8
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Dopamine D2 Receptor-Mediated Regulation of Pancreatic β Cell Mass

      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

          Understanding the molecular mechanisms that regulate β cell mass and proliferation is important for the treatment of diabetes. Here, we identified domperidone (DPD), a dopamine D2 receptor (DRD2) antagonist that enhances β cell mass. Over time, islet β cell loss occurs in dissociation cultures, and this was inhibited by DPD. DPD increased proliferation and decreased apoptosis of β cells through increasing intracellular cAMP. DPD prevented β cell dedifferentiation, which together highly contributed to the increased β cell mass. DRD2 knockdown phenocopied the effects of domperidone and increased the number of β cells. Drd2 overexpression sensitized the dopamine responsiveness of β cells and increased apoptosis. Further analysis revealed that the adenosine agonist 5′-N-ethylcarboxamidoadenosine, a previously identified promoter of β cell proliferation, acted with DPD to increase the number of β cells. In humans, dopamine also modulates β cell mass through DRD2 and exerts an inhibitory effect on adenosine signaling.

          Graphical Abstract

          Highlights

          • Dopamine inhibits dedifferentiation and proliferation, and promotes apoptosis of β cells

          • Dopamine confers its action through binding to DRD2 and decreasing cAMP

          • Dopamine-DRD2 signal also functions through interaction with adenosine-ADORA2A signal

          • Dopamine-DRD2-cAMP signal is a potential target for β cell regeneration

          Abstract

          In this article, Kume and colleagues report on the identification of a dopamine D2 receptor (DRD2) antagonist as a promoter to increase β cell mass. Blockade of dopamine-DRD2 signal prevents dedifferentiation, increases proliferation, and decreases apoptosis of β cells. Dopamine-DRD2 signaling also negatively regulates other signaling pathways such as adenosine-ADORA2A signaling through heterodimer formation.

          Related collections

          Most cited references35

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

          Pancreatic β cell dedifferentiation as a mechanism of diabetic β cell failure.

          Diabetes is associated with β cell failure. But it remains unclear whether the latter results from reduced β cell number or function. FoxO1 integrates β cell proliferation with adaptive β cell function. We interrogated the contribution of these two processes to β cell dysfunction, using mice lacking FoxO1 in β cells. FoxO1 ablation caused hyperglycemia with reduced β cell mass following physiologic stress, such as multiparity and aging. Surprisingly, lineage-tracing experiments demonstrated that loss of β cell mass was due to β cell dedifferentiation, not death. Dedifferentiated β cells reverted to progenitor-like cells expressing Neurogenin3, Oct4, Nanog, and L-Myc. A subset of FoxO1-deficient β cells adopted the α cell fate, resulting in hyperglucagonemia. Strikingly, we identify the same sequence of events as a feature of different models of murine diabetes. We propose that dedifferentiation trumps endocrine cell death in the natural history of β cell failure and suggest that treatment of β cell dysfunction should restore differentiation, rather than promoting β cell replication. Copyright © 2012 Elsevier Inc. All rights reserved.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Beta cells can be generated from endogenous progenitors in injured adult mouse pancreas.

            Novel strategies in diabetes therapy would obviously benefit from the use of beta (beta) cell stem/progenitor cells. However, whether or not adult beta cell progenitors exist is one of the most controversial issues in today's diabetes research. Guided by the expression of Neurogenin 3 (Ngn3), the earliest islet cell-specific transcription factor in embryonic development, we show that beta cell progenitors can be activated in injured adult mouse pancreas and are located in the ductal lining. Differentiation of the adult progenitors is Ngn3 dependent and gives rise to all islet cell types, including glucose responsive beta cells that subsequently proliferate, both in situ and when cultured in embryonic pancreas explants. Multipotent progenitor cells thus exist in the pancreas of adult mice and can be activated cell autonomously to increase the functional beta cell mass by differentiation and proliferation rather than by self-duplication of pre-existing beta cells only.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              The ectopic expression of Pax4 in the mouse pancreas converts progenitor cells into alpha and subsequently beta cells.

              We have previously reported that the loss of Arx and/or Pax4 gene activity leads to a shift in the fate of the different endocrine cell subtypes in the mouse pancreas, without affecting the total endocrine cell numbers. Here, we conditionally and ectopically express Pax4 using different cell-specific promoters and demonstrate that Pax4 forces endocrine precursor cells, as well as mature alpha cells, to adopt a beta cell destiny. This results in a glucagon deficiency that provokes a compensatory and continuous glucagon+ cell neogenesis requiring the re-expression of the proendocrine gene Ngn3. However, the newly formed alpha cells fail to correct the hypoglucagonemia since they subsequently acquire a beta cell phenotype upon Pax4 ectopic expression. Notably, this cycle of neogenesis and redifferentiation caused by ectopic expression of Pax4 in alpha cells is capable of restoring a functional beta cell mass and curing diabetes in animals that have been chemically depleted of beta cells.
                Bookmark

                Author and article information

                Contributors
                Journal
                Stem Cell Reports
                Stem Cell Reports
                Stem Cell Reports
                Elsevier
                2213-6711
                30 June 2016
                12 July 2016
                30 June 2016
                : 7
                : 1
                : 95-109
                Affiliations
                [1 ]Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259-B-25 Nagatsuta, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
                [2 ]Division of Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Honjo 2-2-1, Kumamoto 860-0811, Japan
                [3 ]Department of Chemical Biology, Institute for Chemical Research, Institute for Integrated Cell-Material Sciences, Kyoto University, Uji, Kyoto 611-0011, Japan
                [4 ]Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe Street, Mizuho, Nagoya 467-8603, Japan
                Author notes
                []Corresponding author skume@ 123456bio.titech.ac.jp
                Article
                S2213-6711(16)30070-4
                10.1016/j.stemcr.2016.05.015
                4944721
                27373926
                61adebd2-5fb5-45d1-a20e-c128db5919d1
                © 2016 The Author(s)

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

                History
                : 18 August 2015
                : 27 May 2016
                : 31 May 2016
                Categories
                Article

                pancreas,insulin,proliferation,apoptosis,dedifferentiation,diabetes

                Comments

                Comment on this article