1
|
Modak S, Aktar T, Majumder D, Singha AK, Maiti D. A systemic review on leptin's role in defining cancer: special emphasis on immunomodulation, inflammation, and therapeutic interventions. Genes Immun 2025:10.1038/s41435-025-00333-7. [PMID: 40374921 DOI: 10.1038/s41435-025-00333-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 04/22/2025] [Accepted: 04/30/2025] [Indexed: 05/18/2025]
Abstract
Leptin, an adipokine related to obesity, is mainly known for its role in regulating energy homeostasis and appetite by working via the leptin receptor. Recently, different groups have demonstrated that apart from adipocytes, specific cell types associated with cancer and tumor microenvironments express leptin and leptin receptors. This tumor microenvironment-associated leptin-leptin receptor signaling contributes to the different hallmarks of cancer, ranging from inflammatory changes to metastasis. Eventually, it has also been reported that high serum level of leptin, a characteristic of obese people, is linked to enhanced tumor growth. On the other hand, leptin can influence both innate as well as adaptive immunity related to cancer. Overall, leptin's role in modulating cancer is controversial. So, in this review, we summarized the role of leptin in shaping different forms of cancer that are influenced by leptin-leptin receptor signaling with special emphasis on immunomodulation and inflammatory events and also discussed the possible therapeutic interventions to date. As this review work, with the collection of different updated knowledge, has summarized the role of leptin on cancer, it would be useful material to have on hand for both beginners as well as pioneers of these and related fields.
Collapse
Affiliation(s)
- Snehashish Modak
- Immunology Microbiology Lab, Tripura University, Suryamaninagar, Tripura, India
| | - Tamanna Aktar
- Immunology Microbiology Lab, Tripura University, Suryamaninagar, Tripura, India
| | - Debabrata Majumder
- Immunology Microbiology Lab, Tripura University, Suryamaninagar, Tripura, India
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC, USA
| | - Ashish Kr Singha
- Immunology Microbiology Lab, Tripura University, Suryamaninagar, Tripura, India
- Department of Human Physiology, Holy Cross College, Agartala, West Tripura, India
| | - Debasish Maiti
- Immunology Microbiology Lab, Tripura University, Suryamaninagar, Tripura, India.
| |
Collapse
|
2
|
Cheong LYT, Saipuljumri EN, Loi GWZ, Zeng J, Lo CH. Autolysosomal Dysfunction in Obesity-induced Metabolic Inflammation and Related Disorders. Curr Obes Rep 2025; 14:43. [PMID: 40366502 PMCID: PMC12078456 DOI: 10.1007/s13679-025-00638-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/05/2025] [Indexed: 05/15/2025]
Abstract
PURPOSE OF REVIEW Obesity is a global health crisis affecting individuals across all age groups, significantly increasing the risk of metabolic disorders such as type 2 diabetes (T2D), metabolic dysfunction-associated fatty liver disease (MAFLD), and cardiovascular diseases. The World Health Organization reported in 2022 that 2.5 billion adults were overweight, with 890 million classified as obese, emphasizing the urgent need for effective interventions. A critical aspect of obesity's pathophysiology is meta-inflammation-a chronic, systemic low-grade inflammatory state driven by excess adipose tissue, which disrupts metabolic homeostasis. This review examines the role of autolysosomal dysfunction in obesity-related metabolic disorders, exploring its impact across multiple metabolic organs and evaluating potential therapeutic strategies that target autophagy and lysosomal function. RECENT FINDINGS Emerging research highlights the importance of autophagy in maintaining cellular homeostasis and metabolic balance. Obesity-induced lysosomal dysfunction impairs the autophagic degradation process, contributing to the accumulation of damaged organelles and toxic aggregates, exacerbating insulin resistance, lipotoxicity, and chronic inflammation. Studies have identified autophagic defects in key metabolic tissues, including adipose tissue, skeletal muscle, liver, pancreas, kidney, heart, and brain, linking autophagy dysregulation to the progression of metabolic diseases. Preclinical investigations suggest that pharmacological and nutritional interventions-such as AMPK activation, caloric restriction mimetics, and lysosomal-targeting compounds-can restore autophagic function and improve metabolic outcomes in obesity models. Autolysosomal dysfunction is a pivotal contributor to obesity-associated metabolic disorders , influencing systemic inflammation and metabolic dysfunction. Restoring autophagy and lysosomal function holds promise as a therapeutic strategy to mitigate obesity-driven pathologies. Future research should focus on translating these findings into clinical applications, optimizing targeted interventions to improve metabolic health and reduce obesity-associated complications.
Collapse
Affiliation(s)
- Lenny Yi Tong Cheong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | | | - Gavin Wen Zhao Loi
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Jialiu Zeng
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA.
- Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY, 13244, USA.
| | - Chih Hung Lo
- Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY, 13244, USA.
- Department of Biology, Syracuse University, Syracuse, NY, 13244, USA.
| |
Collapse
|
3
|
Yu S, Zhu X, Zhao X, Li Y, Niu X, Chen Y, Ying J. Improvement of chronic metabolic inflammation and regulation of gut homeostasis: Tea as a potential therapy. Pharmacol Ther 2025; 269:108828. [PMID: 40020787 DOI: 10.1016/j.pharmthera.2025.108828] [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: 04/30/2024] [Revised: 12/27/2024] [Accepted: 02/21/2025] [Indexed: 03/03/2025]
Abstract
Chronic metabolic inflammation is a common mechanism linked to the development of metabolic disorders such as obesity, diabetes, and cardiovascular disease (CVD). Chronic metabolic inflammation often related to alterations in gut homeostasis, and pathological processes involve the activation of endotoxin receptors, metabolic reprogramming, mitochondrial dysfunction, and disruption of intestinal nuclear receptor activity. Recent investigations into homeostasis and chronic metabolic inflammation have revealed a novel mechanism which is characterized by a timing interaction involving multiple components and targets. This article explores the positive impact of tea consumption on metabolic health of populations, with a special focus on the improvement of inflammatory indicators and the regulation of gut microbiota. Studies showed that tea consumption is related to the enrichment of gut microbiota. The relative proportion of Firmicutes/Bacteroidetes (F/B) is altered, while the abundance of Lactobacillus, Bifidobacterium, and A. muciniphila increased significantly in most of the studies. Thus, tea consumption could provide potential protection from the development of chronic diseases by improving gut homeostasis and reducing chronic metabolic inflammation. The direct impact of tea on intestinal homeostasis primarily targets lipopolysaccharide (LPS)-related pathways. This includes reducing the synthesis of intestinal LPS, inhibiting LPS translocation, and preventing the binding of LPS to TLR4 receptors to block downstream inflammatory pathways. The TLR4/MyD88/NF-κB p65 pathway is crucial for anti-metaflammatory responses. The antioxidant properties of tea are linked to enhancing mitochondrial function and mitigating mitochondria-related inflammation by eliminating free radicals, inhibiting NLRP3 inflammasomes, and modulating Nrf2/ARE activity. Tea also contributes to safeguarding the intestinal barrier through various mechanisms, such as promoting the synthesis of short-chain fatty acids in the intestine, activating intestinal aryl hydrocarbon receptor (AhR) and farnesoid X receptor (FXR), and improving enteritis. Functional components that improve chronic metabolic inflammation include tea polyphenols, tea pigments, TPS, etc. Tea metabolites such as 4-Hydroxyphenylacetic acid and 3,4-Dihydroxyflavan derivatives, etc., also contribute to anti-chronic metabolic inflammation effects of tea consumption. The raw materials and processing technologies affect the functional component compositions of tea; therefore, consuming different types of tea may result in varying action characteristics and mechanisms. However, there is currently limited elaboration on this aspect. Future research should conduct in-depth studies on the mechanism of tea and its functional components in improving chronic metabolic inflammation. Researchers should pay attention to whether there are interactions between tea and other foods or drugs, explore safe and effective usage and dosage, and investigate whether there are individual differences in the tea-drinking population leading to different effects of tea intervention. Ultimately, the application of tea drinking could be a universal therapy for regulating intestinal homeostasis, anti-chronic metabolic inflammatory responses, and promoting metabolic health.
Collapse
Affiliation(s)
- Shiyi Yu
- Nutrition and Health Research Institute, School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430000, China
| | - Xuan Zhu
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Xiayu Zhao
- National Institute of Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Yan Li
- National Institute of Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Xinghe Niu
- Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China; COFCO Nutrition and Health Research Institute, Beijing 102209, China
| | - Yinghua Chen
- Nutrition and Health Research Institute, School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430000, China
| | - Jian Ying
- Nutrition and Health Research Institute, School of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430000, China.
| |
Collapse
|
4
|
Sun Y, Shan X, Li M, Niu Y, Sun Z, Ma X, Wang T, Zhang J, Niu D. Autoimmune mechanisms and inflammation in obesity-associated type 2 diabetes, atherosclerosis, and non-alcoholic fatty liver disease. Funct Integr Genomics 2025; 25:84. [PMID: 40205260 DOI: 10.1007/s10142-025-01587-0] [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: 03/11/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/11/2025]
Abstract
Obesity, characterized by the excessive accumulation of white adipose tissue, is a significant global health burden and a major risk factor for a range of diseases, including malignancies and metabolic disorders. Individuals with high visceral fat content are particularly susceptible to severe complications such as type 2 diabetes, cardiovascular diseases, and liver disorders. However, the pathogenesis of obesity-related metabolic diseases extends beyond simple adiposity. Chronic obesity triggers a prolonged inflammatory response, which leads to tissue fibrosis and sustained organ damage, contributing to multi-organ dysfunction. This review explores the autoimmune mechanisms and inflammatory pathways underlying obesity-induced type 2 diabetes, atherosclerosis, and non-alcoholic fatty liver disease, with an emphasis on their interrelated pathophysiology and the potential for therapeutic interventions.
Collapse
Grants
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- LZ22C010003 Key Project of Zhejiang Provincial Natural Science Foundation of China
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 2021R52043 Scientific and Technological Innovation Leading Talents Project of Zhejiang Provincial "High-level Talents Special Support Plan"
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- 32202656, 32402753 National Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- LQ23C170003, LQ23C180003 & LQ24C170001 Zhejiang Provincial Natural Science Foundation of China
- 2021C02068-4 Zhejiang Science and Technology Major Program on Agricultural New Variety Breeding
- 2021C02068-4 Zhejiang Science and Technology Major Program on Agricultural New Variety Breeding
Collapse
Affiliation(s)
- Yuanyuan Sun
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Xueting Shan
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Mingyang Li
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Yifan Niu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Zhongxin Sun
- Department of Plastic, Reconstructive & Hand Microsurgery, Ningbo NO.6 Hospital, Ningbo, 315000, Zhejiang, China
| | - Xiang Ma
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China
| | - Tao Wang
- Nanjing Kgene Genetic Engineering Co., Ltd, Nanjing, 211300, Jiangsu, China.
| | - Jufang Zhang
- Department of Plastic and Aesthetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, Zhejiang, China.
| | - Dong Niu
- College of Animal Science and Technology & College of Veterinary Medicine, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou, 311300, Zhejiang, China.
| |
Collapse
|
5
|
Tilg H, Ianiro G, Gasbarrini A, Adolph TE. Adipokines: masterminds of metabolic inflammation. Nat Rev Immunol 2025; 25:250-265. [PMID: 39511425 DOI: 10.1038/s41577-024-01103-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 11/15/2024]
Abstract
Adipose tissue is an immunologically active organ that controls host physiology, partly through the release of mediators termed adipokines. In obesity, adipocytes and infiltrating leukocytes produce adipokines, which include the hormones adiponectin and leptin and cytokines such as tumour necrosis factor and IL-1β. These adipokines orchestrate immune responses that are collectively referred to as metabolic inflammation. Consequently, metabolic inflammation characterizes metabolic disorders and promotes distinct disease aspects, such as insulin resistance, metabolic dysfunction-associated liver disease and cardiovascular complications. In this unifying concept, adipokines participate in the immunological cross-talk that occurs between metabolically active organs in metabolic diseases, highlighting the fundamental role of adipokines in obesity and their potential for therapeutic intervention. Here, we summarize how adipokines shape metabolic inflammation in mice and humans, focusing on their contribution to metabolic disorders in the setting of obesity and discussing their value as therapeutic targets.
Collapse
Affiliation(s)
- Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Gianluca Ianiro
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
6
|
Fu L, Han X, Wang Y, Hu YQ. Identifying genetic overlaps in obesity and metabolic disorders unlocking unique and shared mechanistic insights. Free Radic Biol Med 2025; 231:80-93. [PMID: 39999931 DOI: 10.1016/j.freeradbiomed.2025.02.033] [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: 01/20/2025] [Revised: 02/19/2025] [Accepted: 02/22/2025] [Indexed: 02/27/2025]
Abstract
OBJECTIVE Obesity has a high heritability and frequently co-occurs with metabolic disorders, indicating shared genetic susceptibility. The underlying causative genes and biological mechanisms of obesity and metabolic disorders remain predominantly elusive. METHODS The FinnGen R11 dataset, including over 450,000 subjects, was employed in conjunction with the Genotype-Tissue Expression Project (GTEx) v8 eQTls dataset to conduct cross-tissue transcriptome association studies, Functional Summary-based Imputation in single tissues, and Gene Analysis combined with Multimarker Analysis of Genomic Annotation, respectively, for identifying distinct and shared genetic architectures of obesity and metabolic disorders. We also employed RHOGE to ascertain the genetic correlation and putative causal directions between them. Subsequent Mendelian randomization, colocalization analyses, and other cell and tissue enrichment analyses were employed to enhance our understanding of the functional implications of these susceptibility genes. RESULTS A total of 35 genes were identified as obesity susceptibility and 10 genes linked to metabolic disorder susceptibility. Of these, three genes (MCM6, MAPRE3 and UBXN4) were identified as being shared. Mendelian randomization and colocalization analyses revealed the three shared genes have causal associations with obesity and metabolic disorders and serve as independent signals. Subsequent analyses indicated MCM6 may influence obesity and metabolic disorder risk by regulating DNA replication, cell proliferation, and interactions with chemical responses. MAPRE3 may confer protective effects against obesity and metabolic disorders through PAK Pathway, while UBXN4 may involve in regulating cholesterol metabolism. CONCLUSION Our study provides insight into the novel shared genetic mechanism between obesity and metabolic disorders and identifies potential targets for pharmacological intervention.
Collapse
Affiliation(s)
- Liwan Fu
- Center for Non-Communicable Disease Management, Department of Neurology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.
| | - Xiaodi Han
- Center for Non-Communicable Disease Management, Department of Neurology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.
| | - Yuquan Wang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Institute of Biostatistics, School of Life Sciences, Fudan University, Shanghai, China.
| | - Yue-Qing Hu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Institute of Biostatistics, School of Life Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Ray S, Huang E, McMullen MR, Jatana S, de la Motte C, Nagy LE. 35k Da specific-sized hyaluronan ameliorates high-fat diet-induced liver injury in murine model of moderate obesity. Matrix Biol 2025; 136:1-8. [PMID: 39732151 DOI: 10.1016/j.matbio.2024.12.010] [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: 06/30/2024] [Revised: 12/07/2024] [Accepted: 12/25/2024] [Indexed: 12/30/2024]
Abstract
Obesity is a growing concern in the US and world-wide, associated with an increased risk for several cardiometabolic diseases, including metabolic associated steatotic liver disease (MASLD). Currently, therapeutic interventions to prevent and/or treat MASLD are limited, and research is needed to identify new therapeutic targets. The specific-sized 35 kDa fragment of hyaluronan (HA35), has gut protective and anti-inflammatory properties and a previous pilot clinical study reported it is well tolerated in healthy individuals. Here we tested the hypothesis that HA35 treatment ameliorates high fat diet-induced liver injury. Five-week-old male C57BL/6 J mice were allowed ad lib access to control chow or high fat fructose and cholesterol (FFC) diet over a period of 12 weeks. HA35 was administered at 15mg/kg via oral gavage on the last 6 days of the study as a therapeutic intervention. Mice on FFC diet-gained more body weight compared to those on chow diet, with final body weights ranging from 30.8 to 45.6 g. FFC diet caused hepatocyte injury, increased expression of inflammatory cytokine/chemokine mRNA, as well as indicators of liver fibrosis. When mice were stratified based on their final body weight, only mice <40 g were protected by treatment with HA35. In this group, treatment with HA35 also restored tight junction integrity in the colon and increased expression of α -defensins in the small intestine. Taken together the data suggests that HA35 is an effective therapeutic in ameliorating high fat diet-induced liver inflammation and fibrosis in moderately obese, but not severe, conditions.
Collapse
Affiliation(s)
- Semanti Ray
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH USA
| | - Emily Huang
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH USA
| | - Megan R McMullen
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH USA
| | - Samreen Jatana
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH USA
| | - Carol de la Motte
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH USA; Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH USA
| | - Laura E Nagy
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH USA; Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH USA.
| |
Collapse
|
8
|
Reinisch I, Enzenhofer S, Prokesch A. Mechanisms of Lipid-Associated Macrophage Accrual in Metabolically Stressed Adipose Tissue. Bioessays 2025; 47:e202400203. [PMID: 39828607 PMCID: PMC11931678 DOI: 10.1002/bies.202400203] [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: 08/21/2024] [Revised: 12/06/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
Adipose tissue (AT) inflammation, a hallmark of the metabolic syndrome, is triggered by overburdened adipocytes sending out immune cell recruitment signals during obesity development. An AT immune landscape persistent throughout weight loss and regain constitutes an immune-obesogenic memory that hinders long-term weight loss management. Lipid-associated macrophages (LAMs) are emerging as major players in diseased, inflamed metabolic tissues and may be key contributors to an obesogenic memory in AT. Our previous study found that LAM abundance increases with weight loss via intermittent fasting (IF) in obese mice, which is driven by adipocyte p53 signalling. However, the specific signals causing LAM accumulation in AT under IF remain unknown. In this piece, we hypothesise on a range of adipocyte-secreted signals that can harbor immune-attractive features upon fasting/refeeding cycles. We highlight possible mechanisms including cell death signalling, matrikines, and other damage-associated molecular patterns (DAMPs), as well as adipo(-cyto)kines, lipid mediators, metabolites, extracellular vesicles, and epigenetic rewiring. Finally, we consider how advances in mechanisms of AT LAM recruitment gleaned from preclinical models might be translatable to long-term weight management in humans. Thus, we provide vantage points to study signals driving monocyte recruitment, polarisation towards LAMs, and LAM retention, to harness the therapeutic potential of modulating AT LAM levels by impacting the immune-obesogenic memory in metabolic disease.
Collapse
Affiliation(s)
- Isabel Reinisch
- Department of Health Sciences and Technology, Institute of Food Nutrition and HealthEidgenössische Technische Hochschule Zürich (ETH)SchwerzenbachSwitzerland
| | - Sarah Enzenhofer
- Gottfried Schatz Research Center for Cell SignalingMetabolism and Aging, Division of Cell Biology, Histology and EmbryologyMedical University of GrazGrazAustria
| | - Andreas Prokesch
- Gottfried Schatz Research Center for Cell SignalingMetabolism and Aging, Division of Cell Biology, Histology and EmbryologyMedical University of GrazGrazAustria
- BioTechMed‐GrazGrazAustria
| |
Collapse
|
9
|
Chen X, Su P, Guan H, Xiang A, Zhang L, Deng Z, Yu Q. Physiochemotherapy alginate-based hydrogel inhibiting lipogenesis through antioxidant and ant-inflammatory effects for obesity treatment. Int J Biol Macromol 2025; 304:140776. [PMID: 39924048 DOI: 10.1016/j.ijbiomac.2025.140776] [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: 11/24/2024] [Revised: 01/07/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
Obesity is strongly associated with hypertension, hyperglycemia, insulin resistance, and other metabolic syndrome symptoms, emphasizing the urgent need for innovative therapeutic strategies. Physiochemotherapy, which combines the advantages of chemical and physical therapies, has attracted significant research interest. However, the precise outcomes and mechanisms of this integrative approach for obesity treatment remain poorly understood. In this study, we developed a physically photothermal nanocomposite hydrogel (termed SDAR) loaded with chemical resveratrol nanoparticles for obesity mitigation. SDAR was synthesized using polydopamine-functionalized sodium alginate, Zn2+, and resveratrol nanoparticles. The resveratrol nanoparticles exhibited a size distribution of 5-15 nm and a minor Zetal potential below 1.3 mV across varying pH conditions. The engineered SDAR hydrogel demonstrated stable photothermal performance with a steady temperature around 44 °C, high biocompatibility, strong antioxidant capacity (antioxidant rate of 93.7 %), and anti-inflammatory properties. In a lipid differentiation model, photothermal SDAR hydrogel administration effectively reduced intracellular reactive oxygen species (ROS) and inflammatory markers by over 50 % in 3T3-L1 preadipocytes. Furthermore, SDAR attenuated CCAAT/enhancer-binding protein beta (C/EBPβ) binding activity to downstream target genes, thereby inhibiting lipogenic differentiation. Collectively, the photothermal-responsive SDAR hydrogel presents a promising physiochemotherapy strategy for obesity treatment by suppressing lipogenesis.
Collapse
Affiliation(s)
- Xiaochang Chen
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an 710021, China
| | - Peihong Su
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an 710021, China
| | - Hua Guan
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an 710021, China
| | - Aoqi Xiang
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an 710021, China
| | - Lusha Zhang
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an 710021, China
| | - Zexing Deng
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China; College of Materials Science and Engineering, Xi'an University of Science and Technology, Xi'an 710054, China.
| | - Qi Yu
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an 710021, China.
| |
Collapse
|
10
|
Abu Rached N, Dietrich JW, Ocker L, Stockfleth E, Haven Y, Myszkowski D, Bechara FG. Endotyping Insulin-Glucose Homeostasis in Hidradenitis Suppurativa: The Impact of Diabetes Mellitus and Inflammation. J Clin Med 2025; 14:2145. [PMID: 40217596 PMCID: PMC11990022 DOI: 10.3390/jcm14072145] [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: 01/29/2025] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 04/14/2025] Open
Abstract
Background: Hidradenitis suppurativa (HS) is a chronic inflammatory skin disease often associated with metabolic disorders such as diabetes mellitus. Recent research suggests a link between systemic inflammation and insulin-glucose dysregulation in HS. This study investigates the relationship between insulin-glucose homeostasis, diabetes mellitus and the haptoglobin concentration in HS patients. Methods: We assessed 95 HS patients and 49 controls using validated fasting-based function tests, including the Structural Parameter Inference Approach (SPINA), Homeostasis Model Assessment (HOMA) and Quantitative Insulin Sensitivity Check Index (QUICKI). Results: The HS patients had a significantly higher fasting insulin concentration (97.2 vs. 69.0 pmol/L, p = 0.035), increased insulin resistance (HOMA-IR: 3.47 vs. 2.57, p = 0.016) and impaired insulin sensitivity (SPINA-GR: 1.34 vs. 1.76 mol/s, p = 0.017). In diabetes, the insulin sensitivity was more strongly reduced (SPINA-GR: 0.61 vs. 1.41 mol/s, p = 0.0057) and the insulin resistance increased (HOMA-IR: 7.3 vs. 3.2, p = 0.017). Higher haptoglobin concentrations were accompanied by worse glycaemic control, demonstrating a significantly elevated fasting glucose (5.77 vs. 5.11 mmol/L, p = 0.043) concentration and HbA1c (5.7% vs. 5.4%, p = 0.0081) fraction. Conclusions: Our findings suggest that chronic inflammation in HS contributes to metabolic dysregulation, worsening insulin resistance and glycaemic control, particularly in those with elevated haptoglobin or diabetes.
Collapse
Affiliation(s)
- Nessr Abu Rached
- International Centre for Hidradenitis Suppurativa/Acne Inversa (ICH), Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, 44791 Bochum, Germany; (L.O.); (E.S.); (Y.H.); (D.M.); (F.G.B.)
- Skin Cancer Center, Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Johannes W. Dietrich
- Diabetes, Endocrinology and Metabolism Section, Department of Internal Medicine I, St. Josef Hospital, Ruhr University Bochum, Gudrunstr. 56, 44791 Bochum, Germany;
- Diabetes Centre Bochum-Hattingen, St. Elisabeth-Hospital Blankenstein, Im Vogelsang 5-11, 45527 Hattingen, Germany
- Centre for Rare Endocrine Diseases, Ruhr Centre for Rare Diseases (CeSER), Ruhr University Bochum and Witten/Herdecke University, Alexandrinenstr. 5, 44791 Bochum, Germany
- Centre for Diabetes Technology, Catholic Hospitals Bochum, Gudrunstr. 56, 44791 Bochum, Germany
| | - Lennart Ocker
- International Centre for Hidradenitis Suppurativa/Acne Inversa (ICH), Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, 44791 Bochum, Germany; (L.O.); (E.S.); (Y.H.); (D.M.); (F.G.B.)
- Skin Cancer Center, Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Eggert Stockfleth
- International Centre for Hidradenitis Suppurativa/Acne Inversa (ICH), Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, 44791 Bochum, Germany; (L.O.); (E.S.); (Y.H.); (D.M.); (F.G.B.)
- Skin Cancer Center, Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Yannik Haven
- International Centre for Hidradenitis Suppurativa/Acne Inversa (ICH), Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, 44791 Bochum, Germany; (L.O.); (E.S.); (Y.H.); (D.M.); (F.G.B.)
- Skin Cancer Center, Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Daniel Myszkowski
- International Centre for Hidradenitis Suppurativa/Acne Inversa (ICH), Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, 44791 Bochum, Germany; (L.O.); (E.S.); (Y.H.); (D.M.); (F.G.B.)
- Skin Cancer Center, Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Falk G. Bechara
- International Centre for Hidradenitis Suppurativa/Acne Inversa (ICH), Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, 44791 Bochum, Germany; (L.O.); (E.S.); (Y.H.); (D.M.); (F.G.B.)
- Skin Cancer Center, Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, 44791 Bochum, Germany
| |
Collapse
|
11
|
Guan F, Wang R, Yi Z, Luo P, Liu W, Xie Y, Liu Z, Xia Z, Zhang H, Cheng Q. Tissue macrophages: origin, heterogenity, biological functions, diseases and therapeutic targets. Signal Transduct Target Ther 2025; 10:93. [PMID: 40055311 PMCID: PMC11889221 DOI: 10.1038/s41392-025-02124-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 11/01/2024] [Accepted: 12/15/2024] [Indexed: 05/04/2025] Open
Abstract
Macrophages are immune cells belonging to the mononuclear phagocyte system. They play crucial roles in immune defense, surveillance, and homeostasis. This review systematically discusses the types of hematopoietic progenitors that give rise to macrophages, including primitive hematopoietic progenitors, erythro-myeloid progenitors, and hematopoietic stem cells. These progenitors have distinct genetic backgrounds and developmental processes. Accordingly, macrophages exhibit complex and diverse functions in the body, including phagocytosis and clearance of cellular debris, antigen presentation, and immune response, regulation of inflammation and cytokine production, tissue remodeling and repair, and multi-level regulatory signaling pathways/crosstalk involved in homeostasis and physiology. Besides, tumor-associated macrophages are a key component of the TME, exhibiting both anti-tumor and pro-tumor properties. Furthermore, the functional status of macrophages is closely linked to the development of various diseases, including cancer, autoimmune disorders, cardiovascular disease, neurodegenerative diseases, metabolic conditions, and trauma. Targeting macrophages has emerged as a promising therapeutic strategy in these contexts. Clinical trials of macrophage-based targeted drugs, macrophage-based immunotherapies, and nanoparticle-based therapy were comprehensively summarized. Potential challenges and future directions in targeting macrophages have also been discussed. Overall, our review highlights the significance of this versatile immune cell in human health and disease, which is expected to inform future research and clinical practice.
Collapse
Affiliation(s)
- Fan Guan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Ruixuan Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wanyao Liu
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yao Xie
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China.
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
12
|
Gordito Soler M, Ramírez-Manent JI, Tárraga López PJ, Martínez-Almoyna Rifá E, Paublini H, López González ÁA. Usefulness of body and visceral fat determined by bioimpedancemetry versus body mass index and waist circumference in the identification of elevated values of different atherogenesis risk scales. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2025:500772. [PMID: 40037953 DOI: 10.1016/j.arteri.2025.500772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 03/06/2025]
Abstract
INTRODUCTION Obesity and atherogenesis are two highly prevalent pathological processes worldwide that also share several pathophysiological mechanisms. OBJECTIVES Descriptive and cross-sectional study in 8,590 Spanish workers (4,104 men and 4,486 women) with an average age of 41.5years, in which the usefulness of four scales of overweight and obesity such as body mass index (BMI), waist circumference and percentages of body and visceral fat determined by bioimpedance measurement to identify high levels of atherogenic risk determined by atherogenic dyslipidemia (AD), lipid triad (LT) and several atherogenic indices is assessed. RESULTS All the overweight and obesity scales show a predictive value between moderate and good, determined by the AUC of the ROC curves, with values ranging from 0.727 to 0.886 in women and 0.676 to 0.885 in men. Of all of them, the one with the highest AUC is visceral fat with values exceeding 0.800 and the lowest are for BMI. In all cases, the AUC is higher in women. CONCLUSIONS The overweight and obesity scales analysed (BMI, waist circumference and percentage of body and visceral fat) show AUCs for predicting atherogenic risk between moderate and high, with visceral fat being the most useful of all.
Collapse
Affiliation(s)
- María Gordito Soler
- Farmacéutica, Sevilla, España; Grupo de Investigación ADEMA-Salud del Instituto Universitario de Investigación en Ciencias de la Salud (IUNICS), Universidad de les Illes Balears, Palma de Mallorca, España
| | - José Ignacio Ramírez-Manent
- Grupo de Investigación ADEMA-Salud del Instituto Universitario de Investigación en Ciencias de la Salud (IUNICS), Universidad de les Illes Balears, Palma de Mallorca, España; Instituto de Investigación Sanitaria de les Illes Balears (IDISBA), Palma de Mallorca, España; Servicio de Salud de les Illes Balears, Palma de Mallorca, España; Facultad de Medicina de les Illes Balears, Palma de Mallorca, España
| | - Pedro Juan Tárraga López
- Facultad de Medicina de Albacete, Universidad de Castilla la Mancha, Gerencia de Atención Integrada de Albacete, SESCAM, España; Servicio de Salud de Castilla-La Mancha, Toledo, España.
| | - Emilio Martínez-Almoyna Rifá
- Grupo de Investigación ADEMA-Salud del Instituto Universitario de Investigación en Ciencias de la Salud (IUNICS), Universidad de les Illes Balears, Palma de Mallorca, España; Facultad de Odontología, Escuela Universitaria ADEMA-UIB, Palma de Mallorca, España
| | - Hernán Paublini
- Grupo de Investigación ADEMA-Salud del Instituto Universitario de Investigación en Ciencias de la Salud (IUNICS), Universidad de les Illes Balears, Palma de Mallorca, España; Facultad de Odontología, Escuela Universitaria ADEMA-UIB, Palma de Mallorca, España
| | - Ángel Arturo López González
- Grupo de Investigación ADEMA-Salud del Instituto Universitario de Investigación en Ciencias de la Salud (IUNICS), Universidad de les Illes Balears, Palma de Mallorca, España; Instituto de Investigación Sanitaria de les Illes Balears (IDISBA), Palma de Mallorca, España; Servicio de Salud de les Illes Balears, Palma de Mallorca, España; Facultad de Odontología, Escuela Universitaria ADEMA-UIB, Palma de Mallorca, España
| |
Collapse
|
13
|
Toshimitsu T, Irie J. An update and overview of the various health-related benefits of probiotics: A focus on clinical trials demonstrating efficacy, tolerability and use in patients with impaired glucose tolerance and type 2 diabetes. Diabetes Obes Metab 2025; 27 Suppl 1:15-22. [PMID: 39989436 PMCID: PMC11894779 DOI: 10.1111/dom.16273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 02/25/2025]
Abstract
Recently, probiotics have been investigated as potential therapeutic agents for various diseases. Clinical studies using probiotics have been conducted in humans with impaired glucose tolerance and type 2 diabetes mellitus. Chronic inflammation plays a pivotal role in initiating insulin resistance in the pathogenesis of type 2 diabetes, leading to cardiovascular diseases. Intestinal dysfunction and inflammation have been postulated to trigger systemic chronic inflammation, and it is assumed that the suppression of inflammation in the intestine is the point of activity of probiotics. Therefore, in this review, among the randomised controlled trials that evaluated the effects of probiotics in patients with impaired glucose tolerance and type 2 diabetes, we selected trials that evaluated the indices of glycaemic control and inflammation-related markers. Some trials have shown that the probiotics administration improved glycaemic indices, such as HbA1c levels, and reduced C-reactive protein levels and proinflammatory cytokines, such as IL-6, in the blood, suggesting the suppression of inflammation. Two trials showed improvements in glycaemic indices, implying that they were mediated by IL-10, an anti-inflammatory cytokine. Although a correlation between the suppression of inflammation by probiotics and improvement in glycaemic control has not been documented, one trial revealed that glycaemic control worsened, accompanied by a decrease in anti-inflammatory cytokine levels, after probiotics were discontinued. Other studies have shown that probiotics can reduce blood endotoxin levels and increase intestinal mucin production. These findings suggest that probiotic administration has enormous potential to suppress chronic inflammation in metabolic disorders, leading to improved glycaemic control. Suppression of chronic inflammation has been speculated to prevent vascular diseases in type 2 diabetes.
Collapse
Affiliation(s)
- Takayuki Toshimitsu
- Health Science Research Unit, Division of Research and DevelopmentMeiji Co., LtdTokyoJapan
| | - Junichiro Irie
- Division of Diabetes, Department of Medicine IIKansai Medical UniversityOsakaJapan
| |
Collapse
|
14
|
Kohutek ZA, Caslin HL, Fehrenbach DJ, Heimlich JB, Brown JD, Madhur MS, Ferrell PB, Doran AC. Bone Marrow Niche in Cardiometabolic Disease: Mechanisms and Therapeutic Potential. Circ Res 2025; 136:325-353. [PMID: 39883790 PMCID: PMC11790260 DOI: 10.1161/circresaha.124.323778] [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] [Indexed: 02/01/2025]
Abstract
Cardiovascular and cardiometabolic diseases are leading causes of morbidity and mortality worldwide, driven in part by chronic inflammation. Emerging research suggests that the bone marrow microenvironment, or marrow niche, plays a critical role in both immune system regulation and disease progression. The bone marrow niche is essential for maintaining hematopoietic stem cells (HSCs) and orchestrating hematopoiesis. Under normal conditions, this niche ensures a return to immune homeostasis after acute stress. However, in the setting of inflammatory conditions such as those seen in cardiometabolic diseases, it becomes dysregulated, leading to enhanced myelopoiesis and immune activation. This review explores the reciprocal relationship between the bone marrow niche and cardiometabolic diseases, highlighting how alterations in the niche contribute to disease development and progression. The niche regulates HSCs through complex interactions with stromal cells, endothelial cells, and signaling molecules. However, in the setting of chronic diseases such as hypertension, atherosclerosis, and diabetes, inflammatory signals disrupt the balance between HSC self-renewal and differentiation, promoting the excessive production of proinflammatory myeloid cells that exacerbate the disease. Key mechanisms discussed include the effects of hyperlipidemia, hyperglycemia, and sympathetic nervous system activation on HSC proliferation and differentiation. Furthermore, the review emphasizes the role of epigenetic modifications and metabolic reprogramming in creating trained immunity, a phenomenon whereby HSCs acquire long-term proinflammatory characteristics that sustain disease states. Finally, we explore therapeutic strategies aimed at targeting the bone marrow niche to mitigate chronic inflammation and its sequelae. Novel interventions that modulate hematopoiesis and restore niche homeostasis hold promise for the treatment of cardiometabolic diseases. By interrupting the vicious cycle of inflammation and marrow dysregulation, such therapies may offer new avenues for reducing cardiovascular risk and improving patient outcomes.
Collapse
Affiliation(s)
- Zachary A. Kohutek
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Heather L. Caslin
- Department of Health and Human Performance, University of Houston, Houston, TX 77204, USA
| | - Daniel J. Fehrenbach
- Department of Medicine, Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - J. Brett Heimlich
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jonathan D. Brown
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Meena S. Madhur
- Department of Medicine, Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - P. Brent Ferrell
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University, Nashville, TN 37212, USA
| | - Amanda C. Doran
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University, Nashville, TN 37212, USA
| |
Collapse
|
15
|
An J, Fu D, Chen X, Guan C, Li L, Bai J, Lv H. Revisiting the role of IL-27 in obesity-related metabolic diseases: safeguard or perturbation? Front Immunol 2025; 15:1498288. [PMID: 39906735 PMCID: PMC11792170 DOI: 10.3389/fimmu.2024.1498288] [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: 09/18/2024] [Accepted: 12/31/2024] [Indexed: 02/06/2025] Open
Abstract
The prevalence of metabolic diseases, such as obesity, has been steadily increasing in recent years, posing a significant threat to public health. Therefore, early identification and intervention play a crucial role. With the deepening understanding of the etiology of metabolic diseases, novel therapeutic targets are emerging for the treatment of obesity, lipid metabolism disorders, cardiovascular and cerebrovascular diseases, glucose metabolism disorders, and other related metabolic conditions. IL-27, as a multi-potent cytokine, holds great promise as a potential candidate target in this regard. This article provides a comprehensive review of the latest findings on IL-27 expression and signal transduction in the regulation of immune inflammatory cells, as well as its implications in obesity and other related metabolic diseases. Furthermore, it explores the potential of IL-27 as a novel therapeutic target for the treatment of obesity and metabolic disorders. Finally, an overview is presented on both the opportunities and challenges associated with targeting IL-27 for therapeutic interventions.
Collapse
Affiliation(s)
- Jinyang An
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Donghua Fu
- Department of Endocrinology, The People’s Hospital of Yuzhong County, Lanzhou, Gansu, China
| | - Ximei Chen
- Department of General Medicine, Zhengzhou Yihe Hospital affiliated to Henan University, Zhengzhou, Henan, China
| | - Conghui Guan
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Lingling Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Jia Bai
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Haihong Lv
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
16
|
Zeng D, Zeng Q, Li S, Lu J, Cheng N. Evaluating body roundness index and systemic immune inflammation index for mortality prediction in MAFLD patients. Sci Rep 2025; 15:330. [PMID: 39747385 PMCID: PMC11695853 DOI: 10.1038/s41598-024-83324-4] [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: 10/19/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is a major cause of liver-related morbidity and mortality, contributing to both cardiovascular and non-cardiovascular deaths. The Body Roundness Index (BRI) and Systemic Immune-Inflammation Index (SII) have emerged as predictors of adverse outcomes in metabolic diseases. This study investigates the association between BRI, SII, and mortality risk in MAFLD patients. A nationwide retrospective cohort study was conducted using data from the NHANES database (January 1999-December 2018), including patients diagnosed with MAFLD. BRI and SII were calculated at baseline. Cox proportional hazards models assessed the association between these indices and all-cause, cardiovascular, and non-cardiovascular mortality, adjusting for confounders. Among 12,435 participants diagnosed with MAFLD, 3,381 (27.2%) were classified into the low BRI and low SII group, 2,889 (23.2%) into the low BRI and high SII group, 2,802 (22.5%) into the high BRI and low SII group, and 3,363 (27.1%) into the high BRI and high SII group. Compared to the low BRI and low SII group, the high BRI and high SII group demonstrated significantly higher all-cause mortality, with an adjusted hazard ratio (HR) of 1.89. For cardiovascular mortality, the HR was 2.31, while for non-cardiovascular mortality, the HR was 1.78. The high BRI and high SII cohort exhibited the highest risk of all-cause mortality, cardiovascular mortality, and non-cardiovascular mortality. BRI and SII are independent predictors of mortality in MAFLD patients, and their combined use enhances risk stratification. Integrating these indices into clinical practice could improve personalized management strategies and outcomes in this high-risk population.
Collapse
Affiliation(s)
- Di Zeng
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qingyue Zeng
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Shuangqing Li
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jiong Lu
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Nansheng Cheng
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
17
|
Huang X, Cheng H, Deng L, Wang S, Li J, Qin A, Chu C, Du W, Liu X. Weight-adjusted-waist index: an innovative indicator of breast cancer hazard. BMC Womens Health 2024; 24:660. [PMID: 39709439 DOI: 10.1186/s12905-024-03507-z] [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: 04/22/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Central obesity and breast cancer (BC) have been identified as relevant by empirical research. The weight-adjusted-waist index (WWI) is a novel methodology for quantifying central obesity. Inspection of the association between WWI and BC in American adult women was the primary goal of the current investigation. METHODS Cross-sectional assessments were conducted on information gathered from 10,193 National Health and Nutrition Examination Survey (NHANES) participants from 2011 to 2018. The waist circumference was divided by the square root of the body's mass to compute WWI. Data were assessed via descriptive statistics to present data distributions according to BC grouping and WWI grouping, receiver operating characteristic curves (ROCs) to evaluate the obesity indicators' applied value, logistic regression to reflect associations between WWI and BC prevalence, and restricted cubic splines (RCSs) and subgroup analysis forest plots to visualise and complement the relationships. RESULTS This study enrolled 10,193 participants whose WWI ranged from 8.38 to 14.41, 259 of whom were diagnosed with BC, and the results revealed significant differences in baseline characteristics between the groups. With an area under the curve (AUC) value (95% confidence interval) (CI)of 0.611 (0.577-0.644), WWI was a promising indicator of BC with good application value rather than waist circumference (WC), body mass index (BMI), or waist-height ratio (WHtR). WWI and BC laid out a substantial relationship, yielding an odds ratio (OR) of 1.54 and a 95% CI of (1.34, 1.79), which remained at 1.19 (1.00, 1.42) after considerable adjustments were made, according to the logistic regression analysis. Compared with the lowest quartile of WWI, the highest quartile had a 62% greater in the probability of suffering from BC. With the RCS's inverted U-shape highlighting the importance of considering the nonlinear nature of the relationship and subgroup analyses reflecting variations among populations, all the results demonstrated that WWI was a well-suggestive indicator of BC hazard. CONCLUSION The current investigation revealed a meaningful association between the prevalence of BC and WWI, which was superior to other obesity indicators, albeit one that was more complex than the positive relationship initially derived. There existed a turning point for BC prevalence at WWI of approximately 12 cm/√kg. Nevertheless, maintaining WWI in the lower range is critical for preventing and administering BC and minimizing disease risk.
Collapse
Affiliation(s)
- Xinyi Huang
- Department of General Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Hengzheng Cheng
- Department of General Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Laifu Deng
- Department of General Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Shuting Wang
- Department of General Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Jiaxiu Li
- Department of General Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - An Qin
- Department of Thyroid and Breast Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Chunqiang Chu
- Department of Thyroid and Breast Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
- Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Wenyi Du
- General Surgery Center, Yixing People's Hospital Affiliated to Jiangsu University, Yixing, China.
- Wuxi Medical Center, Nanjing Medical University, Wuxi, China.
| | - Xiao Liu
- Department of Thyroid and Breast Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China.
- Wuxi Medical Center, Nanjing Medical University, Wuxi, China.
| |
Collapse
|
18
|
Schleh MW, Ameka MK, Rodriguez AS, Hasty AH. Deficiency of the Hemoglobin-Haptoglobin Receptor, CD163, Worsens Insulin Sensitivity in Obese Male Mice. Diabetes 2024; 73:1990-2002. [PMID: 39325576 PMCID: PMC11579413 DOI: 10.2337/db24-0405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024]
Abstract
Excessive iron accumulation in metabolic organs such as the adipose tissue, liver, and skeletal muscle is associated with increased diabetes risk. Tissue-resident macrophages serve multiple roles, including managing inflammatory tone and regulating parenchymal iron homeostasis, thus protecting against metabolic dysfunction upon iron overload. The scavenger receptor CD163 is uniquely present on tissue-resident macrophages and plays a significant role in iron homeostasis by clearing extracellular hemoglobin-haptoglobin complexes, thereby limiting oxidative damage caused by free hemoglobin in metabolic tissues. We show that the absence of CD163 exacerbates glucose intolerance and insulin resistance in male mice with obesity. Additionally, loss of CD163 reduced the expression of iron regulatory genes (Tfr1, Cisd1, Slc40a1) in adipose tissue macrophages and anti-inflammatory (M2-like) bone marrow-derived macrophages (BMDMs). Furthermore, CD163 deficiency mediated a proinflammatory shift and limited hemoglobin scavenging specifically in M2-like BMDMs. To this end, iron buffering was diminished in inguinal white adipose tissue (iWAT) macrophages in vivo, which culminated in iron spillover into adipocytes and CD45+ CD11B- nonmyeloid immune cells in iWAT. These findings show that CD163 on tissue-resident macrophages is critical for their anti-inflammatory and hemoglobin scavenging roles, and its absence results in impaired systemic insulin action in an obese setting. ARTICLE HIGHLIGHTS
Collapse
MESH Headings
- Animals
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Male
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Insulin Resistance/physiology
- Receptors, Cell Surface/metabolism
- Receptors, Cell Surface/genetics
- Mice
- Obesity/metabolism
- Macrophages/metabolism
- Haptoglobins/metabolism
- Haptoglobins/genetics
- Hemoglobins/metabolism
- Iron/metabolism
- Mice, Inbred C57BL
- Mice, Obese
- Receptors, Transferrin/metabolism
- Receptors, Transferrin/genetics
- Adipose Tissue, White/metabolism
- Mice, Knockout
- Glucose Intolerance/metabolism
- Glucose Intolerance/genetics
- Cation Transport Proteins/metabolism
- Cation Transport Proteins/genetics
Collapse
Affiliation(s)
- Michael W. Schleh
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Magdalene K. Ameka
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Alec S. Rodriguez
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Alyssa H. Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
- VA Tennessee Valley Healthcare System, Nashville, TN
| |
Collapse
|
19
|
Sun N, Krauss T, Seeliger C, Kunzke T, Stöckl B, Feuchtinger A, Zhang C, Voss A, Heisz S, Prokopchuk O, Martignoni ME, Janssen KP, Claussnitzer M, Hauner H, Walch A. Inter-organ cross-talk in human cancer cachexia revealed by spatial metabolomics. Metabolism 2024; 161:156034. [PMID: 39299512 DOI: 10.1016/j.metabol.2024.156034] [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: 04/19/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Cancer cachexia (CCx) presents a multifaceted challenge characterized by negative protein and energy balance and systemic inflammatory response activation. While previous CCx studies predominantly focused on mouse models or human body fluids, there's an unmet need to elucidate the molecular inter-organ cross-talk underlying the pathophysiology of human CCx. METHODS Spatial metabolomics were conducted on liver, skeletal muscle, subcutaneous and visceral adipose tissue, and serum from cachectic and control cancer patients. Organ-wise comparisons were performed using component, pathway enrichment and correlation network analyses. Inter-organ correlations in CCx altered pathways were assessed using Circos. Machine learning on tissues and serum established classifiers as potential diagnostic biomarkers for CCx. RESULTS Distinct metabolic pathway alteration was detected in CCx, with adipose tissues and liver displaying the most significant (P ≤ 0.05) metabolic disturbances. CCx patients exhibited increased metabolic activity in visceral and subcutaneous adipose tissues and liver, contrasting with decreased activity in muscle and serum compared to control patients. Carbohydrate, lipid, amino acid, and vitamin metabolism emerged as highly interacting pathways across different organ systems in CCx. Muscle tissue showed decreased (P ≤ 0.001) energy charge in CCx patients, while liver and adipose tissues displayed increased energy charge (P ≤ 0.001). We stratified CCx patients by severity and metabolic changes, finding that visceral adipose tissue is most affected, especially in cases of severe cachexia. Morphometric analysis showed smaller (P ≤ 0.05) adipocyte size in visceral adipose tissue, indicating catabolic processes. We developed tissue-based classifiers for cancer cachexia specific to individual organs, facilitating the transfer of patient serum as minimally invasive diagnostic markers of CCx in the constitution of the organs. CONCLUSIONS These findings support the concept of CCx as a multi-organ syndrome with diverse metabolic alterations, providing insights into the pathophysiology and organ cross-talk of human CCx. This study pioneers spatial metabolomics for CCx, demonstrating the feasibility of distinguishing cachexia status at the organ level using serum.
Collapse
Affiliation(s)
- Na Sun
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Tanja Krauss
- Else Kröner Fresenius Center for Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising-Weihenstephan, Germany
| | - Claudine Seeliger
- Else Kröner Fresenius Center for Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising-Weihenstephan, Germany; ZIEL Institute for Food and Health, Technical University of Munich, Freising-Weihenstephan, Germany
| | - Thomas Kunzke
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Barbara Stöckl
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Neuherberg, Germany; Else Kröner Fresenius Center for Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising-Weihenstephan, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Chaoyang Zhang
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Andreas Voss
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Simone Heisz
- Else Kröner Fresenius Center for Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising-Weihenstephan, Germany
| | - Olga Prokopchuk
- Department of Surgery, Klinikum rechts der Isar, University Hospital of the Technical University of Munich, Munich, Germany
| | - Marc E Martignoni
- Department of Surgery, Klinikum rechts der Isar, University Hospital of the Technical University of Munich, Munich, Germany
| | - Klaus-Peter Janssen
- Department of Surgery, Klinikum rechts der Isar, University Hospital of the Technical University of Munich, Munich, Germany
| | - Melina Claussnitzer
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Institute of Nutritional Science, University of Hohenheim, 70599 Stuttgart, Germany; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Hans Hauner
- Else Kröner Fresenius Center for Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising-Weihenstephan, Germany; ZIEL Institute for Food and Health, Technical University of Munich, Freising-Weihenstephan, Germany; Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, Munich, Germany.
| | - Axel Walch
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
20
|
Paddu NU, Lawrence B, Wong S, Poon SJ, Srivastava G. Weight maintenance on cost-effective antiobesity medications after 1 year of GLP-1 receptor agonist therapy: a real-world study. Obesity (Silver Spring) 2024; 32:2255-2263. [PMID: 39558626 PMCID: PMC11589535 DOI: 10.1002/oby.24177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 11/20/2024]
Abstract
OBJECTIVE The high cost of novel glucagon-like peptide-1 receptor agonist (GLP-1 RA) class agents often limits access and creates barriers to care. This real-world study evaluated the efficacy of older-generation generic antiobesity medications (AOMs) for weight maintenance after 1 year of GLP-1 RA therapy in patients who had achieved successful weight loss. METHODS We prospectively followed patients (N = 105) who had completed 12 months of therapy and were part of a "medical weight loss bundle," which included 12 months of GLP-1 RA therapy followed by 6 months of transition care. The baseline mean BMI was 36.4 kg/m2. Body weight outcomes were measured at 6, 12, 18, and 24 months. RESULTS After the medical weight loss bundle, 40 patients transitioned to generic AOMs. At 12 months, this cohort lost an average of 18.3%, 95% CI [13.0%, 23.6%] body weight from baseline, with a mean BMI of 27.9 kg/m2. At 18 months, they maintained the weight loss, with a mean BMI of 27.9 kg/m2. Subsequent follow-up visits (average 1.5 months later) without GLP-1 RAs showed further reduction, resulting in a total average weight loss of 25.5%, 95% CI [23.1%, 27.9%] compared to the initial visit. CONCLUSIONS Patients successfully treated with GLP-1 RAs can maintain their weight loss using generic older-generation AOMs, suggesting potential cost savings for insurers and implications for policy regarding AOM coverage.
Collapse
Affiliation(s)
- Nina U. Paddu
- Department of Medicine, Division of Diabetes, Endocrinology & MetabolismVanderbilt University School of MedicineNashvilleTennesseeUSA
- Vanderbilt Weight Loss CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Brianna Lawrence
- Vanderbilt Weight Loss CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Sydnee Wong
- Department of Medicine, Division of Diabetes, Endocrinology & MetabolismVanderbilt University School of MedicineNashvilleTennesseeUSA
| | - Sabrina J. Poon
- Department of Population HealthVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Gitanjali Srivastava
- Department of Medicine, Division of Diabetes, Endocrinology & MetabolismVanderbilt University School of MedicineNashvilleTennesseeUSA
- Vanderbilt Weight Loss CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of SurgeryVanderbilt University School of MedicineNashvilleTennesseeUSA
- Department of PediatricsVanderbilt University School of MedicineNashvilleTennesseeUSA
| |
Collapse
|
21
|
Choi A, Xu S, Luong AU, Wise SK. Current Review of Comorbidities in Chronic Rhinosinusitis. Curr Allergy Asthma Rep 2024; 25:4. [PMID: 39560809 DOI: 10.1007/s11882-024-01184-4] [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] [Accepted: 10/21/2024] [Indexed: 11/20/2024]
Abstract
PURPOSE OF REVIEW Chronic rhinosinusitis (CRS) is a heterogenous disease with a significant impact on patient quality of life and a substantial economic burden. CRS is associated with several systemic inflammatory conditions. We provide an updated review of CRS comorbidities as a springboard for future comorbidity mapping and potential therapeutics. RECENT FINDINGS The link between environmental allergies and CRS is most evident for central compartment atopic disease (CCAD) and allergic fungal rhinosinusitis (AFRS) subtypes but remains inconclusive for CRS overall. The association between asthma and CRS, reinforced by the unified airway theory, is evidenced by their response to similar biologic therapies. Another lower respiratory tract disease, COPD, has up to a 50% co-occurrence with CRS and warrants careful screening and treatment. Eosinophilic esophagitis and CRS share eosinophilic inflammation in different sites, meriting further research. Obesity not only presents physiological challenges but also correlates with a more severe subset of CRS. Diabetes mellitus is associated with CRSwNP, possibly secondary to therapeutics with steroids. Autoimmunity may contribute to nasal polyp formation through cytokines such as B-cell activating factor (BAFF), offering potential for future therapeutics. This review illustrates the need to employ a macroscopic approach in clinical decision making and treatment of CRS. Comorbidities may contribute to an overall proinflammatory state, magnify severity of symptoms, be a source of treatment resistance, and even an opportunity for future therapeutics.
Collapse
Affiliation(s)
- Alexander Choi
- Otolaryngology-HNS, Emory University, Atlanta, Georgia, USA
| | - Shuhui Xu
- Otolaryngology-HNS, McGovern Medical School of the University of Texas, Houston, TX, USA
| | - Amber U Luong
- Otolaryngology-HNS, McGovern Medical School of the University of Texas, Houston, TX, USA
| | - Sarah K Wise
- Otolaryngology-HNS, Emory University, Atlanta, Georgia, USA.
| |
Collapse
|
22
|
Jatana S, Abbadi A, West GA, Ponti AK, Braga-Neto MB, Smith JL, Marino-Melendez A, Willard B, Nagy LE, Motte CDL. Hyperglycemic environments directly compromise intestinal epithelial barrier function in an organoid model and hyaluronan (∼35 kDa) protects via a layilin dependent mechanism. Matrix Biol 2024; 133:116-133. [PMID: 39187208 DOI: 10.1016/j.matbio.2024.08.007] [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: 05/01/2024] [Revised: 08/12/2024] [Accepted: 08/23/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Metabolic syndrome and diabetes in obese individuals are strong risk factors for development of inflammatory bowel disease (IBD) and colorectal cancer. The pathogenic mechanisms of low-grade metabolic inflammation, including chronic hyperglycemic stress, in disrupting gut homeostasis are poorly understood. In this study, we sought to understand the impact of a hyperglycemic environment on intestinal barrier integrity and the protective effects of small molecular weight (35 kDa) hyaluronan on epithelial barrier function. METHODS Intestinal organoids derived from mouse colon were grown in normal glucose media (5 mM) or high glucose media (25 mM) to study the impact of hyperglycemic stress on the intestinal barrier. Additionally, organoids were pretreated with 35 kDa hyaluronan (HA35) to investigate the effect of hyaluronan on epithelial barrier under high glucose stress. Immunoblotting as well as confocal imaging was used to understand changes in barrier proteins, quantitative as well as spatial distribution, respectively. Alterations in barrier function were measured using trans-epithelial electrical resistance and fluorescein isothiocyanate flux assays. Untargeted proteomics analysis was performed to elucidate mechanisms by which HA35 exerts a protective effect on the barrier. Intestinal organoids derived from receptor knockout mice specific to various HA receptors were utilized to understand the role of HA receptors in barrier protection under high glucose conditions. RESULTS We found that high glucose stress decreased the protein expression as well as spatial distribution of two key barrier proteins, zona occludens-1 (ZO-1) and occludin. HA35 prevented the degradation or loss of ZO-1 and maintained the spatial distribution of both ZO-1 and occludin under hyperglycemic stress. Functionally, we also observed a protective effect of HA35 on the epithelial barrier under high glucose conditions. We found that HA receptor, layilin, was involved in preventing barrier protein loss (ZO-1) as well as maintaining spatial distribution of ZO-1 and occludin. Additionally, proteomics analysis showed that cell death and survival was the primary pathway upregulated in organoids treated with HA35 under high glucose stress. We found that XIAP associated factor 1 (Xaf1) was modulated by HA35 thereby regulating apoptotic cell death in the intestinal organoid system. Finally, we observed that spatial organization of both focal adhesion kinase (FAK) as well as F-actin was mediated by HA35 via layilin. CONCLUSION Our results highlight the impact of hyperglycemic stress on the intestinal barrier function. This is of clinical relevance, as impaired barrier function has been observed in individuals with metabolic syndrome. Additionally, we demonstrate barrier protective effects of HA35 through its receptor layilin and modulation of cellular apoptosis under high glucose stress.
Collapse
Affiliation(s)
- Samreen Jatana
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - Amina Abbadi
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Gail A West
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - András K Ponti
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Manuel B Braga-Neto
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Jordyn L Smith
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Armando Marino-Melendez
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Belinda Willard
- Proteomics and Metabolomics Core, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Laura E Nagy
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA; Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Carol de la Motte
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio, USA.
| |
Collapse
|
23
|
Xu R, Vujić N, Bianco V, Reinisch I, Kratky D, Krstic J, Prokesch A. Lipid-associated macrophages between aggravation and alleviation of metabolic diseases. Trends Endocrinol Metab 2024; 35:981-995. [PMID: 38705759 DOI: 10.1016/j.tem.2024.04.009] [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: 01/20/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 05/07/2024]
Abstract
Lipid-associated macrophages (LAMs) are phagocytic cells with lipid-handling capacity identified in various metabolic derangements. During disease development, they locate to atherosclerotic plaques, adipose tissue (AT) of individuals with obesity, liver lesions in steatosis and steatohepatitis, and the intestinal lamina propria. LAMs can also emerge in the metabolically demanding microenvironment of certain tumors. In this review, we discuss major questions regarding LAM recruitment, differentiation, and self-renewal, and, ultimately, their acute and chronic functional impact on the development of metabolic diseases. Further studies need to clarify whether and under which circumstances LAMs drive disease progression or resolution and how their phenotype can be modulated to ameliorate metabolic disorders.
Collapse
Affiliation(s)
- Ruonan Xu
- Gottfried Schatz Research Center for Cell Signaling, Metabolism, and Aging, Division of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria
| | - Nemanja Vujić
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Valentina Bianco
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Isabel Reinisch
- Institute of Food Nutrition and Health, Department of Health Sciences and Technology, Eidgenössische Technische Hochschule Zürich (ETH), Schwerzenbach, Switzerland
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Jelena Krstic
- Gottfried Schatz Research Center for Cell Signaling, Metabolism, and Aging, Division of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Andreas Prokesch
- Gottfried Schatz Research Center for Cell Signaling, Metabolism, and Aging, Division of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
24
|
Suder A, Makiel K, Targosz A, Kosowski P, Malina RM. Positive Effects of Aerobic-Resistance Exercise and an Ad Libitum High-Protein, Low-Glycemic Index Diet on Irisin, Omentin, and Dyslipidemia in Men with Abdominal Obesity: A Randomized Controlled Trial. Nutrients 2024; 16:3480. [PMID: 39458475 PMCID: PMC11510197 DOI: 10.3390/nu16203480] [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: 08/19/2024] [Revised: 10/11/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
OBJECTIVES The aim of this research was to evaluate changes in body composition, adipokine levels, and dyslipidemia parameters in males with abdominal obesity following two distinct interventions: exercise alone and exercise combined with an ad libitum diet. METHODS This study included 44 males with abdominal obesity (mean age 34.7 ± 5.5 years, waist circumference [WC] 110.3 ± 8.5, BMI 32.0 ± 3.9), who were randomly assigned to three groups: an experimental group engaging in aerobic-resistance exercise (II, n = 16), an experimental group engaging in aerobic-resistance exercise combined with an ad libitum high-protein, low-glycemic index carbohydrate diet (III, n = 16), both interventions lasting 6 weeks, and a control group without interventions (I, n = 12). Body composition metrics (body mass index [BMI], waist circumference [WC], body fat [BF], abdominal fat [ABD]) and fat-free mass [FFM], along with biochemical blood analyses (irisin [IR], omentin [OMEN], glucose [GLU], insulin [INS], LDL- and HDL-cholesterol), were measured at baseline and after the 6-week intervention. The effects of the interventions on the analyzed variables across groups were assessed using mixed ANOVA tests with post-hoc comparisons. Effect size (ES) was also calculated using partial eta squared (ηp2). RESULTS The intervention in group III resulted in a significant decrease in IR (p < 0.01, ηp2 = 0.03) by 41% and LDL-C (p < 0.01, ηp2 = 0.02) by 14%. These effects were associated with a reduction in BF (p < 0.01, ηp2 = 0.02) by 14%, ABD (p < 0.01, ηp2 = 0.03) by 31%, and WC (p < 0.01, ηp2 = 0.01) by 3%. In group II, decreases after 6 weeks of intervention were noted only in WC (p = 0.02, ηp2 = 0.01) by 1% and in INS (p < 0.01, ηp2 = 0.04) by 47%. No differences were found between groups. The use of low-glycemic index carbohydrates (p < 0.01, ηp2 = 0.06) and increased protein intake (p < 0.01, ηp2 = 0.30) led to changes in the fiber-to-energy value of the diet ratio (p < 0.01, ηp2 = 0.18) and a reduction in dietary energy value (p < 0.01, ηp2 = 0.13) by 23%, resulting in a greater energy deficit than in the II group. CONCLUSIONS These findings highlight the effect of combining dietary and exercise interventions to achieve significant changes in body composition and metabolic parameters, even over a short period of intervention.
Collapse
Affiliation(s)
- Agnieszka Suder
- Department of Anatomy, Faculty of Physical Rehabilitation, University of Physical Education, 31-571 Cracow, Poland
| | - Karol Makiel
- Department of Anatomy, Faculty of Physical Rehabilitation, University of Physical Education, 31-571 Cracow, Poland
| | - Aneta Targosz
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland
| | - Piotr Kosowski
- Department of Petroleum Engineering, AGH University, 30-059 Cracow, Poland
| | - Robert M. Malina
- Department of Kinesiology and Health Education, University of Texas at Austin, Austin, TX 78712, USA
- School of Public Health and Information Sciences, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
25
|
Mohaghegh N, Ahari A, Buttles C, Davani S, Hoang H, Huang Q, Huang Y, Hosseinpour B, Abbasgholizadeh R, Cottingham AL, Farhadi N, Akbari M, Kang H, Khademhosseini A, Jucaud V, Pearson RM, Hassani Najafabadi A. Simvastatin-Loaded Polymeric Nanoparticles: Targeting Inflammatory Macrophages for Local Adipose Tissue Browning in Obesity Treatment. ACS NANO 2024; 18:27764-27781. [PMID: 39342648 DOI: 10.1021/acsnano.4c10742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Obesity is defined as chronic, low-grade inflammation within specific tissues. Given the escalating prevalence of obesity among individuals of all ages, obesity has reached epidemic proportions, posing an important public health challenge. Despite significant advancements in treating obesity, conventional approaches remain largely ineffective or involve severe side effects, thus underscoring the pressing need to explore and develop treatment approaches. Targeted and local immunomodulation using nanoparticles (NPs) can influence fat production and utilization processes. Statins, known for their anti-inflammatory properties, show the potential for mitigating obesity-related inflammation. A localized delivery option offers several advantages over oral and parenteral delivery methods. Here, we developed simvastatin (Sim) encapsulated within PLGA NPs (Sim-NP) for localized delivery of Sim to adipose tissues (ATs) for immunomodulation to treat obesity. In vitro experiments revealed the strong anti-inflammatory effects of Sim-NPs, which resulted in enhanced modulation of macrophage (MΦ) polarization and induction of AT browning. We then extended our investigation to an in vivo mouse model of high-fat-diet (HFD)-induced obesity. Sim-NP administration led to the controlled release of Sim within AT, directly impacting MΦ activity and inducing AT browning while inducing weight loss. Our findings demonstrated that Sim-NP administration effectively inhibited the progression of obesity-related inflammation, controlled white fat production, and enhanced AT modulation. These results highlight the potential of Sim-NP as a potent nanotherapy for treating obesity by modulating the immune system.
Collapse
Affiliation(s)
- Neda Mohaghegh
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Amir Ahari
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
- Department of Surgery, University of California-Los Angeles, Los Angeles, California 90095, United States
| | - Claire Buttles
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
- Department of Biology, Indiana University Bloomington, Bloomington, Indiana 47405, United States
| | - Saya Davani
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
- Department of Neurobiology, Physiology, and Behavior, University of California Davis, Briggs Hall, Davis, California 95616, United States
| | - Hanna Hoang
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California 90024, United States
| | - Qiang Huang
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Yixuan Huang
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Bahareh Hosseinpour
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
- Department of Chemistry, Carleton University, Ottawa, Ontario K1S 5B6, Canada
| | - Reza Abbasgholizadeh
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Andrea L Cottingham
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Neda Farhadi
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Mohsen Akbari
- Department of Mechanical Engineering, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
- Biotechnology Center, Silesian University of Technology, Gliwice 44-100, Poland
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Ryan M Pearson
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | | |
Collapse
|
26
|
Zhu Z, Yu M, Xu M, Ji X, Zong X, Zhang Z, Shang W, Zhang L, Fang P. Baicalin suppresses macrophage JNK-mediated adipose tissue inflammation to mitigate insulin resistance in obesity. JOURNAL OF ETHNOPHARMACOLOGY 2024; 332:118355. [PMID: 38762213 DOI: 10.1016/j.jep.2024.118355] [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: 01/15/2024] [Revised: 05/05/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Radix scutellariae (the root of Scutellaria baicalensis Georgi) is a traditional Chinese medicine (TCM) used to treat a wide range of inflammation-related diseases, such as obesity, diabetes, diabetic kidney disease, and COVID-19-associated inflammatory states in the lung and kidney. Baicalin is the major anti-inflammatory component of Radix scutellariae and has shown the potential to inhibit inflammation in metabolic disorders. In this study, we explored the ability and underlying mechanisms of baicalin to modulate the macrophage to mitigate insulin resistance in obesity. MATERIALS AND METHODS Obese mice were administered baicalin (50 mg/kg/day) intraperitoneally for 3 weeks. RAW264.7 and BMDM cells were stimulated with LPS and treated with baicalin for 24 h, while 3T3-L1 and primary white adipocytes were treated with the supernatants from baicalin-treated RAW264.7 cells for 24 h. RESULTS The results showed that baicalin significantly improved glucose and insulin tolerance as well as decreased fat and adipose tissue macrophage levels in obese mice. Besides, baicalin significantly reduced serum and adipose tissue IL-1β, TNF-α and IL-6 levels in obese mice, as well as suppressed LPS-induced IL-1β, TNF-α and IL-6 expression and release in macrophages. Furthermore, treatment with the supernatant from baicalin-treated RAW264.7 cells increased the levels of PGC-1α, SIRT1, p-IRS-1 and p-AKT in adipocytes. Moreover, baicalin treatment dramatically downregulated macrophage p-p38, p-JNK, and Ac-p65Lys310 levels while increasing SIRT1 both in vivo and in vitro. Importantly, JNK inhibitor SP600125 blocked most of the effects of baicalin on SIRT1, Ac-p65Lys310 and pro-inflammatory factors in macrophages. CONCLUSION Therefore, these results demonstrated for the first time that baicalin exerts its anti-inflammatory effects in obese adipose tissue macrophages mainly through suppressing JNK/SIRT1/p65 signaling. These findings amplified the mechanisms of baicalin and its potential to attenuate insulin resistance.
Collapse
Affiliation(s)
- Ziyue Zhu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Mei Yu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Mengfan Xu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xin Ji
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China
| | - Xicui Zong
- Hanlin College, Nanjing University of Chinese Medicine, Taizhou, 225300, China
| | - Zhenwen Zhang
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China.
| | - Wenbin Shang
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Li Zhang
- Hanlin College, Nanjing University of Chinese Medicine, Taizhou, 225300, China.
| | - Penghua Fang
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
27
|
Li X, Yang W, Ma K, Zheng Z, Liu X, Hu B, Liu H, Zhao Q, Han Y, Xiao Z, Chen R, Li H, Huang S, Liu J, Wang C, Yin L, Meng Y. Circulating B Cell-Derived Small RNA Delivered by Extracellular Vesicles: A Dialogue Mechanism for Long-Range Targeted Renal Mitochondrial Injury in Obesity. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402526. [PMID: 38958071 DOI: 10.1002/smll.202402526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/24/2024] [Indexed: 07/04/2024]
Abstract
The intricate processes that govern the interactions between peripatetic immune cells and distal renal injury in obesity are not fully understood. Employing transcriptomic analysis of circulating extracellular vesicles (EVs), a marked amplification of small RNA (miR-3960) is discerned within CD3-CD19+ B cells. This RNA is found to be preferentially augmented in kidney tissues, contrasting with its subdued expression in other organs. By synthesizing dual-luciferase reporter assay with co-immunoprecipitation analysis, it is pinpointed that miR-3960 specifically targets the nuclear gene TRMT5, a pivotal actor in the methylation of mitochondrial tRNA. This liaison instigates aberrations in the post-transcriptional modifications of mitochondrial tRNA, engendering deficiencies within the electron respiratory chain, primarily attributable to the diminution of the mitochondrial bioenergetic compound (NDUFA7) complex I. Such perturbations lead to a compromised mitochondrial respiratory capacity in renal tubular cells, thereby exacerbating tubular injury. In contrast, EV blockade or miR-3960 depletion markedly alleviates renal tubular injury in obesity. This investigation unveils a hitherto unexplored pathway by which obesity-induced circulating immune cells remotely manipulate mitochondrial metabolism in target organs. The strategic targeting of obese EVs or infiltrative immune cells and their specifically secreted RNAs emerges as a promising therapeutic avenue to forestall obesity-related renal afflictions.
Collapse
Affiliation(s)
- Xiaqing Li
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, 510632, China
- Nephrology department, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, Guangdong, 517000, China
| | - Wah Yang
- Department of Obesity and Metabolic Disorders, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, 510632, China
- Institute of Obesity and Metabolic Disorders, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Ke Ma
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, 510632, China
- Nephrology department, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, Guangdong, 517000, China
| | - Zirun Zheng
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, 510632, China
- Nephrology department, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, Guangdong, 517000, China
| | - Xiayun Liu
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, 510632, China
- Nephrology department, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, Guangdong, 517000, China
| | - Bo Hu
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Huanhuan Liu
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Qian Zhao
- Department of Infectious Diseases and Hepatology Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510400, China
| | - Yi Han
- Traditional Chinese Medicine Department, People's Hospital of Yanjiang District, Ziyang, Sichuan, 641300, China
| | - Zhangzhang Xiao
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, 510632, China
- Department of Nephrology, Houjie Hospital of Dongguan, Dongguan, Guangdong, 523945, China
| | - Ruichang Chen
- Department of Emergency Medicine, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Hongyue Li
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, 510632, China
- Nephrology department, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, Guangdong, 517000, China
| | - Sibo Huang
- Health Management Center, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Jinfeng Liu
- Department of Gastroenterology, Binhaiwan Central Hospital of Dongguan, Dongguan, Guangdong, 523000, China
| | - Cunchuan Wang
- Department of Obesity and Metabolic Disorders, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, 510632, China
- Institute of Obesity and Metabolic Disorders, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Lianghong Yin
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, 510632, China
- Huangpu Institute of Materials, Guangzhou, Guangdong, 510663, China
| | - Yu Meng
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, 510632, China
- Nephrology department, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, Guangdong, 517000, China
- Nephrology Department and Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, Guangdong, 517000, China
| |
Collapse
|
28
|
He M, Cui Q, Zheng Y, Feng B, Liu Z. Increased nuclear receptor subfamily 2, group E, member 1 (NR2E1) concentrations of PBMCs are associated with chronic inflammation in overweight/obesity. Heliyon 2024; 10:e37909. [PMID: 39323832 PMCID: PMC11422594 DOI: 10.1016/j.heliyon.2024.e37909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 09/02/2024] [Accepted: 09/12/2024] [Indexed: 09/27/2024] Open
Abstract
Background Chronic inflammation plays a crucial role in the pathogenesis of overweight/obesity. Nuclear receptor subfamily 2, group E, member 1 (NR2E1) is one of the nuclear receptor family proteins that play crucial roles in regulating numerous life processes. In this study, we attempted to detect NR2E1 levels in peripheral blood mononuclear cells (PBMCs) of overweight/obese people and preliminarily elucidate the regulatory role of NR2E1 in obesity-related chronic inflammation. Methods We conducted a cross-sectional analysis of the clinical and biochemical data from 62 overweight/obese people and 70 control subjects. PBMCs of the participants were collected for detection of NR2E1 levels. PBMCs isolated from the control subjects were treated with different concentrations of palmitic acid (PA). We also transfected p-EGFP-N1-NR2E1 plasmids into PBMCs and treated them with PA, then detected TNF-α and IL-6 concentrations in the supernatant of PBMCs. Results The NR2E1 mRNA and protein levels in overweight/obese people were both significantly higher than those in normal-BMI people (p < 0.01). NR2E1 mRNA levels in PBMCs of overweight/obese people were positively related with TC, FFA, IL-6, TNF-α (r = 0.387, 0.440, 0.610, 0.530, p < 0.01) and LDL-c (r = 0.290, p < 0.05). A similar correlation was also found between NR2E1 protein levels and these parameters. The expression of NR2E1 in PBMCs from the control subjects increased apparently with the treatment of PA in a concentration-depend manner in vitro. Overexpression of NR2E1 in PBMCs decreased TNF-α and IL-6 expression induced by PA (p < 0.01). Conclusion NR2E1 levels are increased in overweight/obese people and have a positive relationship with TC, FFA, LDL-C, TNF-α and IL-6. Overexpression of NR2E1 could alleviate PA-induced chronic inflammation. NR2E1 may be a potential target for regulating chronic inflammation in obesity.
Collapse
Affiliation(s)
- Mingqing He
- Department of Geriatrics, The First Affiliated Hospital of Soochow University, No.188 Shizi Street, Suzhou, Jiangsu, 215006, China
| | - Qiyuan Cui
- Department of Geriatrics, The First Affiliated Hospital of Soochow University, No.188 Shizi Street, Suzhou, Jiangsu, 215006, China
| | - Yun Zheng
- Department of Geriatrics, The First Affiliated Hospital of Soochow University, No.188 Shizi Street, Suzhou, Jiangsu, 215006, China
| | - Bin Feng
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, No.188 Shizi Street, Suzhou, Jiangsu, 215006, China
| | - Zheng Liu
- Department of Endocrinology, The First Affiliated Hospital of Soochow University, No.188 Shizi Street, Suzhou, Jiangsu, 215006, China
| |
Collapse
|
29
|
Marko DM, Conn MO, Schertzer JD. Intermittent fasting influences immunity and metabolism. Trends Endocrinol Metab 2024; 35:821-833. [PMID: 38719726 DOI: 10.1016/j.tem.2024.04.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 09/12/2024]
Abstract
Intermittent fasting (IF) modifies cell- and tissue-specific immunometabolic responses that dictate metabolic flexibility and inflammation during obesity and type 2 diabetes (T2D). Fasting forces periods of metabolic flexibility and necessitates increased use of different substrates. IF can lower metabolic inflammation and improve glucose metabolism without lowering obesity and can influence time-dependent, compartmentalized changes in immunity. Liver, adipose tissue, skeletal muscle, and immune cells communicate to relay metabolic and immune signals during fasting. Here we review the connections between metabolic and immune cells to explain the divergent effects of IF compared with classic caloric restriction (CR) strategies. We also explore how the immunometabolism of metabolic diseases dictates certain IF outcomes, where the gut microbiota triggers changes in immunity and metabolism during fasting.
Collapse
Affiliation(s)
- Daniel M Marko
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada; Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, ON, Canada
| | - Meghan O Conn
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada; Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, ON, Canada
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada; Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
30
|
Clark M, Suur BE, Soták M, Börgeson E. Attenuation of adipose tissue inflammation by pro-resolving lipid mediators. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2024; 36:100539. [PMID: 39619489 PMCID: PMC11602545 DOI: 10.1016/j.coemr.2024.100539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/24/2024] [Accepted: 07/14/2024] [Indexed: 01/03/2025]
Abstract
Adipose tissue inflammation drives systemic pathophysiology, for instance, obesity-related cardiometabolic disease. Specialized pro-resolving lipid mediators are a superfamily of endogenously produced lipids that promote the resolution of inflammation, an actively regulated process. New evidence suggests that such lipids (e.g. lipoxins) could resolve adipose tissue inflammation and, thus, subvert obesity-related diseases. A key feature of pro-resolving lipids is their ability to promote an M2-like macrophage phenotype and enhance efferocytosis while avoiding adverse side-effects typically associated with anti-inflammatory drugs, such as increased sensitivity to infections. This brief review discusses the therapeutic potential of pro-resolving lipid mediators in mitigating systemic disease fueled by adipose tissue inflammation in both experimental and human disease models.
Collapse
Affiliation(s)
- Madison Clark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Bianca E. Suur
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Matúš Soták
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Emma Börgeson
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
31
|
Hu Y, Zou W, Zhang L, Zhang S, Hu L, Song Z, Kong S, Gao Y, Zhang J, Yang Y, Zheng J. TRPV3 facilitates lipolysis and attenuates diet-induced obesity via activation of the NRF2/FSP1 signaling axis. Free Radic Biol Med 2024; 221:155-168. [PMID: 38777204 DOI: 10.1016/j.freeradbiomed.2024.05.035] [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: 02/20/2024] [Revised: 05/17/2024] [Accepted: 05/19/2024] [Indexed: 05/25/2024]
Abstract
Transient receptor potential vanilloid (TRPV) ion channels play a crucial role in various cellular functions by regulating intracellular Ca2+ levels and have been extensively studied in the context of several metabolic diseases. However, the regulatory effects of TRPV3 in obesity and lipolysis are not well understood. In this study, utilizing a TRPV3 gain-of-function mouse model (TRPV3G568V/G568V), we assessed the metabolic phenotype of both TRPV3G568V/G568V mice and their control littermates, which were randomly assigned to either a 12-week high-fat diet or a control diet. We investigated the potential mechanisms underlying the role of TRPV3 in restraining obesity and promoting lipolysis both in vivo and in vitro. Our findings indicate that a high-fat diet led to significant obesity, characterized by increased epididymal and inguinal white adipose tissue weight and higher fat mass. However, the gain-of-function mutation in TRPV3 appeared to counteract these adverse effects by enhancing lipolysis in visceral fat through the upregulation of the major lipolytic enzyme, adipocyte triglyceride lipase (ATGL). In vitro experiments using carvacrol, a TRPV3 agonist, demonstrated the promotion of lipolysis and antioxidation in 3T3-L1 adipocytes after TRPV3 activation. Notably, carvacrol failed to stimulate Ca2+ influx, lipolysis, and antioxidation in 3T3-L1 adipocytes treated with BAPTA-AM, a cell-permeable calcium chelator. Our results revealed that TRPV3 activation induced the action of transcriptional factor nuclear factor erythroid 2-related factor 2 (NRF2), resulting in increased expression of ferroptosis suppressor protein 1 (FSP1) and superoxide dismutase2 (SOD2). Moreover, the inhibition of NRF2 impeded carvacrol-induced lipolysis and antioxidation in 3T3-L1 adipocytes, with downregulation of ATGL, FSP1, and SOD2. In summary, our study suggests that TRPV3 promotes visceral fat lipolysis and inhibits diet-induced obesity through the activation of the NRF2/FSP1 signaling axis. We propose that TRPV3 may be a potential therapeutic target in the treatment of obesity.
Collapse
Affiliation(s)
- Yongyan Hu
- Laboratory Animal Facility, Peking University First Hospital, Beijing, China
| | - Wenyu Zou
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Ling Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Shixuan Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Linghan Hu
- Genetic Skin Disease Center, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Zhongya Song
- Genetic Skin Disease Center, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Shenshen Kong
- Laboratory Animal Facility, Peking University First Hospital, Beijing, China
| | - Ying Gao
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Junqing Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Yong Yang
- Genetic Skin Disease Center, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Jia Zheng
- Department of Endocrinology, Peking University First Hospital, Beijing, China.
| |
Collapse
|
32
|
Zhou Y, Wang Y, Wu T, Zhang A, Li Y. Association between obesity and systemic immune inflammation index, systemic inflammation response index among US adults: a population-based analysis. Lipids Health Dis 2024; 23:245. [PMID: 39127686 DOI: 10.1186/s12944-024-02240-8] [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: 07/01/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Obesity is characterized by a chronic low-grade inflammatory condition. Two emerging inflammatory biomarkers, the systemic immune-inflammation index (SII) and the systemic inflammation response index (SIRI), have gained attention. However, the relationships between obesity and SII/SRI remain unclear. METHODS In this study, we analyzed data from the National Health and Nutrition Examination Survey (NHANES) from 2011 to 2018 among adults. SII-SIRI/SII/SIRI were categorized into three groups based on tertiles. The association between obesity and SII-SIRI/SII/SIRI was assessed by multivariable logistic regression models. Restricted cubic spline (RCS) plots were used to examine the nonlinear association between obesity and SII/SIRI. Finally, potential independent associations between obesity and SII/SIRI were further explored using subgroup analyses. RESULTS The study included 20,011 adults, of whom 7,890 (39.32%) were obesity. In model 1, participants in the high (Q3) level of SII-SIRI had a significantly association with obesity than those in the low (Q1) level group. The high level of SII and SIRI were positively associated with obesity as compared to low levels. Model 2 revealed a positive association between obesity and high levels of SII-SIRI/SII/SIRI. Model 3 demonstrated a similar trend. RCS curves revealed a nonlinear association linking obesity to SII/SIRI. Subgroup analysis showed an interaction between SII/SIRI and age. CONCLUSIONS Our research suggested that obesity was positively associated with SII-SIRI/SII/SIRI in U.S. adults. SII/SIRI may represent a cost-effective and direct approach to assessing obesity.
Collapse
Affiliation(s)
- Yaoyao Zhou
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310051, China
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yaqi Wang
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310051, China
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Taotao Wu
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, 310051, China
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Anqi Zhang
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yingshuai Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
33
|
Zhang Y, Wang W. Probiotics in reducing obesity by reconfiguring the gut microbiota. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2024; 49:1042-1051. [PMID: 39788492 PMCID: PMC11495983 DOI: 10.11817/j.issn.1672-7347.2024.240361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Indexed: 01/05/2025]
Abstract
Obesity, as a global health crisis, is increasingly linked to intestinal microecology. Probiotics colonise the body, effectively regulating the balance of intestinal flora, while strengthening the intestinal barrier, activating the immune response, releasing beneficial substances, and maintaining micro-ecological balance. This process not only enhances the defence against pathogens, but also reduces the production of inflammatory factors and lowers the level of chronic inflammation. However, the specific process and mechanism by which probiotics influence the intestinal microecology through the immune response, improve metabolic disorders caused by obesity, and participate in weight management are not clear. Through multiple neural pathways including the 'gut-brain axis' and their direct interaction with the intestine, probiotics increase the number of beneficial bacteria in the intestine and inhibit the growth of harmful bacteria, thus effectively restructuring the balance of the intestinal flora. This restructuring of the balance can optimise the intestinal environment and enhance the efficiency of food digestion and nutrient absorption. Probiotics show positive effects on obesity management by regulating the metabolic process and reducing fat accumulation, providing individuals with a new way to control body weight and prevent obesity. Therefore, the application of probiotics is of great significance in promoting gut health and weight management.
Collapse
Affiliation(s)
- Yunuo Zhang
- Department of Endocrinology, First Affiliated Hospital of Baotou Medical College, Baotou Inner Mongolia Autonomous Region 014010, China.
| | - Wei Wang
- Department of Endocrinology, First Affiliated Hospital of Baotou Medical College, Baotou Inner Mongolia Autonomous Region 014010, China.
| |
Collapse
|
34
|
Girotto OS, Furlan OO, Moretti Junior RC, Goulart RDA, Baldi Junior E, Barbalho-Lamas C, Fornari Laurindo L, Barbalho SM. Effects of apples ( Malus domestica) and their derivatives on metabolic conditions related to inflammation and oxidative stress and an overview of by-products use in food processing. Crit Rev Food Sci Nutr 2024:1-32. [PMID: 39049560 DOI: 10.1080/10408398.2024.2372690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Apple (Malus domestica) is the third most produced fruit worldwide. It is a well-known source of bioactive compounds mainly represented by hydroxycinnamic acids, flavan-3-ols, dihydrochalcones, dehydroascorbic acid, carotenoids, chlorogenic acid, epicatechin, and phloridzin. Due to the lack of a recent evaluation of the clinical trials associated with apple consumption, this review investigated the effects of this fruit on metabolic conditions related to inflammation and oxidative stress and reviewed the applications of apple waste on food products. Thirty-three studies showed that apples or its derivatives exhibit anti-inflammatory and antioxidant actions, improve blood pressure, body fat, insulin resistance, dyslipidemia, and reduce cardiovascular risks. Apples have a great economic impact due to its several applications in the food industry and as a food supplement since it has impressive nutritional value. Dietary fiber from the fruit pomace can be used as a substitute for fat in food products or as an improver of fiber content in meat products. It can also be used in bakery and confectionary products or be fermented to produce alcohol. Pomace phytocompounds can also be isolated and applied as antioxidants in food products. The potential for the use of apples and by-products in the food industry can reduce environmental damage.
Collapse
Affiliation(s)
- Otávio Simões Girotto
- Department of Biochemistry, School of Medicine, University of Marília (UNIMAR), Marília, SP, Brazil
| | - Otávio Oliveira Furlan
- Department of Biochemistry, School of Medicine, University of Marília (UNIMAR), Marília, SP, Brazil
| | | | - Ricardo de Alvares Goulart
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation - UNIMAR, Marília, SP, Brazil
| | - Edgar Baldi Junior
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation - UNIMAR, Marília, SP, Brazil
| | - Caroline Barbalho-Lamas
- Department of Gerontology, School of Gerontology, Universidade Federal de São Carlos (UFSCar), São Carlos, SP, Brazil
| | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília, SP, Brazil
| | - Sandra M Barbalho
- Department of Biochemistry, School of Medicine, University of Marília (UNIMAR), Marília, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation - UNIMAR, Marília, SP, Brazil
- School of Food and Technonolgy of Marilia (FATEC), São Paulo, Brazil
| |
Collapse
|
35
|
de Jesus Simão J, de Sousa Bispo AF, Plata VTG, Armelin-Correa LM, Alonso-Vale MIC. Fish Oil Supplementation Mitigates High-Fat Diet-Induced Obesity: Exploring Epigenetic Modulation and Genes Associated with Adipose Tissue Dysfunction in Mice. Pharmaceuticals (Basel) 2024; 17:861. [PMID: 39065712 PMCID: PMC11280081 DOI: 10.3390/ph17070861] [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: 04/13/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
This study investigated the effects of fish oil (FO) treatment, particularly enriched with eicosapentaenoic acid (EPA), on obesity induced by a high-fat diet (HFD) in mice. The investigation focused on elucidating the impact of FO on epigenetic modifications in white adipose tissue (WAT) and the involvement of adipose-derived stem cells (ASCs). C57BL/6j mice were divided into two groups: control diet and HFD for 16 weeks. In the last 8 weeks, the HFD group was subdivided into HFD and HFD + FO (treated with FO). WAT was removed for RNA and protein extraction, while ASCs were isolated, cultured, and treated with leptin. All samples were analyzed using functional genomics tools, including PCR-array, RT-PCR, and Western Blot assays. Mice receiving an HFD displayed increased body mass, fat accumulation, and altered gene expression associated with WAT inflammation and dysfunction. FO supplementation attenuated these effects, a potential protective role against HFD-induced obesity. Analysis of H3K27 revealed HFD-induced changes in histone, which were partially reversed by FO treatment. This study further explored leptin signaling in ASCs, suggesting a potential mechanism for ASC dysfunction in the obesity-rich leptin environment of WAT. Overall, FO supplementation demonstrated efficacy in mitigating HFD-induced obesity, influencing epigenetic and molecular pathways, and shedding light on the role of ASCs and leptin signaling in WAT dysfunction associated with obesity.
Collapse
Affiliation(s)
- Jussara de Jesus Simão
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo—UNIFESP, Diadema 09913-030, Brazil; (J.d.J.S.); (A.F.d.S.B.); (V.T.G.P.); (L.M.A.-C.)
| | - Andressa França de Sousa Bispo
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo—UNIFESP, Diadema 09913-030, Brazil; (J.d.J.S.); (A.F.d.S.B.); (V.T.G.P.); (L.M.A.-C.)
| | - Victor Tadeu Gonçalves Plata
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo—UNIFESP, Diadema 09913-030, Brazil; (J.d.J.S.); (A.F.d.S.B.); (V.T.G.P.); (L.M.A.-C.)
| | - Lucia Maria Armelin-Correa
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo—UNIFESP, Diadema 09913-030, Brazil; (J.d.J.S.); (A.F.d.S.B.); (V.T.G.P.); (L.M.A.-C.)
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo—UNIFESP, Diadema 09913-030, Brazil
| | - Maria Isabel Cardoso Alonso-Vale
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo—UNIFESP, Diadema 09913-030, Brazil; (J.d.J.S.); (A.F.d.S.B.); (V.T.G.P.); (L.M.A.-C.)
- Department of Biological Sciences, Institute of Environmental Sciences, Chemical and Pharmaceutical, Federal University of São Paulo—UNIFESP, Diadema 09913-030, Brazil
| |
Collapse
|
36
|
Schleh MW, Ameka M, Rodriguez A, Hasty AH. Deficiency of the hemoglobin-haptoglobin receptor, CD163, worsens insulin sensitivity in obese male mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.31.596887. [PMID: 38895370 PMCID: PMC11185572 DOI: 10.1101/2024.05.31.596887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Excessive iron accumulation in metabolic organs such as the adipose tissue, liver, and skeletal muscle is associated with increased diabetes risk. Tissue-resident macrophages serve multiple roles including managing inflammatory tone and regulating parachymal iron homeostasis; thus protecting against metabolic dysfunction upon iron overload. The scavenger receptor CD163 is uniquely present on tissue-resident macrophages, and plays a significant role in iron homeostasis by clearing extracellular hemoglobin-haptoglobin complexes, thereby limiting oxidative damage caused by free hemoglobin in metabolic tissues. We show that the absence of CD163 exacerbates glucose intolerance and insulin resistance in male mice with obesity. Additionally, loss of CD163 reduced the expression of iron regulatory genes (Tfr1, Cisd1, Slc40a1) in adipose tissue macrophages and anti-inflammatory (M2-like) bone marrow-derived macrophages (BMDMs). Further, CD163 deficiency mediated a pro-inflammatory shift and limited hemoglobin scavenging specifically in M2-like BMDMs. To this end, iron buffering was diminished in inguinal white adipose tissue (iWAT) macrophages in vivo, which culminated in iron spillover into adipocytes and CD45+CD11B- non-myeloid immune cells in iWAT. These findings show that CD163 on tissue-resident macrophages is critical for their anti-inflammatory and hemoglobin scavenging roles, and its absence results in impaired systemic insulin action in an obese setting.
Collapse
Affiliation(s)
- Michael W Schleh
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine; Nashville, TN 37232, USA
| | - Magdalene Ameka
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine; Nashville, TN 37232, USA
| | - Alec Rodriguez
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine; Nashville, TN 37232, USA
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine; Nashville, TN 37232, USA
- VA Tennessee Valley Healthcare System; Nashville, TN 37212, USA
| |
Collapse
|
37
|
Quarleri J, Delpino MV. The interplay of aging, adipose tissue, and COVID-19: a potent alliance with implications for health. GeroScience 2024; 46:2915-2932. [PMID: 38191833 PMCID: PMC11009220 DOI: 10.1007/s11357-023-01058-z] [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: 11/24/2023] [Accepted: 12/27/2023] [Indexed: 01/10/2024] Open
Abstract
Obesity has emerged as a significant public health challenge. With the ongoing increase in life expectancy, the prevalence of obesity is steadily growing, particularly among older age demographics. The extension of life expectancy frequently results in additional years of vulnerability to chronic health issues associated with obesity in the elderly.The concept of SARS-CoV-2 directly infecting adipose tissue stems from the fact that both adipocytes and stromal vascular fraction cells express ACE2, the primary receptor facilitating SARS-CoV-2 entry. It is noteworthy that adipose tissue demonstrates ACE2 expression levels similar to those found in the lungs within the same individual. Additionally, ACE2 expression in the adipose tissue of obese individuals surpasses that in non-obese counterparts. Viral attachment to ACE2 has the potential to disturb the equilibrium of renin-angiotensin system homeostasis, leading to an exacerbated inflammatory response.Consequently, adipose tissue has been investigated as a potential site for active SARS-CoV-2 infection, suggesting its plausible role in virus persistence and contribution to both acute and long-term consequences associated with COVID-19.This review is dedicated to presenting current evidence concerning the presence of SARS-CoV-2 in the adipose tissue of elderly individuals infected with the virus. Both obesity and aging are circumstances that contribute to severe health challenges, heightening the risk of disease and mortality. We will particularly focus on examining the mechanisms implicated in the long-term consequences, with the intention of providing insights into potential strategies for mitigating the aftermath of the disease.
Collapse
Affiliation(s)
- Jorge Quarleri
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, CONICET, Paraguay 2155, Piso 11, C1121ABG, Ciudad Autónoma de Buenos Aires, Argentina.
| | - M Victoria Delpino
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, CONICET, Paraguay 2155, Piso 11, C1121ABG, Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|