43
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Deficient nitric oxide signalling impairs skeletal muscle growth and performance: involvement of mitochondrial dysregulation

      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

          Nitric oxide (NO), generated in skeletal muscle mostly by the neuronal NO synthases (nNOSμ), has profound effects on both mitochondrial bioenergetics and muscle development and function. The importance of NO for muscle repair emerges from the observation that nNOS signalling is defective in many genetically diverse skeletal muscle diseases in which muscle repair is dysregulated. How the effects of NO/nNOSμ on mitochondria impact on muscle function, however, has not been investigated yet.

          Methods

          In this study we have examined the relationship between the NO system, mitochondrial structure/activity and skeletal muscle phenotype/growth/functions using a mouse model in which nNOSμ is absent. Also, NO-induced effects and the NO pathway were dissected in myogenic precursor cells.

          Results

          We show that nNOSμ deficiency in mouse skeletal muscle leads to altered mitochondrial bioenergetics and network remodelling, and increased mitochondrial unfolded protein response (UPR mt) and autophagy. The absence of nNOSμ is also accompanied by an altered mitochondrial homeostasis in myogenic precursor cells with a decrease in the number of myonuclei per fibre and impaired muscle development at early stages of perinatal growth. No alterations were observed, however, in the overall resting muscle structure, apart from a reduced specific muscle mass and cross sectional areas of the myofibres. Investigating the molecular mechanisms we found that nNOSμ deficiency was associated with an inhibition of the Akt-mammalian target of rapamycin pathway. Concomitantly, the Akt-FoxO3-mitochondrial E3 ubiquitin protein ligase 1 (Mul-1) axis was also dysregulated. In particular, inhibition of nNOS/NO/cyclic guanosine monophosphate (cGMP)/cGMP-dependent-protein kinases induced the transcriptional activity of FoxO3 and increased Mul-1 expression. nNOSμ deficiency was also accompanied by functional changes in muscle with reduced muscle force, decreased resistance to fatigue and increased degeneration/damage post-exercise.

          Conclusions

          Our results indicate that nNOSμ/NO is required to regulate key homeostatic mechanisms in skeletal muscle, namely mitochondrial bioenergetics and network remodelling, UPR mt and autophagy. These events are likely associated with nNOSμ-dependent impairments of muscle fibre growth resulting in a deficit of muscle performance.

          Electronic supplementary material

          The online version of this article (doi:10.1186/s13395-014-0022-6) contains supplementary material, which is available to authorized users.

          Related collections

          Most cited references76

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

          XBP1 controls diverse cell type- and condition-specific transcriptional regulatory networks.

          Using genome-wide approaches, we have elucidated the regulatory circuitry governed by the XBP1 transcription factor, a key effector of the mammalian unfolded protein response (UPR), in skeletal muscle and secretory cells. We identified a core group of genes involved in constitutive maintenance of ER function in all cell types and tissue- and condition-specific targets. In addition, we identified a cadre of unexpected targets that link XBP1 to neurodegenerative and myodegenerative diseases, as well as to DNA damage and repair pathways. Remarkably, we found that XBP1 regulates functionally distinct targets through different sequence motifs. Further, we identified Mist1, a critical regulator of differentiation, as an important target of XBP1, providing an explanation for developmental defects associated with XBP1 loss of function. Our results provide a detailed picture of the regulatory roadmap governed by XBP1 in distinct cell types as well as insight into unexplored functions of XBP1.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Mitochondrial biogenesis in mammals: the role of endogenous nitric oxide.

            Nitric oxide was found to trigger mitochondrial biogenesis in cells as diverse as brown adipocytes and 3T3-L1, U937, and HeLa cells. This effect of nitric oxide was dependent on guanosine 3',5'-monophosphate (cGMP) and was mediated by the induction of peroxisome proliferator-activated receptor gamma coactivator 1alpha, a master regulator of mitochondrial biogenesis. Moreover, the mitochondrial biogenesis induced by exposure to cold was markedly reduced in brown adipose tissue of endothelial nitric oxide synthase null-mutant (eNOS-/-) mice, which had a reduced metabolic rate and accelerated weight gain as compared to wild-type mice. Thus, a nitric oxide-cGMP-dependent pathway controls mitochondrial biogenesis and body energy balance.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              cGMP-dependent protein kinases and cGMP phosphodiesterases in nitric oxide and cGMP action.

              To date, studies suggest that biological signaling by nitric oxide (NO) is primarily mediated by cGMP, which is synthesized by NO-activated guanylyl cyclases and broken down by cyclic nucleotide phosphodiesterases (PDEs). Effects of cGMP occur through three main groups of cellular targets: cGMP-dependent protein kinases (PKGs), cGMP-gated cation channels, and PDEs. cGMP binding activates PKG, which phosphorylates serines and threonines on many cellular proteins, frequently resulting in changes in activity or function, subcellular localization, or regulatory features. The proteins that are so modified by PKG commonly regulate calcium homeostasis, calcium sensitivity of cellular proteins, platelet activation and adhesion, smooth muscle contraction, cardiac function, gene expression, feedback of the NO-signaling pathway, and other processes. Current therapies that have successfully targeted the NO-signaling pathway include nitrovasodilators (nitroglycerin), PDE5 inhibitors [sildenafil (Viagra and Revatio), vardenafil (Levitra), and tadalafil (Cialis and Adcirca)] for treatment of a number of vascular diseases including angina pectoris, erectile dysfunction, and pulmonary hypertension; the PDE3 inhibitors [cilostazol (Pletal) and milrinone (Primacor)] are used for treatment of intermittent claudication and acute heart failure, respectively. Potential for use of these medications in the treatment of other maladies continues to emerge.
                Bookmark

                Author and article information

                Contributors
                clara.depalma@unimi.it
                federica.morisi@unimi.it
                sarah.pambianco@unimi.it
                emma.assi@unimi.it
                touvier.thierry@hsr.it
                russo-stefania@hotmail.com
                cristiana.perrotta@unimi.it
                vanina.romanello@unipd.it
                silvia.carnio@unipd.it
                valentina.cappello@iit.it
                paolo.pellegrino@unimi.it
                claudia.moscheni@unimi.it
                mariateresa.bassi@lanostrafamiglia.it
                marco.sandri@unipd.it
                d.cervia@unitus.it
                emilio.clementi@unimi.it
                Journal
                Skelet Muscle
                Skelet Muscle
                Skeletal Muscle
                BioMed Central (London )
                2044-5040
                12 December 2014
                12 December 2014
                2014
                : 4
                : 1
                : 22
                Affiliations
                [ ]Unit of Clinical Pharmacology, National Research Council-Institute of Neuroscience, Department of Biomedical and Clinical Sciences “Luigi Sacco”, University Hospital “Luigi Sacco”, Università di Milano, Milano, Italy
                [ ]Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy
                [ ]Dulbecco Telethon Institute at Venetian Institute of Molecular Medicine, Padova, Italy
                [ ]National Research Council-Institute of Neuroscience, Department of Medical Biotechnology and Translational Medicine, Università di Milano, Milano, Italy
                [ ]CNI@NEST, Italian Institute of Technology, Pisa, Italy
                [ ]Unit of Morphology, Department of Biomedical and Clinical Sciences “Luigi Sacco”, Università di Milano, Milano, Italy
                [ ]Department of Biomedical Science, Università di Padova, Padova, Italy
                [ ]Department for Innovation in Biological, Agro-food and Forest Systems, Università della Tuscia, Viterbo, Italy
                Article
                22
                10.1186/s13395-014-0022-6
                4272808
                25530838
                d7cd651d-bf57-4e7a-b31b-0515aacf6ec8
                © De Palma et al.; licensee BioMed Central Ltd. 2014

                This is an Open Access article distributed under the terms of the Creative Commons Attribution License ( http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. 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
                : 26 June 2014
                : 18 November 2014
                Categories
                Research
                Custom metadata
                © The Author(s) 2014

                Rheumatology
                nitric oxide synthase and signalling,mitochondrial bioenergetics,mitochondrial network,unfolded protein response,autophagy,akt-mtor pathway,akt-foxo3-mul-1 axis,fibre growth,muscle structure,muscle exercise

                Comments

                Comment on this article