16
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      DNA damage response at telomeres contributes to lung aging and chronic obstructive pulmonary disease

      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

          Cellular senescence has been associated with the structural and functional decline observed during physiological lung aging and in chronic obstructive pulmonary disease (COPD). Airway epithelial cells are the first line of defense in the lungs and are important to COPD pathogenesis. However, the mechanisms underlying airway epithelial cell senescence, and particularly the role of telomere dysfunction in this process, are poorly understood. We aimed to investigate telomere dysfunction in airway epithelial cells from patients with COPD, in the aging murine lung and following cigarette smoke exposure. We evaluated colocalization of γ-histone protein 2A.X and telomeres and telomere length in small airway epithelial cells from patients with COPD, during murine lung aging, and following cigarette smoke exposure in vivo and in vitro. We found that telomere-associated DNA damage foci increase in small airway epithelial cells from patients with COPD, without significant telomere shortening detected. With age, telomere-associated foci increase in small airway epithelial cells of the murine lung, which is accelerated by cigarette smoke exposure. Moreover, telomere-associated foci predict age-dependent emphysema, and late-generation Terc null mice, which harbor dysfunctional telomeres, show early-onset emphysema. We found that cigarette smoke accelerates telomere dysfunction via reactive oxygen species in vitro and may be associated with ataxia telangiectasia mutated-dependent secretion of inflammatory cytokines interleukin-6 and -8. We propose that telomeres are highly sensitive to cigarette smoke-induced damage, and telomere dysfunction may underlie decline of lung function observed during aging and in COPD.

          Related collections

          Most cited references36

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

          Persistent DNA damage signaling triggers senescence-associated inflammatory cytokine secretion

          Cellular senescence suppresses cancer by stably arresting the proliferation of damaged cells1. Paradoxically, senescent cells also secrete factors that alter tissue microenvironments2. The pathways regulating this secretion are unknown. We show that damaged human cells develop persistent chromatin lesions bearing hallmarks of DNA double-strand breaks (DSBs), which initiate increased secretion of inflammatory cytokines such as interleukin-6 (IL-6). Cytokine secretion occurred only after establishment of persistent DNA damage signaling, usually associated with senescence, not after transient DNA damage responses (DDR). Initiation and maintenance of this cytokine response required the DDR proteins ATM, NBS1 and CHK2, but not the cell cycle arrest enforcers p53 and pRb. ATM was also essential for IL-6 secretion during oncogene-induced senescence and by damaged cells that bypass senescence. Further, DDR activity and IL-6 were elevated in human cancers, and ATM-depletion suppressed the ability of senescent cells to stimulate IL-6-dependent cancer cell invasiveness. Thus, in addition to orchestrating cell cycle checkpoints and DNA repair, a novel and important role of the DDR is to allow damaged cells to communicate their compromised state to the surrounding tissue.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Chemokine signaling via the CXCR2 receptor reinforces senescence.

            Cells enter senescence, a state of stable proliferative arrest, in response to a variety of cellular stresses, including telomere erosion, DNA damage, and oncogenic signaling, which acts as a barrier against malignant transformation in vivo. To identify genes controlling senescence, we conducted an unbiased screen for small hairpin RNAs that extend the life span of primary human fibroblasts. Here, we report that knocking down the chemokine receptor CXCR2 (IL8RB) alleviates both replicative and oncogene-induced senescence (OIS) and diminishes the DNA-damage response. Conversely, ectopic expression of CXCR2 results in premature senescence via a p53-dependent mechanism. Cells undergoing OIS secrete multiple CXCR2-binding chemokines in a program that is regulated by the NF-kappaB and C/EBPbeta transcription factors and coordinately induce CXCR2 expression. CXCR2 upregulation is also observed in preneoplastic lesions in vivo. These results suggest that senescent cells activate a self-amplifying secretory network in which CXCR2-binding chemokines reinforce growth arrest.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Telomere shortening and tumor formation by mouse cells lacking telomerase RNA.

              To examine the role of telomerase in normal and neoplastic growth, the telomerase RNA component (mTR) was deleted from the mouse germline. mTR-/- mice lacked detectable telomerase activity yet were viable for the six generations analyzed. Telomerase-deficient cells could be immortalized in culture, transformed by viral oncogenes, and generated tumors in nude mice following transformation. Telomeres were shown to shorten at a rate of 4.8+/-2.4 kb per mTR-/- generation. Cells from the fourth mTR-/- generation onward possessed chromosome ends lacking detectable telomere repeats, aneuploidy, and chromosomal abnormalities, including end-to-end fusions. These results indicate that telomerase is essential for telomere length maintenance but is not required for establishment of cell lines, oncogenic transformation, or tumor formation in mice.
                Bookmark

                Author and article information

                Journal
                Am J Physiol Lung Cell Mol Physiol
                Am. J. Physiol. Lung Cell Mol. Physiol
                ajplung
                ajplung
                AJPLUNG
                American Journal of Physiology - Lung Cellular and Molecular Physiology
                American Physiological Society (Bethesda, MD )
                1040-0605
                1522-1504
                18 September 2015
                15 November 2015
                18 September 2015
                : 309
                : 10
                : L1124-L1137
                Affiliations
                [1] 1Newcastle University Institute for Ageing, Institute for Cell and Molecular Biosciences, Newcastle upon Tyne, United Kingdom;
                [2] 2Lung Immunobiology Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom;
                [3] 3Institute of Transplantation, Freeman Hospital, Newcastle upon Tyne, United Kingdom;
                [4] 4Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom;
                [5] 5Respiratory Pharmacology, Airway Disease Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom; and
                [6] 6Department of Pathology, Newcastle upon Tyne Hospitals Trust, Newcastle upon Tyne, United Kingdom
                Author notes
                [*]

                A. De Soyza and J. F. Passos contributed equally to this work.

                Address for reprint requests and other correspondence: J. F. Passos, Institute for Cell and Molecular Biosciences, Newcastle Univ. Institute for Ageing, Campus for Ageing and Vitality, Newcastle Univ., Newcastle upon Tyne NE4 5PL, UK (e-mail: joao.passos@ 123456newcastle.ac.uk ).
                Article
                L-00293-2015
                10.1152/ajplung.00293.2015
                4652155
                26386121
                40385521-92d9-4a8d-aeb9-ad5fa085b818
                Copyright © 2015 the American Physiological Society

                Licensed under Creative Commons Attribution CC-BY 3.0: © the American Physiological Society.

                History
                : 24 August 2015
                : 14 September 2015
                Funding
                Funded by: 501100000268 Biotechnology and Biological Sciences Research Council (BBSRC)
                Award ID: BB/K017314/1
                Categories
                Call for Papers
                Biomarkers in Lung Diseases: from Pathogenesis to Prediction to New Therapies

                Anatomy & Physiology
                senescence,airway epithelial cells,cigarette smoke
                Anatomy & Physiology
                senescence, airway epithelial cells, cigarette smoke

                Comments

                Comment on this article