50
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Arterial “inflammaging” drives vascular calcification in children on dialysis

      research-article

      Read this article at

      ScienceOpenPublisherPMC
      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

          Children on dialysis have a cardiovascular mortality risk equivalent to older adults in the general population, and rapidly develop medial vascular calcification, an age-associated pathology. We hypothesized that premature vascular ageing contributes to calcification in children with advanced chronic kidney disease (CKD). Vessels from children with Stage 5 CKD with and without dialysis had evidence of increased oxidative DNA damage. The senescence markers p16 and p21 were also increased in vessels from children on dialysis. Treatment of vessel rings ex vivo with calcifying media increased oxidative DNA damage in vessels from children with Stage 5 CKD, but not in those from healthy controls. Vascular smooth muscle cells cultured from children on dialysis exhibited persistent DNA damage, impaired DNA damage repair, and accelerated senescence. Under calcifying conditions vascular smooth muscle cells from children on dialysis showed increased osteogenic differentiation and calcification. These changes correlated with activation of the senescence-associated secretory phenotype (SASP), an inflammatory phenotype characterized by the secretion of proinflammatory cytokines and growth factors. Blockade of ataxia-telangiectasia mutated (ATM)-mediated DNA damage signaling reduced both inflammation and calcification. Clinically, children on dialysis had elevated circulating levels of osteogenic SASP factors that correlated with increased vascular stiffness and coronary artery calcification. These data imply that dysregulated mineral metabolism drives vascular “inflammaging” by promoting oxidative DNA damage, premature senescence, and activation of a pro-inflammatory SASP. Drugs that target DNA damage signaling or eliminate senescent cells may have the potential to prevent vascular calcification in patients with advanced CKD.

          Related collections

          Most cited references64

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

          The senescence-associated secretory phenotype: the dark side of tumor suppression.

          Cellular senescence is a tumor-suppressive mechanism that permanently arrests cells at risk for malignant transformation. However, accumulating evidence shows that senescent cells can have deleterious effects on the tissue microenvironment. The most significant of these effects is the acquisition of a senescence-associated secretory phenotype (SASP) that turns senescent fibroblasts into proinflammatory cells that have the ability to promote tumor progression.
            Bookmark
            • 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: not found
              • Article: not found

              Clinical epidemiology of cardiovascular disease in chronic renal disease.

                Bookmark

                Author and article information

                Journal
                0323470
                Kidney Int
                Kidney Int.
                Kidney international
                0085-2538
                1523-1755
                01 April 2019
                01 March 2019
                31 July 2019
                06 August 2019
                : 95
                : 4
                : 958-972
                Affiliations
                [1 ]British Heart Foundation Centre of Excellence, Cardiovascular Division, King’s College London, London, UK
                [2 ]Developmental Biology and Cancer Programme, Great Ormond Street Hospital and University College London Institute of Child Health, London, UK
                [3 ]Nephrology Unit, Great Ormond Street Hospital and University College London Institute of Child Health, London, UK
                Author notes
                Correspondence: Catherine M. Shanahan, King’s College London, Cardio-vascular Division, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK. cathy.shanahan@ 123456kcl.ac.uk
                [5]

                Current affiliation: Son Espases Hospital, Health Research Institute of Balearic Islands, Palma de Mallorca, Spain.

                Article
                EMS83890
                10.1016/j.kint.2018.12.014
                6684370
                30827513
                fe0fa57a-557b-4e9a-ab81-530d1cfeacb3

                This is an open access article under the CC BY-NC-ND license ( http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                Categories
                Article

                Nephrology
                aging,calcification,dialysis,senescence,vascular smooth muscle cells
                Nephrology
                aging, calcification, dialysis, senescence, vascular smooth muscle cells

                Comments

                Comment on this article