Distinct gene expression pathways in islets from individuals with short‐ and long‐duration type 1 diabetes

dc.contributor.authorMastracci, Teresa L.
dc.contributor.authorTuratsinze, Jean-Valery
dc.contributor.authorBook, Benita K.
dc.contributor.authorRestrepo, Ivan A.
dc.contributor.authorPugia, Michael J.
dc.contributor.authorWeibke, Eric A.
dc.contributor.authorPescovitz, Mark D.
dc.contributor.authorEizirik, Decio L.
dc.contributor.authorMirmira, Raghavendra G.
dc.contributor.departmentBiochemistry and Molecular Biology, School of Medicineen_US
dc.date.accessioned2018-04-26T18:56:10Z
dc.date.available2018-04-26T18:56:10Z
dc.date.issued2018
dc.description.abstractAims Our current understanding of the pathogenesis of type 1 diabetes (T1D) arose, in large part, from studies using the non‐obese diabetic (NOD) mouse model. In the present study, we chose a human‐focused method to investigate T1D disease mechanisms and potential targets for therapeutic intervention by directly analysing human donor pancreatic islets from individuals with T1D. Materials and Methods We obtained islets from a young individual with T1D for 3 years and from an older individual with T1D for 27 years and performed unbiased functional genomic analysis by high‐depth RNA sequencing; the T1D islets were compared with islets isolated from 3 non‐diabetic donors. Results The islets procured from these T1D donors represent a unique opportunity to identify gene expression changes in islets after significantly different disease duration. Data analysis identified several inflammatory pathways up‐regulated in short‐duration disease, which notably included many components of innate immunity. As proof of concept for translation, one of the pathways, governed by IL‐23(p19), was selected for further study in NOD mice because of ongoing human trials of biologics against this target for different indications. A mouse monoclonal antibody directed against IL‐23(p19) when administered to NOD mice resulted in a significant reduction in incidence of diabetes. Conclusion While the sample size for this study is small, our data demonstrate that the direct analysis of human islets provides a greater understanding of human disease. These data, together with the analysis of an expanded cohort to be obtained by future collaborative efforts, might result in the identification of promising novel targets for translation into effective therapeutic interventions for human T1D, with the added benefit of repurposing known biologicals for use in different indications.en_US
dc.eprint.versionAuthor's manuscripten_US
dc.identifier.citationMastracci, T. L., Turatsinze, J.-V., Book, B. K., Restrepo, I. A., Pugia, M. J., Wiebke, E. A., … Mirmira, R. G. (2018). Distinct Gene Expression Pathways in Islets from Individuals with Short‐ and Long‐Duration Type 1 Diabetes. Diabetes, Obesity and Metabolism. https://doi.org/10.1111/dom.13298en_US
dc.identifier.urihttps://hdl.handle.net/1805/15922
dc.language.isoenen_US
dc.publisherWileyen_US
dc.relation.isversionof10.1111/dom.13298en_US
dc.relation.journalDiabetes, Obesity and Metabolismen_US
dc.rightsPublisher Policyen_US
dc.sourceAuthoren_US
dc.subjectfunctional genomicsen_US
dc.subjecthuman isletsen_US
dc.subjectIL23en_US
dc.titleDistinct gene expression pathways in islets from individuals with short‐ and long‐duration type 1 diabetesen_US
dc.typeArticleen_US
Files
Original bundle
Now showing 1 - 1 of 1
Loading...
Thumbnail Image
Name:
Mastracci_2018_distinct.pdf
Size:
2.72 MB
Format:
Adobe Portable Document Format
Description:
License bundle
Now showing 1 - 1 of 1
No Thumbnail Available
Name:
license.txt
Size:
1.99 KB
Format:
Item-specific license agreed upon to submission
Description: