14
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      β-arrestin 1 regulates β2-adrenergic receptor-mediated skeletal muscle hypertrophy and contractility

      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

          β 2-adrenergic receptors (β 2ARs) are the target of catecholamines and play fundamental roles in cardiovascular, pulmonary, and skeletal muscle physiology. An important action of β 2AR stimulation on skeletal muscle is anabolic growth, which has led to the use of agonists such as clenbuterol by athletes to enhance muscle performance. While previous work has demonstrated that β 2ARs can engage distinct signaling and functional cascades mediated by either G proteins or the multifunctional adaptor protein, β-arrestin, the precise role of β-arrestin in skeletal muscle physiology is not known. Here, we tested the hypothesis that agonist activation of the β 2AR by clenbuterol would engage β-arrestin as a key transducer of anabolic skeletal muscle growth.

          Methods

          The contractile force of isolated extensor digitorum longus muscle (EDL) and calcium signaling in isolated flexor digitorum brevis (FDB) fibers were examined from the wild-type (WT) and β-arrestin 1 knockout mice (βarr1KO) followed by chronic administration of clenbuterol (1 mg/kg/d). Hypertrophic responses including fiber composition and fiber size were examined by immunohistochemical imaging. We performed a targeted phosphoproteomic analysis on clenbuterol stimulated primary cultured myoblasts from WT and βarr1KO mice. Statistical significance was determined by using a two-way analysis with Sidak’s or Tukey’s multiple comparison test and the Student’s t test.

          Results

          Chronic administration of clenbuterol to WT mice enhanced the contractile force of EDL muscle and calcium signaling in isolated FDB fibers. In contrast, when administered to βarr1KO mice, the effect of clenbuterol on contractile force and calcium influx was blunted. While clenbuterol-induced hypertrophic responses were observed in WT mice, this response was abrogated in mice lacking β-arrestin 1. In primary cultured myoblasts, clenbuterol-stimulated phosphorylation of multiple pro-hypertrophy proteins required the presence of β-arrestin 1.

          Conclusions

          We have identified a previously unappreciated role for β-arrestin 1 in mediating β 2AR-stimulated skeletal muscle growth and strength. We propose these findings could have important implications in the design of future pharmacologic agents aimed at reversing pathological conditions associated with skeletal muscle wasting.

          Electronic supplementary material

          The online version of this article (10.1186/s13395-018-0184-8) contains supplementary material, which is available to authorized users.

          Related collections

          Most cited references58

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Rapid Determination of Myosin Heavy Chain Expression in Rat, Mouse, and Human Skeletal Muscle Using Multicolor Immunofluorescence Analysis

          Skeletal muscle is a heterogeneous tissue comprised of fibers with different morphological, functional, and metabolic properties. Different muscles contain varying proportions of fiber types; therefore, accurate identification is important. A number of histochemical methods are used to determine muscle fiber type; however, these techniques have several disadvantages. Immunofluorescence analysis is a sensitive method that allows for simultaneous evaluation of multiple MHC isoforms on a large number of fibers on a single cross-section, and offers a more precise means of identifying fiber types. In this investigation we characterized pure and hybrid fiber type distribution in 10 rat and 10 mouse skeletal muscles, as well as human vastus lateralis (VL) using multicolor immunofluorescence analysis. In addition, we determined fiber type-specific cross-sectional area (CSA), succinate dehydrogenase (SDH) activity, and α-glycerophosphate dehydrogenase (GPD) activity. Using this procedure we were able to easily identify pure and hybrid fiber populations in rat, mouse, and human muscle. Hybrid fibers were identified in all species and made up a significant portion of the total population in some rat and mouse muscles. For example, rat mixed gastrocnemius (MG) contained 12.2% hybrid fibers whereas mouse white tibialis anterior (WTA) contained 12.1% hybrid fibers. Collectively, we outline a simple and time-efficient method for determining MHC expression in skeletal muscle of multiple species. In addition, we provide a useful resource of the pure and hybrid fiber type distribution, fiber CSA, and relative fiber type-specific SDH and GPD activity in a number of rat and mouse muscles.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Role of beta-adrenoceptor signaling in skeletal muscle: implications for muscle wasting and disease.

            The importance of beta-adrenergic signaling in the heart has been well documented, but it is only more recently that we have begun to understand the importance of this signaling pathway in skeletal muscle. There is considerable evidence regarding the stimulation of the beta-adrenergic system with beta-adrenoceptor agonists (beta-agonists). Although traditionally used for treating bronchospasm, it became apparent that some beta-agonists could increase skeletal muscle mass and decrease body fat. These so-called "repartitioning effects" proved desirable for the livestock industry trying to improve feed efficiency and meat quality. Studying beta-agonist effects on skeletal muscle has identified potential therapeutic applications for muscle wasting conditions such as sarcopenia, cancer cachexia, denervation, and neuromuscular diseases, aiming to attenuate (or potentially reverse) the muscle wasting and associated muscle weakness, and to enhance muscle growth and repair after injury. Some undesirable cardiovascular side effects of beta-agonists have so far limited their therapeutic potential. This review describes the physiological significance of beta-adrenergic signaling in skeletal muscle and examines the effects of beta-agonists on skeletal muscle structure and function. In addition, we examine the proposed beneficial effects of beta-agonist administration on skeletal muscle along with some of the less desirable cardiovascular effects. Understanding beta-adrenergic signaling in skeletal muscle is important for identifying new therapeutic targets and identifying novel approaches to attenuate the muscle wasting concomitant with many diseases.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Seven transmembrane receptors: something old, something new.

              Receptors for hormones, neurotransmitters, drugs, sensory stimuli and many other agents represent the gateway to cellular metabolism and activity. They regulate virtually all physiological processes in mammals. Yet as recently as 40 years ago their very existence was still in question. One class of receptors, those coupled to G proteins (also known as GPCRs or seven transmembrane receptors) comprise by far the largest group (approx. 1000), and are the most important target of clinically used drugs. Here I provide a very personal retrospective of research over the past 35 years which ultimately led to the identification, purification, reconstitution and cloning of the adrenergic receptors; the discovery of their homology with the seven transmembrane spanning visual light receptor rhodopsin and the realization that there was a large gene family of G protein coupled receptors; the elucidation of the molecular mechanisms of receptor desensitization and signalling through G protein-coupled receptor kinases and beta-arrestins; and the appreciation that the structure, signalling, and regulatory mechanisms of the receptors are all highly conserved across the large receptor superfamily.
                Bookmark

                Author and article information

                Contributors
                jiheekim@duke.edu
                chad.grotegut@duke.edu
                jim.wisler@duke.edu
                tianyu.li@gmail.com
                lan.mao@duke.edu
                minyongchen@gmail.com
                w.chen@duke.edu
                paul.b.rosenberg@duke.edu
                rockm001@mc.duke.edu
                lefko001@receptor-biol.duke.edu
                Journal
                Skelet Muscle
                Skelet Muscle
                Skeletal Muscle
                BioMed Central (London )
                2044-5040
                27 December 2018
                27 December 2018
                2018
                : 8
                : 39
                Affiliations
                [1 ]ISNI 0000000100241216, GRID grid.189509.c, Department of Medicine, , Duke University Medical Center, ; Durham, NC USA
                [2 ]ISNI 0000000100241216, GRID grid.189509.c, Department of Obstetrics and Gynecology, , Duke University Medical Center, ; Durham, NC USA
                [3 ]ISNI 0000000100241216, GRID grid.189509.c, Department of Medicine, Division of Cardiology and Duke Cardiovascular Physiology Core, , Duke University Medical Center, ; Durham, NC USA
                [4 ]ISNI 0000000100241216, GRID grid.189509.c, Department of Cell Biology, , Duke University Medical Center, ; Durham, NC USA
                [5 ]ISNI 0000000100241216, GRID grid.189509.c, Departments of Molecular Genetics and Microbiology, , Duke University Medical Center, ; Durham, NC USA
                [6 ]ISNI 0000000100241216, GRID grid.189509.c, Department of Biochemistry, , Duke University Medical Center, ; Durham, NC USA
                [7 ]ISNI 0000000100241216, GRID grid.189509.c, Howard Hughes Medical Institute, , Duke University Medical Center, ; Durham, NC USA
                Author information
                http://orcid.org/0000-0003-1472-7545
                Article
                184
                10.1186/s13395-018-0184-8
                6309084
                30591079
                0e7f45fe-8d99-4b3d-8f1e-4afde7681681
                © The Author(s). 2018

                Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License ( http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

                History
                : 21 September 2018
                : 22 November 2018
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/100000050, National Heart, Lung, and Blood Institute;
                Award ID: HL16037
                Funded by: FundRef http://dx.doi.org/10.13039/100000011, Howard Hughes Medical Institute;
                Award ID: None
                Funded by: FundRef http://dx.doi.org/10.13039/100000002, National Institutes of Health;
                Award ID: HD070872
                Award ID: HL133488
                Award ID: DK109911
                Award ID: R01-CA172570
                Award ID: 5K12-CA100639-08
                Award ID: HL056687
                Award ID: HL075443
                Award Recipient :
                Categories
                Research
                Custom metadata
                © The Author(s) 2018

                Rheumatology
                β-arrestin 1,β2ar,clenbuterol,hypertrophy,contractility,skeletal muscle
                Rheumatology
                β-arrestin 1, β2ar, clenbuterol, hypertrophy, contractility, skeletal muscle

                Comments

                Comment on this article