26
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Enhanced microglial pro‐inflammatory response to lipopolysaccharide correlates with brain infiltration and blood–brain barrier dysregulation in a mouse model of telomere shortening

      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.

          Summary

          Microglia are a proliferative population of resident brain macrophages that under physiological conditions self‐renew independent of hematopoiesis. Microglia are innate immune cells actively surveying the brain and are the earliest responders to injury. During aging, microglia elicit an enhanced innate immune response also referred to as ‘priming’. To date, it remains unknown whether telomere shortening affects the proliferative capacity and induces priming of microglia. We addressed this issue using early (first‐generation G1 mTerc −/−)‐ and late‐generation (third‐generation G3 and G4 mTerc −/−) telomerase‐deficient mice, which carry a homozygous deletion for the telomerase RNA component gene ( mTerc ). Late‐generation mTerc −/− microglia show telomere shortening and decreased proliferation efficiency. Under physiological conditions, gene expression and functionality of G3 mTerc −/− microglia are comparable with microglia derived from G1 mTerc −/− mice despite changes in morphology. However, after intraperitoneal injection of bacterial lipopolysaccharide ( LPS), G3 mTerc −/− microglia mice show an enhanced pro‐inflammatory response. Nevertheless, this enhanced inflammatory response was not accompanied by an increased expression of genes known to be associated with age‐associated microglia priming. The increased inflammatory response in microglia correlates closely with increased peripheral inflammation, a loss of blood–brain barrier integrity, and infiltration of immune cells in the brain parenchyma in this mouse model of telomere shortening.

          Related collections

          Most cited references33

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

          Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a.

          Oncogenic ras can transform most immortal rodent cells to a tumorigenic state. However, transformation of primary cells by ras requires either a cooperating oncogene or the inactivation of tumor suppressors such as p53 or p16. Here we show that expression of oncogenic ras in primary human or rodent cells results in a permanent G1 arrest. The arrest induced by ras is accompanied by accumulation of p53 and p16, and is phenotypically indistinguishable from cellular senescence. Inactivation of either p53 or p16 prevents ras-induced arrest in rodent cells, and E1A achieves a similar effect in human cells. These observations suggest that the onset of cellular senescence does not simply reflect the accumulation of cell divisions, but can be prematurely activated in response to an oncogenic stimulus. Negation of ras-induced senescence may be relevant during multistep tumorigenesis.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The Microglial Sensome Revealed by Direct RNA Sequencing

            Microglia, the principal neuroimmune sentinels of the brain, continuously sense changes in their environment and respond to invading pathogens, toxins and cellular debris. Microglia exhibit plasticity and can assume neurotoxic or neuroprotective priming states that determine their responses to danger. We used direct RNA sequencing, without amplification or cDNA synthesis, to determine the quantitative transcriptomes of microglia of healthy adult and aged mice. We validated our findings by fluorescent dual in-situ hybridization, unbiased proteomic analysis and quantitative PCR. We report here that microglia have a distinct transcriptomic signature and express a unique cluster of transcripts encoding proteins for sensing endogenous ligands and microbes that we term the “sensome”. With aging, sensome transcripts for endogenous ligand recognition are downregulated, whereas those involved in microbe recognition and host defense are upregulated. In addition, aging is associated with an overall increase in expression of microglial genes involved in neuroprotection.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Local self-renewal can sustain CNS microglia maintenance and function throughout adult life.

              Microgliosis is a common response to multiple types of damage in the CNS. However, the origin of the cells involved in this process is still controversial and the relative importance of local expansion versus recruitment of microglia progenitors from the bloodstream is unclear. Here, we investigated the origin of microglia using chimeric animals obtained by parabiosis. We found no evidence of microglia progenitor recruitment from the circulation in denervation or CNS neurodegenerative disease, suggesting that maintenance and local expansion of microglia are solely dependent on the self-renewal of CNS resident cells in these models.
                Bookmark

                Author and article information

                Journal
                Aging Cell
                Aging Cell
                10.1111/(ISSN)1474-9726
                ACEL
                Aging Cell
                John Wiley and Sons Inc. (Hoboken )
                1474-9718
                1474-9726
                03 August 2015
                December 2015
                : 14
                : 6 ( doiID: 10.1111/acel.2015.14.issue-6 )
                : 1003-1013
                Affiliations
                [ 1 ] Section Medical Physiology Department of Neuroscience University of GroningenUniversity Medical Center Groningen Groningen 9713 AVThe Netherlands
                [ 2 ] Department of Critical Care University of GroningenUniversity Medical Center Groningen Groningen 9713 AVThe Netherlands
                [ 3 ] Department of Pathology and Medical Biology University of GroningenUniversity Medical Center Groningen Groningen 9713 AVThe Netherlands
                [ 4 ] Blood‐Brain Barrier Research Group Department of Molecular Cell Biology and Immunology Neuroscience Campus AmsterdamVU University Medical Centre PO Box 7057 1007 MB AmsterdamThe Netherlands
                [ 5 ] Department of Cell Biology European Research Institute on the Biology of Aging University of GroningenUniversity Medical Center Groningen 9713 AVThe Netherlands
                [ 6 ] Department of CardiologyUniversity Medical Center Groningen Groningen 9713 AVThe Netherlands
                [ 7 ] Department of Psychiatry and PsychotherapyUniversity Medical Center Freiburg 79104Germany
                Author notes
                [*] [* ] Correspondence

                Hendrikus W. G. M. Boddeke, Section Medical Physiology, Department of Neuroscience, University of Groningen, University Medical Center Groningen, Groningen 9713 AV, The Netherlands. Tel.: +31 50 363 2701; fax: +31 50 363 2751; e‐mail : h.w.g.m.boddeke@ 123456umcg.nl

                Article
                ACEL12370
                10.1111/acel.12370
                4693462
                26238505
                9b586919-a48d-4376-9bc3-d00ef35fa5c5
                © 2015 The Authors. Aging Cell published by the Anatomical Society and John Wiley & Sons Ltd.

                This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 07 June 2015
                Page count
                Pages: 11
                Funding
                Funded by: NWO
                Award ID: 40‐00506‐98‐9021
                Award ID: 175‐010‐2009‐023
                Funded by: European Community's Seventh Framework Programme (FP7/2007–2013)
                Award ID: HEALTH‐F2‐2010‐ 259893
                Funded by: German Research Council (DFG)
                Award ID: FOR1336
                Award ID: DFG BI 668/5‐1
                Award ID: BI668/2‐2
                Funded by: Mouse Clinic for Cancer and Aging
                Categories
                Original Article
                Original Articles
                Custom metadata
                2.0
                acel12370
                December 2015
                Converter:WILEY_ML3GV2_TO_NLMPMC version:4.7.2 mode:remove_FC converted:22.12.2015

                Cell biology
                aging,blood–brain barrier,microglia,neuroimmune response,priming,telomere,telomerase
                Cell biology
                aging, blood–brain barrier, microglia, neuroimmune response, priming, telomere, telomerase

                Comments

                Comment on this article