11
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Generation and Characterization of a Novel Mouse Model That Allows Spatiotemporal Quantification of Pancreatic β-Cell Proliferation

      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

          Pancreatic β-cell proliferation has been gaining much attention as a therapeutic target for the prevention and treatment of diabetes. In order to evaluate potential β-cell mitogens, accurate and reliable methods for the detection and quantification of the β-cell proliferation rate are indispensable. In this study, we developed a novel tool that specifically labels replicating β-cells as mVenus + cells by using RIP-Cre; R26Fucci2aR mice expressing the fluorescent ubiquitination-based cell cycle indicator Fucci2a in β-cells. In response to β-cell proliferation stimuli, such as insulin receptor antagonist S961 and diet-induced obesity (DIO), the number of 5-ethynyl-2′-deoxyuridine-positive insulin + cells per insulin + cells and the number of mVenus + cells per mCherry + mVenus cells + mCherry mVenus + cells were similarly increased in these mice. Three-dimensional imaging of optically cleared pancreas tissue from these mice enabled quantification of replicating β-cells in the islets and morphometric analysis of the islets after known mitogenic interventions such as S961, DIO, pregnancy, and partial pancreatectomy. Thus, this novel mouse line is a powerful tool for spatiotemporal analysis and quantification of β-cell proliferation in response to mitogenic stimulation.

          Related collections

          Most cited references15

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

          Visualizing spatiotemporal dynamics of multicellular cell-cycle progression.

          The cell-cycle transition from G1 to S phase has been difficult to visualize. We have harnessed antiphase oscillating proteins that mark cell-cycle transitions in order to develop genetically encoded fluorescent probes for this purpose. These probes effectively label individual G1 phase nuclei red and those in S/G2/M phases green. We were able to generate cultured cells and transgenic mice constitutively expressing the cell-cycle probes, in which every cell nucleus exhibits either red or green fluorescence. We performed time-lapse imaging to explore the spatiotemporal patterns of cell-cycle dynamics during the epithelial-mesenchymal transition of cultured cells, the migration and differentiation of neural progenitors in brain slices, and the development of tumors across blood vessels in live mice. These mice and cell lines will serve as model systems permitting unprecedented spatial and temporal resolution to help us better understand how the cell cycle is coordinated with various biological events.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Serotonin Regulates Pancreatic β-Cell Mass during Pregnancy

            During pregnancy, the energy requirements of the fetus impose changes in maternal metabolism. Increasing insulin resistance in the mother maintains nutrient flow to the growing fetus, while prolactin and placental lactogen counterbalance this resistance and prevent maternal hyperglycemia by driving expansion of the maternal population of insulin-producing β-cells1–3. However, the exact mechanisms by which the lactogenic hormones drive β-cell expansion remain uncertain. Here we show that serotonin acts downstream of lactogen signaling to drive β-cell proliferation. Serotonin synthetic enzyme Tph1 and serotonin production increased sharply in β-cells during pregnancy or after treatment with lactogens in vitro. Inhibition of serotonin synthesis by dietary tryptophan restriction or Tph inhibition blocked β-cell expansion and induced glucose intolerance in pregnant mice without affecting insulin sensitivity. Expression of the Gαq-linked serotonin receptor Htr2b in maternal islets increased during pregnancy and normalized just prior to parturition, while expression of the Gαi-linked receptor Htr1d increased at the end of pregnancy and postpartum. Blocking Htr2b signaling in pregnant mice also blocked β-cell expansion and caused glucose intolerance. These studies reveal an integrated signaling pathway linking β-cell mass to anticipated insulin need during pregnancy. Modulators of this pathway, including medications and diet, may affect the risk of gestational diabetes4.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Expansion of beta-cell mass in response to pregnancy.

              Inadequate beta-cell mass can lead to insulin insufficiency and diabetes. During times of prolonged metabolic demand for insulin, the endocrine pancreas can respond by increasing beta-cell mass, both by increasing cell size and by changing the balance between beta-cell proliferation and apoptosis. In this paper, we review recent advances in our understanding of the mechanisms that control the adaptive expansion of beta-cell mass, focusing on the islet's response to pregnancy, a physiological state of insulin resistance. Functional characterization of factors controlling both beta-cell proliferation and survival might not only lead to the development of successful therapeutic strategies to enhance the response of the beta-cell to increased metabolic loads, but also improve islet transplantation regimens. Copyright 2009 Elsevier Ltd. All rights reserved.
                Bookmark

                Author and article information

                Journal
                Diabetes
                Diabetes
                diabetes
                diabetes
                Diabetes
                Diabetes
                American Diabetes Association
                0012-1797
                1939-327X
                November 2020
                07 August 2020
                07 August 2020
                : 69
                : 11
                : 2340-2351
                Affiliations
                [1] 1Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Kyoto, Japan
                [2] 2Department of Diabetes and Endocrinology, Gifu University Graduate School of Medicine, Gifu, Japan
                [3] 3Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
                Author notes
                Corresponding author: Nobuya Inagaki, inagaki@ 123456kuhp.kyoto-u.ac.jp
                Author information
                https://orcid.org/0000-0001-8261-2593
                Article
                200290
                10.2337/db20-0290
                7576556
                32769118
                f52cdd8c-9dca-4a96-a9a7-dab125d52362
                © 2020 by the American Diabetes Association

                Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/content/license.

                History
                : 24 March 2020
                : 02 August 2020
                Page count
                Figures: 8, Tables: 0, Equations: 0, References: 15, Pages: 12
                Categories
                Islet Studies

                Endocrinology & Diabetes
                Endocrinology & Diabetes

                Comments

                Comment on this article