10
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Transcription factor-7–like 2 ( TCF7L2) gene acts downstream of the Lkb1/ Stk11 kinase to control mTOR signaling, β cell growth, and insulin secretion

      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.

          Abstract

          Variants in the transcription factor-7–like 2 ( TCF7L2/ TCF4) gene, involved in Wnt signaling, are associated with type 2 diabetes. Loss of Tcf7l2 selectively from the β cell in mice has previously been shown to cause glucose intolerance and to lower β cell mass. Deletion of the tumor suppressor liver kinase B1 (LKB1/STK11) leads to β cell hyperplasia and enhanced glucose-stimulated insulin secretion, providing a convenient genetic model for increased β cell growth and function. The aim of this study was to explore the possibility that Tcf7l2 may be required for the effects of Lkb1 deletion on insulin secretion in the mouse β cell. Mice bearing floxed Lkb1 and/or Tcf7l2 alleles were bred with knockin mice bearing Cre recombinase inserted at the Ins1 locus ( Ins1Cre), allowing highly β cell–selective deletion of either or both genes. Oral glucose tolerance was unchanged by the further deletion of a single Tcf7l2 allele in these cells. By contrast, mice lacking both Tcf7l2 alleles on this background showed improved oral glucose tolerance and insulin secretion in vivo and in vitro compared with mice lacking a single Tcf7l2 allele. Biallelic Tcf7l2 deletion also enhanced β cell proliferation, increased β cell mass, and caused changes in polarity as revealed by the “rosette-like” arrangement of β cells. Tcf7l2 deletion also increased signaling by mammalian target of rapamycin (mTOR), augmenting phospho-ribosomal S6 levels. We identified a novel signaling mechanism through which a modifier gene, Tcf7l2, lies on a pathway through which LKB1 acts in the β cell to restrict insulin secretion.

          Related collections

          Most cited references37

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

          Peutz-Jeghers syndrome is caused by mutations in a novel serine threonine kinase.

          Peutz-Jeghers (PJ) syndrome is an autosomal-dominant disorder characterized by melanocytic macules of the lips, multiple gastrointestinal hamartomatous polyps and an increased risk for various neoplasms, including gastrointestinal cancer. The PJ gene was recently mapped to chromosome 19p13.3 by linkage analysis, with the highest lod score at marker D19S886. In a distance of 190 kb proximal to D19S886, we identified and characterized a novel human gene encoding the serine threonine kinase STK11. In a three-generation PJ family, we found an STK11 allele with a deletion of exons 4 and 5 and an inversion of exons 6 and 7 segregating with the disease. Sequence analysis of STK11 exons in four unrelated PJ patients has identified three nonsense and one acceptor splice site mutations. All five germline mutations are predicted to disrupt the function of the kinase domain. We conclude that germline mutations in STK11, probably in conjunction with acquired genetic defects of the second allele in somatic cells, cause the manifestations of PJ syndrome.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Adaptive changes in pancreatic beta cell fractional area and beta cell turnover in human pregnancy

            Aims/hypothesis We sought to establish the extent and basis for adaptive changes in beta cell numbers in human pregnancy. Methods Pancreas was obtained at autopsy from women who had died while pregnant (n = 18), post-partum (n = 6) or were not pregnant at or shortly before death (controls; n = 20). Pancreases were evaluated for fractional pancreatic beta cell area, islet size and islet fraction of beta cells, beta cell replication (Ki67) and apoptosis (TUNEL), and indirect markers of beta cell neogenesis (insulin-positive cells in ducts and scattered beta cells in pancreas). Results The pancreatic fractional beta cell area was increased by ∼1.4-fold in human pregnancy, with no change in mean beta cell size. In pregnancy there were more small islets rather than an increase in islet size or beta cells per islet. No increase in beta cell replication or change in beta cell apoptosis was detected, but duct cells positive for insulin and scattered beta cells were increased with pregnancy. Conclusions/interpretation The adaptive increase in beta cell numbers in human pregnancy is not as great as in most reports in rodents. This increase in humans is achieved by increased numbers of beta cells in apparently new small islets, rather than duplication of beta cells in existing islets, which is characteristic of pregnancy in rodents. Electronic supplementary material The online version of this article (doi:10.1007/s00125-010-1809-6) contains supplementary material, which is available to authorised users.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Pancreatic β-cell identity, glucose sensing and the control of insulin secretion.

              Insulin release from pancreatic β-cells is required to maintain normal glucose homoeostasis in man and many other animals. Defective insulin secretion underlies all forms of diabetes mellitus, a disease currently reaching epidemic proportions worldwide. Although the destruction of β-cells is responsible for Type 1 diabetes (T1D), both lowered β-cell mass and loss of secretory function are implicated in Type 2 diabetes (T2D). Emerging results suggest that a functional deficiency, involving de-differentiation of the mature β-cell towards a more progenitor-like state, may be an important driver for impaired secretion in T2D. Conversely, at least in rodents, reprogramming of islet non-β to β-cells appears to occur spontaneously in models of T1D, and may occur in man. In the present paper, we summarize the biochemical properties which define the 'identity' of the mature β-cell as a glucose sensor par excellence. In particular, we discuss the importance of suppressing a group of 11 'disallowed' housekeeping genes, including Ldha and the monocarboxylate transporter Mct1 (Slc16a1), for normal nutrient sensing. We then survey the changes in the expression and/or activity of β-cell-enriched transcription factors, including FOXO1, PDX1, NKX6.1, MAFA and RFX6, as well as non-coding RNAs, which may contribute to β-cell de-differentiation and functional impairment in T2D. The relevance of these observations for the development of new approaches to treat T1D and T2D is considered.
                Bookmark

                Author and article information

                Journal
                J Biol Chem
                J. Biol. Chem
                jbc
                jbc
                JBC
                The Journal of Biological Chemistry
                American Society for Biochemistry and Molecular Biology (11200 Rockville Pike, Suite 302, Rockville, MD 20852-3110, U.S.A. )
                0021-9258
                1083-351X
                7 September 2018
                2 July 2018
                2 July 2018
                : 293
                : 36
                : 14178-14189
                Affiliations
                [1]From the Section of Cell Biology and Functional Genomics and Pancreatic Islet and Diabetes Consortium, Division of Diabetes, Endocrinology and Metabolism, Imperial Centre for Translational and Experimental Medicine, Department of Medicine, Imperial College London, London W12 0NN, United Kingdom
                Author notes
                [1 ]To whom correspondence should be addressed: Imperial College London, Hammersmith Hospital campus, DuCane Rd., London W12 0NN, United Kingdom. Tel.: 44-020759-43351; E-mail: g.rutter@ 123456imperial.ac.uk .

                Edited by Jeffrey E. Pessin

                Article
                RA118.003613
                10.1074/jbc.RA118.003613
                6130960
                29967064
                05c8b8e7-5711-47e7-b1dc-ff04d09b8d97
                © 2018 Nguyen-Tu et al.

                Author's Choice—Final version open access under the terms of the Creative Commons CC-BY license.

                History
                : 20 April 2018
                : 15 June 2018
                Funding
                Funded by: RCUK | Medical Research Council (MRC) , open-funder-registry 10.13039/501100000265;
                Award ID: MR/J0003042/1
                Funded by: Wellcome Trust , open-funder-registry 10.13039/100004440;
                Award ID: WT098424AIA
                Funded by: Diabetes UK , open-funder-registry 10.13039/501100000361;
                Award ID: BDA11/0004210
                Funded by: Royal Society , open-funder-registry 10.13039/501100000288;
                Award ID: Wolfson Research Merit Award
                Categories
                Editors' Picks

                Biochemistry
                pancreatic islet,insulin secretion,cell growth,liver kinase b1 (lkb1),t-cell factor (tcf),pancreatic β cell,tcf7l2

                Comments

                Comment on this article