30
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Changes in the expression of the type 2 diabetes-associated gene VPS13C in the β-cell are associated with glucose intolerance in humans and mice

      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

          Single nucleotide polymorphisms (SNPs) close to the VPS13C, C2CD4A and C2CD4B genes on chromosome 15q are associated with impaired fasting glucose and increased risk of type 2 diabetes. eQTL analysis revealed an association between possession of risk (C) alleles at a previously implicated causal SNP, rs7163757, and lowered VPS13C and C2CD4A levels in islets from female ( n = 40, P < 0.041) but not from male subjects. Explored using promoter-reporter assays in β-cells and other cell lines, the risk variant at rs7163757 lowered enhancer activity. Mice deleted for Vps13c selectively in the β-cell were generated by crossing animals bearing a floxed allele at exon 1 to mice expressing Cre recombinase under Ins1 promoter control (Ins1Cre). Whereas Vps13c fl/fl:Ins1Cre (βVps13cKO) mice displayed normal weight gain compared with control littermates, deletion of Vps13c had little effect on glucose tolerance. Pancreatic histology revealed no significant change in β-cell mass in KO mice vs. controls, and glucose-stimulated insulin secretion from isolated islets was not altered in vitro between control and βVps13cKO mice. However, a tendency was observed in female null mice for lower insulin levels and β-cell function (HOMA-B) in vivo. Furthermore, glucose-stimulated increases in intracellular free Ca 2+ were significantly increased in islets from female KO mice, suggesting impaired Ca 2+ sensitivity of the secretory machinery. The present data thus provide evidence for a limited role for changes in VPS13C expression in conferring altered disease risk at this locus, particularly in females, and suggest that C2CD4A may also be involved.

          Related collections

          Most cited references56

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

          A genome-wide association study identifies novel risk loci for type 2 diabetes.

          Type 2 diabetes mellitus results from the interaction of environmental factors with a combination of genetic variants, most of which were hitherto unknown. A systematic search for these variants was recently made possible by the development of high-density arrays that permit the genotyping of hundreds of thousands of polymorphisms. We tested 392,935 single-nucleotide polymorphisms in a French case-control cohort. Markers with the most significant difference in genotype frequencies between cases of type 2 diabetes and controls were fast-tracked for testing in a second cohort. This identified four loci containing variants that confer type 2 diabetes risk, in addition to confirming the known association with the TCF7L2 gene. These loci include a non-synonymous polymorphism in the zinc transporter SLC30A8, which is expressed exclusively in insulin-producing beta-cells, and two linkage disequilibrium blocks that contain genes potentially involved in beta-cell development or function (IDE-KIF11-HHEX and EXT2-ALX4). These associations explain a substantial portion of disease risk and constitute proof of principle for the genome-wide approach to the elucidation of complex genetic traits.
            Bookmark
            • Record: found
            • Abstract: not found
            • Article: not found

            Diabetes and vascular disease: pathophysiology, clinical consequences, and medical therapy: Part I.

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

              Phosphatidylinositol 4 phosphate regulates targeting of clathrin adaptor AP-1 complexes to the Golgi.

              Phosphatidylinositol 4 phosphate [PI(4)P] is essential for secretion in yeast, but its role in mammalian cells is unclear. Current paradigms propose that PI(4)P acts primarily as a precursor to phosphatidylinositol 4,5 bisphosphate (PIP2), an important plasma membrane regulator. We found that PI(4)P is enriched in the mammalian Golgi, and used RNA interference (RNAi) of PI4KIIalpha, a Golgi resident phosphatidylinositol 4 kinase, to determine whether PI(4)P directly regulates the Golgi. PI4KIIalpha RNAi decreases Golgi PI(4)P, blocks the recruitment of clathrin adaptor AP-1 complexes to the Golgi, and inhibits AP-1-dependent functions. This AP-1 binding defect is rescued by adding back PI(4)P. In addition, purified AP-1 binds PI(4)P, and anti-PI(4)P inhibits the in vitro recruitment of cytosolic AP-1 to normal cellular membranes. We propose that PI4KIIalpha establishes the Golgi's unique lipid-defined organelle identity by generating PI(4)P-rich domains that specify the docking of the AP-1 coat machinery.
                Bookmark

                Author and article information

                Journal
                Am J Physiol Endocrinol Metab
                Am. J. Physiol. Endocrinol. Metab
                ajpendo
                ajpendo
                AJPENDO
                American Journal of Physiology - Endocrinology and Metabolism
                American Physiological Society (Bethesda, MD )
                0193-1849
                1522-1555
                21 June 2016
                1 August 2016
                21 June 2016
                : 311
                : 2
                : E488-E507
                Affiliations
                [1] 1Section of Cell Biology and Functional Genomics, Imperial College London, London, United Kingdom;
                [2] 2Wellcome Trust Centre for Human Genetics, Oxford, United Kingdom; and
                [3] 3Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
                Author notes
                Address for reprint requests and other correspondence: G. A. Rutter, Section of Cell Biology and Functional Genomics, Imperial College London, Du Cane Road W12 0NN, London, UK (e-mail: g.rutter@ 123456imperial.ac.uk ).
                Article
                E-00074-2016
                10.1152/ajpendo.00074.2016
                5005967
                27329800
                4cecf1ca-31ca-4df2-b735-008db0f1b1da
                Copyright © 2016 the American Physiological Society

                Licensed under Creative Commons Attribution CC-BY 3.0: © the American Physiological Society.

                History
                : 26 February 2016
                : 20 June 2016
                Funding
                Funded by: http://doi.org/10.13039/100004440 Wellcome Trust
                Award ID: WT098424AIA
                Funded by: http://doi.org/10.13039/501100000268 Biotechnology and Biological Sciences Research Council (BBSRC)
                Award ID: BB/J015873/1
                Funded by: http://doi.org/10.13039/501100000288 Royal Society
                Award ID: N/A
                Funded by: http://doi.org/10.13039/501100000265 Medical Research Council (MRC)
                Award ID: MR/J0003042/1
                Funded by: http://doi.org/10.13039/501100000273 Diabetes Research & Wellness Foundation (DRWF)
                Award ID: SCA/01/F/12
                Funded by: European Union
                Award ID: FP7/2007-2013
                Categories
                Call for Papers
                Islet Biology

                Endocrinology & Diabetes
                vps13c,c2cd4a,gwas,type 2 diabetes,β-cell
                Endocrinology & Diabetes
                vps13c, c2cd4a, gwas, type 2 diabetes, β-cell

                Comments

                Comment on this article