6
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Antianabolic Effects of Hypercapnia: No Country for Strong Men

      editorial

      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

          Elevated levels of CO2 (hypercapnia) are often observed in acute or chronic lung diseases, such as acute respiratory distress syndrome, chronic obstructive pulmonary disease (COPD), and cystic fibrosis, and reflect the effects of alveolar hypoventilation and altered gas exchange (1). In recent years, it has become increasingly evident that high CO2 levels contribute to pulmonary disease states, and that hypercapnia is an independent risk factor and driver of poor outcomes in patients with the above-mentioned diseases (2–4). We now know that levels of CO2 are sensed by various nonexcitable cells via an as-yet-unknown mechanism, leading to the activation of highly specific signaling cascades (5). Importantly, the primarily detrimental effects of hypercapnia are not limited to the lungs, where high CO2 levels impair alveolar epithelial and bronchial airway function. In systemic tissues, high CO2 levels also negatively affect cell proliferation, repair mechanisms, innate immunity, and skeletal muscle function (6–10). Loss of skeletal muscle mass and function, which is often observed in patients with COPD, correlates with increased morbidity and mortality in chronically ill patients (11). Thus, understanding the mechanisms by which CO2 retention contributes to skeletal muscle wasting is not only interesting as a biological phenomenon, it is also important clinically, as interfering with these events may improve outcomes for hypercapnic patients with COPD. In this issue of the Journal, Korponay and colleagues (pp. 74–86) convincingly demonstrate that elevated levels of CO2 decrease protein anabolism in skeletal muscles by decreasing ribosomal biogenesis (12). By performing a pilot study in human quadriceps muscle biopsies from normo- and hypercapnic patients with a history of lung disease, the authors identified a marked reduction of ribosomal 45S pre-RNA in muscles from the hypercapnic patient group, suggesting decreased protein translation. In subsequent experiments, mice were exposed to 10% CO2 (normoxic hypercapnia) or room air (normoxic normocapnia) for 60 days, and the extensor digitorum longus muscle was processed for an unbiased proteomic study. In line with the initial findings in the human cohort, the authors found a marked downregulation of several components of translation initiation in hypercapnic animals accompanied by a downregulation of “structural constituent of ribosome,” as suggested by ontology enrichment analysis. Further in vivo and in vitro studies showed that protein synthesis in muscle fibers and cultured myotubes was significantly reduced during sustained hypercapnic exposure, as assessed by puromycin incorporation. Puromycin is an aminonucleoside antibiotic and a structural analog of aminoacyl-tRNA, and as such can be incorporated into elongating peptide chains via the formation of a peptide bond (13). Thus, the rate at which puromycin-labeled peptides are formed reflects the overall rate of protein synthesis. Skeletal muscle wasting is a hallmark of various lung diseases and particularly of COPD; however, the nature of the mechanisms that drive muscle loss remains a topic of intense debate. Although it is evident that immobility associated with the disease leads to muscle wasting that is in part reversible with physical exercise, recent evidence shows that more specific mechanisms may cause a distinct COPD myopathy (11). Generally, muscle wasting could be a consequence of activated catabolic functions, such as proteasomal degradation and autophagy, or inhibited anabolic pathways. Korponay and colleagues demonstrate antianabolic effects of hypercapnia in skeletal muscle that are driven by AMP-activated protein kinase α2 (AMPKα2). This is of particular interest because AMPKα2 also promotes myotube degradation during elevated CO2 by activating the ubiquitin proteasome system via the E3-ubiquitin ligase MuRF1 (muscle-specific Ring finger protein 1), which directly targets the myosin heavy chain (14). Interestingly, these catabolic effects of hypercapnia appear to be activated earlier than the antianabolic ones, as a previous study showed that ubiquitination-driven muscle degradation was evident after mice were exposed to 21 days of hypercapnia (14). However, the current study shows that this time course is not sufficient to downregulate protein synthesis that is evident after 60 days of hypercapnic exposure. Of note, it was recently shown that autophagy-driven degradation is also significantly enhanced in locomotor muscles of patients with COPD, which is mediated by AMPK as well (15). Thus, hypercapnia drives both antianabolic and catabolic skeletal muscle wasting by at least two (potentially three) distinct mechanisms. Further research is needed to confirm the involvement of autophagy in CO2-induced muscle loss and to identify the relative contributions of these mechanisms to COPD-associated myopathy. Although hypercapnia induces AMPKα2 in myotubes, it activates AMPKα1 in the lung epithelium, where the kinase markedly downregulates the Na,K-ATPase and thereby impairs alveolar epithelial function (9), further highlighting the specificity of the CO2-induced pathways. It is well known that AMPK, a metabolic sensor, is rapidly activated upon cellular stress or starvation, leading to inhibition of energy-demanding (anabolic) pathways and upregulation of catabolic processes to generate ATP (16). Because muscle fibers require a considerable amount of energy and the Na,K-ATPase accounts for ∼40% of the energy needs of a resting cell, it seems logical that to survive, cells exposed to hypercapnia (mal)adapt by downregulating these energy-demanding processes. This may also explain why the hypercapnia-induced downregulation of protein translation was found to be muscle-fiber-type specific and more pronounced in type IIa and IIb/x fibers than in type I fibers in the current study. Skeletal muscles exhibit significant variability depending on their biochemical, mechanical, and metabolic demands. Although it may be an oversimplification, fast-twitch type II fibers generally contract more powerfully and rapidly than type I fibers, and thus require high energy on demand. Clearly, further studies are warranted to tease out the molecular mechanisms responsible for this specificity. To achieve that goal, it will be necessary to precisely characterize the downstream targets of AMPKα2 in the context of hypercapnia. Korponay and colleagues suggest that mTOR (mammalian target of rapamycin) is probably not involved in the hypercapnia-induced and AMPK-driven downregulation of protein synthesis. However, it is well documented that mTOR complex 1 is directly inhibited by AMPK, leading to inhibition of protein synthesis (17). mTOR is required for initiation of translation through its phosphorylation of substrates such as eukaryotic initiation factor (eIF) 4E binding protein 1 (4E-BP1) and p70 ribosomal protein S6 kinase. AMPK may also downregulate protein synthesis by inhibiting Raptor (regulatory-associated protein of mTOR), blocking ribosomal biogenesis through inhibition of TIF-1A (transcription intermediary factor 1 α) and activating eEF2K (eukaryotic elongation factor 2 kinase), thereby blocking the elongation process by phosphorylating eEF2. Thus, the potential involvement of mTOR in the above-mentioned mechanisms may require future research. Similarly, other AMPKα2-dependent but mTOR-independent regulators of protein synthesis will need to be investigated. Finally, as growing evidence suggests that the metabolic activity of cells impacts chromatin modifications and genome accessibility by inducing methylation, acetylation, phosphorylation, ubiquitination, or SUMOylation of histones (18), further research needs to focus on the potential effects of elevated CO2 levels on these mechanisms in the context of skeletal muscle anabolism. The findings of Korponay and colleagues are timely and important, and foster novel research that may ultimately lead to selective interventions against the maladaptive cellular responses to hypercapnia. The current study clearly shows that, as in the lung, hypercapnia is a nonpermissive environment for skeletal muscle—no country for strong men, indeed.

          Related collections

          Most cited references9

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

          Elevated CO(2) levels cause mitochondrial dysfunction and impair cell proliferation.

          Elevated CO(2) concentrations (hypercapnia) occur in patients with severe lung diseases. Here, we provide evidence that high CO(2) levels decrease O(2) consumption and ATP production and impair cell proliferation independently of acidosis and hypoxia in fibroblasts (N12) and alveolar epithelial cells (A549). Cells exposed to elevated CO(2) died in galactose medium as well as when glucose-6-phosphate isomerase was knocked down, suggesting mitochondrial dysfunction. High CO(2) levels led to increased levels of microRNA-183 (miR-183), which in turn decreased expression of IDH2 (isocitrate dehydrogenase 2). The high CO(2)-induced decrease in cell proliferation was rescued by α-ketoglutarate and overexpression of IDH2, whereas proliferation decreased in normocapnic cells transfected with siRNA for IDH2. Also, overexpression of miR-183 decreased IDH2 (mRNA and protein) as well as cell proliferation under normocapnic conditions, whereas inhibition of miR-183 rescued the normal proliferation phenotype in cells exposed to elevated levels of CO(2). Accordingly, we provide evidence that high CO(2) induces miR-183, which down-regulates IDH2, thus impairing mitochondrial function and cell proliferation. These results are of relevance to patients with hypercapnia such as those with chronic obstructive pulmonary disease, asthma, cystic fibrosis, bronchopulmonary dysplasia, and muscular dystrophies.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            AMP-activated protein kinase regulates CO2-induced alveolar epithelial dysfunction in rats and human cells by promoting Na,K-ATPase endocytosis.

            Hypercapnia (elevated CO(2) levels) occurs as a consequence of poor alveolar ventilation and impairs alveolar fluid reabsorption (AFR) by promoting Na,K-ATPase endocytosis. We studied the mechanisms regulating CO(2)-induced Na,K-ATPase endocytosis in alveolar epithelial cells (AECs) and alveolar epithelial dysfunction in rats. Elevated CO(2) levels caused a rapid activation of AMP-activated protein kinase (AMPK) in AECs, a key regulator of metabolic homeostasis. Activation of AMPK was mediated by a CO(2)-triggered increase in intracellular Ca(2+) concentration and Ca(2+)/calmodulin-dependent kinase kinase-beta (CaMKK-beta). Chelating intracellular Ca(2+) or abrogating CaMKK-beta function by gene silencing or chemical inhibition prevented the CO(2)-induced AMPK activation in AECs. Activation of AMPK or overexpression of constitutively active AMPK was sufficient to activate PKC-zeta and promote Na,K-ATPase endocytosis. Inhibition or downregulation of AMPK via adenoviral delivery of dominant-negative AMPK-alpha(1) prevented CO(2)-induced Na,K-ATPase endocytosis. The hypercapnia effects were independent of intracellular ROS. Exposure of rats to hypercapnia for up to 7 days caused a sustained decrease in AFR. Pretreatment with a beta-adrenergic agonist, isoproterenol, or a cAMP analog ameliorated the hypercapnia-induced impairment of AFR. Accordingly, we provide evidence that elevated CO(2) levels are sensed by AECs and that AMPK mediates CO(2)-induced Na,K-ATPase endocytosis and alveolar epithelial dysfunction, which can be prevented with beta-adrenergic agonists and cAMP.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Risk factors for death of patients with cystic fibrosis awaiting lung transplantation.

              The optimal timing for listing of cystic fibrosis patients for lung transplantation is controversial. We conducted a retrospective cohort study of 343 patients listed for lung transplantation at four academic medical centers to identify risk factors for death while awaiting transplantation. Data on possible risk factors were abstracted from medical records. Time to death, patient demographic characteristics, and risk factors for death while awaiting transplantation were assessed. Univariate and multivariate survival analyses were performed using Cox regression. By univariate analyses, FEV1 or = 50 mm Hg (HR, 1.85; 95% CI, 1.1-3.0), and shorter height (HR, 1.8; 95% CI, 1.1-3.0) were associated with a higher risk of death. Referral from an accredited cystic fibrosis center was associated with a lower risk (HR, 0.53; 95% CI, 0.30-0.92). The final multivariate model included referral from an accredited cystic fibrosis center (HR, 0.5; 95% CI, 0.3-1.0) and listing year after 1996 (HR, 0.4; 95% CI, 0.2-0.7); both were associated with a lower risk of death. FEV1 or = 50 mm Hg (HR, 6.9; 95% CI, 1.5-32.1), and use of a nutritional intervention (HR, 2.3; 95% CI, 1.3-4.1) were associated with increased risk. Patients with FEV1 > 30% predicted had a higher risk of death only when their Pa(CO2) was > or = 50 mm Hg (HR, 7.0; 95% CI, 1.5-32), while the increased risk of death with FEV1 < or = 30% was not further influenced by the presence of hypercapnia. We identified risk factors for waiting list mortality that could impact on transplant listing and allocation guidelines.
                Bookmark

                Author and article information

                Journal
                Am J Respir Cell Mol Biol
                Am. J. Respir. Cell Mol. Biol
                ajrcmb
                American Journal of Respiratory Cell and Molecular Biology
                American Thoracic Society
                1044-1549
                1535-4989
                January 2020
                January 2020
                January 2020
                : 62
                : 1
                : 8-9
                Affiliations
                [ 1 ]Department of Internal Medicine

                Justus Liebig University

                Giessen, Germany
                [ 2 ]Universities of Giessen and Marburg Lung Center

                Giessen, Germany
                [ 3 ]German Center for Lung Research

                Giessen, Germany

                and
                [ 4 ]The Cardio-Pulmonary Institute

                Giessen, Germany
                Author information
                http://orcid.org/0000-0003-1370-9783
                Article
                2019-0225ED
                10.1165/rcmb.2019-0225ED
                6938131
                31290693
                7af22237-c280-408c-9fd4-33c286e3948e
                Copyright © 2020 by the American Thoracic Society

                This article is open access and distributed under the terms of the Creative Commons Attribution Non-Commercial No Derivatives License 4.0 ( http://creativecommons.org/licenses/by-nc-nd/4.0/). For commercial usage and reprints, please contact Diane Gern ( dgern@ 123456thoracic.org ).

                History
                Page count
                Figures: 0, Tables: 0, Pages: 2
                Categories
                Editorials

                Comments

                Comment on this article