Cargando…

Islet transplantation into brown adipose tissue can delay immune rejection

Type 1 diabetes is an autoimmune disease characterized by insulin-producing β cell destruction. Although islet transplantation restores euglycemia and improves patient outcomes, an ideal transplant site remains elusive. Brown adipose tissue (BAT) has a highly vascularized and antiinflammatory microe...

Descripción completa

Detalles Bibliográficos
Autores principales: Kepple, Jessica D., Barra, Jessie M., Young, Martin E., Hunter, Chad S., Tse, Hubert M.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: American Society for Clinical Investigation 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8876467/
https://www.ncbi.nlm.nih.gov/pubmed/35015736
http://dx.doi.org/10.1172/jci.insight.152800
_version_ 1784658181658509312
author Kepple, Jessica D.
Barra, Jessie M.
Young, Martin E.
Hunter, Chad S.
Tse, Hubert M.
author_facet Kepple, Jessica D.
Barra, Jessie M.
Young, Martin E.
Hunter, Chad S.
Tse, Hubert M.
author_sort Kepple, Jessica D.
collection PubMed
description Type 1 diabetes is an autoimmune disease characterized by insulin-producing β cell destruction. Although islet transplantation restores euglycemia and improves patient outcomes, an ideal transplant site remains elusive. Brown adipose tissue (BAT) has a highly vascularized and antiinflammatory microenvironment. Because these tissue features can promote islet graft survival, we hypothesized that islets transplanted into BAT will maintain islet graft and BAT function while delaying immune-mediated rejection. We transplanted syngeneic and allogeneic islets into BAT or under the kidney capsule of streptozotocin-induced diabetic NOD.Rag and NOD mice to investigate islet graft function, BAT function, metabolism, and immune-mediated rejection. Islet grafts within BAT restored euglycemia similarly to kidney capsule controls. Islets transplanted in BAT maintained expression of islet hormones and transcription factors and were vascularized. Compared with those in kidney capsule and euglycemic mock-surgery controls, no differences in glucose or insulin tolerance, thermogenic regulation, or energy expenditure were observed with islet grafts in BAT. Immune profiling of BAT revealed enriched antiinflammatory macrophages and T cells. Compared with the kidney capsule control, there were significant delays in autoimmune and allograft rejection of islets transplanted in BAT, possibly due to increased antiinflammatory immune populations. Our data support BAT as an alternative islet transplant site that may improve graft survival.
format Online
Article
Text
id pubmed-8876467
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher American Society for Clinical Investigation
record_format MEDLINE/PubMed
spelling pubmed-88764672022-03-01 Islet transplantation into brown adipose tissue can delay immune rejection Kepple, Jessica D. Barra, Jessie M. Young, Martin E. Hunter, Chad S. Tse, Hubert M. JCI Insight Research Article Type 1 diabetes is an autoimmune disease characterized by insulin-producing β cell destruction. Although islet transplantation restores euglycemia and improves patient outcomes, an ideal transplant site remains elusive. Brown adipose tissue (BAT) has a highly vascularized and antiinflammatory microenvironment. Because these tissue features can promote islet graft survival, we hypothesized that islets transplanted into BAT will maintain islet graft and BAT function while delaying immune-mediated rejection. We transplanted syngeneic and allogeneic islets into BAT or under the kidney capsule of streptozotocin-induced diabetic NOD.Rag and NOD mice to investigate islet graft function, BAT function, metabolism, and immune-mediated rejection. Islet grafts within BAT restored euglycemia similarly to kidney capsule controls. Islets transplanted in BAT maintained expression of islet hormones and transcription factors and were vascularized. Compared with those in kidney capsule and euglycemic mock-surgery controls, no differences in glucose or insulin tolerance, thermogenic regulation, or energy expenditure were observed with islet grafts in BAT. Immune profiling of BAT revealed enriched antiinflammatory macrophages and T cells. Compared with the kidney capsule control, there were significant delays in autoimmune and allograft rejection of islets transplanted in BAT, possibly due to increased antiinflammatory immune populations. Our data support BAT as an alternative islet transplant site that may improve graft survival. American Society for Clinical Investigation 2022-02-22 /pmc/articles/PMC8876467/ /pubmed/35015736 http://dx.doi.org/10.1172/jci.insight.152800 Text en © 2022 Kepple et al. https://creativecommons.org/licenses/by/4.0/This work is licensed under the Creative Commons Attribution 4.0 International License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Research Article
Kepple, Jessica D.
Barra, Jessie M.
Young, Martin E.
Hunter, Chad S.
Tse, Hubert M.
Islet transplantation into brown adipose tissue can delay immune rejection
title Islet transplantation into brown adipose tissue can delay immune rejection
title_full Islet transplantation into brown adipose tissue can delay immune rejection
title_fullStr Islet transplantation into brown adipose tissue can delay immune rejection
title_full_unstemmed Islet transplantation into brown adipose tissue can delay immune rejection
title_short Islet transplantation into brown adipose tissue can delay immune rejection
title_sort islet transplantation into brown adipose tissue can delay immune rejection
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8876467/
https://www.ncbi.nlm.nih.gov/pubmed/35015736
http://dx.doi.org/10.1172/jci.insight.152800
work_keys_str_mv AT kepplejessicad islettransplantationintobrownadiposetissuecandelayimmunerejection
AT barrajessiem islettransplantationintobrownadiposetissuecandelayimmunerejection
AT youngmartine islettransplantationintobrownadiposetissuecandelayimmunerejection
AT hunterchads islettransplantationintobrownadiposetissuecandelayimmunerejection
AT tsehubertm islettransplantationintobrownadiposetissuecandelayimmunerejection