1
|
Liu X, Qin K, Wang C, Sun X, Li Y, Liu Y, Yang X. Butyric acid reduced lipid deposition in immortalized chicken preadipocyte by inhibiting cell proliferation and differentiation. Poult Sci 2024; 103:104171. [PMID: 39151213 PMCID: PMC11375136 DOI: 10.1016/j.psj.2024.104171] [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: 06/23/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/18/2024] Open
Abstract
The hyperplasia and hypertrophy of preadipocytes were closely related to lipid deposition in animals. Butyric acid was reported to be involved in lipid metabolism. The aim of the current study was to investigate the effect of butyric acid on the proliferation and differentiation of the immortalized chicken preadipocyte 2 (ICP2). ICP2 were treated respectively with 12mM butyric acid for 48h in proliferation trial and 4mM butyric acid plus 200 μM oleic acid for 3 d in differentiation trial. For the proliferation trial, RNA-seq analysis revealed that 2039 genes were significantly up-regulated and 780 genes were significantly down-regulated with 12 mM butyric acid after 48 h treatment. Concurrently, Cell cycle, DNA replication and p53 signaling pathways were down-regulated in Butyric acid group. More importantly, 12 mM butyric acid restrained the expression of cell proliferation genes such as PCNA, CDK1 and CDK2 in Butyric acid group (P < 0.05), and the protein expression levels of PCNA and CDK1 were also significantly decreased (P < 0.05). The Oil red staining revealed a fewer presence of red fat droplets in ICP2 following treatment with 4 mM butyric acid, accompanied by decreased levels of total cholesterol (TC) and triglycerides (TG). RNA-seq analysis shown that the number of up and down-regulated genes were 2095 and 1042 respectively in OAB group (oleic acid+butyric acid) when compared with OA group (oleic acid). Meanwhile the AMPK signaling pathway, FOXO signaling pathway and focal adhesion were significantly enriched in OAB group. Additionally, 4 mM butyric acid inhibited the expression of lipid differentiation genes including FABP4, C/EBPα, PPARγ and LPL in OAB group (P < 0.05), as well as lipogenesis proteins such as FABP4, C/EBP-α and PPARγ (P < 0.05). In conclusion, 12 mM butyric acid effectively inhibited the proliferation of ICP2 by slowing down cell cycle progression, while 4 mM butyric acid alleviated lipid deposition by reducing the production of lipid droplets through inhibiting the expression of lipid differentiation marker genes and proteins.
Collapse
Affiliation(s)
- Xiaoying Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Kailong Qin
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Chaohui Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Xi Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yun Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
2
|
Hu T, Liu CH, Lei M, Zeng Q, Li L, Tang H, Zhang N. Metabolic regulation of the immune system in health and diseases: mechanisms and interventions. Signal Transduct Target Ther 2024; 9:268. [PMID: 39379377 PMCID: PMC11461632 DOI: 10.1038/s41392-024-01954-6] [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: 03/27/2024] [Revised: 07/18/2024] [Accepted: 08/11/2024] [Indexed: 10/10/2024] Open
Abstract
Metabolism, including glycolysis, oxidative phosphorylation, fatty acid oxidation, and other metabolic pathways, impacts the phenotypes and functions of immune cells. The metabolic regulation of the immune system is important in the pathogenesis and progression of numerous diseases, such as cancers, autoimmune diseases and metabolic diseases. The concept of immunometabolism was introduced over a decade ago to elucidate the intricate interplay between metabolism and immunity. The definition of immunometabolism has expanded from chronic low-grade inflammation in metabolic diseases to metabolic reprogramming of immune cells in various diseases. With immunometabolism being proposed and developed, the metabolic regulation of the immune system can be gradually summarized and becomes more and more clearer. In the context of many diseases including cancer, autoimmune diseases, metabolic diseases, and many other disease, metabolic reprogramming occurs in immune cells inducing proinflammatory or anti-inflammatory effects. The phenotypic and functional changes of immune cells caused by metabolic regulation further affect and development of diseases. Based on experimental results, targeting cellular metabolism of immune cells becomes a promising therapy. In this review, we focus on immune cells to introduce their metabolic pathways and metabolic reprogramming, and summarize how these metabolic pathways affect immune effects in the context of diseases. We thoroughly explore targets and treatments based on immunometabolism in existing studies. The challenges of translating experimental results into clinical applications in the field of immunometabolism are also summarized. We believe that a better understanding of immune regulation in health and diseases will improve the management of most diseases.
Collapse
Affiliation(s)
- Tengyue Hu
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Chang-Hai Liu
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Min Lei
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Qingmin Zeng
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Li Li
- Division of Renal and endocrinology, Qin Huang Hospital, Xi'an, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China.
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China.
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
| | - Nannan Zhang
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China.
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China.
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
3
|
Liu S, Zhang H, Lan Z. Associations of obesity with chronic inflammatory airway diseases and mortality in adults: a population-based investigation. BMC Public Health 2024; 24:1300. [PMID: 38741199 DOI: 10.1186/s12889-024-18782-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND The association between obesity and respiratory diseases has been confirmed. However, few studies have reported the relationship between obesity and the risk and mortality of chronic inflammatory airway disease (CIAD). The aim of this study was to reveal the association between obesity and the risk of CIAD, and mortality in patients with CIAD. METHODS The study was conducted using data from the National Health and Nutrition Examination Survey (NHANES) 2013 to 2018 among adults aged 20 years and above. All participants were grouped according to body mass index (BMI) and waist circumference (WC) levels to study the relationship between obesity and CIAD. Multivariate logistic regression analysis was utilized to examine the connection between CIAD and obesity in a cross-sectional study. The association between obesity and all-cause mortality in individuals with CIAD was examined using multiple cox regression models and smooth curve fitting in a prospective cohort study. RESULTS When stratified based on BMI in comparison to the normal weight group, the ORs with 95%CIs of CIAD for underweight and obesity were 1.39 (1.01-1.93) and 1.42 (1.27-1.58), respectively. The OR with 95%CI of CIAD for obesity was 1.20 (1.09-1.31) when stratified according to WC. Additionally, underweight was associated with a higher mortality (HR = 2.44, 95% CI = 1.31-4.55), whereas overweight (HR = 0.58,95% CI = 0.39-0.87) and obesity (HR = 0.59,95% CI = 0.4-0.87) were associated with a lower mortality (P for trend < 0.05). There was a non-linear association between BMI and all-cause mortality (P for non-linear = 0.001). An analysis of a segmentation regression model between BMI and all-cause mortality revealed a BMI turning point value of 32.4 kg/m2. The mortality of CIAD patients was lowest when BMI was 32.4 kg/m2. When BMI ≤ 32.4 kg/m2, BMI was inversely associated with all-cause mortality in patients with CIAD (HR: 0.92, 95%CI:0.88-0.97). However, when BMI > 32.4 kg/m2, there was no association between BMI and all-cause mortality (HR:1.02, 95%CI:0.97-1.06). CONCLUSION Compared to normal weight, underweight and obesity were associated with the increased risk of CIAD. Underweight was associated with increased all-cause mortality, while overweight was associated with reduced all-cause mortality. There was a non-linear association between BMI and all-cause mortality in patients with CIAD. The all-cause mortality was lowest when BMI was 32.4 kg/m2.
Collapse
Affiliation(s)
- Shanshan Liu
- Jiangxi University of Chinese Medicine, No. 1688, Meiling Avenue, Xinjian District, Nanchang City, Jiangxi Province, China
- Department of Cardiology, Donghu District, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, No. 445, Bayi Avenue, Nanchang City, Jiangxi Province, China
| | - Hao Zhang
- Jiangxi University of Chinese Medicine, No. 1688, Meiling Avenue, Xinjian District, Nanchang City, Jiangxi Province, China
| | - Zhihui Lan
- Department of Respiratory and Critical Care, Donghu District, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, No. 445, Bayi Avenue, Nanchang City, Jiangxi Province, China.
| |
Collapse
|
4
|
Rosendo-Silva D, Gomes PB, Rodrigues T, Viana S, da Costa AN, Scherer PE, Reis F, Pereira F, Seiça R, Matafome P. Clinical and molecular profiling of human visceral adipose tissue reveals impairment of vascular architecture and remodeling as an early hallmark of dysfunction. Metabolism 2024; 153:155788. [PMID: 38219974 DOI: 10.1016/j.metabol.2024.155788] [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: 11/03/2023] [Revised: 12/27/2023] [Accepted: 01/05/2024] [Indexed: 01/16/2024]
Abstract
Adipose tissue dysfunction is more related to insulin resistance than body mass index itself and an alteration in adipose tissue function is thought to underlie the shift from metabolically healthy to unhealthy obesity. Herein, we performed a clustering analysis that revealed distinct visceral adipose tissue gene expression patterns in patients with obesity at distinct stages of metabolic dysregulation. We have built a cross-sectional cohort that aims at reflecting the evolution of the metabolic sequelae of obesity with the main objective to map the sequential events that play a role in adipose tissue dysfunction from the metabolically healthy (insulin-sensitive) state to several incremental degrees of metabolic dysregulation, encompassing insulin resistance establishment, pre-diabetes, and type 2 diabetes. We found that insulin resistance is mainly marked by the downregulation of adipose tissue vasculature remodeling-associated gene expression, suggesting that processes like angiogenesis and adaptative expansion/retraction ability suffer early dysregulation. Prediabetes was characterized by compensatory growth factor-dependent signaling and increased response to hypoxia, while type 2 diabetes was associated with loss of cellular response to insulin and hypoxia and concomitant upregulation of inflammatory markers. Our findings suggest a putative sequence of dysregulation of biological processes that is not linear and has multiple distinct phases across the metabolic dysregulation process, ultimately culminating in the climax of adipose tissue dysfunction in type 2 diabetes. Several studies have addressed the transcriptomic changes in adipose tissue of patients with obesity. However, to the best of our knowledge, this is the first study unraveling the potential molecular mechanisms associated with the multi-step evolution of adipose tissue dysfunction along the metabolic sequelae of obesity.
Collapse
Affiliation(s)
- Daniela Rosendo-Silva
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Pedro Bastos Gomes
- Department of Surgery, Universitary Hospital Center of Coimbra, Portugal
| | - Tiago Rodrigues
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Sofia Viana
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; Polytechnic University of Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal
| | - André Nogueira da Costa
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Translational Medicine, Biomedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Flávio Reis
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Francisco Pereira
- Polytechnic University of Coimbra, Coimbra Institute of Engineering, Coimbra, Portugal; Centre for Informatics and Systems of the University of Coimbra (CISUC), University of Coimbra, Coimbra, Portugal
| | - Raquel Seiça
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
| | - Paulo Matafome
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; Polytechnic University of Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal.
| |
Collapse
|
5
|
Vujičić M, Broderick I, Salmantabar P, Perian C, Nilsson J, Sihlbom Wallem C, Wernstedt Asterholm I. A macrophage-collagen fragment axis mediates subcutaneous adipose tissue remodeling in mice. Proc Natl Acad Sci U S A 2024; 121:e2313185121. [PMID: 38300872 PMCID: PMC10861897 DOI: 10.1073/pnas.2313185121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 12/19/2023] [Indexed: 02/03/2024] Open
Abstract
Efficient removal of fibrillar collagen is essential for adaptive subcutaneous adipose tissue (SAT) expansion that protects against ectopic lipid deposition during weight gain. Here, we used mice to further define the mechanism for this collagenolytic process. We show that loss of collagen type-1 (CT1) and increased CT1-fragment levels in expanding SAT are associated with proliferation of resident M2-like macrophages that display increased CD206-mediated engagement in collagen endocytosis compared to chow-fed controls. Blockage of CD206 during acute high-fat diet-induced weight gain leads to SAT CT1-fragment accumulation associated with elevated inflammation and fibrosis markers. Moreover, these SAT macrophages' engagement in collagen endocytosis is diminished in obesity associated with elevated levels collagen fragments that are too short to assemble into triple helices. We show that such short fragments provoke M2-macrophage proliferation and fibroinflammatory changes in fibroblasts. In conclusion, our data delineate the importance of a macrophage-collagen fragment axis in physiological SAT expansion. Therapeutic targeting of this process may be a means to prevent pathological adipose tissue remodeling, which in turn may reduce the risk for obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Milica Vujičić
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg405 30, Sweden
| | - Isabella Broderick
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg405 30, Sweden
| | - Pegah Salmantabar
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg405 30, Sweden
| | - Charlène Perian
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg405 30, Sweden
| | - Jonas Nilsson
- Proteomics Core Facility, The Sahlgrenska Academy at University of Gothenburg, Gothenburg405 30, Sweden
| | - Carina Sihlbom Wallem
- Proteomics Core Facility, The Sahlgrenska Academy at University of Gothenburg, Gothenburg405 30, Sweden
| | - Ingrid Wernstedt Asterholm
- Department of Physiology/Metabolic Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Gothenburg405 30, Sweden
| |
Collapse
|
6
|
Xining Z, Sai L. The Evolving Function of Vasculature and Pro-angiogenic Therapy in Fat Grafting. Cell Transplant 2024; 33:9636897241264976. [PMID: 39056562 PMCID: PMC11282510 DOI: 10.1177/09636897241264976] [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: 03/18/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 07/28/2024] Open
Abstract
Autologous fat grating is a widely-accepted method to correct soft tissue deficiency. Although fat transplantation shows excellent biocompatibility and simple applicability, the relatively low retention rate caused by fat necrosis is still a challenge. The vasculature is integral after fat grafting, serving multiple crucial functions. Rapid and effective angiogenesis within grafts is essential for supplying oxygen necessary for adipocytes' survival. It facilitates the influx of inflammatory cells to remove necrotic adipocytes and aids in the delivery of regenerative cells for adipose tissue regeneration in fat grafts. The vasculature also provides a niche for interaction between adipose progenitor cells and vascular progenitor cells, enhancing angiogenesis and adipogenesis in grafts. Various methods, such as enriching grafts with diverse pro-angiogenic cells or utilizing cell-free approaches, have been employed to enhance angiogenesis. Beige and dedifferentiated adipocytes in grafts could increase vessel density. This review aims to outline the function of vasculature in fat grafting and discuss different cell or cell-free approaches that can enhance angiogenesis following fat grafting.
Collapse
Affiliation(s)
- Zhang Xining
- The Plastic and Aesthetic Center, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Luo Sai
- The Plastic and Aesthetic Center, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
7
|
Bi X, Li B, Zou J, Zhao J, Chen Y, Wang X, Lu F, Li Y, Dong Z, Gao J. Fascia Promotes Adipose Tissue Regeneration by Improving Early Macrophage Infiltration after Fat Grafting in a Mouse Model. Plast Reconstr Surg 2023; 152:446e-457e. [PMID: 36723983 DOI: 10.1097/prs.0000000000010259] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Low early macrophage fat graft infiltration (within a week of surgery) hinders tissue regeneration, suggesting that macrophages play a vital role in early angiogenesis and adipogenesis. However, the source of macrophages during this period is unclear. METHOD C57BL/6 mice were split into fascial removal (FR) group and control groups (CG). Mice had a piece of back fascia removed in the FR group, which was immediately replaced in the CG, and inguinal fat injected into the transplantation site of both groups. Separately, fascia was harvested from green fluorescent protein-expressing mice and transplanted into C57BL/6 mice for tracing macrophage infiltration after fat grafting. RESULTS The number of capillaries in the FR group was lower than that in the CG at days 3 ( P < 0.01) and 7 ( P < 0.05). Moreover, the number of small adipocytes in the FR group was lower than in the CG on days 3, 7, and 14 (all P < 0.05), and the relative expression of several adipogenic proteins was significantly lower in the FR group than in the CG on days 14 and 30. The timeline of macrophage infiltration was consistent with angiogenesis and adipogenesis. The number of macrophages in the FR group was significantly lower than in the CG at days 3 and 7 ( P < 0.05), and there were more fascia-derived macrophages than circulation-derived macrophages infiltrated into fat grafts within 7 days. Finally, the graft retention was lower in the FR group than the CG at day 90 ( P < 0.05). CONCLUSION In the early stage after fat grafting, fascial macrophage infiltration initiates tissue regeneration, thereby improving graft retention by promoting angiogenesis and adipogenesis. CLINICAL RELEVANCE STATEMENT In the clinic, injecting fat close to the fascia may increase fat retention. Fascia is widespread and self-regenerating, which may be a promising alternative source of local macrophages, with implications for tissue-engineering therapies such as correction of soft-tissue defects and breast reconstruction.
Collapse
Affiliation(s)
- Xin Bi
- From the Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University
- Dermatology Department, First People's Hospital of Yunnan Province
| | - Bin Li
- From the Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University
| | - Jialiang Zou
- From the Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University
| | - Jing Zhao
- From the Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University
| | - Yunzi Chen
- From the Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University
| | - Xinhui Wang
- From the Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University
| | - Feng Lu
- From the Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University
| | - Ye Li
- From the Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University
| | - Ziqing Dong
- From the Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University
| | - Jianhua Gao
- From the Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University
| |
Collapse
|
8
|
Musale V, Wasserman DH, Kang L. Extracellular matrix remodelling in obesity and metabolic disorders. LIFE METABOLISM 2023; 2:load021. [PMID: 37383542 PMCID: PMC10299575 DOI: 10.1093/lifemeta/load021] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
Obesity causes extracellular matrix (ECM) remodelling which can develop into serious pathology and fibrosis, having metabolic effects in insulin-sensitive tissues. The ECM components may be increased in response to overnutrition. This review will focus on specific obesity-associated molecular and pathophysiological mechanisms of ECM remodelling and the impact of specific interactions on tissue metabolism. In obesity, complex network of signalling molecules such as cytokines and growth factors have been implicated in fibrosis. Increased ECM deposition contributes to the pathogenesis of insulin resistance at least in part through activation of cell surface integrin receptors and CD44 signalling cascades. These cell surface receptors transmit signals to the cell adhesome which orchestrates an intracellular response that adapts to the extracellular environment. Matrix proteins, glycoproteins, and polysaccharides interact through ligand-specific cell surface receptors that interact with the cytosolic adhesion proteins to elicit specific actions. Cell adhesion proteins may have catalytic activity or serve as scaffolds. The vast number of cell surface receptors and the complexity of the cell adhesome have made study of their roles challenging in health and disease. Further complicating the role of ECM-cell receptor interactions is the variation between cell types. This review will focus on recent insights gained from studies of two highly conserved, ubiquitously axes and how they contribute to insulin resistance and metabolic dysfunction in obesity. These are the collagen-integrin receptor-IPP (ILK-PINCH-Parvin) axis and the hyaluronan-CD44 interaction. We speculate that targeting ECM components or their receptor-mediated cell signalling may provide novel insights into the treatment of obesity-associated cardiometabolic complications.
Collapse
Affiliation(s)
- Vishal Musale
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland DD1 9SY, UK
| | - David H. Wasserman
- Department of Molecular Physiology and Biophysics, Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, TN 37235, USA
| | - Li Kang
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland DD1 9SY, UK
| |
Collapse
|
9
|
Siegel-Axel D, Barroso Oquendo M, Gerst F, Fend F, Wagner R, Heni M, Königsrainer A, Häring HU, Fritsche A, Schleicher E, Birkenfeld AL, Stefan N. Extracellular Matrix Expression in Human Pancreatic Fat Cells of Patients with Normal Glucose Regulation, Prediabetes and Type 2 Diabetes. Int J Mol Sci 2023; 24:11169. [PMID: 37446346 DOI: 10.3390/ijms241311169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Previously, we found that human pancreatic preadipocytes (PPAs) and islets influence each other and that the crosstalk with the fatty liver via the hepatokine fetuin-A/palmitate induces inflammatory responses. Here, we examined whether the mRNA-expression of pancreatic extracellular matrix (ECM)-forming and -degrading components differ in PPAs from individuals with normal glucose regulation (PPAs-NGR), prediabetes (PPAs-PD), and type 2 diabetes (PPAs-T2D), and whether fetuin-A/palmitate impacts ECM-formation/degradation and associated monocyte invasion. Human pancreatic resections were analyzed (immuno)histologically. PPAs were studied for mRNA expression by real-time PCR and protein secretion by Luminex analysis. Furthermore, co-cultures with human islets and monocyte migration assays in Transwell plates were conducted. We found that in comparison with NGR-PPAs, TIMP-2 mRNA levels were lower in PPAs-PD, and TGF-β1 mRNA levels were higher in PPAs-T2D. Fetuin-A/palmitate reduced fibronectin, decorin, TIMP-1/-2 and TGF-ß1 mRNA levels. Only fibronectin was strongly downregulated by fetuin-A/palmitate independently of the glycemic status. Co-culturing of PPAs with islets increased TIMP-1 mRNA expression in islets. Fetuin-A/palmitate increased MMP-1, usherin and dermatopontin mRNA-levels in co-cultured islets. A transmigration assay showed increased monocyte migration towards PPAs, which was enhanced by fetuin-A/palmitate. This was more pronounced in PPAs-T2D. The expression of distinct ECM components differs in PPAs-PD and PPAs-T2D compared to PPAs-NGR, suggesting that ECM alterations can occur even in mild hyperglycemia. Fetuin-A/palmitate impacts on ECM formation/degradation in PPAs and co-cultured islets. Fetuin-A/palmitate also enhances monocyte migration, a process which might impact on matrix turnover.
Collapse
Affiliation(s)
- Dorothea Siegel-Axel
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany
| | - Morgana Barroso Oquendo
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany
- EKU Tübingen, Quantitative Biology Center (QBiC), University of Tübingen, 72076 Tübingen, Germany
| | - Felicia Gerst
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany
| | - Falko Fend
- Department of General Pathology and Pathological Anatomy, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Robert Wagner
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Heinrich Heine University Düsseldorf (HHU), 40225 Düsseldorf, Germany
| | - Martin Heni
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany
- Division of Endocrinology and Diabetology, Department of Internal Medicine I, University Hospital Ulm, 89081 Ulm, Germany
| | - Alfred Königsrainer
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Hans-Ulrich Häring
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany
| | - Erwin Schleicher
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Andreas L Birkenfeld
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany
| | - Norbert Stefan
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Department of Internal Medicine IV, University Hospital of Tübingen, Otfried-Müller Str. 10, 72076 Tübingen, Germany
| |
Collapse
|
10
|
Razi O, Teixeira AM, Tartibian B, Zamani N, Knechtle B. Respiratory issues in patients with multiple sclerosis as a risk factor during SARS-CoV-2 infection: a potential role for exercise. Mol Cell Biochem 2023; 478:1533-1559. [PMID: 36411399 PMCID: PMC9684932 DOI: 10.1007/s11010-022-04610-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 11/04/2022] [Indexed: 11/23/2022]
Abstract
Coronavirus disease-2019 (COVID-19) is associated with cytokine storm and is characterized by acute respiratory distress syndrome (ARDS) and pneumonia problems. The respiratory system is a place of inappropriate activation of the immune system in people with multiple sclerosis (MS), and this may cause damage to the lung and worsen both MS and infections.The concerns for patients with multiple sclerosis are because of an enhance risk of infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The MS patients pose challenges in this pandemic situation, because of the regulatory defect of autoreactivity of the immune system and neurological and respiratory tract symptoms. In this review, we first indicate respiratory issues associated with both diseases. Then, the main mechanisms inducing lung damages and also impairing the respiratory muscles in individuals with both diseases is discussed. At the end, the leading role of physical exercise on mitigating respiratory issues inducing mechanisms is meticulously evaluated.
Collapse
Affiliation(s)
- Omid Razi
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Razi University, Kermanshah, Iran
| | - Ana Maria Teixeira
- Research Center for Sport and Physical Activity, Faculty of Sport Sciences and Physical Education, University of Coimbra, Coimbra, Portugal
| | - Bakhtyar Tartibian
- Department of Exercise Physiology, Faculty of Physical Education and Sports Sciences, Allameh Tabataba’i University, Tehran, Iran
| | - Nastaran Zamani
- Department of Biology, Faculty of Science, Payame-Noor University, Tehran, Iran
| | - Beat Knechtle
- Institute of Primary Care, University of Zurich, Zurich, Switzerland
- Medbase St. Gallen Am Vadianplatz, Vadianstrasse 26, 9001 St. Gallen, Switzerland
| |
Collapse
|
11
|
Santillana N, Astudillo-Guerrero C, D’Espessailles A, Cruz G. White Adipose Tissue Dysfunction: Pathophysiology and Emergent Measurements. Nutrients 2023; 15:nu15071722. [PMID: 37049561 PMCID: PMC10096946 DOI: 10.3390/nu15071722] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/27/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
White adipose tissue (AT) dysfunction plays an important role in the development of cardiometabolic alterations associated with obesity. AT dysfunction is characterized by the loss of the expansion capacity of the AT, an increment in adipocyte hypertrophy, and changes in the secretion profile of adipose cells, associated with accumulation of macrophages and inflammation. Since not all people with an excess of adiposity develop comorbidities, it is necessary to find simple tools that can evidence AT dysfunction and allow the detection of those people with the potential to develop metabolic alterations. This review focuses on the current pathophysiological mechanisms of white AT dysfunction and emerging measurements to assess its functionality.
Collapse
Affiliation(s)
- Natalia Santillana
- Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago 8380453, Chile
| | - Camila Astudillo-Guerrero
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Programa de Doctorado en Ciencias Mención Neurociencia, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Amanda D’Espessailles
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2820000, Chile
| | - Gonzalo Cruz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| |
Collapse
|
12
|
Wu Y, Jiang Y, Guo JQ, Yang ZW, Carvalho A, Qian LL, Ji JJ, Ji ZJ, Ma GS, Yao YY. Visceral adipose tissue-directed human kallistatin gene therapy improves adipose tissue remodeling and metabolic health in obese mice. Cell Signal 2023; 106:110637. [PMID: 36813150 DOI: 10.1016/j.cellsig.2023.110637] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 01/31/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023]
Abstract
OBJECTIVE Adipose tissue remodeling is a dynamic process that is pathologically expedited in the obese state and is closely related to obesity-associated disease progression. This study aimed to explore the effects of human kallistatin (HKS) on adipose tissue remodeling and obesity-related metabolic disorders in mice fed with a high-fat diet (HFD). METHODS Adenovirus-mediated HKS cDNA (Ad.HKS) and a blank adenovirus (Ad.Null) were constructed and injected into the epididymal white adipose tissue (eWAT) of 8-weeks-old male C57B/L mice. The mice were fed normal or HFD for 28 days. The body weight and circulating lipids levels were assessed. Intraperitoneal glucose tolerance test (IGTT) and insulin tolerance test (ITT) were also performed. Oil-red O staining was used to assess the extent of lipid deposition in the liver. Immunohistochemistry and HE staining were used to measure HKS expression, adipose tissue morphology, and macrophage infiltration. Western blot and qRT-PCR were used to evaluate the expression of adipose function-related factors. RESULTS At the end of the experiment, the expression of HKS in the serum and eWAT of the Ad.HKS group was higher than in the Ad.Null group. Furthermore, Ad.HKS mice had lower body weight and decreased serum and liver lipid levels after four weeks of HFD feeding. IGTT and ITT showed that HKS treatment maintained balanced glucose homeostasis. Additionally, inguinal white adipose tissue (iWAT) and eWAT in Ad.HKS mice had a higher number of smaller-size adipocytes and had less macrophage infiltration than Ad.Null group. HKS significantly increased the mRNA levels of adiponectin, vaspin, and eNOS. In contrast, HKS decreased RBP4 and TNFα levels in the adipose tissues. Western blot results showed that local injection of HKS significantly upregulated the protein expressions of SIRT1, p-AMPK, IRS1, p-AKT, and GLUT4 in eWAT. CONCLUSIONS HKS injection in eWAT improves HFD-induced adipose tissue remodeling and function, thus significantly improving weight gain and dysregulation of glucose and lipid homeostasis in mice.
Collapse
Affiliation(s)
- Ya Wu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| | - Yu Jiang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| | - Jia-Qi Guo
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| | - Zi-Wei Yang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| | - Abdlay Carvalho
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| | - Ling-Lin Qian
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| | - Jing-Jing Ji
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| | - Zhen-Jun Ji
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| | - Gen-Shan Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| | - Yu-Yu Yao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
13
|
Asprosin Exerts Pro-Inflammatory Effects in THP-1 Macrophages Mediated via the Toll-like Receptor 4 (TLR4) Pathway. Int J Mol Sci 2022; 24:ijms24010227. [PMID: 36613673 PMCID: PMC9820073 DOI: 10.3390/ijms24010227] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
Adipose tissue is a dynamic endocrine organ, secreting a plethora of adipokines which play a key role in regulating metabolic homeostasis and other physiological processes. An altered adipokine secretion profile from adipose tissue depots has been associated with obesity and related cardio-metabolic diseases. Asprosin is a recently described adipokine that is released in response to fasting and can elicit orexigenic and glucogenic effects. Circulating asprosin levels are elevated in a number of cardio-metabolic diseases, including obesity and type 2 diabetes. In vitro studies have reported pro-inflammatory effects of asprosin in a variety of tissues. The present study aimed to further elucidate the role of asprosin in inflammation by exploring its potential effect(s) in THP-1 macrophages. THP-1 monocytes were differentiated to macrophages by 48 h treatment with dihydroxyvitamin D3. Macrophages were treated with 100 nM recombinant human asprosin, 100 ng/mL lipopolysaccharide (LPS), and 10 μM caffeic acid phenethyl ester (CAPE; an inhibitor of NFκB activation) or 1 µM TAK-242 (a Toll-like receptor 4, TLR4, inhibitor). The expression and secretion of pertinent pro-inflammatory mediators were measured by qPCR, Western blot, ELISA and Bioplex. Asprosin stimulation significantly upregulated the expression and secretion of the pro-inflammatory cytokines: tumour necrosis factor α (TNFα), interleukin-1β (IL-1β), IL-8 and IL-12 in vitro. This pro-inflammatory response in THP-1 macrophages was partly attenuated by the treatments with CAPE and was significantly inhibited by TAK-242 treatment. Asprosin-induced inflammation is significantly counteracted by TLR4 inhibition in THP-1 macrophages, suggesting that asprosin exerts its pro-inflammatory effects, at least in part, via the TLR4 signalling pathway.
Collapse
|
14
|
Qian X, Meng X, Zhang S, Zeng W. Neuroimmune regulation of white adipose tissues. FEBS J 2022; 289:7830-7853. [PMID: 34564950 DOI: 10.1111/febs.16213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/21/2021] [Accepted: 09/24/2021] [Indexed: 01/14/2023]
Abstract
The white adipose tissues (WAT) are located in distinct depots throughout the body. They serve as an energy reserve, providing fatty acids for other tissues via lipolysis when needed, and function as an endocrine organ to regulate systemic metabolism. Their activities are coordinated through intercellular communications among adipocytes and other cell types such as residential and infiltrating immune cells, which are collectively under neuronal control. The adipocytes and immune subtypes including macrophages/monocytes, eosinophils, neutrophils, group 2 innate lymphoid cells (ILC2s), T and B cells, dendritic cells (DCs), and natural killer (NK) cells display cellular and functional diversity in response to the energy states and contribute to metabolic homeostasis and pathological conditions. Accumulating evidence reveals that neuronal innervations control lipid deposition and mobilization via regulating lipolysis, adipocyte size, and cellularity. Vice versa, the neuronal innervations and activity are influenced by cellular factors in the WAT. Though the literature describing adipose tissue cells is too extensive to cover in detail, we strive to highlight a selected list of neuronal and immune components in this review. The cell-to-cell communications and the perspective of neuroimmune regulation are emphasized to enlighten the potential therapeutic opportunities for treating metabolic disorders.
Collapse
Affiliation(s)
- Xinmin Qian
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Xia Meng
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Shan Zhang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Wenwen Zeng
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China.,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| |
Collapse
|
15
|
Wu P, Guo L, Li X, Du Y, Lin X, Ma X, Lin Y, Wen C, Yang C, Liu N, Feng Q, Xue Y, Guan M. Comprehensive analysis of epigenomics and transcriptome data to identify potential target genes associated with obesity. Front Genet 2022; 13:1024300. [PMID: 36313453 PMCID: PMC9614047 DOI: 10.3389/fgene.2022.1024300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
DNA methylation is closely related to the occurrence and development of many diseases, but its role in obesity is still unclear. This study aimed to find the potential differentially methylated genes associated with obesity occurrence and development. By combining methylation and transcriptome analysis, we identified the key genes in adipose tissue affecting the occurrence and development of obesity and revealed the possible molecular mechanisms involved in obesity pathogenesis. We first screened 14 methylation-related differential genes and verified their expression in adipose tissue by quantitative polymerase chain reaction (qPCR). Seven genes with the same expression pattern were identified as key genes, namely, CCRL2, GPT, LGALS12, PC, SLC27A2, SLC4A4, and TTC36. Then, the immune microenvironment of adipose tissue was quantified by CIBERSORT, and we found that the content of M0 macrophages and T follicular helper cells in adipose tissue was significantly increased and decreased, respectively, in the obese group. Furthermore, the relationship between key genes and the immune microenvironment was analyzed. Additionally, the metabolic pathway activity of each sample was calculated based on the ssGSEA algorithm, and the key gene–metabolic network was constructed. Moreover, we performed a CMAP analysis based on the differential genes in adipose tissue to screen out drugs potentially effective in obesity treatment. In conclusion, we identified seven methylation-related key genes closely related to obesity pathogenesis and explored the potential mechanism of their role in obesity. This study provided novel insights into the molecular mechanisms and management of obesity.
Collapse
Affiliation(s)
- Peili Wu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lei Guo
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xuelin Li
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuejun Du
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaochun Lin
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoqin Ma
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yingbei Lin
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Churan Wen
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chuyi Yang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Nannan Liu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qijian Feng
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yaoming Xue
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Meiping Guan
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- *Correspondence: Meiping Guan,
| |
Collapse
|
16
|
Wang H, Xu S, Li D, Xie Z. Structural Characterization and Macrophage Polarization-Modulating Activity of a Novel Polysaccharide from Large Yellow Tea. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:12565-12576. [PMID: 36154025 DOI: 10.1021/acs.jafc.2c05593] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
A novel homogeneous polysaccharide (LYP-S3) that promotes the M2 polarization of macrophages was obtained from large yellow tea by a bioactivity-guided sequential isolation procedure and activity evaluation in the present study. Structural characterization revealed that LYP-S3 has an average molecular weight of 28.6 kDa and is composed of rhamnose, arabinose, galactose, glucose, and galacturonic acid at the molar ratio of 8.08:11.66:11.77:3.96:58.02. The main backbone of LYP-S3 consists of →4)-α-d-GalpA-6-OMe-(1→, β-d-GalpA-(1→, →4)-β-d-Galp-(→1, and →β-d-Galp-(1→, and the branches are composed of α-l-Araf-(→1, →5)-α-l-Araf-(1→, →2,4)-β-l-Rhap-(1→, →2)-β-l-Rhap-(1→, and →4)-β-d-Glcp-(1→. An in vitro bioactivity evaluation assay showed that LYP-S3 remarkably reduced the expression of M1 macrophage markers and increased the expression of M2 macrophage markers. In addition, LYP-S3 inhibited adipocyte differentiation and adipogenesis in 3T3-L1 adipocytes and blocked macrophage migration toward 3T3-L1 adipocytes in the cocultures of bone-marrow-derived monocytes and 3T3-L1 adipocytes. Furthermore, LYP-S3 promoted the M2 polarization of macrophages in cocultures. These findings suggested that LYP-S3 has a potential function in preventing inflammation and obesity.
Collapse
Affiliation(s)
- Hongyan Wang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Shan Xu
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Daxiang Li
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Zhongwen Xie
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| |
Collapse
|
17
|
Bernier E, Lachance A, Plante AS, Lemieux P, Mourabit Amari K, Weisnagel SJ, Gagnon C, Michaud A, Tchernof A, Morisset AS. Trimester-Specific Serum Fructosamine in Association with Abdominal Adiposity, Insulin Resistance, and Inflammation in Healthy Pregnant Individuals. Nutrients 2022; 14:nu14193999. [PMID: 36235652 PMCID: PMC9572673 DOI: 10.3390/nu14193999] [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] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/16/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
This study aimed to (1) characterize the variations in serum fructosamine across trimesters and according to pre-pregnancy BMI (ppBMI), and (2) examine associations between fructosamine and adiposity/metabolic markers (ppBMI, first-trimester adiposity, leptin, glucose homeostasis, and inflammation measurements) during pregnancy. Serum fructosamine, albumin, fasting glucose and insulin, leptin, adiponectin, interleukin-6 (IL-6), and C-reactive protein (CRP) concentrations were measured at each trimester. In the first trimester, subcutaneous (SAT) and visceral (VAT) adipose tissue thicknesses were estimated by ultrasound. In the 101 healthy pregnant individuals included (age: 32.2 ± 3.5 y.o.; ppBMI: 25.5 ± 5.5 kg/m2), fructosamine concentrations decreased during pregnancy whereas albumin-corrected fructosamine concentrations increased (p < 0.0001 for both). Notably, fructosamine concentrations were inversely associated with ppBMI, first-trimester SAT, VAT, and leptin (r = −0.55, r = −0.61, r = −0.48, r = −0.47, respectively; p < 0.0001 for all), first-trimester fasting insulin and HOMA-IR (r = −0.46, r = −0.46; p < 0.0001 for both), and first-trimester IL-6 (r = −0.38, p < 0.01). However, once corrected for albumin, most of the correlations lost strength. Once adjusted for ppBMI, fructosamine concentrations were positively associated with third-trimester fasting glucose and CRP (r = 0.24, r = 0.27; p < 0.05 for both). In conclusion, serum fructosamine is inversely associated with adiposity before and during pregnancy, with markers of glucose homeostasis and inflammation, but the latter associations are partially influenced by albumin concentrations and ppBMI.
Collapse
Affiliation(s)
- Emilie Bernier
- École de Nutrition, l’Université Laval, Québec, QC G1V 0A6, Canada
- Centre Nutrition, Santé et Société (NUTRISS), l’Institut sur la Nutrition et les Aliments Fonctionnels (INAF), l’Université Laval, Québec, QC G1V 0A6, Canada
- Axe Endocrinologie et Néphrologie, Centre de Recherche, CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
| | - Amélie Lachance
- École de Nutrition, l’Université Laval, Québec, QC G1V 0A6, Canada
- Centre Nutrition, Santé et Société (NUTRISS), l’Institut sur la Nutrition et les Aliments Fonctionnels (INAF), l’Université Laval, Québec, QC G1V 0A6, Canada
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie, Québec-Université Laval, Québec, QC G1V 4G5, Canada
| | - Anne-Sophie Plante
- Centre Nutrition, Santé et Société (NUTRISS), l’Institut sur la Nutrition et les Aliments Fonctionnels (INAF), l’Université Laval, Québec, QC G1V 0A6, Canada
- Axe Endocrinologie et Néphrologie, Centre de Recherche, CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
| | - Patricia Lemieux
- Axe Endocrinologie et Néphrologie, Centre de Recherche, CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
- Département de Médecine, l’Université Laval, Québec, QC G1V 0A6, Canada
| | - Karim Mourabit Amari
- Département de Médecine de Laboratoire, CHU de Québec-Université Laval, Québec, QC G1V 4G5, Canada
| | - S. John Weisnagel
- Axe Endocrinologie et Néphrologie, Centre de Recherche, CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
- Département de Médecine, l’Université Laval, Québec, QC G1V 0A6, Canada
| | - Claudia Gagnon
- Axe Endocrinologie et Néphrologie, Centre de Recherche, CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
- Département de Médecine, l’Université Laval, Québec, QC G1V 0A6, Canada
| | - Andréanne Michaud
- École de Nutrition, l’Université Laval, Québec, QC G1V 0A6, Canada
- Centre Nutrition, Santé et Société (NUTRISS), l’Institut sur la Nutrition et les Aliments Fonctionnels (INAF), l’Université Laval, Québec, QC G1V 0A6, Canada
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie, Québec-Université Laval, Québec, QC G1V 4G5, Canada
| | - André Tchernof
- École de Nutrition, l’Université Laval, Québec, QC G1V 0A6, Canada
- Centre Nutrition, Santé et Société (NUTRISS), l’Institut sur la Nutrition et les Aliments Fonctionnels (INAF), l’Université Laval, Québec, QC G1V 0A6, Canada
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie, Québec-Université Laval, Québec, QC G1V 4G5, Canada
| | - Anne-Sophie Morisset
- École de Nutrition, l’Université Laval, Québec, QC G1V 0A6, Canada
- Centre Nutrition, Santé et Société (NUTRISS), l’Institut sur la Nutrition et les Aliments Fonctionnels (INAF), l’Université Laval, Québec, QC G1V 0A6, Canada
- Axe Endocrinologie et Néphrologie, Centre de Recherche, CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
- Correspondence: ; Tel.: +1-418-656-2131 (ext. 13982)
| |
Collapse
|
18
|
Takahashi M, Yamamuro D, Wakabayashi T, Takei A, Takei S, Nagashima S, Okazaki H, Ebihara K, Yagyu H, Takayanagi Y, Onaka T, Goldberg IJ, Ishibashi S. Loss of myeloid lipoprotein lipase exacerbates adipose tissue fibrosis with collagen VI deposition and hyperlipidemia in leptin-deficient obese mice. J Biol Chem 2022; 298:102322. [PMID: 35926714 PMCID: PMC9440390 DOI: 10.1016/j.jbc.2022.102322] [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: 01/21/2022] [Revised: 07/19/2022] [Accepted: 07/19/2022] [Indexed: 11/20/2022] Open
Abstract
During obesity, tissue macrophages increase in number and become proinflammatory, thereby contributing to metabolic dysfunction. Lipoprotein lipase (LPL), which hydrolyzes triglyceride in lipoproteins, is secreted by macrophages. However, the role of macrophage-derived LPL in adipose tissue remodeling and lipoprotein metabolism is largely unknown. To clarify these issues, we crossed leptin-deficient Lepob/ob mice with mice lacking the Lpl gene in myeloid cells (Lplm−/m−) to generate Lplm−/m−;Lepob/ob mice. We found the weight of perigonadal white adipose tissue (WAT) was increased in Lplm−/m−;Lepob/ob mice compared with Lepob/ob mice due to substantial accumulation of both adipose tissue macrophages and collagen that surrounded necrotic adipocytes. In the fibrotic epidydimal WAT of Lplm−/m−;Lepob/ob mice, we observed an increase in collagen VI and high mobility group box 1, while α-smooth muscle cell actin, a marker of myofibroblasts, was almost undetectable, suggesting that the adipocytes were the major source of the collagens. Furthermore, the adipose tissue macrophages from Lplm−/m−;Lepob/ob mice showed increased expression of genes related to fibrosis and inflammation. In addition, we determined Lplm−/m−;Lepob/ob mice were more hypertriglyceridemic than Lepob/ob mice. Lplm−/m−;Lepob/ob mice also showed slower weight gain than Lepob/ob mice, which was primarily due to reduced food intake. In conclusion, we discovered that the loss of myeloid Lpl led to extensive fibrosis of perigonadal WAT and hypertriglyceridemia. In addition to illustrating an important role of macrophage LPL in regulation of circulating triglyceride levels, these data show that macrophage LPL protects against fibrosis in obese adipose tissues.
Collapse
Affiliation(s)
- Manabu Takahashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan.
| | - Daisuke Yamamuro
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Tetsuji Wakabayashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Akihito Takei
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Shoko Takei
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Shuichi Nagashima
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Hiroaki Okazaki
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Ken Ebihara
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Hiroaki Yagyu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Yuki Takayanagi
- Division of Brain and Neurophysiology, Department of Physiology, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Tatsushi Onaka
- Division of Brain and Neurophysiology, Department of Physiology, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan
| | - Ira J Goldberg
- NYU-Langone Medical Center, 435 East 30(th) Street, SB617, New York, NY, 10016
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Jichi Medical University, Tochigi, 329-0498, Japan.
| |
Collapse
|
19
|
Liang W, Qi Y, Yi H, Mao C, Meng Q, Wang H, Zheng C. The Roles of Adipose Tissue Macrophages in Human Disease. Front Immunol 2022; 13:908749. [PMID: 35757707 PMCID: PMC9222901 DOI: 10.3389/fimmu.2022.908749] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/12/2022] [Indexed: 01/02/2023] Open
Abstract
Macrophages are a population of immune cells functioning in antigen presentation and inflammatory response. Research has demonstrated that macrophages belong to a cell lineage with strong plasticity and heterogeneity and can be polarized into different phenotypes under different microenvironments or stimuli. Many macrophages can be recruited by various cytokines secreted by adipose tissue. The recruited macrophages further secrete various inflammatory factors to act on adipocytes, and the interaction between the two leads to chronic inflammation. Previous studies have indicated that adipose tissue macrophages (ATMs) are closely related to metabolic diseases like obesity and diabetes. Here, we will not only conclude the current progress of factors affecting the polarization of adipose tissue macrophages but also elucidate the relationship between ATMs and human diseases. Furthermore, we will highlight its potential in preventing and treating metabolic diseases as immunotherapy targets.
Collapse
Affiliation(s)
- Weizheng Liang
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China.,Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yanxu Qi
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Hongyang Yi
- National Clinical Research Centre for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Chenyu Mao
- School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Qingxue Meng
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Hao Wang
- Shenzhen Key Laboratory, Shenzhen University General Hospital, Shenzhen, China.,Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, China
| | - Chunfu Zheng
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
20
|
Sarsenbayeva A, Pereira MJ, Nandi Jui B, Ahmed F, Dipta P, Fanni G, Almby K, Kristófi R, Hetty S, Eriksson JW. Excess glucocorticoid exposure contributes to adipose tissue fibrosis which involves macrophage interaction with adipose precursor cells. Biochem Pharmacol 2022; 198:114976. [PMID: 35202577 DOI: 10.1016/j.bcp.2022.114976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/17/2022] [Accepted: 02/17/2022] [Indexed: 12/12/2022]
Abstract
Chronic exposure to elevated glucocorticoid levels, as seen in patients with Cushing's syndrome, can induce adipose tissue fibrosis. Macrophages play a pivotal role in adipose tissue remodelling. We used the synthetic glucocorticoid analogue dexamethasone to address glucocorticoid effects on adipose tissue fibrosis, in particular involving macrophage to preadipocyte communication. We analysed the direct effects of dexamethasone at a supra-physiological level, 0.3 µM, on gene expression of pro-fibrotic markers in human subcutaneous adipose tissue. The effects of dexamethasone on the differentiation of human SGBS preadipocytes were assessed in the presence or absence of THP1-macrophages or macrophage-conditioned medium. We measured the expression of different pro-fibrotic factors, including α-smooth muscle actin gene (ACTA2) and protein (α-SMA). Dexamethasone increased the expression of pro-fibrotic genes, e.g. CTGF, COL6A3, FN1, in adipose tissue. Macrophages abolished preadipocyte differentiation and increased the expression of the ACTA2 gene and α-SMA protein in preadipocytes after differentiation. Exposure to dexamethasone during differentiation reduced adipogenesis in preadipocytes, and elevated the expression of pro-fibrotic genes. Moreover, dexamethasone added together with macrophages further increased ACTA2 and α-SMA expression in preadipocytes, making them more myofibroblast-like. Cells differentiated in the presence of conditioned media from macrophages pretreated with or without dexamethasone had a higher expression of profibrotic genes compared to control cells. Our data suggest that macrophages promote adipose tissue fibrosis by directly interfering with preadipocyte differentiation and stimulating gene expression of pro-fibrotic factors. Excess glucocorticoid exposure also has pro-fibrotic effect on adipose tissue, but this requires the presence of macrophages.
Collapse
Affiliation(s)
- Assel Sarsenbayeva
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Maria J Pereira
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Bipasha Nandi Jui
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Fozia Ahmed
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Priya Dipta
- Department of Pharmacology, Faculty of Medicine, Hadassah Medical Centre, Jerusalem, Israel
| | - Giovanni Fanni
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Kristina Almby
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Robin Kristófi
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Susanne Hetty
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Jan W Eriksson
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
21
|
Simas AM, Kramer CD, Genco CA. Diet-Induced Non-alcoholic Fatty Liver Disease and Associated Gut Dysbiosis Are Exacerbated by Oral Infection. FRONTIERS IN ORAL HEALTH 2022; 2:784448. [PMID: 35141703 PMCID: PMC8820505 DOI: 10.3389/froh.2021.784448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/29/2021] [Indexed: 11/13/2022] Open
Abstract
Increasing evidence indicates that chronic inflammation due to periodontal disease is associated with progression of non-alcoholic fatty liver disease (NAFLD) caused by a Western diet. NAFLD has also been associated with oral infection with the etiological agent of periodontal disease, Porphyromonas gingivalis. P. gingivalis oral infection has been shown to induce cardiometabolic disease features including hepatic lipid accumulation while also leading to dysbiosis of the gut microbiome. However, the impact of P. gingivalis infection on the gut microbiota of mice with diet-induced NAFLD and the potential for those changes to mediate NAFLD progression has yet to be determined. In the current study, we have demonstrated that P. gingivalis infection induced sustained alterations of the gut microbiota composition and predicted functions, which was associated with the promotion of NAFLD in steatotic mice. Reduced abundance of short-chain fatty acid-producing microbiota was observed after both acute and chronic P. gingivalis infection. Collectively, our findings demonstrate that P. gingivalis infection produces a persistent change in the gut microbiota composition and predicted functions that promotes steatosis and metabolic disease.
Collapse
Affiliation(s)
- Alexandra M. Simas
- Gerald J. and Dorothy R. Friedman School of Nutrition and Science Policy, Graduate Program in Biochemical and Molecular Nutrition, Tufts University, Boston, MA, United States
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
| | - Carolyn D. Kramer
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
| | - Caroline A. Genco
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
- Graduate Program in Immunology, Graduate School of Biomedical Sciences, Tufts University, Boston, MA, United States
- Graduate Program in Molecular Microbiology, Graduate School of Biomedical Sciences, Tufts University, Boston, MA, United States
- *Correspondence: Caroline A. Genco
| |
Collapse
|
22
|
Castro BBA, Foresto-Neto O, Saraiva-Camara NO, Sanders-Pinheiro H. Renal lipotoxicity: Insights from experimental models. Clin Exp Pharmacol Physiol 2021; 48:1579-1588. [PMID: 34314523 DOI: 10.1111/1440-1681.13556] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 07/13/2021] [Accepted: 07/23/2021] [Indexed: 11/29/2022]
Abstract
In recent decades, there has been a progressive increase in the prevalence of obesity and chronic kidney disease. Renal lipotoxicity has been associated with obesity. Although lipids play fundamental physiological roles, the accumulation of lipids in kidney cells may cause dysfunction and/or renal fibrosis. Adipose tissue that exceeds their lipid storage capacity begins to release triglycerides into the bloodstream that can get stored in several organs, including the kidneys. The mechanisms underlying renal lipotoxicity involve intracellular lipid accumulation and organelle dysfunction, which trigger oxidative stress and inflammation that consequently result in insulin resistance and albuminuria. However, the specific pathways involved in renal lipotoxicity have not yet been fully understood. We aimed to summarize the current knowledge on the mechanisms by which lipotoxicity affects the renal morphology and function in experimental models of obesity. The accumulation of fatty acids in tubular cells has been described as the main mechanism of lipotoxicity; however, lipids and their metabolism also affect the function and the survival of podocytes. In this review, we presented indication of mitochondrial, lysosomal and endoplasmic reticulum alterations involved in kidney damage caused by obesity. The kidney is vulnerable to lipotoxicity, and studies of the mechanisms underlying renal injury caused by obesity can help identify therapeutic targets to control renal dysfunction.
Collapse
Affiliation(s)
- Barbara Bruna Abreu Castro
- Laboratory of Experimental Nephrology, Nucleus of Animal Experimentation (NIDEAL), Centre of Reproductive Biology (CBR), Federal University of Juiz de Fora (UFJF, Juiz de Fora, Minas Gerais, Brazil
- Nephrology Division and Interdisciplinary Nucleus of Studies and Research in Nephrology (NIEPEN), Federal University of Juiz de Fora (UFJF, Juiz de Fora, Minas Gerais, Brazil
| | - Orestes Foresto-Neto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP, São Paulo, São Paulo, Brazil
| | - Niels Olsen Saraiva-Camara
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP, São Paulo, São Paulo, Brazil
| | - Helady Sanders-Pinheiro
- Laboratory of Experimental Nephrology, Nucleus of Animal Experimentation (NIDEAL), Centre of Reproductive Biology (CBR), Federal University of Juiz de Fora (UFJF, Juiz de Fora, Minas Gerais, Brazil
- Nephrology Division and Interdisciplinary Nucleus of Studies and Research in Nephrology (NIEPEN), Federal University of Juiz de Fora (UFJF, Juiz de Fora, Minas Gerais, Brazil
| |
Collapse
|
23
|
Sarsenbayeva A, Jui BN, Fanni G, Barbosa P, Ahmed F, Kristófi R, Cen J, Chowdhury A, Skrtic S, Bergsten P, Fall T, Eriksson JW, Pereira MJ. Impaired HMG-CoA Reductase Activity Caused by Genetic Variants or Statin Exposure: Impact on Human Adipose Tissue, β-Cells and Metabolome. Metabolites 2021; 11:574. [PMID: 34564389 PMCID: PMC8468287 DOI: 10.3390/metabo11090574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 11/20/2022] Open
Abstract
Inhibition of 3-hydroxy-3-methyl-glutaryl-CoA (HMG-CoA) reductase is associated with an increased risk of new-onset type 2 diabetes. We studied the association of genetic or pharmacological HMG-CoA reductase inhibition with plasma and adipose tissue (AT) metabolome and AT metabolic pathways. We also investigated the effects of statin-mediated pharmacological inhibition of HMG-CoA reductase on systemic insulin sensitivity by measuring the HOMA-IR index in subjects with or without statin therapy. The direct effects of simvastatin (20-250 nM) or its active metabolite simvastatin hydroxy acid (SA) (8-30 nM) were investigated on human adipocyte glucose uptake, lipolysis, and differentiation and pancreatic insulin secretion. We observed that the LDL-lowering HMGCR rs12916-T allele was negatively associated with plasma phosphatidylcholines and sphingomyelins, and HMGCR expression in AT was correlated with various metabolic and mitochondrial pathways. Clinical data showed that statin treatment was associated with HOMA-IR index after adjustment for age, sex, BMI, HbA1c, LDL-c levels, and diabetes status in the subjects. Supra-therapeutic concentrations of simvastatin reduced glucose uptake in adipocytes and normalized fatty acid-induced insulin hypersecretion from β-cells. Our data suggest that inhibition of HMG-CoA reductase is associated with insulin resistance. However, statins have a very mild direct effect on AT and pancreas, hence, other tissues as the liver or muscle appear to be of greater importance.
Collapse
Affiliation(s)
- Assel Sarsenbayeva
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden; (A.S.); (B.N.J.); (G.F.); (F.A.); (R.K.); (T.F.); (J.W.E.)
| | - Bipasha Nandi Jui
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden; (A.S.); (B.N.J.); (G.F.); (F.A.); (R.K.); (T.F.); (J.W.E.)
| | - Giovanni Fanni
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden; (A.S.); (B.N.J.); (G.F.); (F.A.); (R.K.); (T.F.); (J.W.E.)
| | - Pedro Barbosa
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal;
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Fozia Ahmed
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden; (A.S.); (B.N.J.); (G.F.); (F.A.); (R.K.); (T.F.); (J.W.E.)
| | - Robin Kristófi
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden; (A.S.); (B.N.J.); (G.F.); (F.A.); (R.K.); (T.F.); (J.W.E.)
| | - Jing Cen
- Department of Medical Cell Biology, Uppsala University, 751 85 Uppsala, Sweden; (J.C.); (A.C.); (P.B.)
| | - Azazul Chowdhury
- Department of Medical Cell Biology, Uppsala University, 751 85 Uppsala, Sweden; (J.C.); (A.C.); (P.B.)
| | - Stanko Skrtic
- Innovation Strategies & External Liaison, Pharmaceutical Technologies & Development, AstraZeneca, 431 83 Gothenburg, Sweden;
- Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Peter Bergsten
- Department of Medical Cell Biology, Uppsala University, 751 85 Uppsala, Sweden; (J.C.); (A.C.); (P.B.)
| | - Tove Fall
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden; (A.S.); (B.N.J.); (G.F.); (F.A.); (R.K.); (T.F.); (J.W.E.)
| | - Jan W. Eriksson
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden; (A.S.); (B.N.J.); (G.F.); (F.A.); (R.K.); (T.F.); (J.W.E.)
| | - Maria J. Pereira
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, 751 85 Uppsala, Sweden; (A.S.); (B.N.J.); (G.F.); (F.A.); (R.K.); (T.F.); (J.W.E.)
| |
Collapse
|
24
|
Yu Q, Xu T, Ding F, Ding Z, Lin R. Decreased infiltration of adipose tissue macrophages and amplified inflammation of adipose tissue in obese mice with severe acute pancreatitis. Pancreatology 2021; 21:S1424-3903(21)00156-3. [PMID: 34088592 DOI: 10.1016/j.pan.2021.05.005] [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: 08/17/2020] [Revised: 04/13/2021] [Accepted: 05/07/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Macrophages are involved in obesity-associated inflammation and severe acute pancreatitis (SAP) development. However, the role of adipose tissue macrophages (ATMs) in obesity-related SAP has not been fully elucidated. We investigated the relationship between ATMs and inflammatory responses in SAP model mice fed a high-fat diet (HFD). METHODS SAP was induced in animal models via intraperitoneal injections of caerulein and lipopolysaccharide (LPS). SAP severity was evaluated, both morphologically and biochemically, and macrophage infiltration in the pancreas and epididymal adipose tissue was measured. We also analyzed apoptosis levels, polarization of the ATMs, and expression of inflammatory mediators in epididymal adipose tissue. RESULTS Obesity increased disease severity in SAP animals. Increased macrophage infiltration in the pancreas induced by SAP was found in both normal diet (ND)- and HFD-fed mice. Total ATM infiltration in epididymal adipose tissue was elevated by HFD, while a significant decrease in infiltration was observed in both the ND + SAP and HFD + SAP groups. The apoptosis levels of ATMs were reduced in the HFD group, but were markedly enhanced in both the ND + SAP and HFD + SAP groups compared to their respective control groups. Higher levels of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and monocyte chemoattractant protein-1 (MCP-1) were observed in the HFD + SAP than in the ND + SAP group. Increased proportion of M1 type ATMs was induced by both HFD and SAP. CONCLUSIONS Total ATM infiltration was decreased in epididymal adipose tissue of SAP animals. ATM polarization to the M1 type resulted in an amplified inflammatory response in obese mice with SAP.
Collapse
Affiliation(s)
- Qiao Yu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tao Xu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Feifei Ding
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, 730030, China
| | - Zhen Ding
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Rong Lin
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
25
|
Lima RS, Mattos RT, Medeiros NI, Kattah FM, Nascimento JRS, Menezes CA, Rios-Santos F, Dutra WO, Gomes JAS, Moreira PR. CXCL8 expression and methylation are correlated with anthropometric and metabolic parameters in childhood obesity. Cytokine 2021; 143:155538. [PMID: 33926776 DOI: 10.1016/j.cyto.2021.155538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 03/02/2021] [Accepted: 04/05/2021] [Indexed: 02/07/2023]
Abstract
Childhood obesity is a global and increasing health issue. Inflammation and dysregulated adipose tissue secretion are common findings in obesity and have been related to poor metabolic function. Given that DNA methylation impacts gene expression and is responsive to environmental changes, we aimed, in addition to characterize the patients in anthropometric and biochemical terms, to determine the expression of cytokines and adipokines, assess the methylation on regulatory regions of the genes that code for these molecules, and investigate the association of the expression and gene methylation with anthropometric and biochemical parameters in childhood obesity. Obese children present dyslipidemia, dysregulated serum levels of adipokines and their ratios, altered leukocytic expression of cytokines, and higher methylation at the CXCL8 promoter as compared to the control group. However, no significant results were observed in the fasting plasma glucose levels or the methylation of TGFB1, LEP, and the enhancer region of ADIPOQ. We also found negative correlations of CXCL8 expression with anthropometric and biochemical parameters, and positive correlation of CXCL8 promoter methylation and the serum levels of hepatic enzymes. Our results indicate that changes in metabolic parameters observed in childhood obesity are associated with the expression of adipokines and cytokines, and the methylation status at the CXCL8 promoter. CXCL8 may be a key factor for these alterations, as it correlates with many of the parameters assessed in the present study.
Collapse
Affiliation(s)
- Rafael S Lima
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Rafael T Mattos
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Nayara I Medeiros
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Fabiana M Kattah
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Julya R S Nascimento
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Carlos A Menezes
- Department of Genetics, State University of Santa Cruz, Bahia, Brazil; Service of Preventive Medicine - Unimed, Sergipe, Brazil
| | - Fabricio Rios-Santos
- Department of Basic and Health Sciences, Federal University of Mato Grosso, Mato Grosso, Brazil
| | - Walderez O Dutra
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil; National Institute of Science and Technology in Tropical Diseases - INCT-DT, Brazil
| | - Juliana A S Gomes
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Paula R Moreira
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil.
| |
Collapse
|
26
|
Abstract
On this 100th anniversary of the discovery of insulin, we recognize the critical role that adipocytes, which are exquisitely responsive to insulin, have played in determining the mechanisms for insulin action at the cellular level. Our understanding of adipose tissue biology has evolved greatly, and it is now clear that adipocytes are far more complicated than simple storage depots for fat. A growing body of evidence documents how adipocytes, in response to insulin, contribute to the control of whole-body nutrient homeostasis. These advances highlight adipocyte plasticity, heterogeneity, and endocrine function, unique features that connect adipocyte metabolism to the regulation of other tissues important for metabolic homeostasis (e.g., liver, muscle, pancreas).
Collapse
Affiliation(s)
- Anna Santoro
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Timothy E McGraw
- Department of Biochemistry, Weill Medical College of Cornell University, New York, NY 10065, USA.
| | - Barbara B Kahn
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
27
|
Early Childhood Fat Tissue Changes-Adipocyte Morphometry, Collagen Deposition, and Expression of CD163 + Cells in Subcutaneous and Visceral Adipose Tissue of Male Children. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18073627. [PMID: 33807325 PMCID: PMC8037722 DOI: 10.3390/ijerph18073627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/06/2021] [Accepted: 03/26/2021] [Indexed: 01/14/2023]
Abstract
Childhood obesity is a complex health problem, and not many studies have been done on adipose tissue remodeling in early childhood. The aim of this study was to examine extracellular matrix remodeling in the adipose tissue of healthy male children depending on their weight status. Subcutaneous and visceral adipose tissue was obtained from 45 otherwise healthy male children who underwent elective surgery for hernia repairs or orchidopexy. The children were divided into overweight/obese (n = 17) or normal weight groups (n = 28) depending on their body mass index (BMI) z-score. Serum was obtained for glucose, testosterone, triglyceride, total cholesterol, high-density lipoprotein (HDL), and low-density lipoprotein (LDL) measurements. Sections of adipose tissue were stained with hematoxylin and eosin to determine the adipocytes' surface area, and Masson's trichrome stain was used to detect the adipocytes' collagen content. Immunohistochemistry for CD163+ cells was also performed. The results showed that male children in the overweight group had higher serum triglyceride levels, greater adipocyte surface area and collagen content in their subcutaneous adipose tissue, more crown-like structures in fat tissues, and more CD163+ cells in their visceral adipose tissue than males in the normal weight group. In conclusion, in male children, obesity can lead to the hypertrophy of adipocytes, increased collagen deposition in subcutaneous adipose tissues, and changes in the polarization and accumulation of macrophages.
Collapse
|
28
|
Ruggiero AD, Key CCC, Kavanagh K. Adipose Tissue Macrophage Polarization in Healthy and Unhealthy Obesity. Front Nutr 2021; 8:625331. [PMID: 33681276 PMCID: PMC7925825 DOI: 10.3389/fnut.2021.625331] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/05/2021] [Indexed: 12/12/2022] Open
Abstract
Over 650 million adults are obese (body mass index ≥ 30 kg/m2) worldwide. Obesity is commonly associated with several comorbidities, including cardiovascular disease and type II diabetes. However, compiled estimates suggest that from 5 to 40% of obese individuals do not experience metabolic or cardiovascular complications. The existence of the metabolically unhealthy obese (MUO) and the metabolically healthy obese (MHO) phenotypes suggests that underlying differences exist in both tissues and overall systemic function. Macrophage accumulation in white adipose tissue (AT) in obesity is typically associated with insulin resistance. However, as plastic cells, macrophages respond to stimuli in their microenvironments, altering their polarization between pro- and anti-inflammatory phenotypes, depending on the state of their surroundings. The dichotomous nature of MHO and MUO clinical phenotypes suggests that differences in white AT function dictate local inflammatory responses by driving changes in macrophage subtypes. As obesity requires extensive AT expansion, we posit that remodeling capacity with adipose expansion potentiates favorable macrophage profiles in MHO as compared with MUO individuals. In this review, we discuss how differences in adipogenesis, AT extracellular matrix deposition and breakdown, and AT angiogenesis perpetuate altered AT macrophage profiles in MUO compared with MHO. We discuss how non-autonomous effects of remote organ systems, including the liver, gastrointestinal tract, and cardiovascular system, interact with white adipose favorably in MHO. Preferential AT macrophage profiles in MHO stem from sustained AT function and improved overall fitness and systemic health.
Collapse
Affiliation(s)
- Alistaire D Ruggiero
- Section on Comparative Medicine, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Chia-Chi Chuang Key
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Kylie Kavanagh
- Section on Comparative Medicine, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, United States.,Department of Biomedicine, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
29
|
Maternal vitamin D deficiency increases the risk of obesity in male offspring mice by affecting the immune response. Nutrition 2021; 87-88:111191. [PMID: 33744641 DOI: 10.1016/j.nut.2021.111191] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 01/08/2021] [Accepted: 02/03/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Recently, many epidemiologic and animal studies have indicated that obesity has its origin in early stages of life, including the inappropriate balance of some nutrients. So the objectives of this study were to determine the risk of obesity in male offspring mice as a consequence of maternal vitamin D (VD) deficiency mediating the disordered immune response. METHODS C57BL/6J female mice 4 wk old were fed VD-deficient or normal reproductive diets during pregnancy and lactation. Their male offspring were given control and high-fat diets for 16 wk after weaning and then weighed and euthanized. The serum was collected for biochemical analyses. Epididymal (eWAT) and inguinal white adipose tissue (iWAT) were excised for histologic examination, immunohistochemistry, gene expression of inflammatory factors, and determination by flow cytometry of the proportions of immune cells. RESULTS Insufficient maternal VD intake exacerbated the development of obesity in male offspring mice that were both obese and non-obese, as evidenced by larger adipose cells and abnormal glucose and lipid metabolisms. Also, the expressions of proinflammatory cytokines were increased and that of anti-inflammatory cytokines was decreased in eWAT and/or iWAT in the maternal VD-deficient group, accompanied by higher levels of tumor necrosis factor-α and/or interferon-γ and lower levels of interleukin-4 and interleukin-10. Insufficient maternal VD intake was also observed to induce a shift in the profiles of immune cells in the eWAT and/or iWAT of male offspring that were both obese and non-obese, resulting in increased percentages of M1 macrophages, adipose tissue dendritic cells, and CD4+ and CD8+ T cells but a significant decrease in the percentages of M2 macrophages. All these changes in the immune cell profile were more obvious in the eWAT than those in the iWAT. CONCLUSIONS Maternal VD deficiency might promote the development of obesity in male offspring mice partly by modulating the immune cell populations and causing a polarization in the adipose depots.
Collapse
|
30
|
Woo J, Koziol-White C, Panettieri R, Jude J. TGF-β: The missing link in obesity-associated airway diseases? CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100016. [PMID: 34909651 PMCID: PMC8663968 DOI: 10.1016/j.crphar.2021.100016] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/15/2021] [Accepted: 01/20/2021] [Indexed: 01/19/2023] Open
Abstract
Obesity is emerging as a global public health epidemic. The co-morbidities associated with obesity significantly contribute to reduced quality of life, mortality, and global healthcare burden. Compared to other asthma comorbidities, obesity prominently engenders susceptibility to inflammatory airway diseases such as asthma and chronic obstructive pulmonary disease (COPD), contributes to greater disease severity and evokes insensitivity to current therapies. Unlike in other metabolic diseases associated with obesity, the mechanistic link between obesity and airway diseases is only poorly defined. Transforming growth factor-β (TGF-β) is a pleiotropic inflammatory cytokine belonging to a family of growth factors with pivotal roles in asthma. In this review, we summarize the role of TGF-β in major obesity-associated co-morbidities to shed light on mechanisms of the diseases. Literature evidence shows that TGF-β mechanistically links many co-morbidities with obesity through its profibrotic, remodeling, and proinflammatory functions. We posit that TGF-β plays a similar mechanistic role in obesity-associated inflammatory airway diseases such as asthma and COPD. Concerning the role of TGF-β on metabolic effects of obesity, we posit that TGF-β has a similar mechanistic role in obesity-associated inflammatory airway diseases in interplay with different comorbidities such as hypertension, metabolic diseases like type 2 diabetes, and cardiomyopathies. Future studies in TGF-β-dependent mechanisms in obesity-associated inflammatory airway diseases will advance our understanding of obesity-induced asthma and help find novel therapeutic targets for prevention and treatment.
Collapse
Affiliation(s)
- Joanna Woo
- Rutgers Institute for Translational Medicine & Science, The State University of New Jersey, 89 French Street, Rutgers, 160 Frelinghuysen Road, Piscataway, NJ08854, United States,Ernest Mario School of Pharmacy, The State University of New Jersey, 89 French Street, Rutgers, 160 Frelinghuysen Road, Piscataway, NJ08854, United States
| | - Cynthia Koziol-White
- Rutgers Institute for Translational Medicine & Science, The State University of New Jersey, 89 French Street, Rutgers, 160 Frelinghuysen Road, Piscataway, NJ08854, United States,Robert Wood Johnson Medical School, The State University of New Jersey, 89 French Street, Rutgers, 160 Frelinghuysen Road, Piscataway, NJ08854, United States
| | - Reynold Panettieri
- Rutgers Institute for Translational Medicine & Science, The State University of New Jersey, 89 French Street, Rutgers, 160 Frelinghuysen Road, Piscataway, NJ08854, United States,Robert Wood Johnson Medical School, The State University of New Jersey, 89 French Street, Rutgers, 160 Frelinghuysen Road, Piscataway, NJ08854, United States,Ernest Mario School of Pharmacy, The State University of New Jersey, 89 French Street, Rutgers, 160 Frelinghuysen Road, Piscataway, NJ08854, United States
| | - Joseph Jude
- Rutgers Institute for Translational Medicine & Science, The State University of New Jersey, 89 French Street, Rutgers, 160 Frelinghuysen Road, Piscataway, NJ08854, United States,Robert Wood Johnson Medical School, The State University of New Jersey, 89 French Street, Rutgers, 160 Frelinghuysen Road, Piscataway, NJ08854, United States,Ernest Mario School of Pharmacy, The State University of New Jersey, 89 French Street, Rutgers, 160 Frelinghuysen Road, Piscataway, NJ08854, United States,Corresponding author. Rutgers Institute for Translational Medicine & Science, Rm# 4276, 89 French Street, New Brunswick, NJ08901, United States.
| |
Collapse
|
31
|
Komatsu Y, Aoyama K, Yoneda M, Ashikawa S, Nakano S, Kawai Y, Cui X, Furukawa N, Ikeda K, Nagata K. The prebiotic fiber inulin ameliorates cardiac, adipose tissue, and hepatic pathology, but exacerbates hypertriglyceridemia in rats with metabolic syndrome. Am J Physiol Heart Circ Physiol 2021; 320:H281-H295. [PMID: 33216624 DOI: 10.1152/ajpheart.00657.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/18/2020] [Indexed: 01/17/2023]
Abstract
Prebiotics ameliorate dysbiosis and influence metabolism and the immune system, but their effects on cardiovascular complications in metabolic disorders remain largely unknown. We here investigated the effects of the soluble fiber inulin on cardiac, adipose tissue, and hepatic pathology as well as on metabolic disorders in DahlS.Z-Leprfa/Leprfa (DS/obese) rats, an animal model of metabolic syndrome (MetS). DS/obese rats and their homozygous lean (DahlS.Z-Lepr+/Lepr+, or DS/lean) littermate controls were fed a purified diet containing 5% or 20% inulin from 9 to 13 wk of age. The high-fiber diet ameliorated hypertension, left ventricular inflammation, fibrosis and diastolic dysfunction; attenuated adipose tissue inflammation and fibrosis; and alleviated the elevation of interleukin-6 levels, without affecting insulin resistance, in DS/obese rats. In addition, high fiber intake ameliorated lipid accumulation, inflammation, and fibrosis; attenuated the reduction in AMPK activity; upregulated sterol regulatory element-binding protein-1c gene expression; and increased the expression of microsomal triglyceride transfer protein gene in the liver of DS/obese rats. It also mitigated increases in total and non-high-density lipoprotein cholesterol levels but increased the triglyceride concentration in serum in these rats. None of these parameters were affected by high dietary fiber in DS/lean rats. The proportion of regulatory T cells in adipose tissue was influenced by dietary fiber but not by genotype. Our results indicate that inulin exacerbates hypertriglyceridemia but alleviates hypertension and cardiac injury as well as adipose tissue and hepatic pathology in MetS rats.NEW & NOTEWORTHY Prebiotics ameliorate dysbiosis and influence metabolism and the immune system, but their effects on cardiovascular complications in metabolic disorders remain largely unknown. Inulin ameliorated hypertension, cardiac injury, and diastolic dysfunction without affecting obesity or insulin resistance in a rat model of metabolic syndrome. The favorable cardiac effects of inulin may be related to inhibition of systemic inflammation associated with a reduction in circulating interleukin-6 levels. Additionally, inulin exacerbated hypertriglyceridemia but alleviates adipose tissue and hepatic pathology in these animals, as well as increased the number of regulatory T cells in adipose tissue.
Collapse
Affiliation(s)
- Yuki Komatsu
- Pathophysiology Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kiyoshi Aoyama
- Pathophysiology Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mamoru Yoneda
- Pathophysiology Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sao Ashikawa
- Pathophysiology Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shiho Nakano
- Pathophysiology Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yumeno Kawai
- Pathophysiology Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Xixi Cui
- Pathophysiology Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nozomi Furukawa
- Pathophysiology Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Katsuhide Ikeda
- Pathophysiology Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kohzo Nagata
- Pathophysiology Sciences, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
32
|
Kalupahana NS, Goonapienuwala BL, Moustaid-Moussa N. Omega-3 Fatty Acids and Adipose Tissue: Inflammation and Browning. Annu Rev Nutr 2020; 40:25-49. [DOI: 10.1146/annurev-nutr-122319-034142] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
White adipose tissue (WAT) and brown adipose tissue (BAT) are involved in whole-body energy homeostasis and metabolic regulation. Changes to mass and function of these tissues impact glucose homeostasis and whole-body energy balance during development of obesity, weight loss, and subsequent weight regain. Omega-3 polyunsaturated fatty acids (ω-3 PUFAs), which have known hypotriglyceridemic and cardioprotective effects, can also impact WAT and BAT function. In rodent models, these fatty acids alleviate obesity-associated WAT inflammation, improve energy metabolism, and increase thermogenic markers in BAT. Emerging evidence suggests that ω-3 PUFAs can also modulate gut microbiota impacting WAT function and adiposity. This review discusses molecular mechanisms, implications of these findings, translation to humans, and future work, especially with reference to the potential of these fatty acids in weight loss maintenance.
Collapse
Affiliation(s)
- Nishan Sudheera Kalupahana
- Department of Physiology, Faculty of Medicine, University of Peradeniya, Peradeniya, 20400, Sri Lanka
- Department of Nutritional Sciences and Obesity Research Institute, Texas Tech University, Lubbock, Texas 79409-1270, USA;,
| | - Bimba Lakmini Goonapienuwala
- Department of Nutritional Sciences and Obesity Research Institute, Texas Tech University, Lubbock, Texas 79409-1270, USA;,
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences and Obesity Research Institute, Texas Tech University, Lubbock, Texas 79409-1270, USA;,
| |
Collapse
|
33
|
Abstract
Obesity is becoming an epidemic in the United States and worldwide and increases risk for many diseases, particularly insulin resistance, type 2 diabetes mellitus, and cardiovascular disease. The mechanisms linking obesity with these diseases remain incompletely understood. Over the past 2 to 3 decades, it has been recognized that in obesity, inflammation, with increased accumulation and inflammatory polarization of immune cells, takes place in various tissues, including adipose tissue, skeletal muscle, liver, gut, pancreatic islet, and brain and may contribute to obesity-linked metabolic dysfunctions, leading to insulin resistance and type 2 diabetes mellitus. Therapies targeting inflammation have shed light on certain obesity-linked diseases, including type 2 diabetes mellitus and atherosclerotic cardiovascular disease, but remain to be tested further and confirmed in clinical trials. This review focuses on inflammation in adipose tissue and its potential role in insulin resistance associated with obesity.
Collapse
Affiliation(s)
- Huaizhu Wu
- From the Department of Medicine (H.W., C.M.B.), Baylor College of Medicine, Houston, TX.,Department of Pediatrics (H.W.), Baylor College of Medicine, Houston, TX
| | - Christie M Ballantyne
- From the Department of Medicine (H.W., C.M.B.), Baylor College of Medicine, Houston, TX.,Department of Molecular and Human Genetics (C.M.B.), Baylor College of Medicine, Houston, TX.,Center for Cardiometabolic Disease Prevention (C.M.B.), Baylor College of Medicine, Houston, TX
| |
Collapse
|
34
|
Zhao XY, Zhou L, Chen Z, Ji Y, Peng X, Qi L, Li S, Lin JD. The obesity-induced adipokine sST2 exacerbates adipose T reg and ILC2 depletion and promotes insulin resistance. SCIENCE ADVANCES 2020; 6:eaay6191. [PMID: 32426492 PMCID: PMC7220368 DOI: 10.1126/sciadv.aay6191] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 03/02/2020] [Indexed: 05/02/2023]
Abstract
Depletion of fat-resident regulatory T cells (Tregs) and group 2 innate lymphoid cells (ILC2s) has been causally linked to obesity-associated insulin resistance. However, the molecular nature of the pathogenic signals suppress adipose Tregs and ILC2s in obesity remains unknown. Here, we identified the soluble isoform of interleukin (IL)-33 receptor ST2 (sST2) as an obesity-induced adipokine that attenuates IL-33 signaling and disrupts Treg/ILC2 homeostasis in adipose tissue, thereby exacerbates obesity-associated insulin resistance in mice. We demonstrated sST2 is a target of TNFα signaling in adipocytes that is countered by Zbtb7b. Fat-specific ablation of Zbtb7b augments adipose sST2 gene expression, leading to diminished fat-resident Tregs/ILC2s, more pronounced adipose tissue inflammation and fibrosis, and impaired glucose homeostasis in mice. Mechanistically, Zbtb7b suppresses NF-κB activation in response to TNFα through destabilizing IκBα. These findings uncover an adipokine-immune signaling pathway that is engaged in obesity to drive the pathological changes of the immunometabolic landscape.
Collapse
Affiliation(s)
- Xu-Yun Zhao
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
- Corresponding author. (J.D.L.); (X.-Y.Z.)
| | - Linkang Zhou
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Zhimin Chen
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Yewei Ji
- Department of Molecular and Integrative Physiology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Xiaoling Peng
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Ling Qi
- Department of Molecular and Integrative Physiology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Siming Li
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Jiandie D. Lin
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
- Corresponding author. (J.D.L.); (X.-Y.Z.)
| |
Collapse
|
35
|
He B, Wang X, Jin X, Xue Z, Ni Y, Zhu J, Wang C, Jin Y, Fu Z. β‐Cypermethrin
Alleviated the Inhibitory Effect of Medium from
RAW
264.7 Cells on
3T3‐L1
Cell Maturation into Adipocytes. Lipids 2020; 55:251-260. [DOI: 10.1002/lipd.12234] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/29/2020] [Accepted: 03/08/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Bingnan He
- College of Biotechnology and BioengineeringZhejiang University of Technology Hangzhou Zhejiang 310032 China
| | - Xia Wang
- College of Biotechnology and BioengineeringZhejiang University of Technology Hangzhou Zhejiang 310032 China
| | - Xini Jin
- College of Biotechnology and BioengineeringZhejiang University of Technology Hangzhou Zhejiang 310032 China
| | - Zimeng Xue
- College of Biotechnology and BioengineeringZhejiang University of Technology Hangzhou Zhejiang 310032 China
| | - Yinhua Ni
- College of Biotechnology and BioengineeringZhejiang University of Technology Hangzhou Zhejiang 310032 China
| | - Jianbo Zhu
- College of Biotechnology and BioengineeringZhejiang University of Technology Hangzhou Zhejiang 310032 China
| | - Caiyun Wang
- College of Biotechnology and BioengineeringZhejiang University of Technology Hangzhou Zhejiang 310032 China
| | - Yuanxiang Jin
- College of Biotechnology and BioengineeringZhejiang University of Technology Hangzhou Zhejiang 310032 China
| | - Zhengwei Fu
- College of Biotechnology and BioengineeringZhejiang University of Technology Hangzhou Zhejiang 310032 China
| |
Collapse
|
36
|
Obesity Promotes Cooperation of Cancer Stem-Like Cells and Macrophages to Enhance Mammary Tumor Angiogenesis. Cancers (Basel) 2020; 12:cancers12020502. [PMID: 32098183 PMCID: PMC7072330 DOI: 10.3390/cancers12020502] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/13/2020] [Accepted: 02/19/2020] [Indexed: 02/06/2023] Open
Abstract
Obesity is correlated with worsened prognosis and treatment resistance in breast cancer. Macrophage-targeted therapies are currently in clinical trials, however, little is known about how obesity may impact treatment efficacy. Within breast adipose tissue, obesity leads to chronic, macrophage-driven inflammation, suggesting that obese breast cancer patients may benefit from these therapies. Using a high fat diet model of obesity, we orthotopically transplanted cancer cell lines into the mammary glands of obese and lean mice. We quantified changes in tumor invasiveness, angiogenesis and metastasis, and examined the efficacy of macrophage depletion to diminish tumor progression in obese and lean mice. Mammary tumors from obese mice grew significantly faster, were enriched for cancer stem-like cells (CSCs) and were more locally invasive and metastatic. Tumor cells isolated from obese mice demonstrated enhanced expression of stem cell-related pathways including Sox2 and Notch2. Despite more rapid growth, mammary tumors from obese mice had reduced necrosis, higher blood vessel density, and greater macrophage recruitment. Depletion of macrophages in obese tumor-bearing mice resulted in increased tumor necrosis, reduced endothelial cells, and enhanced recruitment of CD8+ T cells compared to IgG-treated controls. Macrophages may be an important clinical target to improve treatment options for obese breast cancer patients.
Collapse
|
37
|
|
38
|
Chen CY, Su CW, Kang JX. Endogenous Omega-3 Polyunsaturated Fatty Acids Reduce the Number and Differentiation of White Adipocyte Progenitors in Mice. Obesity (Silver Spring) 2020; 28:235-240. [PMID: 31721479 DOI: 10.1002/oby.22626] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 07/26/2019] [Indexed: 11/09/2022]
Abstract
OBJECTIVES Reducing the increased number of white adipocyte progenitors (WAP) is considered a novel approach to controlling obesity. The role of omega-3 polyunsaturated fatty acids (PUFA) in regulating the WAP resident population is unclear. The objective of this study was to investigate the effect of omega-3 PUFA on the niche composition of adipose-derived stem cells. METHODS Stromal vascular cell fraction (SVF) was collected from subcutaneous fat of wild-type (WT) and transgenic mice carrying a fat-1 gene from Caenorhabditis elegans (Fat-1 mice), which are capable of synthesizing omega-3 PUFA and have much higher tissue levels of omega-3 PUFA relative to WT mice. The isolated SVF cells were cultured and used for the examination of adipocyte differentiation, adipogenic markers, fatty acid composition, and WAP numbers. RESULTS SVF isolated from Fat-1 mice (Fat-1-SVF) exhibited markedly fewer differentiated adipocytes with smaller cell size and less lipid content than that of WT mice (WT-SVF). Accordingly, adipogenesis-related genes and the white adipocyte surface marker ASC-1 were downregulated in Fat-1-SVF relative to WT-SVF. Furthermore, WAP numbers and adipose tissue macrophages were lower in Fat-1-SVF than WT-SVF. CONCLUSIONS Omega-3 PUFA can both limit the WAP resident population and suppress their differentiation to white adipocytes, suggesting a new mechanism for the antiobesity effect of omega-3 PUFA.
Collapse
Affiliation(s)
- Chih-Yu Chen
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Chien-Wen Su
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jing X Kang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
39
|
Daemen S, Schilling JD. The Interplay Between Tissue Niche and Macrophage Cellular Metabolism in Obesity. Front Immunol 2020; 10:3133. [PMID: 32038642 PMCID: PMC6987434 DOI: 10.3389/fimmu.2019.03133] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 12/23/2019] [Indexed: 12/13/2022] Open
Abstract
Obesity is associated with the development of metabolic diseases such as type 2 diabetes and non-alcoholic fatty liver disease. The presence of chronic, low-grade inflammation appears to be an important mechanistic link between excess nutrients and clinical disease. The onset of these metabolic disorders coincides with changes in the number and phenotype of macrophages in peripheral organs, particularly in the liver and adipose tissue. Macrophage accumulation in these tissues has been implicated in tissue inflammation and fibrosis, contributing to metabolic disease progression. Recently, the concept has emerged that changes in macrophage metabolism affects their functional phenotype, possibly triggered by distinct environmental metabolic cues. This may be of particular importance in the setting of obesity, where both liver and adipose tissue are faced with a high metabolic burden. In the first part of this review we will discuss current knowledge regarding macrophage dynamics in both adipose tissue and liver in obesity. Then in the second part, we will highlight data linking macrophage metabolism to functional phenotype with an emphasis on macrophage activation in metabolic disease. The importance of understanding how tissue niche influences macrophage function in obesity will be highlighted. In addition, we will identify important knowledge gaps and outstanding questions that are relevant for future research in this area and will facilitate the identification of novel targets for therapeutic intervention in associated metabolic diseases.
Collapse
Affiliation(s)
- Sabine Daemen
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Joel D Schilling
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
40
|
Kalmukova OO, Yurchenko AV, Savchuk AM, Dzerzhynsky ME. Changes in the Inflammatory Status in White Adipose Tissue of Rats with Diet-Induced Obesity at Different Regimens of Melatonin Administration. CYTOL GENET+ 2020. [DOI: 10.3103/s0095452720010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
41
|
Aging Induces an Nlrp3 Inflammasome-Dependent Expansion of Adipose B Cells That Impairs Metabolic Homeostasis. Cell Metab 2019; 30:1024-1039.e6. [PMID: 31735593 PMCID: PMC6944439 DOI: 10.1016/j.cmet.2019.10.006] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 08/10/2019] [Accepted: 10/15/2019] [Indexed: 12/23/2022]
Abstract
During aging, visceral adiposity is often associated with alterations in adipose tissue (AT) leukocytes, inflammation, and metabolic dysfunction. However, the contribution of AT B cells in immunometabolism during aging is unexplored. Here, we show that aging is associated with an expansion of a unique population of resident non-senescent aged adipose B cells (AABs) found in fat-associated lymphoid clusters (FALCs). AABs are transcriptionally distinct from splenic age-associated B cells (ABCs) and show greater expansion in female mice. Functionally, whole-body B cell depletion restores proper lipolysis and core body temperature maintenance during cold stress. Mechanistically, the age-induced FALC formation, AAB, and splenic ABC expansion is dependent on the Nlrp3 inflammasome. Furthermore, AABs express IL-1R, and inhibition of IL-1 signaling reduces their proliferation and increases lipolysis in aging. These data reveal that inhibiting Nlrp3-dependent B cell accumulation can be targeted to reverse metabolic impairment in aging AT.
Collapse
|
42
|
|
43
|
Liu B, Hutchison AT, Thompson CH, Lange K, Heilbronn LK. Markers of adipose tissue inflammation are transiently elevated during intermittent fasting in women who are overweight or obese. Obes Res Clin Pract 2019; 13:408-415. [PMID: 31302012 DOI: 10.1016/j.orcp.2019.07.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 06/24/2019] [Accepted: 07/01/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE This study compared the effects of daily calorie restriction (DR) versus intermittent fasting (IF) on markers of inflammation and extracellular matrix deposition in adipose tissue and skeletal muscle in a controlled feeding trial in women with overweight or obesity. METHODS Women (N = 76) were randomised to one of three diets and provided with all foods at 100% (IF100) or 70% (IF70 and DR70) of calculated energy requirements for 8 weeks. IF groups ate breakfast prior to fasting for 24-h on 3 non-consecutive days/week. Weight, body composition, serum non-esterified fatty acids (NEFA), tumour necrosis factor-alpha (TNFα), interleukin-6 (IL-6), interleukin-10 (IL-10), M1- and M2-macrophage markers by qPCR and immunohistochemistry in adipose tissue and skeletal muscle were measured following a 12-h overnight fast (fed day, all groups) and a 24-h fast (IF groups only). RESULTS IF70 resulted in greater weight and fat losses and reductions in serum NEFA versus DR70 and IF100 (P < 0.05) after fed days. Markers of inflammation in serum (TNFα, IL6 and IL10), subcutaneous adipose tissue and skeletal muscle (CD68, CD40 and CD163) were unchanged by DR or IF after fed days. After fasting, NEFA, M1-macrophages (CD40+) in adipose tissue, and M2-macrophages (CD163+) in muscle were increased in IF70 and IF100 (all P < 0.05) and the changes in NEFA and mRNA of pan-macrophage marker CD68 in adipose tissue were positively correlated (r = 0.56, P = 0.002). CONCLUSIONS Unlike caloric restriction, IF transiently elevated markers of macrophage infiltration in adipose tissue and skeletal muscle, possibly in response to marked increases in adipose tissue lipolysis.
Collapse
Affiliation(s)
- Bo Liu
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5000, Australia; Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| | - Amy T Hutchison
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5000, Australia; Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| | - Campbell H Thompson
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Kylie Lange
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Leonie K Heilbronn
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5000, Australia; Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia.
| |
Collapse
|
44
|
Yang IH, Rose GE, Ezra DG, Bailly M. Macrophages promote a profibrotic phenotype in orbital fibroblasts through increased hyaluronic acid production and cell contractility. Sci Rep 2019; 9:9622. [PMID: 31270379 PMCID: PMC6610127 DOI: 10.1038/s41598-019-46075-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 06/21/2019] [Indexed: 12/29/2022] Open
Abstract
Graves’ orbitopathy (GO) is an autoimmune inflammatory disease affecting the orbit. Orbital fibroblasts are a key component in GO pathogenesis, which includes inflammation, adipogenesis, hyaluronic acid (HA) secretion, and fibrosis. Macrophages are thought to participate in the immunological stage of GO, but whether they can directly affect the fibroblasts phenotype and modulate disease progression is unknown. We previously showed that GO adipogenic and fibrotic phenotypes could be modelled in a pseudo-physiological 3D environment in vitro. Here, we introduced macrophages in this 3D culture model to investigate role for macrophages in modulating adipogenesis, HA production, and contractility in orbital fibroblasts. Macrophages had a minimal effect on lipid droplet formation in fibroblasts, but significantly increased HA production and cell contractility, suggesting that they may promote the fibrotic phenotype. This effect was found to be mediated at least in part through phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) activation and linked to an increase in actin polymerization and protrusive activity in fibroblasts. Overall our work shows for the first time a direct role for macrophages in modulating the fibroblasts’ phenotype in GO, supporting a role for macrophages in the progression of the fibrotic phenotype through induction of HA production and stimulation of the contractile phenotype in orbital fibroblasts.
Collapse
Affiliation(s)
- I-Hui Yang
- UCL Institute of Ophthalmology, London, EC1V 9EL, UK.,Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Geoffrey E Rose
- Department of Adnexal Surgery, Moorfields Eye Hospital, London, EC1V 2PD, UK
| | - Daniel G Ezra
- UCL Institute of Ophthalmology, London, EC1V 9EL, UK.,Department of Adnexal Surgery, Moorfields Eye Hospital, London, EC1V 2PD, UK.,NIHR Biomedical Research Centre for Ophthalmology, Moorfields Eye Hospital, City Road, London, EC1V 2PD, UK
| | - Maryse Bailly
- UCL Institute of Ophthalmology, London, EC1V 9EL, UK.
| |
Collapse
|
45
|
Affiliation(s)
- Henry Kuang
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Jiandie D Lin
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
46
|
Sun L, Lin JD. Function and Mechanism of Long Noncoding RNAs in Adipocyte Biology. Diabetes 2019; 68:887-896. [PMID: 31010880 PMCID: PMC6477904 DOI: 10.2337/dbi18-0009] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 02/19/2019] [Indexed: 12/17/2022]
Abstract
The last two decades have witnessed an explosion of interest in adipocyte biology, coinciding with the upsurge of obesity and metabolic syndrome. Now we have new perspectives on the distinct developmental origins of white, brown, and beige adipocytes and their role in metabolic physiology and disease. Beyond fuel metabolism, adipocytes communicate with the immune system and other tissues by releasing diverse paracrine and endocrine factors to orchestrate adipose tissue remodeling and maintain systemic homeostasis. Significant progress has been made in delineating the regulatory networks that govern different aspects of adipocyte biology. Here we provide an overview on the emerging role of long noncoding RNAs (lncRNAs) in the regulation of adipocyte development and metabolism and discuss the implications of the RNA-protein regulatory interface in metabolic control.
Collapse
Affiliation(s)
- Lei Sun
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Jiandie D Lin
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
47
|
Li C, Menoret A, Farragher C, Ouyang Z, Bonin C, Holvoet P, Vella AT, Zhou B. Single cell transcriptomics based-MacSpectrum reveals novel macrophage activation signatures in diseases. JCI Insight 2019; 5:126453. [PMID: 30990466 DOI: 10.1172/jci.insight.126453] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Adipose tissue macrophages (ATM) are crucial for maintaining adipose tissue homeostasis and mediating obesity-induced metabolic abnormalities, including prediabetic conditions and type 2 diabetes mellitus. Despite their key functions in regulating adipose tissue metabolic and immunologic homeostasis under normal and obese conditions, a high-resolution transcriptome annotation system that can capture ATM multifaceted activation profiles has not yet been developed. This is primarily attributed to the complexity of their differentiation/activation process in adipose tissue and their diverse activation profiles in response to microenvironmental cues. Although the concept of multifaceted macrophage action is well-accepted, no current model precisely depicts their dynamically regulated in vivo features. To address this knowledge gap, we generated single-cell transcriptome data from primary bone marrow-derived macrophages under polarizing and non-polarizing conditions to develop new high-resolution algorithms. The outcome was creation of a two-index platform, MacSpectrum (https://macspectrum.uconn.edu), that enables comprehensive high-resolution mapping of macrophage activation states from diverse mixed cell populations. MacSpectrum captured dynamic transitions of macrophage subpopulations under both in vitro and in vivo conditions. Importantly, MacSpectrum revealed unique "signature" gene sets in ATMs and circulating monocytes that displayed significant correlation with BMI and homeostasis model assessment of insulin resistance (HOMA-IR) in obese human patients. Thus, MacSpectrum provides unprecedented resolution to decode macrophage heterogeneity and will open new areas of clinical translation.
Collapse
Affiliation(s)
- Chuan Li
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, Connecticut, USA
| | - Antoine Menoret
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, Connecticut, USA.,Institute for Systems Genomics, University of Connecticut, Farmington, Connecticut, USA
| | - Cullen Farragher
- College of Liberal Arts and Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Zhengqing Ouyang
- Institute for Systems Genomics, University of Connecticut, Farmington, Connecticut, USA.,The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA.,Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA.,Department of Genetics and Genome Sciences, University of Connecticut, Farmington, Connecticut, USA
| | - Christopher Bonin
- School of Medicine, University of Connecticut, Farmington, Connecticut, USA
| | - Paul Holvoet
- Experimental Cardiology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Anthony T Vella
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, Connecticut, USA
| | - Beiyan Zhou
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, Connecticut, USA.,Institute for Systems Genomics, University of Connecticut, Farmington, Connecticut, USA
| |
Collapse
|
48
|
Bradley D, Blaszczak A, Yin Z, Liu J, Joseph JJ, Wright V, Anandani K, Needleman B, Noria S, Renton D, Yearsley M, Wong STC, Hsueh WA. Clusterin Impairs Hepatic Insulin Sensitivity and Adipocyte Clusterin Associates With Cardiometabolic Risk. Diabetes Care 2019; 42:466-475. [PMID: 30659075 PMCID: PMC6385696 DOI: 10.2337/dc18-0870] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 12/19/2018] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Components of the adipose tissue (AT) extracellular matrix (ECM) are recently discovered contributors to obesity-related cardiometabolic disease. We identified increased adipocyte expression of ECM-related clusterin (apolipoprotein J) in obese versus lean women by microarray. Our objective was to determine 1) whether subcutaneous AT adipocyte (SAd) clusterin and serum clusterin are associated with insulin resistance (IR) and known markers of cardiometabolic risk and 2) how clusterin may contribute to increased risk. RESEARCH DESIGN AND METHODS We validated increased clusterin expression in adipocytes from a separate group of 18 lean and 54 obese individuals. The relationship of clusterin gene expression and plasma clusterin with IR, cardiovascular biomarkers, and risk of cardiovascular disease (CVD) was then determined. Further investigations in human cultured cells and in aged LDLR-/- mice prone to development of obesity-associated complications were performed. RESULTS SAd clusterin correlated with IR, multiple CVD biomarkers, and CVD risk, independent of traditional risk factors. Circulating human clusterin exhibited similar associations. In human adipocytes, palmitate enhanced clusterin secretion, and in human hepatocytes, clusterin attenuated insulin signaling and APOA1 expression and stimulated hepatic gluconeogenesis. LRP2 (megalin), a clusterin receptor, highly expressed in liver, mediated these effects, which were inhibited by LRP2 siRNA. In response to Western diet feeding, an increase in adipocyte clusterin expression was associated with a progressive increase in liver fat, steatohepatitis, and fibrosis in aged LDLR-/- mice. CONCLUSIONS Adipocyte-derived clusterin is a novel ECM-related protein linking cardiometabolic disease and obesity through its actions in the liver.
Collapse
Affiliation(s)
- David Bradley
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH
| | - Alecia Blaszczak
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH
| | - Zheng Yin
- Department of Systems Medicine and Bioengineering, Institute for Academic Medicine, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX
| | - Joey Liu
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH
| | - Joshua J Joseph
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH
| | - Valerie Wright
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH
| | - Kajol Anandani
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH
| | - Bradley Needleman
- Center for Minimally Invasive Surgery, Department of Surgery, Wexner Medical Center, The Ohio State University, Columbus, OH
| | - Sabrena Noria
- Center for Minimally Invasive Surgery, Department of Surgery, Wexner Medical Center, The Ohio State University, Columbus, OH
| | - David Renton
- Center for Minimally Invasive Surgery, Department of Surgery, Wexner Medical Center, The Ohio State University, Columbus, OH
| | - Martha Yearsley
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH
| | - Stephen T C Wong
- Department of Systems Medicine and Bioengineering, Institute for Academic Medicine, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX
| | - Willa A Hsueh
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH
| |
Collapse
|
49
|
Labrecque J, Michaud A, Gauthier MF, Pelletier M, Julien F, Bouvet-Bouchard L, Tchernof A. Interleukin-1β and prostaglandin-synthesizing enzymes as modulators of human omental and subcutaneous adipose tissue function. Prostaglandins Leukot Essent Fatty Acids 2019; 141:9-16. [PMID: 30661603 DOI: 10.1016/j.plefa.2018.11.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 11/28/2018] [Accepted: 11/28/2018] [Indexed: 02/05/2023]
Abstract
IL-1β stimulates expression of prostaglandin (PG)-synthesizing enzymes cyclooxygenase (COX)-2 and aldo-keto reductase (AKR)1B1 in human preadipocytes. We aimed to examine the impact of IL-1β, COX-2 and AKR1B1 on markers of human visceral and subcutaneous adipose tissue function, and to assess whether PG synthesis by these enzymes mediates IL-1β effects. Omental and subcutaneous fat samples were obtained from bariatric surgery patients. PG release and expression of inflammatory and adipogenic markers were assessed in explants treated with COX-2 inhibitor NS-398 or AKR1B1 inhibitor Statil, with or without IL-1β. Preadipocyte differentiation experiments were also performed. IL-1β decreased expression of PPARγ in both fat depots compared to control and increased expression of NF-κB1, IL-6, CCL-5, ICAM-1 and VEGFA, especially in visceral fat for IL-6, CCL-5 and VEGFA. Adding Statil or NS-398 to IL-1β blunted PGF2α and PGE2 release, but did not alter IL-1β effects on adipose tissue function markers. IL-1β down-regulated adipocyte differentiation whereas NS-398 alone increased this process. However, NS-398 did not prevent IL-1β inhibition of adipogenesis. We conclude that IL-1β induces a pro-inflammatory response in human adipose tissues, particularly in visceral fat, and acts independently of concomitant PG release. IL-1β and COX-2 appear to be critical determinants of adipose tissue pathophysiologic remodeling in obesity.
Collapse
Affiliation(s)
- Jennifer Labrecque
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Québec, QC, Canada; École de nutrition - Université Laval, Québec, QC, Canada; Centre hospitalier universitaire de Québec - Université Laval, Québec, QC, Canada
| | - Andréanne Michaud
- Montreal Neurological Institute - McGill University, Montreal, QC, Canada
| | - Marie-Frédérique Gauthier
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Québec, QC, Canada
| | - Mélissa Pelletier
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Québec, QC, Canada; Centre hospitalier universitaire de Québec - Université Laval, Québec, QC, Canada
| | - François Julien
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Québec, QC, Canada
| | - Léonie Bouvet-Bouchard
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Québec, QC, Canada
| | - André Tchernof
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Québec, QC, Canada; École de nutrition - Université Laval, Québec, QC, Canada; Centre hospitalier universitaire de Québec - Université Laval, Québec, QC, Canada.
| |
Collapse
|
50
|
Wang Q, Sharma VP, Shen H, Xiao Y, Zhu Q, Xiong X, Guo L, Jiang L, Ohta K, Li S, Shi H, Rui L, Lin JD. The hepatokine Tsukushi gates energy expenditure via brown fat sympathetic innervation. Nat Metab 2019; 1:251-260. [PMID: 31535079 PMCID: PMC6750233 DOI: 10.1038/s42255-018-0020-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 11/20/2018] [Indexed: 12/17/2022]
Abstract
Thermogenesis is an important contributor to whole body energy expenditure and metabolic homeostasis. Although circulating factors that promote energy expenditure are known, endocrine molecules that suppress energy expenditure have remained largely elusive. Here we show that Tsukushi (TSK) is a liver-enriched secreted factor that is highly inducible in response to increased energy expenditure. Hepatic Tsk expression and plasma TSK levels are elevated in obesity. TSK deficiency increases sympathetic innervation and norepinephrine release in adipose tissue, leading to enhanced adrenergic signaling and thermogenesis, attenuation of brown fat whitening and protection from diet-induced obesity in mice. Our work reveals TSK as part of a negative feedback mechanism that gates thermogenic energy expenditure and highlights TSK as a potential target for therapeutic intervention in metabolic disease.
Collapse
Affiliation(s)
- Qiuyu Wang
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Vishal P Sharma
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Hong Shen
- Department of Molecular & Integrated Physiology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Yuanyuan Xiao
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Qi Zhu
- Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH, USA
| | - Xuelian Xiong
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Liang Guo
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Lin Jiang
- Department of Molecular & Integrated Physiology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Kunimasa Ohta
- Department of Developmental Neurobiology, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Siming Li
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Haifei Shi
- Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH, USA
| | - Liangyou Rui
- Department of Molecular & Integrated Physiology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Jiandie D Lin
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI, USA.
| |
Collapse
|