Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Diabetes ; 69(12): 2667-2677, 2020 12.
Article in English | MEDLINE | ID: mdl-32994272

ABSTRACT

Animal models are important tools in diabetes research because ethical and logistical constraints limit access to human tissue. ß-Cell dysfunction is a common contributor to the pathogenesis of most types of diabetes. Spontaneous hyperglycemia was developed in a colony of C57BL/6J mice at King's College London (KCL). Sequencing identified a mutation in the Ins2 gene, causing a glycine-to-serine substitution at position 32 on the B chain of the preproinsulin 2 molecule. Mice with the Ins2 +/G32S mutation were named KCL Ins2 G32S (KINGS) mice. The same mutation in humans (rs80356664) causes dominantly inherited neonatal diabetes. Mice were characterized, and ß-cell function was investigated. Male mice became overtly diabetic at ∼5 weeks of age, whereas female mice had only slightly elevated nonfasting glycemia. Islets showed decreased insulin content and impaired glucose-induced insulin secretion, which was more severe in males. Transmission electron microscopy and studies of gene and protein expression showed ß-cell endoplasmic reticulum (ER) stress in both sexes. Despite this, ß-cell numbers were only slightly reduced in older animals. In conclusion, the KINGS mouse is a novel model of a human form of diabetes that may be useful to study ß-cell responses to ER stress.


Subject(s)
Diabetes Mellitus/genetics , Disease Models, Animal , Endoplasmic Reticulum Stress/physiology , Insulin-Secreting Cells/metabolism , Animals , Ecosystem , Female , Glucose Tolerance Test , Humans , Insulin/blood , Male , Mice , Mice, Inbred Strains , Mutation , Polymorphism, Single Nucleotide
3.
Diabetes ; 65(1): 129-39, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26470781

ABSTRACT

We have previously demonstrated that coculture of islets with mesenchymal stromal cells (MSCs) enhanced islet insulin secretory capacity in vitro, correlating with improved graft function in vivo. To identify factors that contribute to MSC-mediated improvements in islet function, we have used an unbiased quantitative RT-PCR screening approach to identify MSC-derived peptide ligands of G-protein-coupled receptors that are expressed by islets cells. We demonstrated high expression of annexin A1 (ANXA1) mRNA by MSCs and confirmed expression at the protein level in lysates and MSC-conditioned media by Western blot analysis and ELISA. Preculturing islets with exogenous ANXA1 enhanced glucose-stimulated insulin secretion (GSIS), thereby mimicking the beneficial influence of MSC preculture in vitro. Small interfering RNA-mediated knockdown of ANXA1 in MSCs reduced their capacity to potentiate GSIS. MSCs derived from ANXA1(-/-) mice had no functional capacity to enhance GSIS, in contrast to wild-type controls. Preculturing islets with ANXA1 had modest effects on their capacity to regulate blood glucose in streptozotocin-induced diabetic mice, indicating that additional MSC-derived factors are required to fully mimic the beneficial effects of MSC preculture in vivo. These findings demonstrate the feasibility of harnessing the MSC secretome as a defined, noncellular strategy to improve the efficiency of clinical islet transplantation protocols.


Subject(s)
Annexin A1/genetics , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/genetics , Insulin/metabolism , Islets of Langerhans/metabolism , Mesenchymal Stem Cells/metabolism , RNA, Messenger/metabolism , Animals , Annexin A1/metabolism , Blotting, Western , Coculture Techniques , Diabetes Mellitus, Experimental/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Knockdown Techniques , In Vitro Techniques , Insulin Secretion , Mesenchymal Stem Cell Transplantation , Mice , RNA, Small Interfering , Real-Time Polymerase Chain Reaction
4.
Chem Commun (Camb) ; 51(53): 10652-5, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26051448

ABSTRACT

A spontaneous multilayer deposition approach for presenting therapeutic proteins onto pancreatic islet surfaces, using a heparin polyaldehyde and glycol chitosan alternating layering scheme, has been developed to enable the nanoscale engineering of a microenvironment for transplanted cells. The nanocoating incorporating α1-antitrypsin, an anti-inflammatory protein, exhibited effective anti-coagulant activities in vitro.


Subject(s)
Islets of Langerhans/chemistry , Nanostructures/chemistry , alpha 1-Antitrypsin/chemistry , Amines/chemistry , Animals , Cell Survival , Chitosan/chemistry , Fluorescein-5-isothiocyanate/chemistry , Heparin/chemistry , Insulin/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Mice , Mice, Inbred ICR , Microscopy, Electron, Transmission , alpha 1-Antitrypsin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...