29
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access
      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

          Metformin, an oral hypoglycemic agent, has been used for decades to treat type 2 diabetes mellitus. Recent studies indicate that mice treated with metformin live longer and have fewer manifestations of age‐related chronic disease. However, the molecular mechanisms underlying this phenotype are unknown. Here, we show that metformin treatment increases the levels of the microRNA‐processing protein DICER1 in mice and in humans with diabetes mellitus. Our results indicate that metformin upregulates DICER1 through a post‐transcriptional mechanism involving the RNA‐binding protein AUF1. Treatment with metformin altered the subcellular localization of AUF1, disrupting its interaction with DICER1 mRNA and rendering DICER1 mRNA stable, allowing DICER1 to accumulate. Consistent with the role of DICER1 in the biogenesis of microRNAs, we found differential patterns of microRNA expression in mice treated with metformin or caloric restriction, two proven life‐extending interventions. Interestingly, several microRNAs previously associated with senescence and aging, including miR‐20a, miR‐34a, miR‐130a, miR‐106b, miR‐125, and let‐7c, were found elevated. In agreement with these findings, treatment with metformin decreased cellular senescence in several senescence models in a DICER1‐dependent manner. Metformin lowered p16 and p21 protein levels and the abundance of inflammatory cytokines and oncogenes that are hallmarks of the senescence‐associated secretory phenotype (SASP). These data lead us to hypothesize that changes in DICER1 levels may be important for organismal aging and to propose that interventions that upregulate DICER1 expression (e.g., metformin) may offer new pharmacotherapeutic approaches for age‐related disease.

          Related collections

          Most cited references30

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

          Impaired microRNA processing enhances cellular transformation and tumorigenesis.

          MicroRNAs (miRNAs) are a new class of small noncoding RNAs that post-transcriptionally regulate the expression of target mRNA transcripts. Many of these target mRNA transcripts are involved in proliferation, differentiation and apoptosis, processes commonly altered during tumorigenesis. Recent work has shown a global decrease of mature miRNA expression in human cancers. However, it is unclear whether this global repression of miRNAs reflects the undifferentiated state of tumors or causally contributes to the transformed phenotype. Here we show that global repression of miRNA maturation promotes cellular transformation and tumorigenesis. Cancer cells expressing short hairpin RNAs (shRNAs) targeting three different components of the miRNA processing machinery showed a substantial decrease in steady-state miRNA levels and a more pronounced transformed phenotype. In animals, miRNA processing-impaired cells formed tumors with accelerated kinetics. These tumors were more invasive than control tumors, suggesting that global miRNA loss enhances tumorigenesis. Furthermore, conditional deletion of Dicer1 enhanced tumor development in a K-Ras-induced mouse model of lung cancer. Overall, these studies indicate that abrogation of global miRNA processing promotes tumorigenesis.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            From birth to death: the complex lives of eukaryotic mRNAs.

            Recent work indicates that the posttranscriptional control of eukaryotic gene expression is much more elaborate and extensive than previously thought, with essentially every step of messenger RNA (mRNA) metabolism being subject to regulation in an mRNA-specific manner. Thus, a comprehensive understanding of eukaryotic gene expression requires an appreciation for how the lives of mRNAs are influenced by a wide array of diverse regulatory mechanisms.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Investigating metformin for cancer prevention and treatment: the end of the beginning.

              Laboratory research and pharmacoepidemiology are providing converging evidence that the widely used antidiabetic drug metformin has antineoplastic activity, but there are caveats. Although population studies suggest that metformin exposure is associated with reduced cancer risk and/or improved prognosis, these data are mostly retrospective and nonrandomized. Laboratory models show antineoplastic activity, but metformin concentrations used in many experiments exceed those achieved with conventional doses used for diabetes treatment. Ongoing translational research should be useful in guiding design of clinical trials, not only to evaluate metformin at conventional antidiabetic doses, where reduction of elevated insulin levels may contribute to antineoplastic activity for certain subsets of patients, but also to explore more aggressive dosing of biguanides, which may lead to reprogramming of energy metabolism in a manner that could provide important opportunities for synthetic lethality through rational drug combinations or in the context of genetic lesions associated with hypersensitivity to energetic stress. There are tantalizing clues that justify the investigation of antineoplastic activities of biguanides. The complexity of their biologic effects requires further translational research to guide clinical trial design.
                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
                17 March 2016
                June 2016
                : 15
                : 3 ( doiID: 10.1111/acel.2016.15.issue-3 )
                : 572-581
                Affiliations
                [ 1 ] Laboratory of Epidemiology and Population Sciences National Institute on AgingNational Institutes of Health 251 Bayview Boulevard Baltimore MD 21224USA
                [ 2 ] Translational Gerontology Branch National Institute on AgingNational Institutes of Health 251 Bayview Boulevard Baltimore MD 21224USA
                [ 3 ] Pancreatic Islet Development and Regeneration Unit Department of Stem CellsCABIMER‐Andalusian Center for Molecular Biology and Regenerative Medicine Avenida Americo Vespucio, Parque Científico y Tecnologico Cartuja 93 41092 SevillaSpain
                [ 4 ] Laboratory of Genetics National Institute on AgingNational Institutes of Health 251 Bayview Boulevard Baltimore MD 21224USA
                Author notes
                [*] [* ] Correspondence

                Michele K. Evans, MD, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA. Tel.: +1 410 558 8573; fax: +1 410 558 8268; e‐mail me42v@ 123456nih.gov

                Article
                ACEL12469
                10.1111/acel.12469
                4854919
                26990999
                a69357b8-37ba-4198-a1f7-9cced4715e53
                Published 2016. This article is a U.S. Government work and is in the public domain in the USA. Aging Cell published by 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
                : 21 February 2016
                Page count
                Pages: 10
                Funding
                Funded by: National Institutes of Health
                Funded by: National Institute on Aging
                Categories
                Original Article
                Original Articles
                Custom metadata
                2.0
                acel12469
                June 2016
                Converter:WILEY_ML3GV2_TO_NLMPMC version:4.9.1 mode:remove_FC converted:10.06.2016

                Cell biology
                aging,auf1,caloric restriction,diabetes mellitus,microrna,rna‐binding proteins
                Cell biology
                aging, auf1, caloric restriction, diabetes mellitus, microrna, rna‐binding proteins

                Comments

                Comment on this article