy to combat the systemic low-grade inflammation experienced in obesity and minimize the impact of obesity on pancreatic homeostasis. Thus, offering an alternative strategy to reduce the risk of developing obesity-related co-morbidities, such as type 2 diabetes mellitus, in individuals at high risk and struggling to control their weight through lifestyle modifications.
Collapse
Affiliation(s)
- Laleh Pezhman
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Sophie J Hopkin
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Jenefa Begum
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Silke Heising
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Daniela Nasteska
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Mussarat Wahid
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - G Ed Rainger
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - David J Hodson
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Asif J Iqbal
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Myriam Chimen
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Helen M McGettrick
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| |
Collapse
|
23
|
Li X, Hager M, McPherson M, Lee M, Hagalwadi R, Skinner ME, Lombard D, Miller RA. Recapitulation of anti-aging phenotypes by global, but not by muscle-specific, deletion of PAPP-A in mice. GeroScience 2023; 45:931-948. [PMID: 36542300 PMCID: PMC9886707 DOI: 10.1007/s11357-022-00692-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 11/15/2022] [Indexed: 12/24/2022] Open
Abstract
Deletion of pregnancy-associated plasma protein-A (PAPP-A), a protease that cleaves some but not all IGF1 binding proteins, postpones late-life diseases and extends lifespan in mice, but the mechanism of this effect is unknown. Here we show that PAPP-A knockout (PKO) mice display a set of changes, in multiple tissues, that are characteristic of other varieties of slow-aging mice with alterations in GH production or GH responsiveness, including Ames dwarf, Snell dwarf, and GHRKO mice. PKO mice have elevated UCP1 in brown and white adipose tissues (WAT), and a change in fat-associated macrophage subsets that leads to diminished production of inflammatory cytokines. PKO mice also show increased levels of muscle FNDC5 and its cleavage product, the myokine irisin, thought to cause changes in fat cell differentiation. PKO mice have elevated production of hepatic GPLD1 and plasma GPLD1, consistent with their elevation of hippocampal BDNF and DCX, used as indices of neurogenesis. In contrast, disruption of PAPP-A limited to muscle ("muPKO" mice) produces an unexpectedly complex set of changes, in most cases opposite in direction from those seen in PKO mice. These include declines in WAT UCP1, increases in inflammatory macrophages and cytokines in WAT, and a decline in muscle FNDC5 and plasma irisin. muPKO mice do, however, resemble global PKO mice in their elevation of hippocampal BDNF and DCX. The data for the PKO mice support the idea that these changes in fat, macrophages, liver, muscle, plasma, and brain are consistent and biologically significant features of the slow-aging phenotype in mice. The results on the muPKO mice provide a foundation for further investigation of the complex, local, and global circuits by which PAPP-A modulates signals ordinarily controlled by GH and/or IGF1.
Collapse
Affiliation(s)
- Xinna Li
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA.
- , Ann Arbor, USA.
| | - Mary Hager
- College of Literature, Sciences, & the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Madaline McPherson
- College of Literature, Sciences, & the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Michael Lee
- College of Literature, Sciences, & the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Riha Hagalwadi
- College of Literature, Sciences, & the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Mary E Skinner
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA
| | - David Lombard
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA
- Sylvester Cancer Center, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Richard A Miller
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA
- University of Michigan Geriatrics Center, Ann Arbor, MI, 48109, USA
| |
Collapse
|
24
|
Barthelemy J, Bogard G, Wolowczuk I. Beyond energy balance regulation: The underestimated role of adipose tissues in host defense against pathogens. Front Immunol 2023; 14:1083191. [PMID: 36936928 PMCID: PMC10019896 DOI: 10.3389/fimmu.2023.1083191] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/09/2023] [Indexed: 03/06/2023] Open
Abstract
Although the adipose tissue (AT) is a central metabolic organ in the regulation of whole-body energy homeostasis, it is also an important endocrine and immunological organ. As an endocrine organ, AT secretes a variety of bioactive peptides known as adipokines - some of which have inflammatory and immunoregulatory properties. As an immunological organ, AT contains a broad spectrum of innate and adaptive immune cells that have mostly been studied in the context of obesity. However, overwhelming evidence supports the notion that AT is a genuine immunological effector site, which contains all cell subsets required to induce and generate specific and effective immune responses against pathogens. Indeed, AT was reported to be an immune reservoir in the host's response to infection, and a site of parasitic, bacterial and viral infections. In addition, besides AT's immune cells, preadipocytes and adipocytes were shown to express innate immune receptors, and adipocytes were reported as antigen-presenting cells to regulate T-cell-mediated adaptive immunity. Here we review the current knowledge on the role of AT and AT's immune system in host defense against pathogens. First, we will summarize the main characteristics of AT: type, distribution, function, and extraordinary plasticity. Second, we will describe the intimate contact AT has with lymph nodes and vessels, and AT immune cell composition. Finally, we will present a comprehensive and up-to-date overview of the current research on the contribution of AT to host defense against pathogens, including the respiratory viruses influenza and SARS-CoV-2.
Collapse
Affiliation(s)
| | | | - Isabelle Wolowczuk
- Univ. Lille, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (Inserm), Centre Hospitalier Universitaire de Lille (CHU Lille), Institut Pasteur de Lille, U1019 - UMR 9017 - Center for Infection and Immunity of Lille (CIIL), Lille, France
| |
Collapse
|
25
|
Kim JW, Shin SK, Kwon EY. Luteolin Protects Against Obese Sarcopenia in Mice with High-Fat Diet-Induced Obesity by Ameliorating Inflammation and Protein Degradation in Muscles. Mol Nutr Food Res 2023; 67:e2200729. [PMID: 36708177 DOI: 10.1002/mnfr.202200729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/04/2023] [Indexed: 01/29/2023]
Abstract
SCOPE Although sarcopenia is mainly caused by aging, sarcopenia due to obesity has become an emerging issue given the increase in obesity among people of various ages. There are studies on obesity or sarcopenia, our understanding of obesity-mediated sarcopenia is insufficient. Luteolin (LU) has exhibited antiobesity effects, but no studies have investigated the LU effects on antisarcopenia. This study therefore investigated the effects of LU on obese sarcopenia in mice with high-fat diet (HFD)-induced obesity. METHODS AND RESULTS To evaluate its inhibitory efficacy against obese sarcopenia, 5-week-old mice are fed an HFD supplemented with LU for 20 weeks. LU exerts suppressive effects on obesity, inflammation, and protein degradation in the HFD-fed obese mice. It also inhibits lipid infiltration into the muscle and decreases p38 activity and the mRNA expression of inflammatory factors, including TNF-α, Tlr2, Tlr4, MCP1, and MMP2, in the muscle. The suppression of muscle inflammation by LU leads to the inhibition of myostatin, FoxO, atrogin, and MuRF expression. These effects of LU affect inhibition of protein degradation and improvement of muscle function. CONCLUSION Here, it demonstrates that LU's antiobesity and antiinflammatory functionality affect inhibition of muscle protein degradation, and consequently, these interactions by LU exerts a protective effect against obese sarcopenia.
Collapse
Affiliation(s)
- Ji-Won Kim
- Department of Food Science and Nutrition, Kyungpook National University, 80, Daehak-ro, Buk-Ku, Daegu, 41566, Republic of Korea
- Center for Food and Nutritional Genomics Research, Kyungpook National University, 80, Daehak-ro, Buk-Ku, Daegu, 41566, Republic of Korea
| | - Su-Kyung Shin
- Department of Food Science and Nutrition, Kyungpook National University, 80, Daehak-ro, Buk-Ku, Daegu, 41566, Republic of Korea
- Center for Food and Nutritional Genomics Research, Kyungpook National University, 80, Daehak-ro, Buk-Ku, Daegu, 41566, Republic of Korea
| | - Eun-Young Kwon
- Department of Food Science and Nutrition, Kyungpook National University, 80, Daehak-ro, Buk-Ku, Daegu, 41566, Republic of Korea
- Center for Food and Nutritional Genomics Research, Kyungpook National University, 80, Daehak-ro, Buk-Ku, Daegu, 41566, Republic of Korea
| |
Collapse
|
26
|
Latteri S, Sofia M, Puleo S, Di Vincenzo A, Cinti S, Castorina S. Mechanisms linking bariatric surgery to adipose tissue, glucose metabolism, fatty liver disease and gut microbiota. Langenbecks Arch Surg 2023; 408:101. [PMID: 36826628 PMCID: PMC9957865 DOI: 10.1007/s00423-023-02821-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 01/25/2023] [Indexed: 02/25/2023]
Abstract
PURPOSE In the last 20 years, bariatric surgery has achieved an important role in translational and clinical research because of obesity comorbidities. Initially, a tool to lose weight, bariatric surgery now has been shown to be involved in several metabolic pathways. METHODS We conducted a narrative review discussing the underlying mechanisms that could explain the impact of bariatric surgery and the relationship between obesity and adipose tissue, T2D, gut microbiota, and NAFLD. RESULTS Bariatric surgery has an impact in the relation between obesity and type 2 diabetes, but in addition it induces the white-to-brown adipocyte trans-differentiation, by enhancing thermogenesis. Another issue is the connection of bariatric surgery with the gut microbiota and its role in the complex mechanism underlying weight gain. CONCLUSION Bariatric surgery modifies gut microbiota, and these modifications influence lipid metabolism, leading to improvement of non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Saverio Latteri
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy
| | - Maria Sofia
- Department of General Surgery, Cannizzaro Hospital, Via Messina 829, 95126, Catania, Italy.
| | - Stefano Puleo
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| | - Angelica Di Vincenzo
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Via Tronto 10A, 60020, Ancona, Italy
| | - Saverio Cinti
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Via Tronto 10A, 60020, Ancona, Italy
| | - Sergio Castorina
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| |
Collapse
|
27
|
Thangavel H, Dhanyalayam D, Lizardo K, Oswal N, Dolgov E, Perlin DS, Nagajyothi JF. Susceptibility of Fat Tissue to SARS-CoV-2 Infection in Female hACE2 Mouse Model. Int J Mol Sci 2023; 24:1314. [PMID: 36674830 PMCID: PMC9863100 DOI: 10.3390/ijms24021314] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/30/2022] [Accepted: 12/24/2022] [Indexed: 01/12/2023] Open
Abstract
The coronavirus disease (COVID-19) is a highly contagious viral illness caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). COVID-19 has had a catastrophic effect globally causing millions of deaths worldwide and causing long-lasting health complications in COVID-19 survivors. Recent studies including ours have highlighted that adipose tissue can act as a reservoir where SARS-CoV-2 can persist and cause long-term health problems. Here, we evaluated the effect of SARS-CoV-2 infection on adipose tissue physiology and the pathogenesis of fat loss in a murine COVID-19 model using humanized angiotensin-converting enzyme 2 (hACE2) mice. Since epidemiological studies reported a higher mortality rate of COVID-19 in males than in females, we examined hACE2 mice of both sexes and performed a comparative analysis. Our study revealed for the first time that: (a) viral loads in adipose tissue and the lungs differ between males and females in hACE2 mice; (b) an inverse relationship exists between the viral loads in the lungs and adipose tissue, and it differs between males and females; and (c) CoV-2 infection alters immune signaling and cell death signaling differently in SARS-CoV-2 infected male and female mice. Overall, our data suggest that adipose tissue and loss of fat cells could play important roles in determining susceptibility to CoV-2 infection in a sex-dependent manner.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jyothi F. Nagajyothi
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| |
Collapse
|
28
|
Ohm H, Abdel-Rahman O. Impact of Patient Characteristics on the Outcomes of Patients with Gastrointestinal Cancers Treated with Immune Checkpoint Inhibitors. Curr Oncol 2023; 30:786-802. [PMID: 36661709 PMCID: PMC9858132 DOI: 10.3390/curroncol30010060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/01/2023] [Accepted: 01/03/2023] [Indexed: 01/11/2023] Open
Abstract
Gastrointestinal (GI) cancers are a group of malignancies that globally account for a significant portion of cancer incidence and cancer-related death. Survival outcomes for esophageal, gastric, pancreatic, and hepatobiliary cancers remain poor, but new treatment paradigms are emerging with the advent of immune checkpoint inhibitor (ICI) therapy. This review characterizes patient-related prognostic factors that influence the response to ICI therapy. We performed an analysis of the landmark randomized clinical trials in esophageal, gastric, colorectal, hepatocellular, pancreatic, and biliary tract cancers in terms of patient demographic factors. A literature review of smaller retrospective studies investigating patient-related factors was completed. The immunological bases for these associations were further explored. The key predictive factors identified include age, sex, performance status, geography, body mass index, sarcopenia, gut microbiome, various biochemical factors, and disease distribution.
Collapse
Affiliation(s)
- Hyejee Ohm
- Department of Medicine, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Omar Abdel-Rahman
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| |
Collapse
|
29
|
|
30
|
Lin WY, Chen JD. The association between eosinophil count, serum lipids and metabolic syndrome in Taiwanese. Am J Med Sci 2023; 365:37-41. [PMID: 35917948 DOI: 10.1016/j.amjms.2022.07.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 04/24/2022] [Accepted: 07/27/2022] [Indexed: 01/04/2023]
Abstract
BACKGROUND Eosinophil count, dyslipidemia, and metabolic syndrome (MetS) are associated with systemic inflammation. We conducted this large population-based study to investigate the association between elevated eosinophil count, serum lipids, and MetS in the Taiwanese population. METHODS A cross-sectional study of 10,357 adults who underwent health checkups at Shin Kong Wu Ho-Su Memorial Hospital in Taiwan between January 2006 and December 2016 was conducted. MetS was defined according to criteria modified by the International Diabetes Federation specifically for the Chinese population. The measurement of serum lipids included high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), apolipoprotein A-I (ApoA-I), and apolipoprotein B (ApoB). Pearson's correlation and linear regression were used to determine the association of eosinophil count with waist circumference, blood pressure, fasting glucose, and serum lipids. Multivariate logistic regression analysis was used to determine the odds ratio of MetS and abnormal serum lipid levels in each eosinophil count quartile. RESULTS Eosinophil count was positively associated with triglycerides levels and negatively associated with HDL-C levels; however, it was not significantly associated with ApoA-I and ApoB. The odds ratio of MetS increased significantly across eosinophil count quartiles. The adjusted odds ratios of MetS for the second, third, and fourth quartiles were 1.28, 1.38, and 1.42, respectively, with reference to the first quartile. CONCLUSIONS High eosinophil count is an independent risk factor for MetS. Hypertriglyceridemia and hypo-HDL-cholesterolemia might partly contribute to this result.
Collapse
Affiliation(s)
- Wei-Yu Lin
- Department of Family Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan.
| | - Jong-Dar Chen
- Department of Family Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan; Department of Medicine, School of Medicine, Fu Jen Catholic University, Taipei, Taiwan.
| |
Collapse
|
31
|
Willemsen ACH, De Moor N, Van Dessel J, Baijens LWJ, Bila M, Hauben E, van den Hout MFCM, Vander Poorten V, Hoeben A, Clement PM, Schols AMWJ. The predictive and prognostic value of weight loss and body composition prior to and during immune checkpoint inhibition in recurrent or metastatic head and neck cancer patients. Cancer Med 2022; 12:7699-7712. [PMID: 36484469 PMCID: PMC10134381 DOI: 10.1002/cam4.5522] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Response rates of immune checkpoint inhibitor (ICI) therapy for recurrent and/or metastatic head and neck squamous cell carcinoma (R/M HNSCC) are low. PATIENTS AND METHODS This retrospective multicentre cohort study evaluates the predictive and prognostic value of weight loss and changes in body composition prior and during therapy. Patient, tumor, and treatment characteristics of 98 patients were retrieved, including neutrophil and platelet-lymphocyte-ratio (NLR and PLR). Programmed death-ligand 1 (PD-L1) expression was determined on residual material. Cachexia was defined according to Fearon et al. (2011). Skeletal muscle (SM), visceral adipose tissue (VAT), and subcutaneous adipose tissue (SAT) were evaluated on computed tomography scans at the third lumbar vertebrae level. Univariable and multivariable regression analyses were performed for 6 months progression free survival (PFS6m) and overall survival (OS). RESULTS Significant early weight loss (>2%) during the first 6 weeks of therapy was shown in 34 patients (35%). This patient subgroup had a significantly higher NLR and PLR at baseline. NLR and PLR were inversely correlated with SM and VAT index. Independent predictors of PFS6m were lower World Health Organization performance status (HR 0.16 [0.04-0.54] p = 0.003), higher baseline SAT index (HR 1.045 [1.02-1.08] p = 0.003), and weight loss <2% (HR 0.85 [0.74-0.98] p = 0.03). Baseline cachexia in combination with >2% early weight loss remained a predictor of OS, independent of PD-L1 expression (HR 2.09 [1.11-3.92] p = 0.02, HR 2.18 [1.13-4.21] p = 0.02). CONCLUSION We conclude that the combination of cachexia at baseline and weight loss during ICI therapy is associated with worse OS in R/M HNSCC patients, independent of PD-L1 expression.
Collapse
Affiliation(s)
- Anna C. H. Willemsen
- Division of Medical Oncology, Department of Internal Medicine Maastricht University Medical Center+ Maastricht The Netherlands
- GROW‐School of Oncology and Developmental Biology Maastricht University Medical Center+ Maastricht The Netherlands
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism Maastricht University Medical Center+ Maastricht The Netherlands
| | - Nina De Moor
- Department of Oncology Leuven Cancer Institute, KU Leuven Leuven Belgium
| | - Jeroen Van Dessel
- Department of Biomedical Sciences, KU Leuven & Oral and Maxillofacial Surgery University Hospitals Leuven Leuven Belgium
| | - Laura W. J. Baijens
- GROW‐School of Oncology and Developmental Biology Maastricht University Medical Center+ Maastricht The Netherlands
- Department of Otorhinolaryngology, Head and Neck Surgery Maastricht University Medical Center+ Maastricht The Netherlands
| | - Michel Bila
- Department of Biomedical Sciences, KU Leuven & Oral and Maxillofacial Surgery University Hospitals Leuven Leuven Belgium
| | - Esther Hauben
- Department of Pathology University Hospitals Leuven Leuven Belgium
| | - Mari F. C. M. van den Hout
- GROW‐School of Oncology and Developmental Biology Maastricht University Medical Center+ Maastricht The Netherlands
- Department of Pathology Maastricht University Medical Center+ Maastricht The Netherlands
| | - Vincent Vander Poorten
- Department of Oncology Leuven Cancer Institute, KU Leuven Leuven Belgium
- Otorhinolaryngology Head and Neck Surgery University Hospitals Leuven, Leuven Cancer Institute, KU Leuven Leuven Belgium
| | - Ann Hoeben
- Division of Medical Oncology, Department of Internal Medicine Maastricht University Medical Center+ Maastricht The Netherlands
- GROW‐School of Oncology and Developmental Biology Maastricht University Medical Center+ Maastricht The Netherlands
| | - Paul M. Clement
- Department of Oncology Leuven Cancer Institute, KU Leuven Leuven Belgium
| | - Annemie M. W. J. Schols
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism Maastricht University Medical Center+ Maastricht The Netherlands
| |
Collapse
|
32
|
Role of NKT cells in cancer immunotherapy-from bench to bed. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 40:29. [PMID: 36460881 DOI: 10.1007/s12032-022-01888-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 11/08/2022] [Indexed: 12/04/2022]
Abstract
Natural killer T (NKT) cells are a specific T cell subset known to express the αβ-T cell receptor (TCR) for antigens identification and express typical NK cell specifications, such as surface expression of CD56 and CD16 markers as well as production of granzyme. Human NKT cells are divided into two subgroups based on their cytokine receptor and TCR repertoire. Both of them are CD1-restricted and recognize lipid antigens presented by CD1d molecules. Studies have demonstrated that these cells are essential in defense against malignancies. These cells secret proinflammatory and regulatory cytokines that stimulate or suppress immune system responses. In several murine tumor models, activation of type I NKT cells induces tumor rejection and inhibits metastasis's spread. However, type II NKT cells are associated with an inhibitory and regulatory function during tumor immune responses. Variant NKT cells may suppress tumor immunity via different mechanisms that require cross-talk with other immune-regulatory cells. NKT-like cells display high tumor-killing abilities against many tumor cells. In the recent decade, different studies have been performed based on the application of NKT-based immunotherapy for cancer therapy. Moreover, manipulation of NKT cells through administering autologous dendritic cell (DC) loaded with α-galactosylceramide (α-GalCer) and direct α-GalCer injection has also been tested. In this review, we described different subtypes of NKT cells, their function in the anti-tumor immune responses, and the application of NKT cells in cancer immunotherapy from bench to bed.
Collapse
|
33
|
Morais JBS, Dias TMDS, Cardoso BEP, de Paiva Sousa M, Sousa TGVD, Araújo DSCD, Marreiro DDN. Adipose Tissue Dysfunction: Impact on Metabolic Changes? Horm Metab Res 2022; 54:785-794. [PMID: 35952684 DOI: 10.1055/a-1922-7052] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Adipose tissue is a metabolically dynamic organ that is the primary site of storage for excess energy, but it serves as an endocrine organ capable of synthesizing a number of biologically active compounds that regulate metabolic homeostasis. However, when the capacity of expansion of this tissue exceeds, dysfunction occurs, favoring ectopic accumulation of fat in the visceral, which has been implicated in several disease states, most notably obesity. This review highlights the mechanisms involved in the structure of adipose tissue, tissue expandability, adipocyte dysfunction, as well as the impact of these events on the manifestation of important metabolic disorders associated with adipose tissue dysfunction. A literature search using Pubmed, Web of Science, Scopus, and Cochrane databases were used to identify relevant studies, using clinical trials, experimental studies in animals and humans, case-control studies, case series, letters to the editor, and review articles published in English, without restrictions on year of publication. The excessive ectopic lipid accumulation leads to local inflammation and insulin resistance. Indeed, overnutrition triggers uncontrolled inflammatory responses white adipose tissue, leading to chronic low-grade inflammation, therefore fostering the progression of important metabolic disorders. Thus, it is essential to advance the understanding of the molecular mechanisms involved in adipose tissue dysfunction in order to mitigate the negative metabolic consequences of obesity.
Collapse
|
34
|
Shree N, Ding Z, Flaws J, Choudhury M. Role of microRNA in Endocrine Disruptor-Induced Immunomodulation of Metabolic Health. Metabolites 2022; 12:1034. [PMID: 36355117 PMCID: PMC9695656 DOI: 10.3390/metabo12111034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 01/22/2025] Open
Abstract
The prevalence of poor metabolic health is growing exponentially worldwide. This condition is associated with complex comorbidities that lead to a compromised quality of life. One of the contributing factors recently gaining attention is exposure to environmental chemicals, such as endocrine-disrupting chemicals (EDCs). Considerable evidence suggests that EDCs can alter the endocrine system through immunomodulation. More concerning, EDC exposure during the fetal development stage has prominent adverse effects later in life, which may pass on to subsequent generations. Although the mechanism of action for this phenomenon is mostly unexplored, recent reports implicate that non-coding RNAs, such as microRNAs (miRs), may play a vital role in this scenario. MiRs are significant contributors in post-transcriptional regulation of gene expression. Studies demonstrating the immunomodulation of EDCs via miRs in metabolic health or towards the Developmental Origins of Health and Disease (DOHaD) Hypothesis are still deficient. The aim of the current review was to focus on studies that demonstrate the impact of EDCs primarily on innate immunity and the potential role of miRs in metabolic health.
Collapse
Affiliation(s)
- Nitya Shree
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University (TAMU), College Station, TX 77843, USA
| | - Zehuan Ding
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University (TAMU), College Station, TX 77843, USA
| | - Jodi Flaws
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Mahua Choudhury
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University (TAMU), College Station, TX 77843, USA
| |
Collapse
|
35
|
Raza GS, Sodum N, Kaya Y, Herzig KH. Role of Circadian Transcription Factor Rev-Erb in Metabolism and Tissue Fibrosis. Int J Mol Sci 2022; 23:12954. [PMID: 36361737 PMCID: PMC9655416 DOI: 10.3390/ijms232112954] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/21/2022] [Accepted: 10/22/2022] [Indexed: 09/12/2023] Open
Abstract
Circadian rhythms significantly affect metabolism, and their disruption leads to cardiometabolic diseases and fibrosis. The clock repressor Rev-Erb is mainly expressed in the liver, heart, lung, adipose tissue, skeletal muscles, and brain, recognized as a master regulator of metabolism, mitochondrial biogenesis, inflammatory response, and fibrosis. Fibrosis is the response of the body to injuries and chronic inflammation with the accumulation of extracellular matrix in tissues. Activation of myofibroblasts is a key factor in the development of organ fibrosis, initiated by hormones, growth factors, inflammatory cytokines, and mechanical stress. This review summarizes the importance of Rev-Erb in ECM remodeling and tissue fibrosis. In the heart, Rev-Erb activation has been shown to alleviate hypertrophy and increase exercise capacity. In the lung, Rev-Erb agonist reduced pulmonary fibrosis by suppressing fibroblast differentiation. In the liver, Rev-Erb inhibited inflammation and fibrosis by diminishing NF-κB activity. In adipose tissue, Rev- Erb agonists reduced fat mass. In summary, the results of multiple studies in preclinical models demonstrate that Rev-Erb is an attractive target for positively influencing dysregulated metabolism, inflammation, and fibrosis, but more specific tools and studies would be needed to increase the information base for the therapeutic potential of these substances interfering with the molecular clock.
Collapse
Affiliation(s)
- Ghulam Shere Raza
- Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Nalini Sodum
- Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Yagmur Kaya
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Marmara University, 34854 Istanbul, Turkey
| | - Karl-Heinz Herzig
- Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
- Oulu University Hospital, University of Oulu, 90220 Oulu, Finland
- Pediatric Gastroenterology and Metabolic Diseases, Pediatric Institute, Poznan University of Medical Sciences, 60-572 Poznań, Poland
| |
Collapse
|
36
|
Liu X, Jiang Y, Ye J, Wang X. Helminth infection and helminth-derived products: A novel therapeutic option for non-alcoholic fatty liver disease. Front Immunol 2022; 13:999412. [PMID: 36263053 PMCID: PMC9573989 DOI: 10.3389/fimmu.2022.999412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is closely related to obesity, diabetes, and metabolic syndrome (MetS), and it has become the most common chronic liver disease. Helminths have co-evolved with humans, inducing multiple immunomodulatory mechanisms to modulate the host's immune system. By using their immunomodulatory ability, helminths and their products exhibit protection against various autoimmune and inflammatory diseases, including obesity, diabetes, and MetS, which are closely associated with NAFLD. Here, we review the pathogenesis of NAFLD from abnormal glycolipid metabolism, inflammation, and gut dysbiosis. Correspondingly, helminths and their products can treat or relieve these NAFLD-related diseases, including obesity, diabetes, and MetS, by promoting glycolipid metabolism homeostasis, regulating inflammation, and restoring the balance of gut microbiota. Considering that a large number of clinical trials have been carried out on helminths and their products for the treatment of inflammatory diseases with promising results, the treatment of NAFLD and obesity-related diseases by helminths is also a novel direction and strategy.
Collapse
Affiliation(s)
- Xi Liu
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yuyun Jiang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jixian Ye
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xuefeng Wang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Department of Nuclear Medicine and Institute of Digestive Diseases, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
37
|
Harris BHL, Macaulay VM, Harris DA, Klenerman P, Karpe F, Lord SR, Harris AL, Buffa FM. Obesity: a perfect storm for carcinogenesis. Cancer Metastasis Rev 2022; 41:491-515. [PMID: 36038791 PMCID: PMC9470699 DOI: 10.1007/s10555-022-10046-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/08/2022] [Indexed: 12/14/2022]
Abstract
Obesity-related cancers account for 40% of the cancer cases observed in the USA and obesity is overtaking smoking as the most widespread modifiable risk factor for carcinogenesis. Here, we use the hallmarks of cancer framework to delineate how obesity might influence the carcinogenic hallmarks in somatic cells. We discuss the effects of obesity on (a) sustaining proliferative signaling; (b) evading growth suppressors; (c) resisting cell death; (d) enabling replicative immortality; (e) inducing angiogenesis; (f) activating invasion and metastasis; (g) reprogramming energy metabolism; and (h) avoiding immune destruction, together with its effects on genome instability and tumour-promoting inflammation. We present the current understanding and controversies in this evolving field, and highlight some areas in need of further cross-disciplinary focus. For instance, the relative importance of the many potentially causative obesity-related factors is unclear for each type of malignancy. Even within a single tumour type, it is currently unknown whether one obesity-related factor consistently plays a predominant role, or if this varies between patients or, even in a single patient with time. Clarifying how the hallmarks are affected by obesity may lead to novel prevention and treatment strategies for the increasingly obese population.
Collapse
Affiliation(s)
- Benjamin H L Harris
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK.
- St Anne's College, 56 Woodstock Rd, Oxford, OX2 6HS, UK.
| | - Valentine M Macaulay
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, OX3 9DU, UK
| | | | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, OX1 3SY, UK
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Oxford, Oxford, OX3 7LE, UK
| | - Simon R Lord
- Early Phase Clinical Trials Unit, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Adrian L Harris
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | | |
Collapse
|
38
|
Velluzzi F, Pisanu S, Galletta M, Fosci M, Secci G, Deledda A, Boi F, Rodia R, Fanciulli G, Delitala AP, Sainas G, Loviselli A. Association between High Normal TSH Levels and Obesity in Women with Anti-Thyroid Autoantibodies (ATAs). J Clin Med 2022; 11:jcm11175125. [PMID: 36079055 PMCID: PMC9457091 DOI: 10.3390/jcm11175125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/06/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
A positive correlation between Thyroid-Stimulating Hormone (TSH) and Body Mass Index (BMI) has been reported in many studies, but data on this topic remain controversial, especially when TSH values are in the normal range. Moreover, few studies have evaluated the co-existence of thyroid autoimmunity. This study investigated the role of thyroid autoimmunity in the interconnection between TSH, BMI, and waist circumference (WC) in euthyroid patients with overweight or obesity. We enrolled 902 patients (213 males; mean age ± SD: 45 ± 14 years; mean BMI ± SD: 35.8 ± 6.5 kg/m2), with normal serum TSH concentration; anti-thyroid autoantibodies (ATAs) were evaluated in 752 patients (186 males). Patients were divided into four BMI classes, based on WHO criteria, and the relationship between BMI, WC, and TSH was evaluated in the whole sample and compared to ATAs positivity, observed in 235 patients (44 males). No significant difference was found between TSH levels in the BMI classes. A statistically significant correlation between TSH and BMI was found only in ATAs-positive females (N = 191, Spearman rho: 0.149; p-value: 0.040). However, this finding was not confirmed when considering the WC. Our study shows a positive correlation only between TSH and BMI in obese women with positive ATAs, suggesting that in these patients, the high normal levels of TSH could be attributed to a mild thyroid failure with a possible worsening obesity-related effect, and both need a careful evaluation.
Collapse
Affiliation(s)
- Fernanda Velluzzi
- Endocrinology, and Obesity Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Silvia Pisanu
- Endocrinology, and Obesity Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Maura Galletta
- Hygiene Section, Department of Medical Sciences and Public Health, University of Cagliari, 09042 Cagliari, Italy
| | - Michele Fosci
- Endocrinology, and Obesity Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
- Correspondence:
| | - Gianni Secci
- Endocrinology, and Obesity Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Andrea Deledda
- Endocrinology, and Obesity Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Francesco Boi
- Endocrinology, and Obesity Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Rossella Rodia
- Endocrinology, and Obesity Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Giuseppe Fanciulli
- Department of Surgical, Medical and Experimental Sciences, University of Sassari-Endocrine Unit, AOU Sassari, 07100 Sassari, Italy
| | - Alessandro Palmerio Delitala
- Department of Surgical, Medical and Experimental Sciences, University of Sassari-Endocrine Unit, AOU Sassari, 07100 Sassari, Italy
| | - Gianmarco Sainas
- Endocrinology, and Obesity Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Andrea Loviselli
- Endocrinology, and Obesity Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| |
Collapse
|
39
|
Hertzel AV, Yong J, Chen X, Bernlohr DA. Immune Modulation of Adipocyte Mitochondrial Metabolism. Endocrinology 2022; 163:6618136. [PMID: 35752995 PMCID: PMC9653008 DOI: 10.1210/endocr/bqac094] [Citation(s) in RCA: 4] [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: 05/06/2022] [Indexed: 11/19/2022]
Abstract
Immune cells infiltrate adipose tissue as a function of age, sex, and diet, leading to a variety of regulatory processes linked to metabolic disease and dysfunction. Cytokines and chemokines produced by resident macrophages, B cells, T cells and eosinophils play major role(s) in fat cell mitochondrial functions modulating pyruvate oxidation, electron transport and oxidative stress, branched chain amino acid metabolism, fatty acid oxidation, and apoptosis. Indeed, cytokine-dependent downregulation of numerous genes affecting mitochondrial metabolism is strongly linked to the development of the metabolic syndrome, whereas the potentiation of mitochondrial metabolism represents a counterregulatory process improving metabolic outcomes. In contrast, inflammatory cytokines activate mitochondrially linked cell death pathways such as apoptosis, pyroptosis, necroptosis, and ferroptosis. As such, the adipocyte mitochondrion represents a major intersection point for immunometabolic regulation of central metabolism.
Collapse
Affiliation(s)
- Ann V Hertzel
- Department of Biochemistry, Molecular Biology and Biophysics, The University of Minnesota, Minneapolis, MN 55455, USA
| | - Jeongsik Yong
- Department of Biochemistry, Molecular Biology and Biophysics, The University of Minnesota, Minneapolis, MN 55455, USA
| | - Xiaoli Chen
- Department of Food Science and Nutrition, The University of Minnesota, Minneapolis, MN 55455, USA
| | - David A Bernlohr
- Correspondence: David A. Bernlohr, PhD, Department of Biochemistry, Molecular Biology and Biophysics, The University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
40
|
Sen T, Thummer RP. The Impact of Human Microbiotas in Hematopoietic Stem Cell and Organ Transplantation. Front Immunol 2022; 13:932228. [PMID: 35874759 PMCID: PMC9300833 DOI: 10.3389/fimmu.2022.932228] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/06/2022] [Indexed: 11/18/2022] Open
Abstract
The human microbiota heavily influences most vital aspects of human physiology including organ transplantation outcomes and transplant rejection risk. A variety of organ transplantation scenarios such as lung and heart transplantation as well as hematopoietic stem cell transplantation is heavily influenced by the human microbiotas. The human microbiota refers to a rich, diverse, and complex ecosystem of bacteria, fungi, archaea, helminths, protozoans, parasites, and viruses. Research accumulating over the past decade has established the existence of complex cross-species, cross-kingdom interactions between the residents of the various human microbiotas and the human body. Since the gut microbiota is the densest, most popular, and most studied human microbiota, the impact of other human microbiotas such as the oral, lung, urinary, and genital microbiotas is often overshadowed. However, these microbiotas also provide critical and unique insights pertaining to transplantation success, rejection risk, and overall host health, across multiple different transplantation scenarios. Organ transplantation as well as the pre-, peri-, and post-transplant pharmacological regimens patients undergo is known to adversely impact the microbiotas, thereby increasing the risk of adverse patient outcomes. Over the past decade, holistic approaches to post-transplant patient care such as the administration of clinical and dietary interventions aiming at restoring deranged microbiota community structures have been gaining momentum. Examples of these include prebiotic and probiotic administration, fecal microbial transplantation, and bacteriophage-mediated multidrug-resistant bacterial decolonization. This review will discuss these perspectives and explore the role of different human microbiotas in the context of various transplantation scenarios.
Collapse
Affiliation(s)
| | - Rajkumar P. Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| |
Collapse
|
41
|
Crosstalk between macrophages and innate lymphoid cells (ILCs) in diseases. Int Immunopharmacol 2022; 110:108937. [PMID: 35779490 DOI: 10.1016/j.intimp.2022.108937] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/15/2022]
Abstract
Innate lymphoid cells (ILCs) and macrophages are tissue-resident cells that play important roles in tissue-immune homeostasis and immune regulation. ILCs are mainly distributed on the barrier surfaces of mammals to ensure immunity or tissue homeostasis following host, microbial, or environmental stimulation. Their complex relationships with different organs enable them to respond quickly to disturbances in environmental conditions and organ homeostasis, such as during infections and tissue damage. Gradually emerging evidence suggests that ILCs also play complex and diverse roles in macrophage development, homeostasis, polarization, inflammation, and viral infection. In turn, macrophages also determine the fate of ILCs to some extent, which indicates that network crossover between these interactions is a key determinant of the immune response. More work is needed to better define the crosstalk of ILCs with macrophages in different tissues and demonstrate how it is affected during inflammation and other diseases. Here, we summarize current research on the functional interactions between ILCs and macrophages and consider the potential therapeutic utility of these interactions for the benefit of human health.
Collapse
|
42
|
Abstract
Adipose tissue is a complex dynamic organ with whole-body immunometabolic influence. Much of the work into understanding the role of immune cells in adipose tissue has been in the context of obesity. These investigations have also uncovered a range of typical (immune) and non-typical functions exerted by adipose tissue leukocytes. Here we provide an overview of the adipose tissue immune system, including its role as an immune reservoir in the whole-body response to infection and as a site of parasitic and viral infections. We also describe the functional roles of specialized immunological structures found within adipose tissue. However, our main focus is on the recently discovered 'non-immune' functions of adipose tissue immune cells, which include the regulation of adipocyte homeostasis, as well as responses to changing nutrient status and body temperature. In doing so, we outline the therapeutic potential of the adipose tissue immune system in health and disease.
Collapse
|
43
|
Low-grade inflammation, CoVID-19, and obesity: clinical aspect and molecular insights in childhood and adulthood. Int J Obes (Lond) 2022; 46:1254-1261. [PMID: 35393519 PMCID: PMC8988546 DOI: 10.1038/s41366-022-01111-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 02/21/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022]
Abstract
The new 2019 coronavirus 19 disease (CoVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a serious threat to health systems. As a global health problem, this pandemic poses a huge threat to people and is responsible for significant morbidity and mortality worldwide. On the other hand, obesity has also reached epidemic proportions and poses another challenge to the healthcare system. There is increasing evidence of a strong association between obesity and CoVID-19 disease, but the mechanisms underlying the link between the two remain unclear and the role of obesity also remains to be elucidated. In particular obesity-related low-grade inflammation has been hypothesized as the Achille's heel that could predispose subjects with obesity to a more severe CoVID-19 compared to subjects with normal weight. Hence, we summarized recent evidence on the role of low-grade inflammation in clinical aspects of CoVID-19 in subjects with obesity in both childhood and adulthood. Further, we provide molecular insights to explain this link.
Collapse
|
44
|
Sbierski-Kind J, Grenkowitz S, Schlickeiser S, Sandforth A, Friedrich M, Kunkel D, Glauben R, Brachs S, Mai K, Thürmer A, Radonić A, Drechsel O, Turnbaugh PJ, Bisanz JE, Volk HD, Spranger J, von Schwartzenberg RJ. Effects of caloric restriction on the gut microbiome are linked with immune senescence. MICROBIOME 2022; 10:57. [PMID: 35379337 PMCID: PMC8978410 DOI: 10.1186/s40168-022-01249-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/07/2022] [Indexed: 05/05/2023]
Abstract
BACKGROUND Caloric restriction can delay the development of metabolic diseases ranging from insulin resistance to type 2 diabetes and is linked to both changes in the composition and metabolic function of the gut microbiota and immunological consequences. However, the interaction between dietary intake, the microbiome, and the immune system remains poorly described. RESULTS We transplanted the gut microbiota from an obese female before (AdLib) and after (CalRes) an 8-week very-low-calorie diet (800 kcal/day) into germ-free mice. We used 16S rRNA sequencing to evaluate taxa with differential abundance between the AdLib- and CalRes-microbiota recipients and single-cell multidimensional mass cytometry to define immune signatures in murine colon, liver, and spleen. Recipients of the CalRes sample exhibited overall higher alpha diversity and restructuring of the gut microbiota with decreased abundance of several microbial taxa (e.g., Clostridium ramosum, Hungatella hathewayi, Alistipi obesi). Transplantation of CalRes-microbiota into mice decreased their body fat accumulation and improved glucose tolerance compared to AdLib-microbiota recipients. Finally, the CalRes-associated microbiota reduced the levels of intestinal effector memory CD8+ T cells, intestinal memory B cells, and hepatic effector memory CD4+ and CD8+ T cells. CONCLUSION Caloric restriction shapes the gut microbiome which can improve metabolic health and may induce a shift towards the naïve T and B cell compartment and, thus, delay immune senescence. Understanding the role of the gut microbiome as mediator of beneficial effects of low calorie diets on inflammation and metabolism may enhance the development of new therapeutic treatment options for metabolic diseases. TRIAL REGISTRATION NCT01105143 , "Effects of negative energy balance on muscle mass regulation," registered 16 April 2010. Video Abstract.
Collapse
Affiliation(s)
- Julia Sbierski-Kind
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Chariteplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Sophia Grenkowitz
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Stephan Schlickeiser
- BIH Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin and Berlin Institute of Health (BIH), Berlin, Germany
| | - Arvid Sandforth
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Chariteplatz 1, 10117, Berlin, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
| | - Marie Friedrich
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Chariteplatz 1, 10117, Berlin, Germany
| | - Désirée Kunkel
- Berlin Institute of Health (BIH), Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Flow & Mass Cytometry Core Facility, Berlin, Germany
| | - Rainer Glauben
- Medical Department for Gastroenterology, Infectious Diseases and Rheumatology, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sebastian Brachs
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Chariteplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Knut Mai
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Chariteplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | | | | | | | - Peter J Turnbaugh
- Department of Microbiology & Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Jordan E Bisanz
- Department of Microbiology & Immunology, University of California San Francisco, San Francisco, CA, USA
- Department for Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, USA
| | - Hans-Dieter Volk
- Berlin Institute of Health (BIH), Berlin, Germany
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Joachim Spranger
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Chariteplatz 1, 10117, Berlin, Germany.
- Berlin Institute of Health (BIH), Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
| | - Reiner Jumpertz von Schwartzenberg
- Department of Endocrinology and Metabolism, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Chariteplatz 1, 10117, Berlin, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology and Nephrology, Eberhard-Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| |
Collapse
|
45
|
Moreno-Fernandez J, Ochoa J, Ojeda ML, Nogales F, Carreras O, Díaz-Castro J. Inflammation and oxidative stress, the links between obesity and COVID-19: a narrative review. J Physiol Biochem 2022; 78:581-591. [PMID: 35316507 PMCID: PMC8938224 DOI: 10.1007/s13105-022-00887-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 03/03/2022] [Indexed: 02/06/2023]
Abstract
COVID-19, an acute respiratory disease caused by SARS-CoV-2, has rapidly become a pandemic. On the other hand, obesity is also reaching dramatic dimensions and it is a risk factor for morbidity and premature mortality. Obesity has been linked to a high risk of serious-associated complications to COVID-19, due to the increased risk of concomitant chronic diseases, which highlights the health public relevance of the topic. Obese subjects have a pro-inflammatory environment, which can further exacerbate COVID-19-induced inflammation and oxidative stress, explaining the increased risk of serious complications in these patients. Another factor that favors infection in obese patients is the high expression of ACE2 receptors in the adipose tissue. The negative impact of COVID-19 in obesity is also associated with a decrease in respiratory function, the concurrence of multiple comorbidities, a low-degree chronic inflammatory state, immunocompromised situation, and therefore a higher rate of hospitalization, mechanical ventilation, in-hospital complications such as pneumonia, and death. In this review, the link between obesity and COVID-19 was analyzed, exploring the potential common mechanisms in both diseases, with special attention to oxidative stress and inflammation, due to the crucial role of both pathways in the development of the disease.
Collapse
Affiliation(s)
- Jorge Moreno-Fernandez
- Department of Physiology, University of Granada, Granada, Spain.
- Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, Avenida del Conocimiento s/n, , 18071, Armilla, Granada, Spain.
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Seville, Seville, Spain.
- Instituto de Investigación Biosanitaria (IBS), 18016, Granada, Spain.
| | - Julio Ochoa
- Department of Physiology, University of Granada, Granada, Spain.
- Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, Avenida del Conocimiento s/n, , 18071, Armilla, Granada, Spain.
| | - María Luisa Ojeda
- Department of Physiology, University of Seville University, Seville, Spain
| | - Fátima Nogales
- Department of Physiology, University of Seville University, Seville, Spain
| | - Olimpia Carreras
- Department of Physiology, University of Seville University, Seville, Spain
| | - Javier Díaz-Castro
- Department of Physiology, University of Granada, Granada, Spain
- Institute of Nutrition and Food Technology "José Mataix Verdú", University of Granada, Avenida del Conocimiento s/n, , 18071, Armilla, Granada, Spain
- Instituto de Investigación Biosanitaria (IBS), 18016, Granada, Spain
| |
Collapse
|
46
|
Li Q, Wang L, Wu J, Wang J, Wang Y, Zeng X. Role of age, gender and ethnicity in the association between visceral adiposity index and non-alcoholic fatty liver disease among US adults (NHANES 2003-2018): cross-sectional study. BMJ Open 2022; 12:e058517. [PMID: 35314476 PMCID: PMC8938699 DOI: 10.1136/bmjopen-2021-058517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVES The association between visceral adiposity index (VAI) and the prevalence of non-alcoholic fatty liver disease (NAFLD) has not been fully determined. Here, we aimed to explore the association between VAI and NAFLD in the general US population, and further investigate whether the association involves population differences. DESIGN Cross-sectional population-based study. SETTING The National Health and Nutrition Examination Survey (2003-2018). PARTICIPANTS A total of 7522 participants aged 20 years or older who have complete information for NAFLD assessment test were included in this study. PRIMARY AND SECONDARY OUTCOME MEASURES NAFLD was assessed by the modified fatty liver index for the US population (USFLI) using a cut-off point of 30. Correlation between VAI and NAFLD prediction scores was calculated using the partial correlation analysis. Logistic regression models were further used to estimate ORs and 95% CIs. RESULTS Insulin resistance (IR), inflammation and waist circumference-adjusted partial correlation analysis indicated that VAI scores were positively correlated with USFLI (r=0.404 for men, and r=0.395 for women; p<0.001). In a comparison of the highest versus the lowest quartiles of VAI, multivariable logistic regression analysis demonstrated a positive association between VAI and NAFLD (OR (95% CI)=1.97 (1.12 to 3.47) for men, OR (95% CI)=4.03 (1.98 to 8.20) for women). The stratified analyses revealed that the positive association involves age/gender-specific and ethnic differences. As for the impact of metabolic disorders, our results revealed that the association was independent of IR and diabetes, but it would be confounded by other metabolic disorders. However, no significant association was found between VAI and hepatic fibrosis. CONCLUSION VAI is positively associated with the prevalence of NAFLD, but not hepatic fibrosis among US adults, and the association involves age/gender-specific and ethnic differences. The results reported here have important public health implications in NAFLD screening in the future.
Collapse
Affiliation(s)
- Qianwen Li
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Ling Wang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
- Faculty of Medicine, Macau University of Science and Technology, Macau SAR, People's Republic of China
| | - Jian Wu
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Jing Wang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Yanjie Wang
- School of Management, Xinxiang Medical University, Xinxiang, China
| | - Xin Zeng
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
47
|
Satoh M, Iizuka M, Majima M, Ohwa C, Hattori A, Van Kaer L, Iwabuchi K. Adipose invariant NKT cells interact with CD1d-expressing macrophages to regulate obesity-related inflammation. Immunology 2022; 165:414-427. [PMID: 35137411 DOI: 10.1111/imm.13447] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 01/06/2022] [Accepted: 01/11/2022] [Indexed: 11/28/2022] Open
Abstract
Obesity is accompanied by and accelerated with chronic inflammation in adipose tissue, especially visceral adipose tissue (VAT). This low-level inflammation predisposes the host to the development of metabolic disease, most notably type 2 diabetes. We have focused on the capacity of glycolipid-reactive, CD1d-restricted natural killer T (NKT) cells to modulate obesity and its associated metabolic sequelae. We previously reported that CD1d knockout (KO) mice are partially protected against the development of obesity-associated insulin-resistance, and these findings were recapitulated in mice with an adipocyte-specific CD1d deficiency, suggesting that NKT cell-adipocyte interactions play a critical role in exacerbating disease. However, many other CD1d-expressing cells contribute to the in vivo responses of NKT cells to lipid antigens. In the present study, we examined the role of CD1d expression by macrophages (Mϕ) to the development of obesity-associated metabolic inflammation using LysMcre-cd1d1f/f mice where the CD1d1 gene is disrupted in a Mϕ-specific manner. Unexpectedly, these animals contained a higher frequency of T-bet+ CD4+ T cells in VAT with increased production of Th1-cytokines that aggravated VAT inflammation. Mϕ from mutant mice displayed increased production of IL-12p40, suggesting M1 polarization. These findings indicate that interactions of CD1d on Mϕ with NKT cells play a beneficial role in obesity-associated VAT inflammation and insulin resistance with a sharp contrast to an aggravating role of CD1d on another type of antigen presenting cell, dendritic cells.
Collapse
Affiliation(s)
- Masashi Satoh
- Department of Immunology, Kitasato University School of Medicine.,Program in Cellular Immunology, Graduate School of Medical Sciences, Kitasato University
| | - Misao Iizuka
- Department of Immunology, Kitasato University School of Medicine
| | - Masataka Majima
- Department of Pharmacology, Kitasato University School of Medicine.,Program in Molecular Pharmacology, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan.,School of Health and Medical Sciences, Kanagawa Institute of Technology (KAIT), Atsugi, Japan
| | - Chizuru Ohwa
- Program in Cellular Immunology, Graduate School of Medical Sciences, Kitasato University
| | - Akito Hattori
- Program in Cellular Immunology, Graduate School of Medical Sciences, Kitasato University
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Kazuya Iwabuchi
- Department of Immunology, Kitasato University School of Medicine.,Program in Cellular Immunology, Graduate School of Medical Sciences, Kitasato University
| |
Collapse
|
48
|
Mashayekhi M, Wanjalla CN, Warren CM, Simmons JD, Ghoshal K, Pilkinton M, Bailin SS, Gabriel CL, Pozzi A, Koethe JR, Brown NJ, Kalams SA, Luther JM. The soluble epoxide hydrolase inhibitor GSK2256294 decreases the proportion of adipose pro-inflammatory T cells. Prostaglandins Other Lipid Mediat 2022; 158:106604. [PMID: 34922004 PMCID: PMC8742790 DOI: 10.1016/j.prostaglandins.2021.106604] [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] [Received: 08/15/2021] [Revised: 11/12/2021] [Accepted: 12/11/2021] [Indexed: 02/03/2023]
Abstract
Adipose tissue contains a complex immune environment and is a central contributor to heightened systemic inflammation in obese persons. Epoxyeicosatrienoic acids (EETs) are lipid signaling molecules that decrease inflammation in obese animals, but their effect on inflammation in humans is unknown. The enzyme soluble epoxide hydrolase (sEH) hydrolyzes EETs to less active diols, and we hypothesized that pharmacologic sEH inhibition would decrease adipose inflammation in obese individuals. We treated obese prediabetic adults with the sEH inhibitor GSK2256294 versus placebo in a crossover design, collected subcutaneous abdominal adipose tissue via lipoaspiration and characterized the tissue T cell profile. Treatment with GSK2256294 decreased the percentage of pro-inflammatory T cells producing interferon-gamma (IFNγ), but not interleukin (IL)-17A, and decreased the amount of secreted tumor necrosis factor-alpha (TNFα). Understanding the contribution of the EET/sEH pathway to inflammation in obesity could lead to new strategies to modulate adipose and systemic inflammation.
Collapse
Affiliation(s)
- Mona Mashayekhi
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Nashville, TN, United States.
| | - Celestine N Wanjalla
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Diseases, Nashville, TN, United States
| | - Christian M Warren
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Diseases, Nashville, TN, United States
| | - Joshua D Simmons
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Diseases, Nashville, TN, United States
| | - Kakali Ghoshal
- Vanderbilt University Medical Center, Department of Medicine, Division of Nephrology, Nashville, TN, United States
| | - Mark Pilkinton
- Saint Thomas Hospital West, Nashville, TN, United States
| | - Samuel S Bailin
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Diseases, Nashville, TN, United States
| | - Curtis L Gabriel
- Vanderbilt University Medical Center, Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Nashville, TN, United States
| | - Ambra Pozzi
- Vanderbilt University Medical Center, Department of Medicine, Division of Nephrology, Nashville, TN, United States; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, United States
| | - John R Koethe
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Diseases, Nashville, TN, United States; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, United States
| | - Nancy J Brown
- Yale School of Medicine, Nashville, TN, United States
| | - Spyros A Kalams
- Vanderbilt University Medical Center, Department of Medicine, Division of Infectious Diseases, Nashville, TN, United States
| | - J Matthew Luther
- Vanderbilt University Medical Center, Department of Medicine, Division of Clinical Pharmacology, Nashville, TN, United States
| |
Collapse
|
49
|
Han T, Yuan T, Liang X, Chen N, Song J, Zhao X, Weng Y, Hu Y. Sarcopenic Obesity with Normal Body Size May Have Higher Insulin Resistance in Elderly Patients with Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes 2022; 15:1197-1206. [PMID: 35469341 PMCID: PMC9034890 DOI: 10.2147/dmso.s360942] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/08/2022] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE Data are limited regarding how body composition is linked to insulin resistance in elderly patients with type 2 diabetes mellitus (T2DM). We examined the association between body composition and insulin resistance in elderly T2DM patients. METHODS The cross-sectional study included 488 Chinese elderly patients wth T2DM. Subjects were classified into four groups based on body composition: normal body composition (NBC), low muscle mass alone (LMM), high body fat alone (HBF), both low muscle mass and high body fat (LMMHBF). RESULTS The percentage of subjects with LMMHBF was 14.5% (11.9% in men and 17.7% in women). Homeostasis model assessment of insulin resistance (HOMA2-IR) was higher in the LMMHBF group than in the HBF group (p = 0.045), and was also significantly higher in the LMMHBF or HBF group than in the NBC or LMM group. The HBF group showed the highest body mass index (BMI) of the four groups of different body compositions, and the LMMHBF group showed lower BMI than the HBF group; however, there was no significant difference in BMI or waist to hip ratio (WHR) between the LMMHBF group and the NBC group. The LMMHBF and HBF groups were significantly associated with increased risk of insulin resistance compared to the NBC group, with odds ratios (ORs) of 4.47 [95% confidence interval (CI) 2.06-9.68, p < 0.001] and 1.76 (95% CI 1.02-3.02, p = 0.041) respectively, even after the adjustment for covariates. CONCLUSION In China, though elderly T2DM patients with the body composition of sarcopenic obesity (as defined by coexistence of low muscle mass and high body fat) seemed to have normal body size, they exhibited the most severe degree and the highest risk of insulin resistance.
Collapse
Affiliation(s)
- Tingting Han
- Department of Geriatrics, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
| | - Ting Yuan
- Department of Geriatrics, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
| | - Xinyue Liang
- Department of Geriatrics, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
| | - Ningxin Chen
- Department of Geriatrics, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
| | - Jia Song
- Department of Geriatrics, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
| | - Xin Zhao
- Department of Geriatrics, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
| | - Yurong Weng
- Department of Geriatrics, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
| | - Yaomin Hu
- Department of Geriatrics, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, People’s Republic of China
- Correspondence: Yaomin Hu, Department of Geriatrics, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, No. 160 Pujian Road, Shanghai, 200127, People’s Republic of China,Tel +86 02168383815, Email
| |
Collapse
|
50
|
Zaborowski AM, Winter DC, Lynch L. The therapeutic and prognostic implications of immunobiology in colorectal cancer: a review. Br J Cancer 2021; 125:1341-1349. [PMID: 34302062 PMCID: PMC8575924 DOI: 10.1038/s41416-021-01475-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 05/13/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer represents the second leading cause of cancer-related death worldwide. The therapeutic field of immuno-oncology has rapidly gained momentum, with strikingly promising results observed in clinical practice. Increasing emphasis has been placed on the role of the immune response in tumorigenesis, therapy and predicting prognosis. Enhanced understanding of the dynamic and complex tumour-immune microenvironment has enabled the development of molecularly directed, individualised treatment. Analysis of intra-tumoural lymphocyte infiltration and the dichotomisation of colorectal cancer into microsatellite stable and unstable disease has important therapeutic and prognostic implications, with potential to capitalise further on this data. This review discusses the latest evidence surrounding the tumour biology and immune landscape of colorectal cancer, novel immunotherapies and the interaction of the immune system with each apex of the tripartite of cancer management (oncotherapeutics, radiotherapy and surgery). By utilising the synergy of chemotherapeutic agents and immunotherapies, and identifying prognostic and predictive immunological biomarkers, we may enter an era of unprecedented disease control, survivorship and cure rates.
Collapse
Affiliation(s)
- Alexandra M. Zaborowski
- grid.412751.40000 0001 0315 8143Centre for Colorectal Disease, St. Vincent’s University Hospital, Dublin 4, Ireland ,grid.8217.c0000 0004 1936 9705School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Des C. Winter
- grid.412751.40000 0001 0315 8143Centre for Colorectal Disease, St. Vincent’s University Hospital, Dublin 4, Ireland ,grid.7886.10000 0001 0768 2743School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Lydia Lynch
- grid.8217.c0000 0004 1936 9705School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland ,grid.38142.3c000000041936754XHarvard Institutes of Medicine, Harvard Medical School, Boston, MA USA
| |
Collapse
|