3
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Abnormal brown adipose tissue mitochondrial structure and function in IL10 deficiency

      research-article

      Read this article at

      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

          Background

          Inflammation is the most relevant mechanism linking obesity with insulin-resistance and metabolic disease. It impacts the structure and function of tissues and organs involved in metabolism, such as the liver, pancreatic islets and the hypothalamus. Brown adipose tissue has emerged as an important component of whole body energy homeostasis, controlling caloric expenditure through the regulation of non-shivering thermogenesis. However, little is known about the impact of systemic inflammation on the structure and function of brown adipose tissue.

          Methods

          The relations between IL10 and mitochondria structure/function and also with thermogenesis were evaluated by bioinformatics using human and rodent data. Real-time PCR, immunoblot, fluorescence and transmission electron microscopy were employed to determine the effect of IL10 in the brown adipose tissue of wild type and IL10 knockout mice.

          Findings

          IL10 knockout mice, a model of systemic inflammation, present severe structural abnormalities of brown adipose tissue mitochondria, which are round-shaped with loss of cristae structure and increased fragmentation. IL10 deficiency leads to newborn cold intolerance and impaired UCP1-dependent brown adipose tissue mitochondrial respiration. The reduction of systemic inflammation with an anti-TNFα monoclonal antibody partially rescued the structural but not the functional abnormalities of brown adipose tissue mitochondria. Using bioinformatics analyses we show that in both humans and mice, IL10 transcripts correlate with mitochondrial lipid metabolism and caspase gene expression.

          Interpretation

          IL10 and systemic inflammation play a central role in the regulation of brown adipose tissue by controlling mitochondrial structure and function.

          Fund

          Sao Paulo Research Foundation grant 2013/07607-8.

          Related collections

          Most cited references48

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

          A creatine-driven substrate cycle enhances energy expenditure and thermogenesis in beige fat.

          Thermogenic brown and beige adipose tissues dissipate chemical energy as heat, and their thermogenic activities can combat obesity and diabetes. Herein the functional adaptations to cold of brown and beige adipose depots are examined using quantitative mitochondrial proteomics. We identify arginine/creatine metabolism as a beige adipose signature and demonstrate that creatine enhances respiration in beige-fat mitochondria when ADP is limiting. In murine beige fat, cold exposure stimulates mitochondrial creatine kinase activity and induces coordinated expression of genes associated with creatine metabolism. Pharmacological reduction of creatine levels decreases whole-body energy expenditure after administration of a β3-agonist and reduces beige and brown adipose metabolic rate. Genes of creatine metabolism are compensatorily induced when UCP1-dependent thermogenesis is ablated, and creatine reduction in Ucp1-deficient mice reduces core body temperature. These findings link a futile cycle of creatine metabolism to adipose tissue energy expenditure and thermal homeostasis. PAPERCLIP.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Mice lacking mitochondrial uncoupling protein are cold-sensitive but not obese.

            The mitochondrial uncoupling protein (UCP) in the mitochondrial inner membrane of mammalian brown adipose tissue generates heat by uncoupling oxidative phosphorylation. This process protects against cold and regulates energy balance. Manipulation of thermogenesis could be an effective strategy against obesity. Here we determine the role of UCP in the regulation of body mass by targeted inactivation of the gene encoding it. We find that UCP-deficient mice consume less oxygen after treatment with a beta3-adrenergic-receptor agonist and that they are sensitive to cold, indicating that their thermoregulation is defective. However, this deficiency caused neither hyperphagia nor obesity in mice fed on either a standard or a high-fat diet. We propose that the loss of UCP may be compensated by UCP2, a newly discovered homologue of UCP; this gene is ubiquitously expressed and is induced in the brown fat of UCP-deficient mice.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Brown adipose tissue as a secretory organ.

              Brown adipose tissue (BAT) is the main site of adaptive thermogenesis and experimental studies have associated BAT activity with protection against obesity and metabolic diseases, such as type 2 diabetes mellitus and dyslipidaemia. Active BAT is present in adult humans and its activity is impaired in patients with obesity. The ability of BAT to protect against chronic metabolic disease has traditionally been attributed to its capacity to utilize glucose and lipids for thermogenesis. However, BAT might also have a secretory role, which could contribute to the systemic consequences of BAT activity. Several BAT-derived molecules that act in a paracrine or autocrine manner have been identified. Most of these factors promote hypertrophy and hyperplasia of BAT, vascularization, innervation and blood flow, processes that are all associated with BAT recruitment when thermogenic activity is enhanced. Additionally, BAT can release regulatory molecules that act on other tissues and organs. This secretory capacity of BAT is thought to be involved in the beneficial effects of BAT transplantation in rodents. Fibroblast growth factor 21, IL-6 and neuregulin 4 are among the first BAT-derived endocrine factors to be identified. In this Review, we discuss the current understanding of the regulatory molecules (the so-called brown adipokines or batokines) that are released by BAT that influence systemic metabolism and convey the beneficial metabolic effects of BAT activation. The identification of such adipokines might also direct drug discovery approaches for managing obesity and its associated chronic metabolic diseases.
                Bookmark

                Author and article information

                Contributors
                Journal
                EBioMedicine
                EBioMedicine
                EBioMedicine
                Elsevier
                2352-3964
                27 November 2018
                January 2019
                27 November 2018
                : 39
                : 436-447
                Affiliations
                [a ]Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas, Campinas, São Paulo 13084-970, Brazil
                [b ]Obesity and Comorbidities Research Center, University of Campinas, Campinas, São Paulo 13084-970, Brazil
                [c ]CEPECE - Research Center of Sport Sciences, School of Applied Sciences, University of Campinas, Limeira, SP, Brazil .
                [d ]Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, SP 13083-970, Brazil
                [e ]Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, São Paulo, Brazil
                [f ]National Institute of Photonics Applied to Cell Biology (INFABiC), Campinas, São Paulo, Brazil
                [g ]Laboratory of Exercise Physiology, School of Physical Education, University of Campinas, Campinas, SP 13083-970, Brazil
                [h ]Laboratory of Investigation in Metabolism and Diabetes (LIMED)/Gastrocentro, Department of Surgery, University of Campinas (UNICAMP), Campinas, SP 13081-970, Brazil
                [i ]Biomedical Engineering Center, University of Campinas (UNICAMP), Campinas, SP, Brazil
                [j ]Department of Radiology, University of Campinas, Campinas, São Paulo 13084-970, Brazil
                [k ]Department of Pediatrics, Federal University of Uberlandia, Uberlandia, Brazil
                Author notes
                [* ]Corresponding author at: Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas, Campinas, São Paulo 13084-970, Brazil. lavelloso@ 123456fcm.unicamp.br
                [1]

                The Laboratory of Cell Signaling belongs to the National Institute of Science and Technology – Neuroimmunomodulation (INCT-NIM).

                Article
                S2352-3964(18)30545-0
                10.1016/j.ebiom.2018.11.041
                6355943
                30502051
                128f7f19-f368-4507-a71e-b70e05cbc6d1
                © 2018 The Authors

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                : 14 August 2018
                : 13 November 2018
                : 19 November 2018
                Categories
                Research paper

                interleukin-10,inflammation,thermogenesis,mitochondria,respiration,obesity

                Comments

                Comment on this article