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

      Lipodystrophy in methylmalonic acidemia associated with elevated FGF21 and abnormal methylmalonylation

      research-article
      1 , 1 , 1 , 1 , 2 , 3 , 1 , 1 , 1 , 1 , 3 , 1 , 4 , 4 , 5 , 6 , 7 , 8 , 9 , 10 , 6 , 6 , 5 , 11 , 12 , 13 , 3 , 1 ,
      JCI Insight
      American Society for Clinical Investigation
      Genetics, Therapeutics, Amino acid metabolism, Mouse models, Obesity

      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

          A distinct adipose tissue distribution pattern was observed in patients with methylmalonyl-CoA mutase deficiency, an inborn error of branched-chain amino acid (BCAA) metabolism, characterized by centripetal obesity with proximal upper and lower extremity fat deposition and paucity of visceral fat, that resembles familial multiple lipomatosis syndrome. To explore brown and white fat physiology in methylmalonic acidemia (MMA), body composition, adipokines, and inflammatory markers were assessed in 46 patients with MMA and 99 matched controls. Fibroblast growth factor 21 levels were associated with acyl-CoA accretion, aberrant methylmalonylation in adipose tissue, and an attenuated inflammatory cytokine profile. In parallel, brown and white fat were examined in a liver-specific transgenic MMA mouse model ( Mmut –/– Tg INS-Alb-Mmut). The MMA mice exhibited abnormal nonshivering thermogenesis with whitened brown fat and had an ineffective transcriptional response to cold stress. Treatment of the MMA mice with bezafibrates led to clinical improvement with beiging of subcutaneous fat depots, which resembled the distribution seen in the patients. These studies defined what we believe to be a novel lipodystrophy phenotype in patients with defects in the terminal steps of BCAA oxidation and demonstrated that beiging of subcutaneous adipose tissue in MMA could readily be induced with small molecules.

          Abstract

          Abstract

          <p>Subcutaneous adipose tissue expansion with beige fat properties is observed in methylmalonic acidemia associated with aberrant methylmalonylation and high FGF21.</p>

          Related collections

          Most cited references104

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

          Brown adipose tissue: function and physiological significance.

          The function of brown adipose tissue is to transfer energy from food into heat; physiologically, both the heat produced and the resulting decrease in metabolic efficiency can be of significance. Both the acute activity of the tissue, i.e., the heat production, and the recruitment process in the tissue (that results in a higher thermogenic capacity) are under the control of norepinephrine released from sympathetic nerves. In thermoregulatory thermogenesis, brown adipose tissue is essential for classical nonshivering thermogenesis (this phenomenon does not exist in the absence of functional brown adipose tissue), as well as for the cold acclimation-recruited norepinephrine-induced thermogenesis. Heat production from brown adipose tissue is activated whenever the organism is in need of extra heat, e.g., postnatally, during entry into a febrile state, and during arousal from hibernation, and the rate of thermogenesis is centrally controlled via a pathway initiated in the hypothalamus. Feeding as such also results in activation of brown adipose tissue; a series of diets, apparently all characterized by being low in protein, result in a leptin-dependent recruitment of the tissue; this metaboloregulatory thermogenesis is also under hypothalamic control. When the tissue is active, high amounts of lipids and glucose are combusted in the tissue. The development of brown adipose tissue with its characteristic protein, uncoupling protein-1 (UCP1), was probably determinative for the evolutionary success of mammals, as its thermogenesis enhances neonatal survival and allows for active life even in cold surroundings.
            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
              • Record: found
              • Abstract: found
              • Article: not found

              A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance.

              Metabolomic profiling of obese versus lean humans reveals a branched-chain amino acid (BCAA)-related metabolite signature that is suggestive of increased catabolism of BCAA and correlated with insulin resistance. To test its impact on metabolic homeostasis, we fed rats on high-fat (HF), HF with supplemented BCAA (HF/BCAA), or standard chow (SC) diets. Despite having reduced food intake and a low rate of weight gain equivalent to the SC group, HF/BCAA rats were as insulin resistant as HF rats. Pair-feeding of HF diet to match the HF/BCAA animals or BCAA addition to SC diet did not cause insulin resistance. Insulin resistance induced by HF/BCAA feeding was accompanied by chronic phosphorylation of mTOR, JNK, and IRS1Ser307 and by accumulation of multiple acylcarnitines in muscle, and it was reversed by the mTOR inhibitor, rapamycin. Our findings show that in the context of a dietary pattern that includes high fat consumption, BCAA contributes to development of obesity-associated insulin resistance.
                Bookmark

                Author and article information

                Contributors
                Journal
                JCI Insight
                JCI Insight
                JCI Insight
                JCI Insight
                American Society for Clinical Investigation
                2379-3708
                22 February 2024
                22 February 2024
                22 February 2024
                : 9
                : 4
                : e174097
                Affiliations
                [1 ]Metabolic Medicine Branch, National Human Genome Research Institute;
                [2 ]Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases;
                [3 ]Section on Growth and Obesity, Eunice Kennedy Shriver National Institute of Child Health and Human Development; and
                [4 ]Office of Research Services, Division of Veterinary Resources, NIH, Bethesda, Maryland, USA.
                [5 ]Department of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland, USA.
                [6 ]Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases;
                [7 ]Radiology and Imaging Sciences Department, Clinical Center;
                [8 ]Ultrastructural Pathology Section, Center for Cancer Research; and
                [9 ]Section on Endocrinology & Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA.
                [10 ]Department of Radiology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.
                [11 ]Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, Massachusetts, USA.
                [12 ]Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA.
                [13 ]Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, NIH, Bethesda, Maryland, USA.
                Author notes
                Address correspondence to: Charles P. Venditti, National Human Genome Research Institute, National Institutes of Health, Bldg 10, Room 7N248A, Bethesda, Maryland 20892-4442, USA. Phone: 301.496.6213; Email: venditti@ 123456mail.nih.gov .
                Author information
                http://orcid.org/0000-0003-1543-2941
                http://orcid.org/0000-0002-9656-725X
                http://orcid.org/0000-0003-4520-0949
                http://orcid.org/0000-0002-6398-4401
                http://orcid.org/0000-0001-8072-1358
                http://orcid.org/0000-0002-4058-5520
                http://orcid.org/0000-0002-2589-7382
                http://orcid.org/0000-0001-8542-1637
                http://orcid.org/0000-0001-6599-1253
                Article
                174097
                10.1172/jci.insight.174097
                10967474
                38271099
                fd09bafb-d162-47b5-879e-f47c72f096dc
                © 2024 Manoli et al.

                This work is licensed under the Creative Commons Attribution 4.0 International License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 24 July 2023
                : 9 January 2024
                Funding
                Funded by: National Human Genome Research Institute, https://doi.org/10.13039/100000051;
                Award ID: ZIAHG200318-16
                Funded by: Eunice Kennedy Shriver National Institute of Child Health and Human Development, https://doi.org/10.13039/100009633;
                Award ID: ZIAHD00641
                Funded by: National Institute of Diabetes, Digestive and Kidney Diseases
                Award ID: ZIADK071014
                NHGRI, NIH to C.P.V.
                NICHD, NIH to J.A.Y
                NIDDK, NIH to K.Y.C.
                Categories
                Research Article

                genetics,therapeutics,amino acid metabolism,mouse models,obesity

                Comments

                Comment on this article