1
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Adipocyte-Specific Hnrnpa1 Knockout Aggravates Obesity-Induced Metabolic Dysfunction via Upregulation of CCL2

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Heterogeneous nuclear ribonucleoprotein A1 (HNRNPA1) is involved in lipid and glucose metabolism via mRNA processing. However, whether and how HNRNPA1 alters adipocyte function in obesity remain obscure. Here, we found that the obese state downregulated HNRNPA1 expression in white adipose tissue (WAT). The depletion of adipocyte HNRNPA1 promoted markedly increased macrophage infiltration and expression of proinflammatory and fibrosis genes in WAT of obese mice, eventually leading to exacerbated insulin sensitivity, glucose tolerance, and hepatic steatosis. Mechanistically, HNRNPA1 interacted with Ccl2 and regulated its mRNA stability. Intraperitoneal injection of CCL2-CCR2 signaling antagonist improved adipose tissue inflammation and systemic glucose homeostasis. Furthermore, HNRNPA1 expression in human WAT was negatively correlated with BMI, fat percentage, and subcutaneous fat area. Among individuals with 1-year metabolic surgery follow-up, HNRNPA1 expression was positively related to percentage of total weight loss. These findings identify adipocyte HNRNPA1 as a link between adipose tissue inflammation and systemic metabolic homeostasis, which might be a promising therapeutic target for obesity-related disorders.

          Article Highlights
          • Heterogeneous nuclear ribonucleoprotein A1 (HNRNPA1) is decreased during obesity, but the specific role in adipose tissue remains unclear.

          • Adipocyte-specific depletion of Hnrnpa1 accelerates adipose tissue inflammation and systemic metabolic disorders via enhancing mRNA stability of chemokine CCL2.

          • CCL2 receptor antagonist rescues metabolic dysfunction elicited by Hnrnpa1 depletion.

          • HNRNPA1 expression in human adipose tissue is closely related to BMI, fat percentage, and subcutaneous fat area and is positively associated with percentage of total weight loss 1 year after bariatric surgery.

          Related collections

          Most cited references59

          • Record: found
          • Abstract: found
          • Article: not found

          Obesity: global epidemiology and pathogenesis

          The prevalence of obesity has increased worldwide in the past ~50 years, reaching pandemic levels. Obesity represents a major health challenge because it substantially increases the risk of diseases such as type 2 diabetes mellitus, fatty liver disease, hypertension, myocardial infarction, stroke, dementia, osteoarthritis, obstructive sleep apnoea and several cancers, thereby contributing to a decline in both quality of life and life expectancy. Obesity is also associated with unemployment, social disadvantages and reduced socio-economic productivity, thus increasingly creating an economic burden. Thus far, obesity prevention and treatment strategies - both at the individual and population level - have not been successful in the long term. Lifestyle and behavioural interventions aimed at reducing calorie intake and increasing energy expenditure have limited effectiveness because complex and persistent hormonal, metabolic and neurochemical adaptations defend against weight loss and promote weight regain. Reducing the obesity burden requires approaches that combine individual interventions with changes in the environment and society. Therefore, a better understanding of the remarkable regional differences in obesity prevalence and trends might help to identify societal causes of obesity and provide guidance on which are the most promising intervention strategies.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity.

            Adipocytes secrete a variety of bioactive molecules that affect the insulin sensitivity of other tissues. We now show that the abundance of monocyte chemoattractant protein-1 (MCP-1) mRNA in adipose tissue and the plasma concentration of MCP-1 were increased both in genetically obese diabetic (db/db) mice and in WT mice with obesity induced by a high-fat diet. Mice engineered to express an MCP-1 transgene in adipose tissue under the control of the aP2 gene promoter exhibited insulin resistance, macrophage infiltration into adipose tissue, and increased hepatic triglyceride content. Furthermore, insulin resistance, hepatic steatosis, and macrophage accumulation in adipose tissue induced by a high-fat diet were reduced extensively in MCP-1 homozygous KO mice compared with WT animals. Finally, acute expression of a dominant-negative mutant of MCP-1 ameliorated insulin resistance in db/db mice and in WT mice fed a high-fat diet. These findings suggest that an increase in MCP-1 expression in adipose tissue contributes to the macrophage infiltration into this tissue, insulin resistance, and hepatic steatosis associated with obesity in mice.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Adipokines in inflammation and metabolic disease.

              The worldwide epidemic of obesity has brought considerable attention to research aimed at understanding the biology of adipocytes (fat cells) and the events occurring in adipose tissue (fat) and in the bodies of obese individuals. Accumulating evidence indicates that obesity causes chronic low-grade inflammation and that this contributes to systemic metabolic dysfunction that is associated with obesity-linked disorders. Adipose tissue functions as a key endocrine organ by releasing multiple bioactive substances, known as adipose-derived secreted factors or adipokines, that have pro-inflammatory or anti-inflammatory activities. Dysregulated production or secretion of these adipokines owing to adipose tissue dysfunction can contribute to the pathogenesis of obesity-linked complications. In this Review, we focus on the role of adipokines in inflammatory responses and discuss their potential as regulators of metabolic function.
                Bookmark

                Author and article information

                Journal
                Diabetes
                Diabetes
                diabetes
                Diabetes
                American Diabetes Association
                0012-1797
                1939-327X
                May 2024
                06 February 2024
                06 February 2024
                : 73
                : 5
                : 713-727
                Affiliations
                [1]Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People’s Hospital, Shanghai, China
                Author notes

                X.Li, Y.S., and Y.X. contributed equally to this work.

                Corresponding authors: Yuqian Bao, yqbao@ 123456sjtu.edu.cn , Ying Yang, yangyingsh@ 123456sjtu.edu.cn , and Xiaojing Ma, maxiaojing@ 123456sjtu.edu.cn
                Author information
                https://orcid.org/0000-0002-1534-2279
                https://orcid.org/0000-0001-9607-161X
                https://orcid.org/0000-0001-8355-6642
                https://orcid.org/0000-0002-4754-3470
                Article
                230609
                10.2337/db23-0609
                11043064
                38320300
                b3ca0f5b-45e2-4123-b11f-16e2e35ff999
                © 2024 by the American Diabetes Association

                Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/journals/pages/license.

                History
                : 01 August 2023
                : 28 January 2024
                Funding
                Funded by: National Science Foundation of China;
                Award ID: 82170889
                Funded by: Shanghai Research Center for Endocrine and Metabolic Diseases;
                Award ID: 2022ZZ01002
                Funded by: National Natural Science Foundation of China, DOI 10.13039/501100001809;
                Award ID: 82200897
                This research was supported by National Natural Science Foundation of China grants 82200897 and 82170889 and Shanghai Research Center for Endocrine and Metabolic Diseases grant 2022ZZ01002.
                Categories
                Obesity Studies

                Endocrinology & Diabetes
                Endocrinology & Diabetes

                Comments

                Comment on this article