1
|
Zong J, Wu X, Huang X, Yuan L, Yuan K, Zhang Z, Jiang M, Ping Z, Cheong LY, Xu A, Hoo RLC. Adipocyte-derived shed Syndecan-4 suppresses lipolysis contributing to impaired adipose tissue browning and adaptive thermogenesis. Mol Metab 2025; 96:102133. [PMID: 40180176 PMCID: PMC12004711 DOI: 10.1016/j.molmet.2025.102133] [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: 01/08/2025] [Revised: 03/17/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025] Open
Abstract
Lipolysis in white adipose tissue (WAT) provides fatty acids as energy substrates for thermogenesis to increase energy expenditure. Syndecan-4 (Sdc4) is a transmembrane proteoglycan bearing heparan sulfate chains. Although single nucleotide polymorphisms (SNPs) of the Sdc4 gene have been identified linking to metabolic syndromes, its specific function in adipose tissue remains obscure. Here, we show that Sdc4 serves as a regulator of lipid metabolism and adaptive thermogenesis. Sdc4 expression and shedding are elevated in the white adipose tissue (WAT) of diet-induced obese mice. Adipocyte-specific deletion of Sdc4 promotes lipolysis and WAT browning, thereby raising whole-body energy expenditure to protect against diet-induced obesity. Mechanistically, fibroblast growth factor 2 (FGF2) is a paracrine factor that maintains energy homeostasis. Elevated shed Sdc4 concentrates and delivers FGF2 to fibroblast growth factor receptor 1 (FGFR1) on adipocytes, which in turn suppresses lipolysis by reducing hormone-sensitive lipase (HSL) activity, thus exaggerating adipose tissue dysfunction upon high-fat diet induction. Sdc4-deficient adipocytes show higher lipolytic and thermogenic capacity by enhancing HSL phosphorylation and UCP1 expression. Overall, our study reveals that adipocyte-derived shed Sdc4 is a novel suppressor of lipolysis, contributing to decreased energy expenditure, thus exaggerating obesity. Targeting shed Sdc4 is a potential therapeutic strategy for obesity.
Collapse
Affiliation(s)
- Jiuyu Zong
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Xiaoping Wu
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Xiaowen Huang
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Lufengzi Yuan
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Kai Yuan
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Zixuan Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Mengxue Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Zhihui Ping
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Lai Yee Cheong
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Ruby Lai Chong Hoo
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| |
Collapse
|
2
|
Zhang Y, Su X, Li Y, Cai Y, Kang N, Duan J, Chen F, Xue F, Chen X. Evaluation of salivary interleukin-17 and developmental endothelial locus-1 in patients with periodontitis with and without type 2 diabetes mellitus. J Periodontol 2025; 96:407-417. [PMID: 39311693 DOI: 10.1002/jper.23-0720] [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: 11/30/2023] [Revised: 05/10/2024] [Accepted: 06/27/2024] [Indexed: 10/27/2024]
Abstract
BACKGROUND Bidirectional positive relationship between periodontitis and type 2 diabetes mellitus (T2DM) has been recognized, interleukin 17 (IL-17) and developmental endothelial locus-1 (Del-1) are proposed to play roles in periodontitis and T2DM. This study aims to investigate the association of IL-17 and Del-1 in patients with periodontitis with and without T2DM by measuring their salivary levels. METHODS A total of 80 participants were enrolled in a cross-sectional study and divided into healthy (H, n = 27), periodontitis (P, n = 29) and periodontitis with diabetes (PDM, n = 24) groups based on their periodontal and diabetic examination results. Periodontal parameters (plaque index [PI], bleeding on probing [BOP], probing depth [PD], and clinical attachment level [CAL]) as well as diabetic parameters (fasting plasma glucose [FG] and glycated hemoglobin [HbA1c]) were documented and unstimulated saliva was collected. Salivary IL-1β, active-matrix metalloproteinase-8 (aMMP-8), tumor necrosis factor-α (TNF-α), IL-6, IL-8, IL-17, and Del-1 were determined through enzyme-linked immunosorbent assay (ELISA) and their relationships with periodontal and diabetic parameters were examined. RESULTS The periodontitis and periodontitis with diabetes groups showed significantly higher levels of IL-17 and lower levels of Del-1 compared with healthy group. The periodontitis with diabetes group exhibited higher levels of IL-17 and lower levels of Del-1 compared with the periodontitis group. After correlation analysis, there were significant correlations between salivary IL-17 and Del-1 and clinical parameters, IL-17 and Del-1 were correlated with PD (r = 0.36, -0.39, p < 0.01), CAL (r = 0.40, -0.42, p < 0.01) and BOP (r = 0.35, -0.37, p < 0.01), they were correlated with FG (r = 0.26, -0.25, p < 0.05) and HbA1c (r = 0.28, -0.40, p < 0.05). Positive relationships were observed between IL-17 and IL-1β and between IL-17 and aMMP-8 (r = 0.80, 0.77, p < 0.01), while Del-1 exhibited negative correlations with IL-1β and aMMP-8 (r = 0.59, 0.69 p < 0.01). Comparison between IL-17 and Del-1 confirmed an inverse relationship (r = -0.71, p < 0.01). Salivary Del-1 levels in the older group were lower compared with young group across the H, P and PDM groups, although these differences were not statistically significant (p > 0.05). CONCLUSIONS Salivary IL-17 and Del-1 levels in the periodontitis with diabetes group showed significant changes compared with the periodontitis group, they exhibited an inverse relationship and were both correlated with periodontal parameters (PD, CAL, and BOP) and diabetic parameters (FG and HbA1c). PLAIN LANGUAGE SUMMARY Periodontitis and type 2 diabetes mellitus (T2DM) are two common diseases all over the world, some inflammatory mediators (interleukin 17 [IL-17] and developmental endothelial locus-1 [Del-1]) regulate neutrophil production, recruitment and clearance when the body becomes infected and believed to be involved in the progress of diseases of periodontitis and diabetes. In this study, we enrolled healthy subjects, patients with periodontitis, patients with periodontitis and diabetes. We performed dental examinations and evaluated their blood glucose levels, collected their saliva, and detected IL-17 and Del-1 levels in their saliva. We found both patients with periodontitis and patients with periodontitis and diabetes showed higher IL-17 levels and lower Del-1 levels compared with healthy subjects. Patients with periodontitis and diabetes showed higher IL-17 levels and lower Del-1 levels compared with patients with periodontitis. Salivary IL-17 and Del-1 levels were both correlated with dental examination results and blood glucose levels, and salivary IL-17 and Del-1 displayed an inverse relationship.
Collapse
Affiliation(s)
- Yong Zhang
- First Clinical Division, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Xu Su
- Department of Stomatology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Yuzhi Li
- Department of Stomatology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Yu Cai
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Ni Kang
- Department of Periodontology & Hunan Key Laboratory of Oral Health Research, Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Jinyu Duan
- First Clinical Division, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Fan Chen
- Department of Stomatology, People's Hospital of Peking University, Beijing, China
| | - Fei Xue
- First Clinical Division, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Xiaotao Chen
- Department of Stomatology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| |
Collapse
|
3
|
Rigual MDM, Angulo-Aguado M, Zagorac S, Álvarez-Díaz R, Benítez-Mondéjar M, Yi F, Martínez-Garay C, Santos-de-Frutos K, Kim E, Campos-Olivas R, Djouder N. Macrophages harness hepatocyte glutamate to boost liver regeneration. Nature 2025; 641:1005-1016. [PMID: 40140584 DOI: 10.1038/s41586-025-08778-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 02/12/2025] [Indexed: 03/28/2025]
Abstract
Liver regeneration after hepatectomy follows accurate coordination with the body's specific requirements1-3. However, the molecular mechanisms, factors and particular hepatocyte population influencing its efficiency remain unclear. Here we report on a unique regeneration mechanism involving unconventional RPB5 prefoldin interactor 1 (URI1), which exclusively colocalizes with, binds to and activates glutamine synthase (GS) in pericentral hepatocytes. Genetic GS or URI1 depletion in mouse pericentral hepatocytes increases circulating glutamate levels, accelerating liver regeneration after two-third hepatectomy. Conversely, mouse hepatocytic URI1 overexpression hinders liver restoration, which can be reversed by elevating glutamate through supplementation or genetic GS depletion. Glutamate metabolically reprograms bone-marrow-derived macrophages, stabilizing HIF1α, which transcriptionally activates WNT3 to promote YAP1-dependent hepatocyte proliferation, boosting liver regeneration. GS regulation by URI1 is a mechanism that maintains optimal glutamate levels, probably to spatiotemporally fine-tune liver growth in accordance with the body's homeostasis and nutrient supply. Accordingly, in acute and chronic injury models, including in cirrhotic mice with low glutamate levels and in early mortality after liver resection, as well as in mice undergoing 90% hepatectomy, glutamate addition enhances hepatocyte proliferation and survival. Furthermore, URI1 and GS expression co-localize in human hepatocytes and correlate with WNT3 in immune cells across liver disease stages. Glutamate supplementation may therefore support liver regeneration, benefiting patients awaiting transplants or recovering from hepatectomy.
Collapse
Affiliation(s)
- María Del Mar Rigual
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Mariana Angulo-Aguado
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Sladjana Zagorac
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Ruth Álvarez-Díaz
- Bioinformatic Unit, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Marta Benítez-Mondéjar
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Fengming Yi
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Carlos Martínez-Garay
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Karla Santos-de-Frutos
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Eunjeong Kim
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
- KNU G-LAMP Research Center, KNU Institute of Basic Sciences, BK21 FOUR KNU Creative BioResearch Group, Department of Biology, College of Natural Sciences, Kyungpook National University, Daegu, South Korea
| | - Ramón Campos-Olivas
- Spectroscopy and Nuclear Magnetic Resonance Unit, Structural Biology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Nabil Djouder
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain.
| |
Collapse
|
4
|
Luo J, Chen K, Nong X. Potential regulation of artesunate on bone metabolism through suppressing inflammatory infiltration in type 2 diabetes mellitus. Immunopharmacol Immunotoxicol 2025; 47:147-158. [PMID: 39762719 DOI: 10.1080/08923973.2024.2444953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 12/15/2024] [Indexed: 03/29/2025]
Abstract
OBJECTIVE Osteoimmunology is an emerging field that explores the interplay between bone and the immune system. The immune system plays a critical role in the pathogenesis of diabetes and significantly affects bone homeostasis. Artesunate, a first-line treatment for malaria, is known for its low toxicity and multifunctional properties. Increasing evidence suggests that artesunate possesses anti-inflammatory, immunoregulatory, and osteogenic effects. This review aims to explore the relationship between immune regulation and bone metabolism in type 2 diabetes (T2DM) and to investigate the potential therapeutic application of artesunate. METHODS This review systematically examines literature from PubMed/Medline, Elsevier, Web of Science, Embase, the International Diabetes Federation, and other relevant databases. RESULTS This review synthesizes evidence from multiple sources to delineate the relationship between T lymphocytes and T2DM, the regulation of T lymphocyte subsets in bone metabolism, and the effects of artesunate on both T lymphocytes and bone metabolism. Recent studies suggest a bidirectional regulatory relationship between T2DM and T lymphocytes (CD4+ T and CD8+ T) during the onset and progression of the disease, with inflammatory and anti-inflammatory cytokines serving as key mediators. T lymphocyte subsets and their cytokines play a pivotal role in regulating osteogenesis and osteoclastogenesis in pathological conditions. Furthermore, artesunate has shown promise in modulating inflammatory infiltration and bone metabolism. CONCLUSION The accumulated evidence indicates that artesunate exerts regulatory effects on bone metabolism in T2DM by influencing T lymphocyte differentiation.
Collapse
Affiliation(s)
- Jinghong Luo
- Department of Oral & Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Kun Chen
- Department of Oral & Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaolin Nong
- Department of Oral & Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
5
|
Sinton MC, Kajimura S. From fat storage to immune hubs: the emerging role of adipocytes in coordinating the immune response to infection. FEBS J 2025; 292:1868-1883. [PMID: 39428707 PMCID: PMC12001177 DOI: 10.1111/febs.17302] [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: 06/14/2024] [Revised: 08/21/2024] [Accepted: 10/04/2024] [Indexed: 10/22/2024]
Abstract
Adipose tissue is a rich source of diverse cell populations, including immune cells, adipocytes and stromal cells. Interactions between these different cell types are now appreciated to be critical for maintaining tissue structure and function, by governing processes such as adipogenesis, lipolysis and differentiation of white to beige adipocytes. Interactions between these cells also drive inflammation in obesity, leading to an expansion of adipose tissue immune cells, and the secretion of proinflammatory cytokines from immune cells and from adipocytes themselves. However, in evolutionary terms, obesity is a recent phenomenon, raising the question of why adipocytes evolved to express factors that influence the immune response. Studies of various pathogens indicate that adipocytes are highly responsive to infection, altering their metabolic profiles in a way that can be used to release nutrients and fuel the immune response. In the case of infection with the extracellular parasite Trypanosoma brucei, attenuating the ability of adipocytes to sense the cytokine IL-17 results in a loss of control of the local immune response and an increased pathogen load. Intriguingly, comparisons of the adipocyte response to infection suggest that the immune responses of these cells occur in a pathogen-dependent manner, further confirming their complexity. Here, with a focus on murine adipose tissue, we discuss the emerging concept that, in addition to their canonical function, adipocytes are immune signalling hubs that integrate and disseminate signals from the immune system to generate a local environment conducive to pathogen clearance.
Collapse
Affiliation(s)
- Matthew C. Sinton
- Division of Immunology, Immunity to Infection and Respiratory MedicineUniversity of ManchesterUK
- Lydia Becker Institute of Immunology and InflammationUniversity of ManchesterUK
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes and MetabolismBeth Israel Deaconess Medical Center and Harvard Medical SchoolBostonMAUSA
- Howard Hughes Medical InstituteChevy ChaseMDUSA
| |
Collapse
|
6
|
Zhang Y, Zhang X, Jin X, Li Z, Li L, Zhu Y, Kong F, Wang D. Structural characterization of polysaccharide isolated from Inonotus hispidus and its anti-obesity effect based on regulation of the interleukin-17-mediated inflammatory response. Int J Biol Macromol 2025; 291:138975. [PMID: 39706397 DOI: 10.1016/j.ijbiomac.2024.138975] [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: 06/25/2024] [Revised: 11/19/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
A heteropolysaccharide (IHP3) with a molecular weight of 22.0 kDa was isolated from Inonotus hispidus (Bull.: Fr.) P. Karst using column chromatography purification from water extraction. Its backbone was predominantly composed of →6)-α-D-Galp-(1→, →2,6)-α-D-Galp-(1→,→6)-α-D-O-Me-Galp-(1→, →3)-α-D-Manp-(1→, and →3,4,6) -β-D-Galp-(1→ residues, branched at C2 of partial α-D-Galp, or C3 and C4 of β-D-Galp, and terminated by α-D-Manp, and α-L-Fucp. In high-fat diet (HFD)-fed obese mice, IHP3 effectively suppressed body weight and plasma glucose gain, decreased fat accumulation, ameliorated lipid metabolism, and protected liver function from HFD-induced damage. Combining the analysis of gut microbiota metabolomics, hepatic proteomics and biochemical detection revealed, IHP3 significantly altered cecum fecal metabolite abundances, inhibited the phosphorylation of peroxisome proliferator-activated receptor gamma, and promoted the browning of white adipose tissue and the activation of brown adipose tissue. These changes collectively contributed to alleviating obesity symptoms by suppressing the interleukin (IL)-17-mediated inflammatory response in obese mice. Therefore, these findings suggest that IHP3 could be a potential candidate for the development of anti-obesity drugs.
Collapse
Affiliation(s)
- Yongfeng Zhang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, 130118, China; School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Xin Zhang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, 130118, China; Shenyang Agricultural University, Shenyang 110866, China.
| | - Xinghui Jin
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Zhige Li
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Lanzhou Li
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, 130118, China.
| | - Yanfeng Zhu
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Fange Kong
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, 130118, China.
| | - Di Wang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun, 130118, China; School of Life Sciences, Jilin University, Changchun 130012, China.
| |
Collapse
|
7
|
Wang X, Tian R, Liang C, Jia Y, Zhao L, Xie Q, Huang F, Yuan H. Biomimetic nanoplatform with microbiome modulation and antioxidant functions ameliorating insulin resistance and pancreatic β-cell dysfunction for T2DM management. Biomaterials 2025; 313:122804. [PMID: 39236631 DOI: 10.1016/j.biomaterials.2024.122804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/29/2024] [Accepted: 09/01/2024] [Indexed: 09/07/2024]
Abstract
Insulin resistance and pancreatic β-cell dysfunction are the main pathogenesis of type 2 diabetes mellitus (T2DM). However, insulin therapy and diabetes medications do not effectively solve the two problems simultaneously. In this study, a biomimetic oral hydrogen nanogenerator that leverages the benefits of edible plant-derived exosomes and hydrogen therapy was constructed to overcome this dilemma by modulating gut microbiota and ameliorating oxidative stress and inflammatory responses. Hollow mesoporous silica (HMS) nanoparticles encapsulating ammonia borane (A) were used to overcome the inefficiency of H2 delivery in traditional hydrogen therapy, and exosomes originating from ginger (GE) were employed to enhance biocompatibility and regulate intestinal flora. Our study showed that HMS/A@GE not only considerably ameliorated insulin resistance and liver steatosis, but inhibited the dedifferentiation of islet β-cell and enhanced pancreatic β-cell proportion in T2DM model mice. In addition to its antioxidant and anti-inflammatory effects, HMS/A@GE augmented the abundance of Lactobacilli spp. and tryptophan metabolites, such as indole and indole acetic acid, which further activated the AhR/IL-22 pathway to improve intestinal-barrier function and metabolic impairments. This study offers a potentially viable strategy for addressing the current limitations of diabetes treatment by integrating gut-microbiota remodelling with antioxidant therapies.
Collapse
Affiliation(s)
- Xiudan Wang
- Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, Henan Provincial People's Hospital, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China; Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, People's Hospital of Zhengzhou University, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China; Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, People's Hospital of Henan University, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China
| | - Rui Tian
- Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, Henan Provincial People's Hospital, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China; Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, People's Hospital of Zhengzhou University, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China; Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, People's Hospital of Henan University, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China
| | - Chenghong Liang
- Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, Henan Provincial People's Hospital, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China; Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, People's Hospital of Zhengzhou University, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China; Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, People's Hospital of Henan University, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China
| | - Yifan Jia
- Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, Henan Provincial People's Hospital, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China; Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, People's Hospital of Zhengzhou University, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China; Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, People's Hospital of Henan University, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China
| | - Lingyun Zhao
- Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, Henan Provincial People's Hospital, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China; Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, People's Hospital of Zhengzhou University, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China; Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, People's Hospital of Henan University, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China
| | - Qinyuan Xie
- Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, Henan Provincial People's Hospital, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China; Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, People's Hospital of Zhengzhou University, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China; Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, People's Hospital of Henan University, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China
| | - Fenglian Huang
- Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, Henan Provincial People's Hospital, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China; Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, People's Hospital of Zhengzhou University, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China; Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, People's Hospital of Henan University, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China
| | - Huijuan Yuan
- Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, Henan Provincial People's Hospital, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China; Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, People's Hospital of Zhengzhou University, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China; Department of Endocrinology, Henan Provincial Key Medicine Laboratory of Intestinal Microecology and Diabetes, People's Hospital of Henan University, Weiwu Road 7, Zhengzhou, 450003, Henan, PR China.
| |
Collapse
|
8
|
Yildiz R, Ganbold K, Sparman NZR, Rajbhandari P. Immune Regulatory Crosstalk in Adipose Tissue Thermogenesis. Compr Physiol 2025; 15:e70001. [PMID: 39921241 DOI: 10.1002/cph4.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/10/2025]
Abstract
Brown adipose tissue (BAT) and thermogenic beige fat within white adipose tissue (WAT), collectively known as adaptive thermogenic fat, dissipate energy as heat, offering promising therapeutic potential to combat obesity and metabolic disorders. The specific biological functions of these fat depots are determined by their unique interaction with the microenvironments, composed of immune cells, endothelial cells, pericytes, and nerve fibers. Immune cells residing in these depots play a key role in regulating energy expenditure and systemic energy homeostasis. The dynamic microenvironment of thermogenic fat depots is essential for maintaining tissue health and function. Immune cells infiltrate both BAT and beige WAT, contributing to their homeostasis and activation through intricate cellular communications. Emerging evidence underscores the importance of various immune cell populations in regulating thermogenic adipose tissue, though many remain undercharacterized. This review provides a comprehensive overview of the immune cells that regulate adaptive thermogenesis and their complex interactions within the adipose niche, highlighting their potential to influence metabolic health and contribute to therapeutic interventions for obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Ramazan Yildiz
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Khatanzul Ganbold
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Njeri Z R Sparman
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Prashant Rajbhandari
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Disease Mechanism and Therapeutics Program, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
9
|
Grazioso TP, del Mar Rigual M, Perna C, Caleiras EJ, Djouder N. Cold exposure reinstates NAD + levels and attenuates hepatocellular carcinoma. Cell Stress 2024; 8:125-139. [PMID: 39781363 PMCID: PMC11708783 DOI: 10.15698/cst2024.12.302] [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: 11/28/2023] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025] Open
Abstract
Cold exposure has been historically used for medicinal purposes, but its benefits and associated mechanisms in mammalian organisms still remain unclear. Here, we explore the chemoprotective properties of cold temperature using a mouse model of hepatocellular carcinoma (HCC) that recapitulates several human features. Chronic cold exposure is shown to prolong lifespan in diseased mice, enhance liver health, and suppress the development of aggressive HCC, preventing hepatocellular hypertrophy, high-grade oval cell hyperplasia, liver steatosis, and aberrant hepatocyte hyperproliferation. Mechanistically, exposure to cold temperatures reinstates NAD+ levels in the HCC mouse models that originally exhibited low NAD+ levels, a contributing process to the development of liver tumors. These findings uncover the role of cold therapy to attenuate HCC development and potentially other existing malignancies involving NAD+ modulation.
Collapse
Affiliation(s)
- Tatiana P Grazioso
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO)Madrid, ES28029Spain
- Instituto de Investigación Sanitaria HM Hospitales (IISHM)MadridSpain
- Laboratory of Innovation in Oncology, Gynecological, Genitourinary and Skin Cancer Unit, HM CIOCC, Centro Integral Oncológico Clara Campal, Hospital Universitario HM Sanchinarro, HM HospitalesMadrid, ES-28050Spain
| | - Maria del Mar Rigual
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO)Madrid, ES28029Spain
| | - Cristian Perna
- Department of Pathology, Hospital Universitario Ramón y Cajal, IRYCISMadrid, ES28034Spain
- Universidad de AlcaláMadrid, ES28801Spain
| | | | - Nabil Djouder
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO)Madrid, ES28029Spain
| |
Collapse
|
10
|
Douglas A, Stevens B, Rendas M, Kane H, Lynch E, Kunkemoeller B, Wessendorf-Rodriguez K, Day EA, Sutton C, Brennan M, O'Brien K, Kohlgruber AC, Prendeville H, Garza AE, O'Neill LAJ, Mills KHG, Metallo CM, Veiga-Fernandes H, Lynch L. Rhythmic IL-17 production by γδ T cells maintains adipose de novo lipogenesis. Nature 2024; 636:206-214. [PMID: 39478228 PMCID: PMC11618085 DOI: 10.1038/s41586-024-08131-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/27/2024] [Indexed: 11/06/2024]
Abstract
The circadian rhythm of the immune system helps to protect against pathogens1-3; however, the role of circadian rhythms in immune homeostasis is less well understood. Innate T cells are tissue-resident lymphocytes with key roles in tissue homeostasis4-7. Here we use single-cell RNA sequencing, a molecular-clock reporter and genetic manipulations to show that innate IL-17-producing T cells-including γδ T cells, invariant natural killer T cells and mucosal-associated invariant T cells-are enriched for molecular-clock genes compared with their IFNγ-producing counterparts. We reveal that IL-17-producing γδ (γδ17) T cells, in particular, rely on the molecular clock to maintain adipose tissue homeostasis, and exhibit a robust circadian rhythm for RORγt and IL-17A across adipose depots, which peaks at night. In mice, loss of the molecular clock in the CD45 compartment (Bmal1∆Vav1) affects the production of IL-17 by adipose γδ17 T cells, but not cytokine production by αβ or IFNγ-producing γδ (γδIFNγ) T cells. Circadian IL-17 is essential for de novo lipogenesis in adipose tissue, and mice with an adipocyte-specific deficiency in IL-17 receptor C (IL-17RC) have defects in de novo lipogenesis. Whole-body metabolic analysis in vivo shows that Il17a-/-Il17f-/- mice (which lack expression of IL-17A and IL-17F) have defects in their circadian rhythm for de novo lipogenesis, which results in disruptions to their whole-body metabolic rhythm and core-body-temperature rhythm. This study identifies a crucial role for IL-17 in whole-body metabolic homeostasis and shows that de novo lipogenesis is a major target of IL-17.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Adipose Tissue/metabolism
- Adipose Tissue/immunology
- Circadian Rhythm/genetics
- Circadian Rhythm/immunology
- Homeostasis
- Interferon-gamma/metabolism
- Interleukin-17/genetics
- Interleukin-17/immunology
- Interleukin-17/metabolism
- Lipogenesis
- Mice, Inbred C57BL
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Single-Cell Gene Expression Analysis
- Biological Clocks/genetics
- Biological Clocks/immunology
- Receptors, Interleukin-17/deficiency
- Receptors, Interleukin-17/metabolism
- Body Temperature
- Leukocyte Common Antigens/metabolism
- ARNTL Transcription Factors/genetics
- ARNTL Transcription Factors/metabolism
Collapse
Affiliation(s)
- Aaron Douglas
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Brenneth Stevens
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Miguel Rendas
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Harry Kane
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Evan Lynch
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | - Emily A Day
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Caroline Sutton
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Martin Brennan
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Katie O'Brien
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | | | - Hannah Prendeville
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Amanda E Garza
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Kingston H G Mills
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Christian M Metallo
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | - Lydia Lynch
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland.
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Molecular Biology, Princeton University, Princeton, NJ, USA.
- Ludwig Cancer Research Institute, Princeton Branch, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
11
|
Reguero M, Reglero G, Quintela JC, Ramos-Ruiz R, Ramírez de Molina A, Gómez de Cedrón M. Silymarin-Enriched Extract from Milk Thistle Activates Thermogenesis in a Preclinical Model of High-Fat-Diet-Induced Obesity to Relieve Systemic Meta-Inflammation. Nutrients 2024; 16:4166. [PMID: 39683558 DOI: 10.3390/nu16234166] [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: 11/03/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Obesity and aging are associated with the progressive loss of brown adipose tissue (BAT), an increase in visceral white adipose tissue (vWAT), and a reduction in subcutaneous white adipose tissue (sWAT). The progressive expansion of visceral obesity promotes a low grade of systemic chronic inflammation (meta-inflammation), contributing to the onset of comorbidities such as type 2 diabetes mellitus (T2DM), metabolic syndrome, and even cancer. Thus, preserving the thermogenic activity of adipose tissue and improving the metabolic flexibility of sWAT could be an effective strategy to prevent the development of metabolic chronic diseases and promote healthy aging. Precision nutrition has emerged as a complementary approach to control the metabolic alterations associated with unhealthy obesity and aging. In a previous work, we described that a silymarin-enriched extract from milk thistle (Mthistle) increased markers of browning and thermogenesis in vitro in human differentiated adipocytes (SGBS). OBJECTIVES/METHODS Therefore, this study aims to evaluate the potential of Mthistle to activate thermogenesis in a preclinical model of high-fat diet (HFD)-induced obesity (DIO). RESULTS Our results demonstrate that Mthistle increases systemic energy expenditure (EE), preserves body temperature after cold exposure, improves insulin resistance, and reduces inflammatory markers in WAT. CONCLUSIONS Based on these results, silymarin-enriched extract from Mthistle may be proposed as a nutraceutical for the management of metabolic chronic diseases and/or accelerated aging.
Collapse
Affiliation(s)
- Marina Reguero
- Molecular Oncology Group, IMDEA Food Institute, CEI UAM + CSIC, E28049 Madrid, Spain
- NATAC BIOTECH, Electronica 7, E28923 Madrid, Spain
| | - Guillermo Reglero
- Production and Characterization of Novel Foods Department, Institute of Food Science Research CIAL, CEI UAM + CSIC, E28049 Madrid, Spain
| | | | - Ricardo Ramos-Ruiz
- Molecular Oncology Group, IMDEA Food Institute, CEI UAM + CSIC, E28049 Madrid, Spain
| | - Ana Ramírez de Molina
- Molecular Oncology Group, IMDEA Food Institute, CEI UAM + CSIC, E28049 Madrid, Spain
| | - Marta Gómez de Cedrón
- Molecular Oncology Group, IMDEA Food Institute, CEI UAM + CSIC, E28049 Madrid, Spain
- Cell Metabolism Unit, IMDEA Food Institute, CEI UAM + CSIC, E28049 Madrid, Spain
| |
Collapse
|
12
|
Ren Y, Huang P, Zhang L, Tang Y, He S, Li H, Huang X, Ding Y, Liu L, Liu L, He X. Multi-omics landscape of childhood simple obesity: novel insights into pathogenesis and biomarkers discovery. Cell Biosci 2024; 14:145. [PMID: 39609876 PMCID: PMC11606102 DOI: 10.1186/s13578-024-01322-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND The increasing incidence of childhood obesity annually has led to a surge in physical and mental health risks, making it a significant global public health concern. This study aimed to discover novel biomarkers of childhood simple obesity through integrative multi-omics analysis, uncovering their potential connections and providing fresh research directions for the complex pathogenesis and treatment strategies. METHODS Transcriptome, untargeted metabolome, and 16 S rDNA sequencing were conducted on subjects to examine transcripts, metabolites in blood, and gut microflora in stool. RESULTS Transcriptomic analysis identified 599 differentially expressed genes (DEGs), of which 25 were immune-related genes, and participated in immune pathways such as antimicrobial peptides, neutrophil degranulation, and interferons. The optimal random forest model based on these genes exhibited an AUC of 0.844. The metabolomic analysis examined 71 differentially expressed metabolites (DEMs), including 12 immune-related metabolites. Notably, lauric acid showed an extremely strong positive correlation with BMI and showed a good discriminative power for obesity (AUC = 0.82). DEMs were found to be significantly enriched in four metabolic pathways, namely "Aminoacyl-tRNA biosynthesis", "Valine leucine and isoleucine biosynthesis, and Glycine", "Serine and threonine metabolism", and "Biosynthesis of unsaturated fatty acids". Microbiome analysis revealed 12 differential gut microbiotas (DGMs) at the phylum and genus levels, with p_Firmicutes dominating in the obese group and g_Escherichia-Shigella in the normal group. Subsequently, a Random Forest model was developed based on the DEMs, immune-related DEGs, and metabolites with an AUC value of 0.912. The 14 indicators identified by this model could potentially serve as a set of biomarkers for obesity. The analysis of the inter-omics correlation network found 233 pairs of significant correlations. DEGs BPIFA1, BPI, and SAA1, DEMs Dimethy(tetradecyl)amine, Deoxycholic acid, Pathalic anhydride, and DL-Alanine, and DGMs g_Intestinimonas and g_Turicibacter showed strong connectivity within the network, constituting a large proportion of interactions. CONCLUSION This study presents the first comprehensive description of the multi-omics characteristics of childhood simple obesity, recognizing promising biomarkers. Immune-related markers offer a new perspective for researching the immunological mechanisms underlying obesity and its associated complications. The revealed interactions among these biomarkers contribute to a deeper understanding the intricate biological regulatory networks associated with obesity.
Collapse
Affiliation(s)
- Yi Ren
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
- Department of Pediatrics, Haikou Hospital of the Maternal and Child Health, Haikou, 570100, China
- Department of Pediatrics, Hainan Modern Women and Children's. Medical, Haikou, 570100, China
| | - Peng Huang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Lu Zhang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Yufen Tang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Siyi He
- Department of Anesthesiology, Hainan General Hospital, Haikou, Hainan, 570311, China
- Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - HaiDan Li
- Department of Pediatrics, Hainan Women and Children's Medical Center, Hainan, 570100, China
| | - XiaoYan Huang
- Department of Pediatrics, Hainan Women and Children's Medical Center, Hainan, 570100, China
| | - Yan Ding
- Department of Dermatology, Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Lingjuan Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Liqun Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| | - Xiaojie He
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Laboratory of Pediatric Nephrology, Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| |
Collapse
|
13
|
Fat keeps metabolism in tune and on time using an inflammatory immune protein. Nature 2024:10.1038/d41586-024-03602-z. [PMID: 39506160 DOI: 10.1038/d41586-024-03602-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
|
14
|
Papaporfyriou A, Bartziokas K, Papachatzopoulou E, Grapatsas K, Kallieri M, Spathis A, Steiropoulos P, Bakakos P, Papiris S, Loukides S, Papaioannou AI. Effects of menopause and fat mass in asthmatic inflammation. J Asthma 2024; 61:1488-1496. [PMID: 38900498 DOI: 10.1080/02770903.2024.2362859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024]
Abstract
INTRODUCTION Female hormones and obesity have an impact on women with asthma. We aimed to describe how these components affect asthma inflammatory processes. METHODS Sex hormones [FSH, LH, estradiol (E2), estrone (E1), testosterone and Δ4 androstenedione (A4)] and serum IL1β, IL13, IL17a, IL-5, IL6, TNF-a were measured from 11 to18 pre- and postmenopausal women with asthma. RESULTS Premenopausal normal weight women revealed higher levels of IL5 and IL17a than obese women on both days of the menstrual cycle (IL5: D1: 6.4 vs 1.4 pg/ml, p = .036 and D14: 3 vs 1.4 pg/ml, p = .045 and IL17a: D1: 13.7 pg/ml vs 10.6 pg/ml and D14: 12.4 pg/ml vs 10.6 pg/ml, p = .009, respectively). In premenopausal women on D1, Δ4 Androstenedione was positively correlated with IL1β (p = .016, r = 0.733), whereas on D14, Estradiol with IL1β (p = .009, r = -.768) and TNF-a with Testosterone (p = .004, r = -0.816), and Δ4 Androstenedione (p = .002, r = -0.841) negatively. In postmenopausal women, TNF-a was negatively associated with FSH (p = .004, r = -0.638), but positively with Testosterone (p = .025, r = 0.526) and IL10 also positively with Estradiol (p = .007, r = 0.610). CONCLUSION Obesity shows a protective role in asthma through the suppression of IL5 and IL17. Estrogens seem to inhibit Th1 and Th2 inflammation, while androgens have a dual role with negative and positive correlations with neutrophilic biomarkers.
Collapse
Affiliation(s)
- Anastasia Papaporfyriou
- Division of Pulmonology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | | | - Eftychia Papachatzopoulou
- 1st Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Alexandra Hospital, Athens, Greece
| | - Kostas Grapatsas
- Department of Thoracic Surgery and Thoracic Endoscopy, Ruhrlandklinik, West German Lung Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Maria Kallieri
- 2nd Respiratory Medicine Department, Attikon University Hospital, Athens, Greece
| | - Aris Spathis
- Department of Pathology, Attikon University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Paschalis Steiropoulos
- Department of Respiratory Medicine, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Petros Bakakos
- 1st University Department of Respiratory Medicine, Sotiria Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Spyros Papiris
- 2nd Respiratory Medicine Department, Attikon University Hospital, Athens, Greece
| | - Stelios Loukides
- 2nd Respiratory Medicine Department, Attikon University Hospital, Athens, Greece
| | - Andriana I Papaioannou
- 1st University Department of Respiratory Medicine, Sotiria Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
15
|
Ononamadu CJ, Seidel V. Exploring the Antidiabetic Potential of Salvia officinalis Using Network Pharmacology, Molecular Docking and ADME/Drug-Likeness Predictions. PLANTS (BASEL, SWITZERLAND) 2024; 13:2892. [PMID: 39458839 PMCID: PMC11510882 DOI: 10.3390/plants13202892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/04/2024] [Accepted: 10/06/2024] [Indexed: 10/28/2024]
Abstract
A combination of network pharmacology, molecular docking and ADME/drug-likeness predictions was employed to explore the potential of Salvia officinalis compounds to interact with key targets involved in the pathogenesis of T2DM. These were predicted using the SwissTargetPrediction, Similarity Ensemble Approach and BindingDB databases. Networks were constructed using the STRING online tool and Cytoscape (v.3.9.1) software. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways analysis and molecular docking were performed using DAVID, SHINEGO 0.77 and MOE suite, respectively. ADME/drug-likeness parameters were computed using SwissADME and Molsoft L.L.C. The top-ranking targets were CTNNB1, JUN, ESR1, RELA, NR3C1, CREB1, PPARG, PTGS2, CYP3A4, MMP9, UGT2B7, CYP2C19, SLCO1B1, AR, CYP19A1, PARP1, CYP1A2, CYP1B1, HSD17B1, and GSK3B. Apigenin, caffeic acid, oleanolic acid, rosmarinic acid, hispidulin, and salvianolic acid B showed the highest degree of connections in the compound-target network. Gene enrichment analysis identified pathways involved in insulin resistance, adherens junctions, metabolic processes, IL-17, TNF-α, cAMP, relaxin, and AGE-RAGE in diabetic complications. Rosmarinic acid, caffeic acid, and salvianolic acid B showed the most promising interactions with PTGS2, DPP4, AMY1A, PTB1B, PPARG, GSK3B and RELA. Overall, this study enhances understanding of the antidiabetic activity of S. officinalis and provides further insights for future drug discovery purposes.
Collapse
Affiliation(s)
- Chimaobi J. Ononamadu
- Natural Products Research Laboratory, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK;
- Natural Product Research Group, Department of Biochemistry and Forensic Science, Nigeria Police Academy, Wudil P.M.B. 3474, Kano, Nigeria
| | - Veronique Seidel
- Natural Products Research Laboratory, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK;
| |
Collapse
|
16
|
Zhong C, Li N, Wang S, Li D, Yang Z, Du L, Huang G, Li H, Yeung WS, He S, Ma S, Wang Z, Jiang H, Zhang H, Li Z, Wen X, Xue S, Tao X, Li H, Xie D, Zhang Y, Chen Z, Wang J, Yan J, Liang Z, Zhang Z, Zhong Z, Wu Z, Wan C, Liang C, Wang L, Yu S, Ma Y, Yu Y, Li F, Chen Y, Zhang B, Lyu A, Ren F, Zhou H, Liu J, Zhang G. Targeting osteoblastic 11β-HSD1 to combat high-fat diet-induced bone loss and obesity. Nat Commun 2024; 15:8588. [PMID: 39362888 PMCID: PMC11449908 DOI: 10.1038/s41467-024-52965-4] [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: 03/16/2024] [Accepted: 09/27/2024] [Indexed: 10/05/2024] Open
Abstract
Excessive glucocorticoid (GC) action is linked to various metabolic disorders. Recent findings suggest that disrupting skeletal GC signaling prevents bone loss and alleviates metabolic disorders in high-fat diet (HFD)-fed obese mice, underpinning the neglected contribution of skeletal GC action to obesity and related bone loss. Here, we show that the elevated expression of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), the enzyme driving local GC activation, and GC signaling in osteoblasts, are associated with bone loss and obesity in HFD-fed male mice. Osteoblast-specific 11β-HSD1 knockout male mice exhibit resistance to HFD-induced bone loss and metabolic disorders. Mechanistically, elevated 11β-HSD1 restrains glucose uptake and osteogenic activity in osteoblast. Pharmacologically inhibiting osteoblastic 11β-HSD1 by using bone-targeted 11β-HSD1 inhibitor markedly promotes bone formation, ameliorates glucose handling and mitigated obesity in HFD-fed male mice. Taken together, our study demonstrates that osteoblastic 11β-HSD1 directly contributes to HFD-induced bone loss, glucose handling impairment and obesity.
Collapse
Affiliation(s)
- Chuanxin Zhong
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Nanxi Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Shengzheng Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Dijie Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangxi Universities Key Laboratory of Stem cell and Biopharmaceutical Technology, College of Life Sciences, Guangxi Normal University, Gui Lin, China
| | - Zhihua Yang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lin Du
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Guangxin Huang
- Department of Joint Surgery, The Third Affiliated Hospital of Southern Medical University, The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Haitian Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Wing Sze Yeung
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Shan He
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Shuting Ma
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Zhuqian Wang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Hewen Jiang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Huarui Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhanghao Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Xiaoxin Wen
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Song Xue
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xiaohui Tao
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Haorui Li
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Duoli Xie
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Yihao Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Zefeng Chen
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Junqin Wang
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jianfeng Yan
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Zhengming Liang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Zongkang Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhigang Zhong
- Sports Medicine Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Zeting Wu
- International Medical Service Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Chao Wan
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Chao Liang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Luyao Wang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Sifan Yu
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yuan Ma
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
| | - Yuanyuan Yu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Fangfei Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Yang Chen
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Baoting Zhang
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Aiping Lyu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China.
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Hong Kong, China.
| | - Fuzeng Ren
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Hong Zhou
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, Australia.
| | - Jin Liu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China.
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China.
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong SAR, China.
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
| |
Collapse
|
17
|
Díaz-Casado ME, González-García P, López-Herrador S, Hidalgo-Gutiérrez A, Jiménez-Sánchez L, Barriocanal-Casado E, Bakkali M, van de Lest CHA, Corral-Sarasa J, Zaal EA, Berkers CR, López LC. Oral β-RA induces metabolic rewiring leading to the rescue of diet-induced obesity. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167283. [PMID: 38851305 DOI: 10.1016/j.bbadis.2024.167283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/23/2024] [Accepted: 05/31/2024] [Indexed: 06/10/2024]
Abstract
Obesity represents a significant health challenge, intricately linked to conditions such as type II diabetes, metabolic syndrome, and hepatic steatosis. Several existing obesity treatments exhibit limited efficacy, undesirable side effects or a limited capability to maintain therapeutics effects in the long-term. Recently, modulation Coenzyme Q (CoQ) metabolism has emerged as a promising target for treatment of metabolic syndrome. This potential intervention could involve the modulation of endogenous CoQ biosynthesis by the use of analogs of the precursor of its biosynthesis, such as β-resorcylic acid (β-RA). Here, we show that oral supplementation with β-RA, incorporated into the diet of diet-induced obese (DIO) mice, leads to substantial weight loss. The anti-obesity effects of β-RA are partially elucidated through the normalization of mitochondrial CoQ metabolism in white adipose tissue (WAT). Additionally, we identify an HFN4α/LXR-dependent transcriptomic activation of the hepatic lipid metabolism that contributes to the anti-obesity effects of β-RA. Consequently, β-RA mitigates WAT hypertrophy, prevents hepatic steatosis, counteracts metabolic abnormalities in WAT and liver, and enhances glucose homeostasis by reducing the insulin/glucagon ratio and plasma levels of gastric inhibitory peptide (GIP). Moreover, pharmacokinetic evaluation of β-RA supports its translational potential. Thus, β-RA emerges as an efficient, safe, and translatable therapeutic option for the treatment and/or prevention of obesity, metabolic dysfunction-associated steatotic liver disease (MASLD).
Collapse
Affiliation(s)
- María Elena Díaz-Casado
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, 18016 Granada, Spain; Instituto de Investigación Biosanitaria Ibs. Granada, 18016 Granada, Spain
| | - Pilar González-García
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, 18016 Granada, Spain; Instituto de Investigación Biosanitaria Ibs. Granada, 18016 Granada, Spain
| | - Sergio López-Herrador
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, 18016 Granada, Spain
| | - Agustín Hidalgo-Gutiérrez
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, 18016 Granada, Spain; Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA
| | | | - Eliana Barriocanal-Casado
- Department of Neurology, Columbia University Medical Center, New York, NY 10032, USA; GENYO, Centre for Genomics and Oncological Research, Genomic Medicine Department, Pfizer-University of Granada-Andalusian Regional Government, 18016 Granada, Spain
| | - Mohammed Bakkali
- Departamento de Genética, Facultad de Ciencias, Universidad de Granada, 18071 Granada, Spain
| | - Chris H A van de Lest
- Division of Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3508 TD Utrecht, the Netherlands
| | | | - Esther A Zaal
- Division of Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3508 TD Utrecht, the Netherlands
| | - Celia R Berkers
- Division of Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3508 TD Utrecht, the Netherlands
| | - Luis C López
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, 18016 Granada, Spain; Instituto de Biotecnología, Centro de Investigación Biomédica, Universidad de Granada, 18016 Granada, Spain; Instituto de Investigación Biosanitaria Ibs. Granada, 18016 Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), 18016 Granada, Spain.
| |
Collapse
|
18
|
Liu J, Chen Y. Cell-cell crosstalk between fat cells and immune cells. Am J Physiol Endocrinol Metab 2024; 327:E371-E383. [PMID: 39082899 DOI: 10.1152/ajpendo.00024.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 09/06/2024]
Abstract
Obesity is a metabolic disorder with pandemic-like implications, lacking viable pharmaceutical treatments currently. Thermogenic adipose tissues, including brown and beige adipose tissues, play an essential role in regulating systemic energy homeostasis and have emerged as appealing therapeutic targets for the treatment of obesity and obesity-related diseases. The function of adipocytes is subject to complex regulation by a cellular network of immune signaling pathways in response to environmental signals. However, the specific regulatory roles of immune cells in thermogenesis and relevant involving mechanisms are still not well understood. Here, we concentrate on our present knowledge of the interaction between thermogenic adipocytes and immune cells and present an overview of cellular and molecular mechanisms underlying immunometabolism in adipose tissues. We discuss cytokines, especially interleukins, which originate from widely variable sources, and their impacts on the development and function of thermogenic adipocytes. Moreover, we summarize the neuroimmune regulation in heat production and expand a new mode of intercellular communication mediated by mitochondrial transfer. The crosstalk between immune cells and adipocytes achieves adipose tissue homeostasis and systemic energy balance. A deep understanding of this intricate interaction would provide evidence for improving thermogenic efficiency by remodeling the immune microenvironment. Interventions based on these factors show a high potential to prevent adverse metabolic outcomes in patients with obesity.
Collapse
Affiliation(s)
- Jiadai Liu
- Department of Endocrinology, Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Laboratory of Endocrinology and Metabolism, Ministry of Education Key Laboratory of Vascular Aging, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yong Chen
- Department of Endocrinology, Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Laboratory of Endocrinology and Metabolism, Ministry of Education Key Laboratory of Vascular Aging, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, People's Republic of China
| |
Collapse
|
19
|
Chen L, Liu L. Adipose thermogenic mechanisms by cold, exercise and intermittent fasting: Similarities, disparities and the application in treatment. Clin Nutr 2024; 43:2043-2056. [PMID: 39088961 DOI: 10.1016/j.clnu.2024.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 08/03/2024]
Abstract
Given its nonnegligible role in metabolic homeostasis, adipose tissue has been the target for treating metabolic disorders such as obesity, diabetes and cardiovascular diseases. Besides its lipolytic function, adipose thermogenesis has gained increased interest due to the irreplaceable contribution to dissipating energy to restore equilibrium, and its therapeutic effects have been testified in various animal models. In this review, we will brief about the canonical cold-stimulated adipose thermogenic mechanisms, elucidate on the exercise- and intermittent fasting-induced adipose thermogenic mechanisms, with a focus on the similarities and disparities among these signaling pathways, in an effort to uncover the overlapped and specific targets that may yield potent therapeutic efficacy synergistically in improving metabolic health.
Collapse
Affiliation(s)
- Linshan Chen
- School of Exercise and Health, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Longhua Liu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, People's Republic of China.
| |
Collapse
|
20
|
Gallo R, Teijeiro A, Angulo-Aguado M, Djouder N. IL-17A produced by POMC neurons regulates diet-induced obesity. iScience 2024; 27:110259. [PMID: 39027371 PMCID: PMC11255842 DOI: 10.1016/j.isci.2024.110259] [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: 04/27/2023] [Revised: 08/29/2023] [Accepted: 06/10/2024] [Indexed: 07/20/2024] Open
Abstract
Overeating leads to obesity, a low-grade inflammatory condition involving interleukin-17A (IL-17A). While pro-opiomelanocortin (POMC) neurons regulate feeding, their connection with IL-17A is not well understood. To impair IL-17A signaling in POMC neurons, IL-17A receptor (Il17ra) was deleted by crossing IL17ra-flox and Pomc-Cre mice. Despite effective deletion, these mice showed no differences in body weight or adiposity compared to control mice, challenging the idea that IL-17A induces obesity through POMC neuron regulation. However, both groups exhibited reduced weight gain and adiposity upon high-fat diet compared to mice carrying only the floxed alleles, suggesting independent effects of Pomc-Cre transgene on body weight. Further analysis reveals that POMC neurons express IL-17A, and reduction in number of POMC neurons in Pomc-Cre mice could be linked to decreased IL-17A expression, which correlates with reduced adipocyte gene expression associated with obesity. Our data underscore an unexpected crosstalk between IL-17A-producing POMC neurons and the endocrine system in obesity regulation.
Collapse
Affiliation(s)
- Rosa Gallo
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, CNIO, 28029 Madrid, Spain
| | - Ana Teijeiro
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, CNIO, 28029 Madrid, Spain
| | - Mariana Angulo-Aguado
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, CNIO, 28029 Madrid, Spain
| | - Nabil Djouder
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, CNIO, 28029 Madrid, Spain
| |
Collapse
|
21
|
Gélineau A, Marcelin G, Ouhachi M, Dussaud S, Voland L, Manuel R, Baba I, Rouault C, Yvan-Charvet L, Clément K, Tussiwand R, Huby T, Gautier EL. Fructooligosaccharides benefits on glucose homeostasis upon high-fat diet feeding require type 2 conventional dendritic cells. Nat Commun 2024; 15:5413. [PMID: 38926424 PMCID: PMC11208547 DOI: 10.1038/s41467-024-49820-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Diet composition impacts metabolic health and is now recognized to shape the immune system, especially in the intestinal tract. Nutritional imbalance and increased caloric intake are induced by high-fat diet (HFD) in which lipids are enriched at the expense of dietary fibers. Such nutritional challenge alters glucose homeostasis as well as intestinal immunity. Here, we observed that short-term HFD induced dysbiosis, glucose intolerance and decreased intestinal RORγt+ CD4 T cells, including peripherally-induced Tregs and IL17-producing (Th17) T cells. However, supplementation of HFD-fed male mice with the fermentable dietary fiber fructooligosaccharides (FOS) was sufficient to maintain RORγt+ CD4 T cell subsets and microbial species known to induce them, alongside having a beneficial impact on glucose tolerance. FOS-mediated normalization of Th17 cells and amelioration of glucose handling required the cDC2 dendritic cell subset in HFD-fed animals, while IL-17 neutralization limited FOS impact on glucose tolerance. Overall, we uncover a pivotal role of cDC2 in the control of the immune and metabolic effects of FOS in the context of HFD feeding.
Collapse
Affiliation(s)
- Adélaïde Gélineau
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Geneviève Marcelin
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Nutrition and Obesities: Systemic approaches research group, NutriOmics, Paris, France
| | - Melissa Ouhachi
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Sébastien Dussaud
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Lise Voland
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Nutrition and Obesities: Systemic approaches research group, NutriOmics, Paris, France
| | - Raoul Manuel
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Ines Baba
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Christine Rouault
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Nutrition and Obesities: Systemic approaches research group, NutriOmics, Paris, France
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale, Inserm, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Karine Clément
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Nutrition and Obesities: Systemic approaches research group, NutriOmics, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital de la Pitié-Salpêtrière, service de Nutrition, Paris, France
| | - Roxane Tussiwand
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Thierry Huby
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Emmanuel L Gautier
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France.
| |
Collapse
|
22
|
Wei Z, Wang J. Exploration of the core pathway of inflammatory bowel disease complicated with metabolic fatty liver and two-sample Mendelian randomization study of the causal relationships behind the disease. Front Immunol 2024; 15:1375654. [PMID: 38698841 PMCID: PMC11063260 DOI: 10.3389/fimmu.2024.1375654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/05/2024] [Indexed: 05/05/2024] Open
Abstract
Background Inflammatory bowel disease (IBD) is often associated with complex extraintestinal manifestations. The incidence of nonalcoholic fatty liver disease (NAFLD) in IBD populations is increasing yearly. However, the mechanism of interaction between NAFLD and IBD is not clear. Consequently, this study aimed to explore the common genetic characteristics of IBD and NAFLD and identify potential therapeutic targets. Materials and methods Gene chip datasets for IBD and NAFLD were obtained from the Gene Expression Omnibus (GEO) database. Weighted gene co-expression network analysis (WGCNA) was performed to identify modules in those datasets related to IBD and NAFLD. ClueGO was used for biological analysis of the shared genes between IBD and NAFLD. Based on the Human MicroRNA Disease Database (HMDD), microRNAs (miRNAs) common to NAFLD and IBD were obtained. Potential target genes for the miRNAs were predicted using the miRTarbase, miRDB, and TargetScan databases. Two-sample Mendelian randomization (MR) and two-way MR were used to explore the causal relationship between Interleukin-17 (IL-17) and the risk of IBD and NAFLD using data from GWAS retrieved from an open database. Results Through WGCNA, gene modules of interest were identified. GO enrichment analysis using ClueGO suggested that the abnormal secretion of chemokines may be a common pathophysiological feature of IBD and NAFLD, and that the IL-17-related pathway may be a common key pathway for the pathological changes that occur in IBD and NAFLD. The core differentially expressed genes (DEGs) in IBD and NAFLD were identified and included COL1A1, LUM, CCL22, CCL2, THBS2, COL1A2, MMP9, and CXCL8. Another cohort was used for validation. Finally, analysis of the miRNAs identified potential therapeutic targets. The MR results suggested that although there was no causal relationship between IBD and NAFLD, there were causal relationships between IL-17 and IBD and NAFLD. Conclusion We established a comorbid model to explain the potential mechanism of IBD with NAFLD and identified the chemokine-related pathway mediated by cytokine IL-17 as the core pathway in IBD with NAFLD, in which miRNA also plays a role and thus provides potential therapeutic targets.
Collapse
Affiliation(s)
| | - Jiangbin Wang
- Department of Digestive, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
23
|
Warger J, Lucas M, Lucas A. Assessing the contribution of plastic-associated obesogenic compounds to cardiometabolic diseases. Curr Opin Endocrinol Diabetes Obes 2024; 31:98-103. [PMID: 38054472 PMCID: PMC10911259 DOI: 10.1097/med.0000000000000852] [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] [Indexed: 12/07/2023]
Abstract
PURPOSE OF REVIEW To present recent evidence that strengthens the concept that exogenous pollutants contribute to adipose dysfunction and increased rates of disease and to highlight the ineffective regulation of this risk as industry switches to related but similarly toxic variants. RECENT FINDINGS Substitutes for common phthalates and the highly regulated bisphenol A (BPA) show similar deleterious effects on adipocytes. The well tolerated limit for BPA exposure has been reduced in Europe to below the level detected in recent population studies. Additionally, the role for BPA-induced inflammation mediated by interleukin 17a has been described in animal and human studies. SUMMARY Despite experimental and associative evidence that supports plastics and plastic associated chemicals deleteriously influencing adipose homeostatasis and contributing to metabolic diseases, structurally related alternate chemicals are being substituted by manufacturers to circumvent trailing regulatory actions.
Collapse
Affiliation(s)
- Jacob Warger
- Medical School, The University of Western Australia
| | - Michaela Lucas
- Medical School, The University of Western Australia
- Department of Immunology PathWest
- Department of Immunology, Sir Charles Gairdner Hospital & Perth Childrens Hospital
| | - Andrew Lucas
- School of Biomedical Sciences, The University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
24
|
Xiao YL, Gong Y, Qi YJ, Shao ZM, Jiang YZ. Effects of dietary intervention on human diseases: molecular mechanisms and therapeutic potential. Signal Transduct Target Ther 2024; 9:59. [PMID: 38462638 PMCID: PMC10925609 DOI: 10.1038/s41392-024-01771-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 02/05/2024] [Accepted: 02/18/2024] [Indexed: 03/12/2024] Open
Abstract
Diet, serving as a vital source of nutrients, exerts a profound influence on human health and disease progression. Recently, dietary interventions have emerged as promising adjunctive treatment strategies not only for cancer but also for neurodegenerative diseases, autoimmune diseases, cardiovascular diseases, and metabolic disorders. These interventions have demonstrated substantial potential in modulating metabolism, disease trajectory, and therapeutic responses. Metabolic reprogramming is a hallmark of malignant progression, and a deeper understanding of this phenomenon in tumors and its effects on immune regulation is a significant challenge that impedes cancer eradication. Dietary intake, as a key environmental factor, can influence tumor metabolism. Emerging evidence indicates that dietary interventions might affect the nutrient availability in tumors, thereby increasing the efficacy of cancer treatments. However, the intricate interplay between dietary interventions and the pathogenesis of cancer and other diseases is complex. Despite encouraging results, the mechanisms underlying diet-based therapeutic strategies remain largely unexplored, often resulting in underutilization in disease management. In this review, we aim to illuminate the potential effects of various dietary interventions, including calorie restriction, fasting-mimicking diet, ketogenic diet, protein restriction diet, high-salt diet, high-fat diet, and high-fiber diet, on cancer and the aforementioned diseases. We explore the multifaceted impacts of these dietary interventions, encompassing their immunomodulatory effects, other biological impacts, and underlying molecular mechanisms. This review offers valuable insights into the potential application of these dietary interventions as adjunctive therapies in disease management.
Collapse
Affiliation(s)
- Yu-Ling Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yue Gong
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ying-Jia Qi
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
25
|
Zhang Y, Zheng L, Cheng D, Lei C, Li H, Zhou J, Zhang C, Song F, Zeng T, Zhao X. Chronic di(2-ethylhexyl) phthalate exposure at environmental-relevant doses induces osteoporosis by disturbing the differentiation of bone marrow mesenchymal stem cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 914:169918. [PMID: 38190899 DOI: 10.1016/j.scitotenv.2024.169918] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/02/2024] [Accepted: 01/02/2024] [Indexed: 01/10/2024]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is a widely used plastic additive with persistent characteristics in the environment. This study was designed to investigate the detrimental effects of chronic DEHP exposure at environmental-relevant doses on bone metabolism and the underlying mechanisms. It was found that exposure to 25 μg/kg bw and 50 μg/kg bw DEHP for 29 weeks led to a reduction of whole-body bone mineral density (BMD), femur microstructure damage, decreased femur new bone formation, and increased femur bone marrow adipogenesis in C57BL/6 female mice, which was not observed in mice exposed to 5000 μg/kg bw DEHP. Further in vitro study showed that DEHP treatment robustly promoted adipogenic differentiation and suppressed osteogenic differentiation of the bone marrow mesenchymal stem cells (BMSCs). Mechanistically, DEHP exposure resulted in elevated expressions of DYRK1B, CDK5, PPARγ, and p-PPARγSer273 in both bone tissue and BMSCs. Interestingly, co-IP analysis showed potential interactions among DYRK1B, PPARγ, and CDK5. Lastly, antagonists of DYRK1B and CDK5 effectively alleviated the BMSCs differentiation disturbance induced by DEHP. These results suggest that DEHP may disturb the BMSCs differentiation by upregulating the PPARγ signaling which may be associated with the activation of DYRK1B and CDK5.
Collapse
Affiliation(s)
- Yifan Zhang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Liugen Zheng
- Hainan Provincial Center for Disease Control and Prevention, Haikou 570203, China
| | - Dong Cheng
- Shandong Center for Disease Control and Prevention, Jinan 250014, China
| | - Changting Lei
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Hui Li
- Shandong Center for Disease Control and Prevention, Jinan 250014, China
| | - Jun Zhou
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Cuili Zhang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Fuyong Song
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Tao Zeng
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| | - Xiulan Zhao
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
26
|
Liu Y, Ouyang Y, You W, Liu W, Cheng Y, Mai X, Shen Z. Physiological roles of human interleukin-17 family. Exp Dermatol 2024; 33:e14964. [PMID: 37905720 DOI: 10.1111/exd.14964] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/08/2023] [Accepted: 10/13/2023] [Indexed: 11/02/2023]
Abstract
Interleukin-17 s (IL-17s) are well-known proinflammatory cytokines, and their antagonists perform excellently in the treatment of inflammatory skin diseases such as psoriasis. However, their physiological functions have not been given sufficient attention by clinicians. IL-17s can protect the host from extracellular pathogens, maintain epithelial integrity, regulate cognitive processes and modulate adipocyte activity through distinct mechanisms. Here, we present a systematic review concerning the physiological functions of IL-17s. Our goal is not to negate the therapeutic effect of IL-17 antagonists, but to ensure their safe use and reasonably explain the possible adverse events that may occur in their application.
Collapse
Affiliation(s)
- Yucong Liu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Ye Ouyang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Wanchun You
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Wenqi Liu
- Department of Dermatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yufan Cheng
- Department of Dermatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xinming Mai
- Medical School, Shenzhen University, Shenzhen, China
| | - Zhu Shen
- Department of Dermatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
27
|
Sinton MC, Chandrasegaran PRG, Capewell P, Cooper A, Girard A, Ogunsola J, Perona-Wright G, M Ngoyi D, Kuispond N, Bucheton B, Camara M, Kajimura S, Bénézech C, Mabbott NA, MacLeod A, Quintana JF. IL-17 signalling is critical for controlling subcutaneous adipose tissue dynamics and parasite burden during chronic murine Trypanosoma brucei infection. Nat Commun 2023; 14:7070. [PMID: 37923768 PMCID: PMC10624677 DOI: 10.1038/s41467-023-42918-8] [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: 05/26/2023] [Accepted: 10/25/2023] [Indexed: 11/06/2023] Open
Abstract
In the skin, Trypanosoma brucei colonises the subcutaneous white adipose tissue, and is proposed to be competent for forward transmission. The interaction between parasites, adipose tissue, and the local immune system is likely to drive the adipose tissue wasting and weight loss observed in cattle and humans infected with T. brucei. However, mechanistically, events leading to subcutaneous white adipose tissue wasting are not fully understood. Here, using several complementary approaches, including mass cytometry by time of flight, bulk and single cell transcriptomics, and in vivo genetic models, we show that T. brucei infection drives local expansion of several IL-17A-producing cells in the murine WAT, including TH17 and Vγ6+ cells. We also show that global IL-17 deficiency, or deletion of the adipocyte IL-17 receptor protect from infection-induced WAT wasting and weight loss. Unexpectedly, we find that abrogation of adipocyte IL-17 signalling results in a significant accumulation of Dpp4+ Pi16+ interstitial preadipocytes and increased extravascular parasites in the WAT, highlighting a critical role for IL-17 signalling in controlling preadipocyte fate, subcutaneous WAT dynamics, and local parasite burden. Taken together, our study highlights the central role of adipocyte IL-17 signalling in controlling WAT responses to infection, suggesting that adipocytes are critical coordinators of tissue dynamics and immune responses to T. brucei infection.
Collapse
Affiliation(s)
- Matthew C Sinton
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK.
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK.
- Division of Cardiovascular Science, University of Manchester, Manchester, UK.
| | - Praveena R G Chandrasegaran
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Paul Capewell
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Anneli Cooper
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Alex Girard
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - John Ogunsola
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Georgia Perona-Wright
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Dieudonné M Ngoyi
- Department of Parasitology, National Institute of Biomedical Research, Kinshasa, Democratic Republic of Congo
- Member of TrypanoGEN, Kinshasa, Democratic Republic of Congo
| | - Nono Kuispond
- Department of Parasitology, National Institute of Biomedical Research, Kinshasa, Democratic Republic of Congo
- Member of TrypanoGEN, Kinshasa, Democratic Republic of Congo
| | - Bruno Bucheton
- Member of TrypanoGEN, Kinshasa, Democratic Republic of Congo
- Institut de Recherche pour le Développement, Unité Mixte de Recherche IRD-CIRAD 177, Campus International de Baillarguet, Montpellier, France
| | - Mamadou Camara
- Member of TrypanoGEN, Kinshasa, Democratic Republic of Congo
- Programme National de Lutte contre la Trypanosomiase Humaine Africaine, Ministère de la Santé, Conakry, Guinea
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Cécile Bénézech
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4TJ, Scotland, UK
| | - Neil A Mabbott
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Annette MacLeod
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Member of TrypanoGEN, Kinshasa, Democratic Republic of Congo
| | - Juan F Quintana
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK.
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK.
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK.
- Division of Immunology, Immunity to Infection and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.
| |
Collapse
|
28
|
Kang GS, Jo HJ, Lee YR, Oh T, Park HJ, Ahn GO. Sensing the oxygen and temperature in the adipose tissues - who's sensing what? Exp Mol Med 2023; 55:2300-2307. [PMID: 37907745 PMCID: PMC10689767 DOI: 10.1038/s12276-023-01113-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 11/02/2023] Open
Abstract
Adipose tissues, composed of various cell types, including adipocytes, endothelial cells, neurons, and immune cells, are organs that are exposed to dynamic environmental challenges. During diet-induced obesity, white adipose tissues experience hypoxia due to adipocyte hypertrophy and dysfunctional vasculature. Under these conditions, cells in white adipose tissues activate hypoxia-inducible factor (HIF), a transcription factor that activates signaling pathways involved in metabolism, angiogenesis, and survival/apoptosis to adapt to such an environment. Exposure to cold or activation of the β-adrenergic receptor (through catecholamines or chemicals) leads to heat generation, mainly in brown adipose tissues through activating uncoupling protein 1 (UCP1), a proton uncoupler in the inner membrane of the mitochondria. White adipose tissues can undergo a similar process under this condition, a phenomenon known as 'browning' of white adipose tissues or 'beige adipocytes'. While UCP1 expression has largely been confined to adipocytes, HIF can be expressed in many types of cells. To dissect the role of HIF in specific types of cells during diet-induced obesity, researchers have generated tissue-specific knockout (KO) mice targeting HIF pathways, and many studies have commonly revealed that intact HIF-1 signaling in adipocytes and adipose tissue macrophages exacerbates tissue inflammation and insulin resistance. In this review, we highlight some of the key findings obtained from these transgenic mice, including Ucp1 KO mice and other models targeting the HIF pathway in adipocytes, macrophages, or endothelial cells, to decipher their roles in diet-induced obesity.
Collapse
Affiliation(s)
- Gi-Sue Kang
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Hye-Ju Jo
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Ye-Rim Lee
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Taerim Oh
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Hye-Joon Park
- College of Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - G-One Ahn
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea.
- College of Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea.
| |
Collapse
|
29
|
Jena J, García-Peña LM, Weatherford ET, Marti A, Bjorkman SH, Kato K, Koneru J, Chen JH, Seeley RJ, Abel ED, Pereira RO. GDF15 is required for cold-induced thermogenesis and contributes to improved systemic metabolic health following loss of OPA1 in brown adipocytes. eLife 2023; 12:e86452. [PMID: 37819027 PMCID: PMC10567111 DOI: 10.7554/elife.86452] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 09/15/2023] [Indexed: 10/13/2023] Open
Abstract
We previously reported that mice lacking the protein optic atrophy 1 (OPA1 BKO) in brown adipose tissue (BAT) display induction of the activating transcription factor 4 (ATF4), which promotes fibroblast growth factor 21 (FGF21) secretion as a batokine. FGF21 increases metabolic rates under baseline conditions but is dispensable for the resistance to diet-induced obesity (DIO) reported in OPA1 BKO mice (Pereira et al., 2021). To determine alternative mediators of this phenotype, we performed transcriptome analysis, which revealed increased levels of growth differentiation factor 15 (GDF15), along with increased protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) levels in BAT. To investigate whether ATF4 induction was mediated by PERK and evaluate the contribution of GDF15 to the resistance to DIO, we selectively deleted PERK or GDF15 in OPA1 BKO mice. Mice with reduced OPA1 and PERK levels in BAT had preserved ISR activation. Importantly, simultaneous deletion of OPA1 and GDF15 partially reversed the resistance to DIO and abrogated the improvements in glucose tolerance. Furthermore, GDF15 was required to improve cold-induced thermogenesis in OPA1 BKO mice. Taken together, our data indicate that PERK is dispensable to induce the ISR, but GDF15 contributes to the resistance to DIO, and is required for glucose homeostasis and thermoregulation in OPA1 BKO mice by increasing energy expenditure.
Collapse
Affiliation(s)
- Jayashree Jena
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of IowaIowa CityUnited States
| | - Luis Miguel García-Peña
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of IowaIowa CityUnited States
| | - Eric T Weatherford
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of IowaIowa CityUnited States
| | - Alex Marti
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of IowaIowa CityUnited States
| | - Sarah H Bjorkman
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of IowaIowa CityUnited States
| | - Kevin Kato
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of IowaIowa CityUnited States
| | - Jivan Koneru
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of IowaIowa CityUnited States
| | - Jason H Chen
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of IowaIowa CityUnited States
| | - Randy J Seeley
- Department of Internal Medicine, University of MichiganAnn ArborUnited States
| | - E Dale Abel
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of IowaIowa CityUnited States
| | - Renata O Pereira
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of IowaIowa CityUnited States
| |
Collapse
|
30
|
Pestel J, Blangero F, Watson J, Pirola L, Eljaafari A. Adipokines in obesity and metabolic-related-diseases. Biochimie 2023; 212:48-59. [PMID: 37068579 DOI: 10.1016/j.biochi.2023.04.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/03/2023] [Accepted: 04/13/2023] [Indexed: 04/19/2023]
Abstract
The discovery of leptin in the 1990s led to a reconsideration of adipose tissue (AT) as not only a fatty acid storage organ, but also a proper endocrine tissue. AT is indeed capable of secreting bioactive molecules called adipokines for white AT or batokines for brown/beige AT, which allow communication with numerous organs, especially brain, heart, liver, pancreas, and/or the vascular system. Adipokines exert pro or anti-inflammatory activities. An equilibrated balance between these two sets ensures homeostasis of numerous tissues and organs. During the development of obesity, AT remodelling leads to an alteration of its endocrine activity, with increased secretion of pro-inflammatory adipokines relative to the anti-inflammatory ones, as shown in the graphical abstract. Pro-inflammatory adipokines take part in the initiation of local and systemic inflammation during obesity and contribute to comorbidities associated to obesity, as detailed in the present review.
Collapse
Affiliation(s)
- Julien Pestel
- INSERM U1060-CarMeN /Université Claude Bernard Lyon 1/INRAE/ Université Claude Bernard Lyon 1: Laboratoire CarMeN, 165 chemin du Grand Revoyet, CHLS, 69310 Pierre Bénite, France
| | - Ferdinand Blangero
- INSERM U1060-CarMeN /Université Claude Bernard Lyon 1/INRAE/ Université Claude Bernard Lyon 1: Laboratoire CarMeN, 165 chemin du Grand Revoyet, CHLS, 69310 Pierre Bénite, France
| | - Julia Watson
- INSERM U1060-CarMeN /Université Claude Bernard Lyon 1/INRAE/ Université Claude Bernard Lyon 1: Laboratoire CarMeN, 165 chemin du Grand Revoyet, CHLS, 69310 Pierre Bénite, France
| | - Luciano Pirola
- INSERM U1060-CarMeN /Université Claude Bernard Lyon 1/INRAE/ Université Claude Bernard Lyon 1: Laboratoire CarMeN, 165 chemin du Grand Revoyet, CHLS, 69310 Pierre Bénite, France
| | - Assia Eljaafari
- INSERM U1060-CarMeN /Université Claude Bernard Lyon 1/INRAE/ Université Claude Bernard Lyon 1: Laboratoire CarMeN, 165 chemin du Grand Revoyet, CHLS, 69310 Pierre Bénite, France; Hospices Civils de Lyon: 2 quai des Célestins, 69001 Lyon, France.
| |
Collapse
|
31
|
Yan L, Yu C, Zhao Z, Zhang Y, Wang R, Li C. Higher IL-9 Level is Associated with Psoriasis Vulgaris Complicated by Metabolic Syndrome. Clin Cosmet Investig Dermatol 2023; 16:2297-2307. [PMID: 37641663 PMCID: PMC10460594 DOI: 10.2147/ccid.s422355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/15/2023] [Indexed: 08/31/2023]
Abstract
Purpose The underlying pathophysiology linking psoriasis vulgaris (PV) and metabolic syndrome (MetS) is not fully understood. The present study aimed to investigate the serum level of interleukin (IL)-9 and tissue levels of IL-9 and its receptor in PV patients with MetS and analyze the correlation of IL-9 levels with psoriasis disease severity and MetS. Methods This study enrolled 75 PV patients with MetS, 57 PV patients without MetS, 20 healthy blood donors, and 7 healthy skin donors. Clinical, socio-demographic, and anthropometric data were obtained from all individuals. Fasting blood glucose, insulin, lipid profile levels, and serum levels of IL-9 and IL-17A were measured. The expression of IL-9 and its receptor in skin specimens in PV patients and healthy controls was determined using immunohistochemistry. Normal human epidermal keratinocytes were stimulated with five pro-inflammatory cytokines (tumor necrosis factor-α, oncostatin M, IL-22, IL-17A, and IL-1α) to establish a psoriatic keratinocyte model and subsequently treated with IL-9. Their mRNA levels of antimicrobial peptides and chemokines were measured using quantitative real-time polymerase chain reaction. Results Serum level of IL-9 and tissue levels of IL-9 and its receptor were upregulated in PV patients with MetS. IL-9 level was positively correlated to IL-17A level; however, no significant correlation of IL-9 level with psoriasis area severity index was observed. IL-9 level had a positive correlation with the presence of MetS and its components. Correspondingly, IL-9 level positively correlated with waist circumference, body mass index, homeostasis model assessment-insulin resistance, blood pressure, and triglyceride level and negatively correlated with high-density lipoprotein cholesterol level. Additionally, IL-9 stimulated the expression of antimicrobial peptides and chemokines in a psoriatic keratinocyte model. Conclusion Our findings confirmed that higher IL-9 level is associated with PV complicated by MetS, suggesting that IL-9 may be a link between PV and MetS.
Collapse
Affiliation(s)
- Liang Yan
- Department of Dermatology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
- Department of Dermatology, General Hospital of Central Theater Command of PLA, Wuhan, Hubei, 430070, People’s Republic of China
| | - Chongli Yu
- Department of Dermatology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
| | - Zhenkai Zhao
- Department of Dermatology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
| | - Yuan Zhang
- Department of Dermatology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
| | - Rui Wang
- Department of Dermatology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
| | - Chengxin Li
- Department of Dermatology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
| |
Collapse
|
32
|
Douglas A, Stevens B, Lynch L. Interleukin-17 as a key player in neuroimmunometabolism. Nat Metab 2023; 5:1088-1100. [PMID: 37488456 PMCID: PMC10440016 DOI: 10.1038/s42255-023-00846-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/14/2023] [Indexed: 07/26/2023]
Abstract
In mammals, interleukin (IL)-17 cytokines are produced by innate and adaptive lymphocytes. However, the IL-17 family has widespread expression throughout evolution, dating as far back as cnidaria, molluscs and worms, which predate lymphocytes. The evolutionary conservation of IL-17 suggests that it is involved in innate defence strategies, but also that this cytokine family has a fundamental role beyond typical host defence. Throughout evolution, IL-17 seems to have a major function in homeostatic maintenance at barrier sites. Most recently, a pivotal role has been identified for IL-17 in regulating cellular metabolism, neuroimmunology and tissue physiology, particularly in adipose tissue. Here we review the emerging role of IL-17 signalling in regulating metabolic processes, which may shine a light on the evolutionary role of IL-17 beyond typical immune responses. We propose that IL-17 helps to coordinate the cross-talk among the nervous, endocrine and immune systems for whole-body energy homeostasis as a key player in neuroimmunometabolism.
Collapse
Affiliation(s)
- Aaron Douglas
- School of Biochemistry and Immunology, TBSI, Trinity College Dublin, Dublin, Ireland
| | - Brenneth Stevens
- School of Biochemistry and Immunology, TBSI, Trinity College Dublin, Dublin, Ireland
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lydia Lynch
- School of Biochemistry and Immunology, TBSI, Trinity College Dublin, Dublin, Ireland.
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
33
|
Li JH, Hepworth MR, O'Sullivan TE. Regulation of systemic metabolism by tissue-resident immune cell circuits. Immunity 2023; 56:1168-1186. [PMID: 37315533 PMCID: PMC10321269 DOI: 10.1016/j.immuni.2023.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/11/2023] [Accepted: 05/02/2023] [Indexed: 06/16/2023]
Abstract
Recent studies have demonstrated that tissue homeostasis and metabolic function are dependent on distinct tissue-resident immune cells that form functional cell circuits with structural cells. Within these cell circuits, immune cells integrate cues from dietary contents and commensal microbes in addition to endocrine and neuronal signals present in the tissue microenvironment to regulate structural cell metabolism. These tissue-resident immune circuits can become dysregulated during inflammation and dietary overnutrition, contributing to metabolic diseases. Here, we review the evidence describing key cellular networks within and between the liver, gastrointestinal tract, and adipose tissue that control systemic metabolism and how these cell circuits become dysregulated during certain metabolic diseases. We also identify open questions in the field that have the potential to enhance our understanding of metabolic health and disease.
Collapse
Affiliation(s)
- Joey H Li
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 900953, USA; Medical Scientist Training Program, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Matthew R Hepworth
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, Manchester Collaborative Centre for Inflammation Research, Lydia Becker Institute of Immunology and Inflammation, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Timothy E O'Sullivan
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 900953, USA.
| |
Collapse
|
34
|
Gourronc FA, Chimenti MS, Lehmler HJ, Ankrum JA, Klingelhutz AJ. Hydroxylation markedly alters how the polychlorinated biphenyl (PCB) congener, PCB52, affects gene expression in human preadipocytes. Toxicol In Vitro 2023; 89:105568. [PMID: 36804509 PMCID: PMC10081964 DOI: 10.1016/j.tiv.2023.105568] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 12/23/2022] [Accepted: 02/07/2023] [Indexed: 02/17/2023]
Abstract
Polychlorinated biphenyls (PCBs) accumulate in adipose tissue and are linked to obesity and diabetes. The congener, PCB52 (2,2',5,5'-tetrachorobiphenyl), is found at high levels in school air. Hydroxylation of PCB52 to 4-OH-PCB52 (4-hydroxy-2,2',5,5'-tetrachorobiphenyl) may increase its toxicity. To understand PCB52's role in causing adipose dysfunction, we exposed human preadipocytes to PCB52 or 4-OH-PCB52 across a time course and assessed transcript changes using RNAseq. 4-OH-PCB52 caused considerably more changes in the number of differentially expressed genes as compared to PCB52. Both PCB52 and 4-OH-PCB52 upregulated transcript levels of the sulfotransferase SULT1E1 at early time points, but cytochrome P450 genes were generally not affected. A set of genes known to be transcriptionally regulated by PPARα were consistently downregulated by PCB52 at all time points. In contrast, 4-OH-PCB52 affected a variety of pathways, including those involving cytokine responses, hormone responses, focal adhesion, Hippo, and Wnt signaling. Sets of genes known to be transcriptionally regulated by IL17A or parathyroid hormone (PTH) were found to be consistently downregulated by 4-OH-PCB52. Most of the genes affected by PCB52 and 4-OH-PCB52 were different and, of those that were the same, many were changed in an opposite direction. These studies provide insight into how PCB52 or its metabolites may cause adipose dysfunction to cause disease.
Collapse
Affiliation(s)
| | - Michael S Chimenti
- Iowa Institute of Human Genetics, Bioinformatics Division, University of Iowa, United States
| | - Hans-Joachim Lehmler
- Department of Occupational and Environmental Health, University of Iowa, United States
| | - James A Ankrum
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, United States; Fraternal Order of Eagles Diabetes Research Center, University of Iowa, United States
| | - Aloysius J Klingelhutz
- Department of Microbiology and Immunology, University of Iowa, United States; Fraternal Order of Eagles Diabetes Research Center, University of Iowa, United States.
| |
Collapse
|
35
|
González-Casanova JE, Bermúdez V, Caro Fuentes NJ, Angarita LC, Caicedo NH, Rivas Muñoz J, Rojas-Gómez DM. New Evidence on BPA's Role in Adipose Tissue Development of Proinflammatory Processes and Its Relationship with Obesity. Int J Mol Sci 2023; 24:ijms24098231. [PMID: 37175934 PMCID: PMC10179730 DOI: 10.3390/ijms24098231] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/19/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Bisphenol A (BPA) is a xenobiotic with endocrine disruptor properties which interacts with various receptors, eliciting a cellular response. In the plastic industry, BPA is widely used in the production of polycarbonate and epoxy-phenolic resins to provide elastic properties. It can be found in the lining of canned foods, certain plastic containers, thermal printing papers, composite dental fillings, and medical devices, among other things. Therefore, it is a compound that, directly or indirectly, is in daily contact with the human organism. BPA is postulated to be a factor responsible for the global epidemic of obesity and non-communicable chronic diseases, belonging to the obesogenic and diabetogenic group of compounds. Hence, this endocrine disruptor may be responsible for the development of metabolic disorders, promoting in fat cells an increase in proinflammatory pathways and upregulating the expression and release of certain cytokines, such as IL6, IL1β, and TNFα. These, in turn, at a systemic and local level, are associated with a chronic low-grade inflammatory state, which allows the perpetuation of the typical physiological complications of obesity.
Collapse
Affiliation(s)
| | - Valmore Bermúdez
- Centro de Investigaciones en Ciencias de la Vida, Universidad Simón Bolívar, Barranquilla 080002, Colombia
| | - Nelson Javier Caro Fuentes
- Centro de Investigación Austral Biotech, Facultad de Ciencias, Universidad Santo Tomás, Avda. Ejército 146, Santiago 8320000, Chile
| | - Lissé Chiquinquirá Angarita
- Escuela de Nutrición y Dietética, Facultad de Medicina, Universidad Andres Bello, Sede Concepción, Talcahuano 4260000, Chile
| | - Nelson Hernando Caicedo
- Departamento de Ciencias Biológicas, Bioprocesos y Biotecnología, Facultad de Ingeniería, Diseño y Ciencias Aplicadas, Universidad Icesi, Calle 18 No. 122-135 Pance, Cali 760031, Colombia
| | - Jocelyn Rivas Muñoz
- Escuela de Nutrición y Dietética, Facultad de Medicina, Universidad Andres Bello, Santiago 8370321, Chile
| | - Diana Marcela Rojas-Gómez
- Escuela de Nutrición y Dietética, Facultad de Medicina, Universidad Andres Bello, Santiago 8370321, Chile
| |
Collapse
|
36
|
Dashti F, Jamshed F, Ouyang X, Mehal WZ, Banini BA. Digoxin as an emerging therapy in noncardiac diseases. Trends Pharmacol Sci 2023; 44:199-203. [PMID: 36396496 DOI: 10.1016/j.tips.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/12/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022]
Abstract
The cardiac glycoside (CG) digoxin is a generic drug approved for the treatment of heart failure and supraventricular arrhythmias. Over the past few decades, substantial strides have been made toward repurposing digoxin to treat various noncardiac diseases. Here, we evaluate recent insights into basic and clinical work related to noncardiac use of digoxin.
Collapse
Affiliation(s)
- Farzaneh Dashti
- Section of Digestive Diseases, Yale School of Medicine, PO Box 208019, New Haven, CT 06520, USA
| | - Fatima Jamshed
- Section of Digestive Diseases, Yale School of Medicine, PO Box 208019, New Haven, CT 06520, USA; Griffin Hospital-Yale University, 130 Division Street, Derby, CT 06418, USA
| | - Xinshou Ouyang
- Section of Digestive Diseases, Yale School of Medicine, PO Box 208019, New Haven, CT 06520, USA
| | - Wajahat Z Mehal
- Section of Digestive Diseases, Yale School of Medicine, PO Box 208019, New Haven, CT 06520, USA; West Haven Veterans Medical Center, West Haven, CT 06516, USA
| | - Bubu A Banini
- Section of Digestive Diseases, Yale School of Medicine, PO Box 208019, New Haven, CT 06520, USA.
| |
Collapse
|
37
|
Jamshed F, Dashti F, Ouyang X, Mehal WZ, Banini BA. New uses for an old remedy: Digoxin as a potential treatment for steatohepatitis and other disorders. World J Gastroenterol 2023; 29:1824-1837. [PMID: 37032732 PMCID: PMC10080697 DOI: 10.3748/wjg.v29.i12.1824] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/12/2023] [Accepted: 03/14/2023] [Indexed: 03/28/2023] Open
Abstract
Repurposing of the widely available and relatively cheap generic cardiac gly-coside digoxin for non-cardiac indications could have a wide-ranging impact on the global burden of several diseases. Over the past several years, there have been significant advances in the study of digoxin pharmacology and its potential non-cardiac clinical applications, including anti-inflammatory, antineoplastic, metabolic, and antimicrobial use. Digoxin holds promise in the treatment of gastrointestinal disease, including nonalcoholic steatohepatitis and alcohol-associated steatohepatitis as well as in obesity, cancer, and treatment of viral infections, among other conditions. In this review, we provide a summary of the clinical uses of digoxin to date and discuss recent research on its emerging applications.
Collapse
Affiliation(s)
- Fatima Jamshed
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT 06510, United States
- Griffin Hospital-Yale University, Derby, CT 06418, United States
| | - Farzaneh Dashti
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT 06510, United States
| | - Xinshou Ouyang
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT 06510, United States
| | - Wajahat Z Mehal
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT 06510, United States
- West Haven Veterans Medical Center, West Haven, CT 06516, United States
| | - Bubu A Banini
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT 06510, United States
| |
Collapse
|
38
|
Zhou Y, Duan S, Wang R, Chen J, Yao S. Nonlinear correlation between fatty liver index and carotid intima media thickness among individuals undergoing health examination. Front Endocrinol (Lausanne) 2023; 14:1120581. [PMID: 37056670 PMCID: PMC10086365 DOI: 10.3389/fendo.2023.1120581] [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: 12/10/2022] [Accepted: 03/01/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Fatty liver index (FLI) is a predictor of non-alcohol fatty liver disease (NAFLD). This study aimed to assess the association between FLI and carotid intima media thickness (CIMT). METHODS In this cross-sectional study, we enrolled 277 individuals for health examination from the China-Japan Friendship Hospital. Blood sampling and ultrasound examinations were conducted. Multivariate logistic regression and restricted cubic spline analyses were performed to evaluate the association between FLI and CIMT. RESULTS Overall, 175 (63.2%) and 105 (37.9%) individuals had NAFLD and CIMT, respectively. The multivariate logistic regression analyses results showed that high FLI was independently associated with a high risk of increased CIMT, T2 vs. T1 (odds ratio [OR], 95% confidence interval [CI]): 2.41, 1.10-5.25, p = 0.027; T3 vs. T1 (OR, 95% CI): 1.58, 0.68-3.64, p = 0.285. The association between FLI and increased CIMT exhibited a J-shaped curve (nonlinear, p = 0.019). In the threshold analysis, the OR for developing increased CIMT was 1.031 (95% CI: 1.011-1.051, p = 0.0023) in participants with FLI < 64.247. CONCLUSION The relationship between FLI and increased CIMT in the health examination population is J-shaped, with an inflection point of 64.247.
Collapse
Affiliation(s)
- Yuanchen Zhou
- Peking University China-Japan Friendship School of Clinical Medicine, Peking University, Beijing, China
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, China
| | - Shaojie Duan
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Rongrui Wang
- Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Jialiang Chen
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Shukun Yao
- Peking University China-Japan Friendship School of Clinical Medicine, Peking University, Beijing, China
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
39
|
Hu Q, Zhang Y, Liu M, Wang B, Wang X, Wang T. Neonatal cytokines associated with infant overweight and obesity at 1 year of age. Obes Res Clin Pract 2023; 17:102-107. [PMID: 36935337 DOI: 10.1016/j.orcp.2023.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 02/14/2023] [Accepted: 02/23/2023] [Indexed: 03/19/2023]
Abstract
OBJECTIVE The incidence of childhood overweight and obesity has been increasing in recent years. Immune dysregulation has been demonstrated as a condition related to childhood obesity. Whether the neonatal immune status is related to infant overweight and obesity at 1 year of age is unclear. METHODS To explore the relationship between neonatal cytokines and infant overweight and obesity, we conducted a prospective study in Suzhou Municipal Hospital Affiliated to Nanjing Medical University from 2015 to 2016. 514 neonates were recruited and their dried blood spots were collected after birth. Infants were grouped into normal size groups and overweight and obesity groups based on BMI at 1 year of age. 27 neonatal cytokines levels were compared between the two groups. RESULTS 370 infants were included in final analysis. Granulocyte colony stimulating factor (GCSF), interleukin-17A (IL17A) and platelet derived growth factor-BB (PDGF-BB) levels were independently associated with childhood overweight and obesity (OR =1.27, 95%CI 1.03, 1.57; OR =1.29, 95%CI: 1.06, 1.60; OR =0.69, 95%CI: 0.49, 0.96). Additionally, neonatal GCSF and IL17A levels were positively associated with increased BMI (β = 0.11, 95%CI: 0.02, 0.19; β = 0.07, 95%CI 0.01, 013) and BMI z-scores (β = 0.10, 95%CI: 0.02, 0.18; β = 0.06, 95%CI 0.01, 0.13). Neonatal PDGF-BB levels were negatively associated with BMI (β = -0.12, 95%CI: -0.23, -0.01) and BMI z-scores (β = -0.12, 95%CI: -0.23, -0.01). The inverse probability weighting (IPW) was performed to account for potential selection bias of this study, and the results were consistent with the above mentioned findings. CONCLUSIONS Neonatal GCSF, IL17A and PDGF-BB levels were correlated with infant overweight and obesity at 1 year of age, suggesting that early life immune status play a significant role of late obesity.
Collapse
Affiliation(s)
- Qi Hu
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Beilun District Center for Disease Control and Prevention, Ningbo 315899, China
| | - Yuqing Zhang
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Minjuan Liu
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215000, China
| | - Benjing Wang
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215000, China
| | - Xu Wang
- Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Ting Wang
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215000, China.
| |
Collapse
|
40
|
Oates EH, Antoniewicz MR. 13C-Metabolic flux analysis of 3T3-L1 adipocytes illuminates its core metabolism under hypoxia. Metab Eng 2023; 76:158-166. [PMID: 36758664 DOI: 10.1016/j.ymben.2023.02.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 01/20/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023]
Abstract
Hypoxia has been identified as a major factor in the pathogenesis of adipose tissue inflammation, which is a hallmark of obesity and obesity-linked type 2 diabetes mellitus. In this study, we have investigated the impact of hypoxia (1% oxygen) on the physiology and metabolism of 3T3-L1 adipocytes, a widely used cell culture model of adipose. Specifically, we applied parallel labeling experiments, isotopomer spectral analysis, and 13C-metabolic flux analysis to quantify the impact of hypoxia on adipogenesis, de novo lipogenesis and metabolic flux reprogramming in adipocytes. We found that 3T3-L1 cells can successfully differentiate into lipid-accumulating adipocytes under hypoxia, although the production of lipids was reduced by about 40%. Quantitative flux analysis demonstrated that short-term (1 day) and long-term (7 days) exposure to hypoxia resulted in similar reprogramming of cellular metabolism. Overall, we found that hypoxia: 1) reduced redox and energy generation by more than 2-fold and altered the patterns of metabolic pathway contributions to production and consumption of energy and redox cofactors; 2) redirected glucose metabolism from pentose phosphate pathway and citric acid cycle to lactate production; 3) rewired glutamine metabolism, from net glutamine production to net glutamine catabolism; 4) suppressed branched chain amino acid consumption; and 5) reduced biosynthesis of odd-chain fatty acids and mono-unsaturated fatty acids, while synthesis of saturated even-chain fatty acids was not affected. Together, these results highlight the profound impact of extracellular microenvironment on adipocyte metabolic activity and function.
Collapse
Affiliation(s)
- Eleanor H Oates
- Department of Chemical and Biomolecular Engineering, Metabolic Engineering and Systems Biology Laboratory, University of Delaware, Newark, DE 19716, USA
| | - Maciek R Antoniewicz
- Department of Chemical and Biomolecular Engineering, Metabolic Engineering and Systems Biology Laboratory, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
41
|
Sun Y, Wang B, Hu Q, Zhang H, Lai X, Wang T, Zhao C, Wang J, Zhang X, Niu Q, He B, Jiang E, Shi M, Feng X, Luo Y. Loss of Lkb1 in CD11c + myeloid cells protects mice from diet-induced obesity while enhancing glucose intolerance and IL-17/IFN-γ imbalance. Cell Mol Life Sci 2023; 80:63. [PMID: 36781473 PMCID: PMC9925521 DOI: 10.1007/s00018-023-04707-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/04/2023] [Accepted: 01/22/2023] [Indexed: 02/15/2023]
Abstract
Adipose tissue CD11c+ myeloid cell is an independent risk factor associated with obesity and metabolic disorders. However, the underlying molecular basis remains elusive. Here, we demonstrated that liver kinase B1 (Lkb1), a key bioenergetic sensor, is involved in CD11c+ cell-mediated immune responses in diet-induced obesity. Loss of Lkb1 in CD11c+ cells results in obesity resistance but lower glucose tolerance, which accompanies tissue-specific immune abnormalities. The accumulation and CD80's expression of Lkb1 deficient adipose-tissue specific dendritic cells but not macrophages is restrained. Additionally, the balance of IL-17A and IFN-γ remarkably tips towards the latter in fat T cells and CD11c- macrophages. Mechanistically, IFN-γ promotes apoptosis of preadipocytes and inhibits their adipogenesis while IL-17A promotes the adipogenesis in vitro, which might account in part for the fat gain resistant phenotype. In summary, these findings reveal that Lkb1 is essential for fat CD11c+ dendritic cells responding to HFD exposure and provides new insights into the IL-17A/IFN-γ balance in HFD-induced obesity.
Collapse
Affiliation(s)
- Yunyan Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China.,Tianjin Institutes of Health Science, Tianjin, 301600, China.,Department of Hematology, Hematology Research Center of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, China.,Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Center, Kunming, China
| | - Bing Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China.,Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Qianwen Hu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China.,Tianjin Institutes of Health Science, Tianjin, 301600, China.,Department of Hematology, Hematology Research Center of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haixiao Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China.,Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Xun Lai
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Center, Kunming, China
| | - Tier Wang
- Department of Hematology, Hematology Research Center of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Chunxiao Zhao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China.,Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Jiali Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China.,Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Xi Zhang
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Center, Kunming, China
| | - Qing Niu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China.,Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Baolin He
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China.,Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Erlie Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China. .,Tianjin Institutes of Health Science, Tianjin, 301600, China.
| | - Mingxia Shi
- Department of Hematology, Hematology Research Center of Yunnan Province, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Xiaoming Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China. .,Tianjin Institutes of Health Science, Tianjin, 301600, China.
| | - Yuechen Luo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, 300020, China. .,Tianjin Institutes of Health Science, Tianjin, 301600, China.
| |
Collapse
|
42
|
Rigual MDM, Sánchez Sánchez P, Djouder N. Is liver regeneration key in hepatocellular carcinoma development? Trends Cancer 2023; 9:140-157. [PMID: 36347768 DOI: 10.1016/j.trecan.2022.10.005] [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: 07/13/2022] [Revised: 10/05/2022] [Accepted: 10/10/2022] [Indexed: 11/08/2022]
Abstract
The liver is the largest organ of the mammalian body and has the remarkable ability to fully regenerate in order to maintain tissue homeostasis. The adult liver consists of hexagonal lobules, each with a central vein surrounded by six portal triads localized in the lobule border containing distinct parenchymal and nonparenchymal cells. Because the liver is continuously exposed to diverse stress signals, several sophisticated regenerative processes exist to restore its functional status following impairment. However, these stress signals can affect the liver's capacity to regenerate and may lead to the development of hepatocellular carcinoma (HCC), one of the most aggressive liver cancers. Here, we review the mechanisms of hepatic regeneration and their potential to influence HCC development.
Collapse
Affiliation(s)
- María Del Mar Rigual
- Molecular Oncology Programme, Growth Factors, Nutrients and Cancer Group, Centro Nacional de Investigaciones Oncológicas, CNIO, Madrid, ES-28029, Spain
| | - Paula Sánchez Sánchez
- Molecular Oncology Programme, Growth Factors, Nutrients and Cancer Group, Centro Nacional de Investigaciones Oncológicas, CNIO, Madrid, ES-28029, Spain
| | - Nabil Djouder
- Molecular Oncology Programme, Growth Factors, Nutrients and Cancer Group, Centro Nacional de Investigaciones Oncológicas, CNIO, Madrid, ES-28029, Spain.
| |
Collapse
|
43
|
Hong X, Zhou Y, Zhu Z, Li Y, Li Z, Zhang Y, Hu X, Zhu F, Wang Y, Fang M, Huang Y, Shen T. Environmental endocrine disruptor Bisphenol A induces metabolic derailment and obesity via upregulating IL-17A in adipocytes. ENVIRONMENT INTERNATIONAL 2023; 172:107759. [PMID: 36696794 DOI: 10.1016/j.envint.2023.107759] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/15/2022] [Accepted: 01/14/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Bisphenol A (BPA), a ubiquitous environmental endocrine disruptor, has been extensively demonstrated to be associated with metabolic disorders, including obesity and type 2 diabetes mellitus. However, the underlying mechanism underpinning the environmental etiology of chronic metabolic disorders has not been sufficiently elucidated. OBJECTIVES This study is designed to explore the toxicological pathogenesis of chronic inflammation in BPA exposure during obesity. METHODS We investigated the role of IL-17A in the association of BPA exposure and obesity from human cross-sectional study to animal models, including genetically modified IL-17A-/- mice. RESULTS Here, our work started from case-control observation that BPA exposure was significantly associated with risk of obesity (odds ratio = 4.72, 95%CI: 3.18 - 11.18, P < 0.01), metabolic disorder and levels of interleukin-17A (IL-17A) in human adipose (estimated changes β = 0.46, 95%CI: 0.15 - 1.01, P < 0.01) with bariatric surgery. Animal model fed with high-fat diet (HFD) confirmed that BPA exposure aggravated body weight gain and insulin resistance, concurrent with much heightened inflammatory responses in the adipose tissue including increase in IL-17A and macrophage polarization towards M1 stage. Genetically modified IL-17A ablated mice (IL-17A-/-) showed reversed adipose tissue inflammation response, improved macrophage polarization homeostasis, along with insulin sensitivity in both HFD group alone or much more significantly the HFD + BPA group. Moreover, mediation analysis in human epidemiological investigation demonstrated that plasma IL-17A attributed up to 30.01% mediating role in the associations between BPA exposure and obesity risk. DISCUSSION This research paradigm from human to animal provides strong evidence for the elucidation of IL-17A moderating inflammation and insulin resistance in obesity. Such findings reiterate the obesogenic role of environmental endocrine disruptor BPA in metabolic disorders and unveils the potential toxicological mechanisms underpinning such effect.
Collapse
Affiliation(s)
- Xu Hong
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, PR China
| | - Yi Zhou
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, PR China
| | - Zhiyuan Zhu
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, PR China
| | - Yuting Li
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, PR China
| | - Zuo Li
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, PR China
| | - Yuheng Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, PR China
| | - Xinxin Hu
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, PR China
| | - Fuhai Zhu
- Health Management Center, Second Affiliated Hospital, Anhui Medical University, Hefei 230032, Anhui, PR China
| | - Yong Wang
- Department of General Surgery, Second Affiliated Hospital, Anhui Medical University, Hefei 230032, Anhui, PR China
| | - Mingliang Fang
- Department of Environmental Science and Engineering, Fudan University, Shanghai 200433, China
| | - Yichao Huang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, PR China.
| | - Tong Shen
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei 230032, Anhui, PR China.
| |
Collapse
|
44
|
Chaffey L, Roberti A, Greaves DR. Drug repurposing in cardiovascular inflammation: Successes, failures, and future opportunities. Front Pharmacol 2022; 13:1046406. [PMID: 36339576 PMCID: PMC9634418 DOI: 10.3389/fphar.2022.1046406] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/10/2022] [Indexed: 12/15/2022] Open
Abstract
Drug repurposing is an attractive, pragmatic approach to drug discovery that has yielded success across medical fields over the years. The use of existing medicines for novel indications enables dramatically reduced development costs and timescales compared with de novo drug discovery and is therefore a promising strategy in cardiovascular disease, where new drug approvals lag significantly behind that of other fields. Extensive evidence from pre-clinical and clinical studies show that chronic inflammation is a driver of pathology in cardiovascular disease, and many efforts have been made to target cardiovascular inflammation therapeutically. This approach has been met with significant challenges however, namely off-target effects associated with broad-spectrum immunosuppression, particularly in long-term conditions such as cardiovascular disease. Nevertheless, multiple anti-inflammatory medicines have been assessed for efficacy in cardiovascular clinical trials, with most of these being repurposed from their original indications in autoimmune conditions like rheumatoid arthritis. In this review, we discuss the mixed successes of clinical trials investigating anti-inflammatory drugs in cardiovascular disease, with examples such as anti-cytokine monoclonal antibodies, colchicine, and methotrexate. Looking to the future, we highlight potential new directions for drug repurposing in cardiovascular inflammation, including the emerging concepts of drug re-engineering and chrono-pharmacology.
Collapse
Affiliation(s)
| | | | - David R. Greaves
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
45
|
Interleukin-17 Family Cytokines in Metabolic Disorders and Cancer. Genes (Basel) 2022; 13:genes13091643. [PMID: 36140808 PMCID: PMC9498678 DOI: 10.3390/genes13091643] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 02/07/2023] Open
Abstract
Interleukin-17 (IL-17) family cytokines are potent drivers of inflammatory responses. Although IL-17 was originally identified as a cytokine that induces protective effects against bacterial and fungal infections, IL-17 can also promote chronic inflammation in a number of autoimmune diseases. Research in the last decade has also elucidated critical roles of IL-17 during cancer development and treatment. Intriguingly, IL-17 seems to play a role in the risk of cancers that are associated with metabolic disorders. In this review, we summarize our current knowledge on the biochemical basis of IL-17 signaling, IL-17′s involvement in cancers and metabolic disorders, and postulate how IL-17 family cytokines may serve as a bridge between these two types of diseases.
Collapse
|
46
|
Jeerawattanawart S, Siripurkpong P, Roytrakul S, Angkasekwinai P. IL-25 directly modulates adipocyte function and inflammation through the regulation of adiponectin. Inflamm Res 2022; 71:1229-1244. [PMID: 35819460 DOI: 10.1007/s00011-022-01606-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/28/2022] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE This study aimed to investigate the direct role of IL-25 in modulating adipocyte function during homeostasis and low-grade inflammation induced by lipopolysaccharide (LPS). METHODS The 3T3-L1 preadipocyte cell lines and primary cultures of adipose-derived stromal vascular precursor cells of wild-type and IL-17RB-deficient mice were used to determine the direct function of IL-25. The expression of IL-17RB in differentiating adipocyte was determined using real-time PCR and flow cytometry analysis. The effect of IL-25 on lipid accumulation, triglyceride content, lipolysis, glucose uptake, and adipokine expression in the mature adipocytes was evaluated. IL-25 modulating the expression of inflammatory cytokines in adipocytes induced by low dose LPS was determined using real-time PCR and ELISA. RESULTS The receptor for IL-25 was up-regulated during adipocyte differentiation and IL-25 directly modulated adipocyte function by reducing lipid accumulation and triglyceride concentration and enhancing lipolysis without affecting an insulin-stimulated glucose uptake. Interestingly, IL-25 induced adiponectin secretion through the PI3K/AKT signaling pathway. In 3T3-L1 adipocytes under low-grade inflammation, IL-25 attenuated the expression of IL-6 and CCL5 through the induction of adiponectin. CONCLUSION Our studies suggest that IL-25 directly regulates adipocyte function by maintaining the adiponectin level during homeostasis and by alleviating inflammatory response through the regulation of adiponectin during low-grade inflammation in adipocytes.
Collapse
Affiliation(s)
- Siranart Jeerawattanawart
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathumthani, 12120, Thailand.,Graduate Program in Biomedical Sciences, Faculty of Allied Health Sciences, Thammasat University, Pathumthani, 12120, Thailand
| | - Pilaiwan Siripurkpong
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathumthani, 12120, Thailand
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, 12120, Thailand
| | - Pornpimon Angkasekwinai
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathumthani, 12120, Thailand.
| |
Collapse
|
47
|
Schmidt V, Hogan AE, Fallon PG, Schwartz C. Obesity-Mediated Immune Modulation: One Step Forward, (Th)2 Steps Back. Front Immunol 2022; 13:932893. [PMID: 35844529 PMCID: PMC9279727 DOI: 10.3389/fimmu.2022.932893] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 05/27/2022] [Indexed: 11/15/2022] Open
Abstract
Over the past decades, the relationship between the immune system and metabolism has become a major research focus. In this arena of immunometabolism the capacity of adipose tissue to secrete immunomodulatory molecules, including adipokines, within the underlying low-grade inflammation during obesity brought attention to the impact obesity has on the immune system. Adipokines, such as leptin and adiponectin, influence T cell differentiation into different T helper subsets and their activation during immune responses. Furthermore, within the cellular milieu of adipose tissue nutrient availability regulates differentiation and activation of T cells and changes in cellular metabolic pathways. Upon activation, T cells shift from oxidative phosphorylation to oxidative glycolysis, while the differential signaling of the kinase mammalian target of rapamycin (mTOR) and the nuclear receptor PPARγ, amongst others, drive the subsequent T cell differentiation. While the mechanisms leading to a shift from the typical type 2-dominated milieu in lean people to a Th1-biased pro-inflammatory environment during obesity are the subject of extensive research, insights on its impact on peripheral Th2-dominated immune responses become more evident. In this review, we will summarize recent findings of how Th2 cells are metabolically regulated during obesity and malnutrition, and how these states affect local and systemic Th2-biased immune responses.
Collapse
Affiliation(s)
- Viviane Schmidt
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Andrew E. Hogan
- Kathleen Lonsdale Human Health Institute, Maynooth University, Maynooth, Ireland
- Obesity Immunology Research, St. Vincent’s University Hospital and University College Dublin, Dublin, Ireland
| | - Padraic G. Fallon
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Christian Schwartz
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- *Correspondence: Christian Schwartz,
| |
Collapse
|
48
|
Macchi C, Moregola A, Greco M, Svecla M, Bonacina F, Dhup S, Dadhich R, Audano M, Sonveaux P, Mauro C, Mitro N, Ruscica M, Norata G. Monocarboxylate transporter 1 deficiency impacts CD8 + T lymphocytes proliferation and recruitment to adipose tissue during obesity. iScience 2022; 25:104435. [PMID: 35707720 PMCID: PMC9189020 DOI: 10.1016/j.isci.2022.104435] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 04/13/2022] [Accepted: 05/17/2022] [Indexed: 12/11/2022] Open
Abstract
Lactate sits at the crossroad of metabolism, immunity, and inflammation. The expression of cellular lactate transporter MCT1 (known as Slc16a1) increases during immune cell activation to cope with the metabolic reprogramming. We investigated the impact of MCT1 deficiency on CD8+ T cell function during obesity-related inflammatory conditions. The absence of MCT1 impaired CD8+ T cell proliferation with a shift of ATP production to mitochondrial oxidative phosphorylation. In Slc16a1 f/f Tcell cre mice fed a high-fat diet, a reduction in the number of CD8+ T cells, which infiltrated epididymal visceral adipose tissue (epiWAT) or subcutaneous adipose tissue, was observed. Adipose tissue weight and adipocyte area were significantly reduced together with downregulation of adipogenic genes only in the epiWAT. Our findings highlight a distinct effect of MCT1 deficiency in CD8+ T cells in the crosstalk with adipocytes and reinforce the concept that targeting immunometabolic reprogramming in lymphocyte could impact the immune-adipose tissue axis in obesity.
Collapse
Affiliation(s)
- C. Macchi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - A. Moregola
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - M.F. Greco
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - M. Svecla
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - F. Bonacina
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - S. Dhup
- Pole of Pharmacology, Institut de Recherche Experimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - R.K. Dadhich
- Pole of Pharmacology, Institut de Recherche Experimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - M. Audano
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - P. Sonveaux
- Pole of Pharmacology, Institut de Recherche Experimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - C. Mauro
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - N. Mitro
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - M. Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - G.D. Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
- SISA Center for the Study of Atherosclerosis, Bassini Hospital, Via M. Gorki 50, 20092 Milan, Cinisello Balsamo, Italy
| |
Collapse
|
49
|
Huang T, Song J, Gao J, Cheng J, Xie H, Zhang L, Wang YH, Gao Z, Wang Y, Wang X, He J, Liu S, Yu Q, Zhang S, Xiong F, Zhou Q, Wang CY. Adipocyte-derived kynurenine promotes obesity and insulin resistance by activating the AhR/STAT3/IL-6 signaling. Nat Commun 2022; 13:3489. [PMID: 35715443 PMCID: PMC9205899 DOI: 10.1038/s41467-022-31126-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 06/02/2022] [Indexed: 02/05/2023] Open
Abstract
Aberrant amino acid metabolism is a common event in obesity. Particularly, subjects with obesity are characterized by the excessive plasma kynurenine (Kyn). However, the primary source of Kyn and its impact on metabolic syndrome are yet to be fully addressed. Herein, we show that the overexpressed indoleamine 2,3-dioxygenase 1 (IDO1) in adipocytes predominantly contributes to the excessive Kyn, indicating a central role of adipocytes in Kyn metabolism. Depletion of Ido1 in adipocytes abrogates Kyn accumulation, protecting mice against obesity. Mechanistically, Kyn impairs lipid homeostasis in adipocytes via activating the aryl hydrocarbon receptor (AhR)/Signal transducer and activator of transcription 3 /interleukin-6 signaling. Genetic ablation of AhR in adipocytes abolishes the effect of Kyn. Moreover, supplementation of vitamin B6 ameliorated Kyn accumulation, protecting mice from obesity. Collectively, our data support that adipocytes are the primary source of increased circulating Kyn, while elimination of accumulated Kyn could be a viable strategy against obesity.
Collapse
Affiliation(s)
- Teng Huang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Song
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Gao
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Cheng
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Xie
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Zhang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Han Wang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhichao Gao
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Wang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohui Wang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinhan He
- Department of Pharmacy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Shiwei Liu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Qilin Yu
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shu Zhang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Xiong
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Qing Zhou
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Cong-Yi Wang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
50
|
Dai W, Liu X, Su H, Li X, Xu Y, Yu Y. Influence of adipose tissue immune dysfunction on childhood obesity. Cytokine Growth Factor Rev 2022; 65:27-38. [PMID: 35595599 DOI: 10.1016/j.cytogfr.2022.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 11/17/2022]
Abstract
In recent decades, a dramatic rise has been observed in the prevalence of obesity in childhood and adolescence, along with an increase in fetal microsomia rates. The increased risk of obesity during this key period in development negatively affects the health of the individual later in life. Immune cells residing and recruited to white adipose tissue have been highlighted as important factors contributing to the pathogenesis of childhood obesity. Immune dysfunction in the context of obesity begins early in childhood, which is different from the pathological characteristics and influencing factors of adipose immunity in adults. Here, we explore the current understanding of the roles of childhood and early life events that result in high risks for obesity by influencing adipose tissue immune dysfunction under the pathological condition of obesity. Such knowledge will help in determining the mechanisms of childhood and early life obesity in efforts to ameliorate chronic inflammation-related metabolic diseases.
Collapse
Affiliation(s)
- Wanlin Dai
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China; Innovation Institute, China Medical University, China Medical University, Shenyang 110122, Liaoning, China
| | - Xiyan Liu
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Han Su
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China
| | - Xuan Li
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China; Innovation Institute, China Medical University, China Medical University, Shenyang 110122, Liaoning, China
| | - Yingxi Xu
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Yang Yu
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang 110122, Liaoning, China; College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, Key Laboratory of Liaoning Province, China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|