• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 1
  • 1
  • Tagged with
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

Starch-binding domain-containing protein 1: a novel participant in glycogen metabolism

Jiang, Sixin 23 August 2011 (has links)
Indiana University-Purdue University Indianapolis (IUPUI) / Glycogen, a branched polymer of glucose, acts as an intracellular carbon and energy reserve in many tissues and cell types. The breakdown of glycogen by hormonally regulated degradation involving the coordinated action of glycogen phosphorylase and debranching enzyme has been well studied. However, the importance of lysosomal disposal of glycogen has been underscored by a glycogen storage disorder, Pompe disease. This disease destroys tissues by over-accumulating glycogen in lysosomes due to a genetic defect in the lysosomal acid α-glucosidase. Details of the intracellular trafficking of glycogen are not well understood. Starch-binding domain-containing protein 1 (Stbd1) is a protein of previously unknown function with predicted hydrophobic N-terminus and C-terminal CBM20 carbohydrate binding domain. The protein is highly expressed in the liver and muscle, the major repositories of glycogen. Stbd1 binds to glycogen in vitro and in vivo with a preference for less branched and more phosphorylated polysaccharides. In animal models, the protein level of Stbd1 correlates with the genetic depletion of glycogen. Endogenous Stbd1 is found in perinuclear compartments in cultured mouse and rat cells. When over-expressed in cells, Stbd1 accumulates and coincides with glycogen and GABARAPL1, the autophagy protein. They form enlarged perinuclear structures which are abolished by removing the hydrophobic N-terminus of Stbd1. Stbd1, with point mutations in the CBM20 domain, retains the perinuclear localization but without concentration of glycogen in this compartment. In cells that are stably over-expressing glycogen synthase, glycogen exists as large perinuclear deposits, where Stbd1 can also be present. Removing glucose from the culture leads to a breakdown of the massive glycogen accumulation into numerous smaller and scattered deposits which are still positive for Stbd1. Furthermore, the autophagy protein GABARAPL1 co-immunoprecipates and co-localizes with Stbd1 when co-expressed in cells. Point mutation or deletion of the autophagy protein interacting region on Stbd1 eliminates the interaction and co-localization with GABARAPL1 but not the characteristic perinuclear distribution of Stbd1. We propose that Stbd1 is involved in glycogen metabolism. In particular, it participates in the vesicular transfer of glycogen to the lysosome with the recruitment of autophagy related proteins GABARAPL1 and/or GABARAP, as these vesicles mature prior to lysosomal fusion.
2

The effects of laforin, malin, Stbd1, and Ptg deficiencies on heart glycogen levels in Pompe disease mouse models

Conway, Betsy Ann 08 1900 (has links)
Indiana University-Purdue University Indianapolis (IUPUI) / Pompe disease (PD) is a rare metabolic myopathy characterized by loss of acid alpha-glucosidase (GAA), the enzyme responsible for breaking down glycogen to glucose within the lysosomes. PD cells accumulate massive quantities of glycogen within their lysosomes, and as such, PD is classified as a “lysosomal storage disease” (LSD). GAA-deficient cells also exhibit accumulation of autophagic debris. Symptoms of severe infantile PD include extreme muscle weakness, hypotonia, and hypertrophic cardiomyopathy, resulting in death before one year of age. Certain LSDs are currently being successfully treated with enzyme replacement therapy (ERT), which involves intravenous infusion of a recombinant enzyme to counteract the endogenous deficiency. ERT has been less successful in PD, however, due to ineffective delivery of the recombinant enzyme. Alternatively, specific genes deletion may reduce lysosomal glycogen load, and could thus be targeted in PD therapy development. Absence of malin (EPM2B) or laforin (EPM2A) has been proposed to impair autophagy, which could reduce lysosomal glycogen levels. Additionally, deficiency of Stbd1 has been postulated to disable lysosomal glycogen import. Furthermore, Ptg deficiency was previously reported to abrogate Lafora body formation and correct neurological abnormalities in Lafora disease mouse models and could have similar effects on PD pathologies. The goal of this study was to characterize the effects of homozygous disruption of Epm2a, Epm2b, Stbd1, and Ptg loci on total glycogen levels in PD mouse model heart tissue, as in severe infantile PD, it is accumulation of glycogen in the heart that results in fatal hypertrophic cardiomyopathy. Gaa-/- mice were intercrossed with Epm2a-/-, Epm2b-/-, Stbd1-/-, and Ptg-/- mice to generate wildtype (WT), single knockout, and double knockout mice. The results indicated that Gaa-/- hearts accumulated up to 100-fold more glycogen than the WT. These mice also displayed cardiac hypertrophy. However, deficiency of Epm2a, Epm2b, Stbd1, or PTG in the Gaa-/- background did not reveal changes of statistical significance in either heart glycogen or cardiac hypertrophy. Nevertheless, since total glycogen was measured, these deficiencies should not be discarded in future discussions of PD therapy, as increasing sample sizes and/or distinguishing cytosolic from lysosomal glycogen content may yet reveal differences of greater significance.

Page generated in 0.0208 seconds