12
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Pathogenesis of sarcopenia and the relationship with fat mass: descriptive review

      review-article

      Read this article at

          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

          Age‐associated obesity and muscle atrophy (sarcopenia) are intimately connected and are reciprocally regulated by adipose tissue and skeletal muscle dysfunction. During ageing, adipose inflammation leads to the redistribution of fat to the intra‐abdominal area (visceral fat) and fatty infiltrations in skeletal muscles, resulting in decreased overall strength and functionality. Lipids and their derivatives accumulate both within and between muscle cells, inducing mitochondrial dysfunction, disturbing β‐oxidation of fatty acids, and enhancing reactive oxygen species (ROS) production, leading to lipotoxicity and insulin resistance, as well as enhanced secretion of some pro‐inflammatory cytokines. In turn, these muscle‐secreted cytokines may exacerbate adipose tissue atrophy, support chronic low‐grade inflammation, and establish a vicious cycle of local hyperlipidaemia, insulin resistance, and inflammation that spreads systemically, thus promoting the development of sarcopenic obesity (SO). We call this the metabaging cycle. Patients with SO show an increased risk of systemic insulin resistance, systemic inflammation, associated chronic diseases, and the subsequent progression to full‐blown sarcopenia and even cachexia. Meanwhile in many cardiometabolic diseases, the ostensibly protective effect of obesity in extremely elderly subjects, also known as the ‘obesity paradox’, could possibly be explained by our theory that many elderly subjects with normal body mass index might actually harbour SO to various degrees, before it progresses to full‐blown severe sarcopenia. Our review outlines current knowledge concerning the possible chain of causation between sarcopenia and obesity, proposes a solution to the obesity paradox, and the role of fat mass in ageing.

          Related collections

          Most cited references118

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

          Sarcopenia: European consensus on definition and diagnosis

          The European Working Group on Sarcopenia in Older People (EWGSOP) developed a practical clinical definition and consensus diagnostic criteria for age-related sarcopenia. EWGSOP included representatives from four participant organisations, i.e. the European Geriatric Medicine Society, the European Society for Clinical Nutrition and Metabolism, the International Association of Gerontology and Geriatrics—European Region and the International Association of Nutrition and Aging. These organisations endorsed the findings in the final document. The group met and addressed the following questions, using the medical literature to build evidence-based answers: (i) What is sarcopenia? (ii) What parameters define sarcopenia? (iii) What variables reflect these parameters, and what measurement tools and cut-off points can be used? (iv) How does sarcopenia relate to cachexia, frailty and sarcopenic obesity? For the diagnosis of sarcopenia, EWGSOP recommends using the presence of both low muscle mass + low muscle function (strength or performance). EWGSOP variously applies these characteristics to further define conceptual stages as ‘presarcopenia’, ‘sarcopenia’ and ‘severe sarcopenia’. EWGSOP reviewed a wide range of tools that can be used to measure the specific variables of muscle mass, muscle strength and physical performance. Our paper summarises currently available data defining sarcopenia cut-off points by age and gender; suggests an algorithm for sarcopenia case finding in older individuals based on measurements of gait speed, grip strength and muscle mass; and presents a list of suggested primary and secondary outcome domains for research. Once an operational definition of sarcopenia is adopted and included in the mainstream of comprehensive geriatric assessment, the next steps are to define the natural course of sarcopenia and to develop and define effective treatment.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The NALP3/NLRP3 Inflammasome Instigates Obesity-Induced Autoinflammation and Insulin Resistance

            Emergence of chronic ‘sterile’ inflammation during obesity in absence of overt infection or autoimmune process is a puzzling phenomenon. The Nod Like Receptor (NLR) family of innate immune cell sensors like the Nlrp3 inflammasome are implicated in recognizing certain non-microbial origin ‘danger–signals’ leading to caspase-1 activation and subsequent IL-1β and IL-18 secretion. We show that reduction in adipose tissue expression of Nlrp3 is coupled with decreased inflammation and improved insulin–sensitivity in obese type-2 diabetic patients. The Nlrp3 inflammasome senses the lipotoxicity–associated ceramide to induce caspase-1 cleavage in macrophages and adipose tissue. Ablation of Nlrp3 prevented the obesity–induced inflammasome activation in fat depots and liver together with enhanced insulin–signalling. Furthermore, elimination of Nlrp3 in obesity reduced IL-18 and adipose tissue IFNγ along with an increase in naïve and reduction in effector adipose tissue T cells. Collectively, these data establish that Nlrp3 inflammasome senses obesity–associated ‘danger–signals’ and contributes to obesity–induced inflammation and insulin–resistance.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Adipogenesis and metabolic health

              Obesity is characterized by increased adipose tissue mass and has been associated with a strong predisposition towards metabolic diseases and cancer. Thus, it constitutes a public health issue of major proportion. The expansion of adipose depots can be driven either by the increase in adipocyte size (hypertrophy) or by the formation of new adipocytes from precursor differentiation in the process of adipogenesis (hyperplasia). Notably, adipocyte expansion through adipogenesis can offset the negative metabolic effects of obesity, and the mechanisms and regulators of this adaptive process are now emerging. Over the past several years, we have learned a considerable amount about how adipocyte fate is determined and how adipogenesis is regulated by signalling and systemic factors. We have also gained appreciation that the adipogenic niche can influence tissue adipogenic capability. Approaches aimed at increasing adipogenesis over adipocyte hypertrophy can now be explored as a means to treat metabolic diseases.
                Bookmark

                Author and article information

                Contributors
                yuk1997@sina.com
                huangsq@ioz.ac.cn
                Journal
                J Cachexia Sarcopenia Muscle
                J Cachexia Sarcopenia Muscle
                10.1007/13539.2190-6009
                JCSM
                Journal of Cachexia, Sarcopenia and Muscle
                John Wiley and Sons Inc. (Hoboken )
                2190-5991
                2190-6009
                02 February 2022
                April 2022
                : 13
                : 2 ( doiID: 10.1002/jcsm.v13.2 )
                : 781-794
                Affiliations
                [ 1 ] Department of Clinical Nutrition & Health Medicine, Peking Union Medical College Hospital Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
                [ 2 ] State Key Laboratory of Stem Cell and Reproductive Biology, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Institute of Zoology Chinese Academy of Sciences Beijing China
                [ 3 ] University of Chinese Academy of Sciences Beijing China
                [ 4 ] Medical Research Center, Peking Union Medical College Hospital Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
                [ 5 ] Department of Clinical Laboratory, Peking Union Medical College Hospital Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
                [ 6 ] Department of General Surgery, Tianjin Union Medical Center, The Affiliated Hospital of Nankai University China (Tianjin Union Medical Center Tianjin China
                [ 7 ] Department of nursing & Clinical Nutrition, Dongzhimen Hospital Beijing University of Traditional Chinese Medicine Beijing China
                [ 8 ] Department of Sport Physiatry, Peking Union Medical College Hospital Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
                [ 9 ] Department of Pharmacy, Peking Union Medical College Hospital Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
                [ 10 ] Department of stomatology, Peking Union Medical College Hospital Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
                [ 11 ] Department of Health Education Shijingshan Center for Disease Prevention and Control Beijing China
                Author notes
                [*] [* ] Correspondence to: Kang Yu, Department of Clinical Nutrition & Health Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng District. Beijing 100730, China. Email: yuk1997@ 123456sina.com

                Ng Shyh‐Chang, Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, No. 1 yuan Beichen west road, Chaoyang District. Beijing 100101, China. Email: huangsq@ 123456ioz.ac.cn

                Author information
                https://orcid.org/0000-0002-6939-7275
                Article
                JCSM12901 JCSM-D-21-00330
                10.1002/jcsm.12901
                8977978
                35106971
                baca0569-cfd1-416d-813c-67c3afda94b0
                © 2022 The Authors. Journal of Cachexia, Sarcopenia and Muscle published by John Wiley & Sons Ltd on behalf of Society on Sarcopenia, Cachexia and Wasting Disorders.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc-nd/4.0/ License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made.

                History
                : 26 October 2021
                : 08 June 2021
                : 28 November 2021
                Page count
                Figures: 2, Tables: 0, Pages: 14, Words: 7524
                Funding
                Funded by: National Natural Science Foundation of China , doi 10.13039/501100001809;
                Award ID: 1191957202
                Award ID: 81900782
                Funded by: Strategic Priority Research Program of the CAS
                Award ID: XDA16010109
                Funded by: National Key R&D Program of China , doi 10.13039/501100001809;
                Award ID: 2018YFE0201103
                Award ID: 2018YFC1004102
                Award ID: 2019YFA0801701
                Award ID: 2021YFE0111800
                Funded by: Whole People Nutrition Research Fund
                Award ID: CNSNNSRG2021‐129
                Funded by: Key Research of Program of the CAS
                Award ID: KJZD‐SW‐L04
                Funded by: CAS Project for Young Scientists in Basic Research
                Award ID: YSBR‐012
                Funded by: Strategic Collaborative Research Program of the Ferring Institute of Reproductive Medicine
                Award ID: FIRMA180301
                Award ID: FIRMA200507
                Funded by: Nutrition Scientific Research Foundation of BY‐HEALTH
                Award ID: TY0171102
                Funded by: Foundation of Tianjin Union Medical Center
                Award ID: 2017YJ023
                Categories
                Review
                Reviews
                Custom metadata
                2.0
                April 2022
                Converter:WILEY_ML3GV2_TO_JATSPMC version:6.1.3 mode:remove_FC converted:04.04.2022

                Orthopedics
                sarcopenia,obesity,proto‐sarcopenia,myosteatosis,insulin resistance,inflammation
                Orthopedics
                sarcopenia, obesity, proto‐sarcopenia, myosteatosis, insulin resistance, inflammation

                Comments

                Comment on this article