The University of Southampton
University of Southampton Institutional Repository

Detection of experimental and clinical immune complexes by measuring SHIP-1 recruitment to the inhibitory FcγRIIB

Detection of experimental and clinical immune complexes by measuring SHIP-1 recruitment to the inhibitory FcγRIIB
Detection of experimental and clinical immune complexes by measuring SHIP-1 recruitment to the inhibitory FcγRIIB
Fc gamma receptors (FcγR) are involved in multiple aspects of immune cell regulation, are central to the success of monoclonal antibody (mAb) therapeutics and underpin the pathology of several autoimmune diseases. However, reliable assays capable of accurately measuring FcγR interactions with their physiological ligands, immunoglobulin G (IgG) immune complexes (IC), are limited. A method to study and detect IC interactions with FcγRs was therefore developed. This method, designed to model the signalling pathway of the inhibitory FcγRIIB (CD32B), utilised NanoLuc® Binary Interaction Technology (NanoBiT™) to measure recruitment of the Src homology 2 (SH2) domain-containing inositol phosphatase 1 (SHIP-1) to the immunoreceptor tyrosine-based inhibitory motif (ITIM) of this receptor. Such recruitment required prior crosslinking of an immunoreceptor tyrosine-based activation motif (ITAM)-containing activatory receptor, and evoked luciferase activity in discrete clusters at the cell surface, recapitulating the known biology of CD32B signalling. The assay detected varying forms of experimental IC, including heat-aggregated IgG, Rituximab:anti-idiotype complexes and anti-trinitrophenol (TNP)-TNP complexes in a sensitive manner (≤1μg/ml), and discriminated between complexes of varying size and isotype. Proof-of-concept for the detection of circulating ICs in autoimmune disease was provided, as responses to sera from patients with systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) were detected in small pilot studies. Finally, the method was translated to a stable cell line system. In conclusion, a rapid and robust method for the detection of IC was developed, which has numerous potential applications including the monitoring of IC in autoimmune diseases and the study of underlying FcγR biology.
Stopforth, R.J.
2d3e18ff-5563-4247-9150-f0f337fb585f
Oldham, R.J.
844b9bff-16f0-4577-abba-b35afd02b923
Tutt, A.L.
46ce577b-aea1-412d-84ea-fc4dab794469
Duriez, P.
523004b2-256b-4e62-b394-100bda55adf0
Chan, H.T.C.
b109c93f-7e9a-44ee-ad12-da757b1b11fc
Binkowski, B.
ac3aa2a0-cac3-4770-9f5f-9e9f7201da75
Zimprich, C.
4db401a0-262b-4c1e-b6db-376d3c197d8b
Li, D.
098466a4-161c-4078-a7ad-497ccbb4104f
Hargreaves, P.
019c11f6-4787-4c4d-9998-02fbf22cf391
Cong, M.
a2c15390-7b20-49f0-afd7-201313bbbc0d
Reddy, V.
09bf2bf4-cfd1-4e1c-9785-4e1cd1fcde61
Leandro, M.
423fe911-a4ff-40ff-a85e-a3afc3841e9b
Cambridge, G.
fcd93a1b-2d17-45b6-be9d-cc1d8101e947
Lux, A.
ea74e934-65ff-4bca-b9cb-fc53a237cafc
Nimmerjahn, F.
93b8ab1f-5420-4630-9199-845339580509
Cragg, M.S.
ec97f80e-f3c8-49b7-a960-20dff648b78c
Stopforth, R.J.
2d3e18ff-5563-4247-9150-f0f337fb585f
Oldham, R.J.
844b9bff-16f0-4577-abba-b35afd02b923
Tutt, A.L.
46ce577b-aea1-412d-84ea-fc4dab794469
Duriez, P.
523004b2-256b-4e62-b394-100bda55adf0
Chan, H.T.C.
b109c93f-7e9a-44ee-ad12-da757b1b11fc
Binkowski, B.
ac3aa2a0-cac3-4770-9f5f-9e9f7201da75
Zimprich, C.
4db401a0-262b-4c1e-b6db-376d3c197d8b
Li, D.
098466a4-161c-4078-a7ad-497ccbb4104f
Hargreaves, P.
019c11f6-4787-4c4d-9998-02fbf22cf391
Cong, M.
a2c15390-7b20-49f0-afd7-201313bbbc0d
Reddy, V.
09bf2bf4-cfd1-4e1c-9785-4e1cd1fcde61
Leandro, M.
423fe911-a4ff-40ff-a85e-a3afc3841e9b
Cambridge, G.
fcd93a1b-2d17-45b6-be9d-cc1d8101e947
Lux, A.
ea74e934-65ff-4bca-b9cb-fc53a237cafc
Nimmerjahn, F.
93b8ab1f-5420-4630-9199-845339580509
Cragg, M.S.
ec97f80e-f3c8-49b7-a960-20dff648b78c

Stopforth, R.J., Oldham, R.J., Tutt, A.L., Duriez, P., Chan, H.T.C., Binkowski, B., Zimprich, C., Li, D., Hargreaves, P., Cong, M., Reddy, V., Leandro, M., Cambridge, G., Lux, A., Nimmerjahn, F. and Cragg, M.S. (2018) Detection of experimental and clinical immune complexes by measuring SHIP-1 recruitment to the inhibitory FcγRIIB. The Journal of Immunology. (doi:10.4049/jimmunol.1700832).

Record type: Article

Abstract

Fc gamma receptors (FcγR) are involved in multiple aspects of immune cell regulation, are central to the success of monoclonal antibody (mAb) therapeutics and underpin the pathology of several autoimmune diseases. However, reliable assays capable of accurately measuring FcγR interactions with their physiological ligands, immunoglobulin G (IgG) immune complexes (IC), are limited. A method to study and detect IC interactions with FcγRs was therefore developed. This method, designed to model the signalling pathway of the inhibitory FcγRIIB (CD32B), utilised NanoLuc® Binary Interaction Technology (NanoBiT™) to measure recruitment of the Src homology 2 (SH2) domain-containing inositol phosphatase 1 (SHIP-1) to the immunoreceptor tyrosine-based inhibitory motif (ITIM) of this receptor. Such recruitment required prior crosslinking of an immunoreceptor tyrosine-based activation motif (ITAM)-containing activatory receptor, and evoked luciferase activity in discrete clusters at the cell surface, recapitulating the known biology of CD32B signalling. The assay detected varying forms of experimental IC, including heat-aggregated IgG, Rituximab:anti-idiotype complexes and anti-trinitrophenol (TNP)-TNP complexes in a sensitive manner (≤1μg/ml), and discriminated between complexes of varying size and isotype. Proof-of-concept for the detection of circulating ICs in autoimmune disease was provided, as responses to sera from patients with systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) were detected in small pilot studies. Finally, the method was translated to a stable cell line system. In conclusion, a rapid and robust method for the detection of IC was developed, which has numerous potential applications including the monitoring of IC in autoimmune diseases and the study of underlying FcγR biology.

Text
resubmitted smaller - Accepted Manuscript
Restricted to Repository staff only
Request a copy

More information

Accepted/In Press date: 20 December 2017
e-pub ahead of print date: 19 January 2018

Identifiers

Local EPrints ID: 417550
URI: http://eprints.soton.ac.uk/id/eprint/417550
PURE UUID: 6da40d56-fa9d-46cc-be4b-1e0196b3051a
ORCID for R.J. Stopforth: ORCID iD orcid.org/0000-0002-0054-7503
ORCID for R.J. Oldham: ORCID iD orcid.org/0000-0002-8007-1145
ORCID for H.T.C. Chan: ORCID iD orcid.org/0000-0003-0530-9480
ORCID for M.S. Cragg: ORCID iD orcid.org/0000-0003-2077-089X

Catalogue record

Date deposited: 02 Feb 2018 17:30
Last modified: 16 Mar 2024 04:28

Export record

Altmetrics

Contributors

Author: R.J. Stopforth ORCID iD
Author: R.J. Oldham ORCID iD
Author: A.L. Tutt
Author: P. Duriez
Author: H.T.C. Chan ORCID iD
Author: B. Binkowski
Author: C. Zimprich
Author: D. Li
Author: P. Hargreaves
Author: M. Cong
Author: V. Reddy
Author: M. Leandro
Author: G. Cambridge
Author: A. Lux
Author: F. Nimmerjahn
Author: M.S. Cragg ORCID iD

Download statistics

Downloads from ePrints over the past year. Other digital versions may also be available to download e.g. from the publisher's website.

View more statistics

Atom RSS 1.0 RSS 2.0

Contact ePrints Soton: eprints@soton.ac.uk

ePrints Soton supports OAI 2.0 with a base URL of http://eprints.soton.ac.uk/cgi/oai2

This repository has been built using EPrints software, developed at the University of Southampton, but available to everyone to use.

We use cookies to ensure that we give you the best experience on our website. If you continue without changing your settings, we will assume that you are happy to receive cookies on the University of Southampton website.

×