Nuclear targeting of IGF-1 receptor in orbital fibroblasts from Graves' disease: apparent role of ADAM17.

Insulin-like growth factor-1 receptor (IGF-1R) comprises two subunits, including a ligand binding domain on extra- cellular IGF-1Rα and a tyrosine phosphorylation site located on IGF-1Rβ. IGF-1R is over-expressed by orbital fibroblasts in the autoimmune syndrome, Graves' disease (GD). When acti...

Full description

Bibliographic Details
Main Authors: Neil Hoa, Shanli Tsui, Nikoo F Afifiyan, Amiya Sinha Hikim, Bin Li, Raymond S Douglas, Terry J Smith
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2012-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC3323600?pdf=render
_version_ 1811209451999330304
author Neil Hoa
Shanli Tsui
Nikoo F Afifiyan
Amiya Sinha Hikim
Bin Li
Raymond S Douglas
Terry J Smith
author_facet Neil Hoa
Shanli Tsui
Nikoo F Afifiyan
Amiya Sinha Hikim
Bin Li
Raymond S Douglas
Terry J Smith
author_sort Neil Hoa
collection DOAJ
description Insulin-like growth factor-1 receptor (IGF-1R) comprises two subunits, including a ligand binding domain on extra- cellular IGF-1Rα and a tyrosine phosphorylation site located on IGF-1Rβ. IGF-1R is over-expressed by orbital fibroblasts in the autoimmune syndrome, Graves' disease (GD). When activated by IGF-1 or GD-derived IgG (GD-IgG), these fibroblasts produce RANTES and IL-16, while those from healthy donors do not. We now report that IGF-1 and GD-IgG provoke IGF-1R accumulation in the cell nucleus of GD fibroblasts where it co-localizes with chromatin. Nuclear IGF-1R is detected with anti-IGF-1Rα-specific mAb and migrates to approximately 110 kDa, consistent with its identity as an IGF-1R fragment. Nuclear IGF-1R migrating as a 200 kDa protein and consistent with an intact receptor was undetectable when probed with either anti-IGF-1Rα or anti-IGF-1Rβ mAbs. Nuclear redistribution of IGF-1R is absent in control orbital fibroblasts. In GD fibroblasts, it can be abolished by an IGF-1R-blocking mAb, 1H7 and by physiological concentrations of glucocorticoids. When cell-surface IGF-1R is cross-linked with (125)I IGF-1, (125)I-IGF-1/IGF-1R complexes accumulate in the nuclei of GD fibroblasts. This requires active ADAM17, a membrane associated metalloproteinase, and the phosphorylation of IGF-1R. In contrast, virally encoded IGF-1Rα/GFP fusion protein localizes equivalently in nuclei in both control and GD fibroblasts. This result suggests that generation of IGF-1R fragments may limit the accumulation of nuclear IGF-1R. We thus identify a heretofore-unrecognized behavior of IGF-1R that appears limited to GD-derived fibroblasts. Nuclear IGF-1R may play a role in disease pathogenesis.
first_indexed 2024-04-12T04:39:38Z
format Article
id doaj.art-051ac3add0bf4b06b94429bd217a6900
institution Directory Open Access Journal
issn 1932-6203
language English
last_indexed 2024-04-12T04:39:38Z
publishDate 2012-01-01
publisher Public Library of Science (PLoS)
record_format Article
series PLoS ONE
spelling doaj.art-051ac3add0bf4b06b94429bd217a69002022-12-22T03:47:42ZengPublic Library of Science (PLoS)PLoS ONE1932-62032012-01-0174e3417310.1371/journal.pone.0034173Nuclear targeting of IGF-1 receptor in orbital fibroblasts from Graves' disease: apparent role of ADAM17.Neil HoaShanli TsuiNikoo F AfifiyanAmiya Sinha HikimBin LiRaymond S DouglasTerry J SmithInsulin-like growth factor-1 receptor (IGF-1R) comprises two subunits, including a ligand binding domain on extra- cellular IGF-1Rα and a tyrosine phosphorylation site located on IGF-1Rβ. IGF-1R is over-expressed by orbital fibroblasts in the autoimmune syndrome, Graves' disease (GD). When activated by IGF-1 or GD-derived IgG (GD-IgG), these fibroblasts produce RANTES and IL-16, while those from healthy donors do not. We now report that IGF-1 and GD-IgG provoke IGF-1R accumulation in the cell nucleus of GD fibroblasts where it co-localizes with chromatin. Nuclear IGF-1R is detected with anti-IGF-1Rα-specific mAb and migrates to approximately 110 kDa, consistent with its identity as an IGF-1R fragment. Nuclear IGF-1R migrating as a 200 kDa protein and consistent with an intact receptor was undetectable when probed with either anti-IGF-1Rα or anti-IGF-1Rβ mAbs. Nuclear redistribution of IGF-1R is absent in control orbital fibroblasts. In GD fibroblasts, it can be abolished by an IGF-1R-blocking mAb, 1H7 and by physiological concentrations of glucocorticoids. When cell-surface IGF-1R is cross-linked with (125)I IGF-1, (125)I-IGF-1/IGF-1R complexes accumulate in the nuclei of GD fibroblasts. This requires active ADAM17, a membrane associated metalloproteinase, and the phosphorylation of IGF-1R. In contrast, virally encoded IGF-1Rα/GFP fusion protein localizes equivalently in nuclei in both control and GD fibroblasts. This result suggests that generation of IGF-1R fragments may limit the accumulation of nuclear IGF-1R. We thus identify a heretofore-unrecognized behavior of IGF-1R that appears limited to GD-derived fibroblasts. Nuclear IGF-1R may play a role in disease pathogenesis.http://europepmc.org/articles/PMC3323600?pdf=render
spellingShingle Neil Hoa
Shanli Tsui
Nikoo F Afifiyan
Amiya Sinha Hikim
Bin Li
Raymond S Douglas
Terry J Smith
Nuclear targeting of IGF-1 receptor in orbital fibroblasts from Graves' disease: apparent role of ADAM17.
PLoS ONE
title Nuclear targeting of IGF-1 receptor in orbital fibroblasts from Graves' disease: apparent role of ADAM17.
title_full Nuclear targeting of IGF-1 receptor in orbital fibroblasts from Graves' disease: apparent role of ADAM17.
title_fullStr Nuclear targeting of IGF-1 receptor in orbital fibroblasts from Graves' disease: apparent role of ADAM17.
title_full_unstemmed Nuclear targeting of IGF-1 receptor in orbital fibroblasts from Graves' disease: apparent role of ADAM17.
title_short Nuclear targeting of IGF-1 receptor in orbital fibroblasts from Graves' disease: apparent role of ADAM17.
title_sort nuclear targeting of igf 1 receptor in orbital fibroblasts from graves disease apparent role of adam17
url http://europepmc.org/articles/PMC3323600?pdf=render
work_keys_str_mv AT neilhoa nucleartargetingofigf1receptorinorbitalfibroblastsfromgravesdiseaseapparentroleofadam17
AT shanlitsui nucleartargetingofigf1receptorinorbitalfibroblastsfromgravesdiseaseapparentroleofadam17
AT nikoofafifiyan nucleartargetingofigf1receptorinorbitalfibroblastsfromgravesdiseaseapparentroleofadam17
AT amiyasinhahikim nucleartargetingofigf1receptorinorbitalfibroblastsfromgravesdiseaseapparentroleofadam17
AT binli nucleartargetingofigf1receptorinorbitalfibroblastsfromgravesdiseaseapparentroleofadam17
AT raymondsdouglas nucleartargetingofigf1receptorinorbitalfibroblastsfromgravesdiseaseapparentroleofadam17
AT terryjsmith nucleartargetingofigf1receptorinorbitalfibroblastsfromgravesdiseaseapparentroleofadam17