Cargando…

Glycogen Synthase Kinase 3 Inactivation Compensates for the Lack of CD28 in the Priming of CD8(+) Cytotoxic T-Cells: Implications for anti-PD-1 Immunotherapy

The rescue of exhausted CD8(+) cytolytic T-cells (CTLs) by anti-Programmed Cell Death-1 (anti-PD-1) blockade has been found to require CD28 expression. At the same time, we have shown that the inactivation of the serine/threonine kinase glycogen synthase kinase (GSK)-3α/β with small-interfering RNAs...

Descripción completa

Detalles Bibliográficos
Autores principales: Taylor, Alison, Rudd, Christopher E.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5732207/
https://www.ncbi.nlm.nih.gov/pubmed/29312284
http://dx.doi.org/10.3389/fimmu.2017.01653
_version_ 1783286641518968832
author Taylor, Alison
Rudd, Christopher E.
author_facet Taylor, Alison
Rudd, Christopher E.
author_sort Taylor, Alison
collection PubMed
description The rescue of exhausted CD8(+) cytolytic T-cells (CTLs) by anti-Programmed Cell Death-1 (anti-PD-1) blockade has been found to require CD28 expression. At the same time, we have shown that the inactivation of the serine/threonine kinase glycogen synthase kinase (GSK)-3α/β with small-interfering RNAs (siRNAs) and small molecule inhibitors (SMIs) specifically down-regulates PD-1 expression for enhanced CD8(+) CTL function and clearance of tumors and viral infections. Despite this, it has been unclear whether the GSK-3α/β pathway accounts for CD28 costimulation of CD8(+) CTL function. In this article, we show that inactivation of GSK-3α/β through siRNA or by SMIs during priming can substitute CD28 co-stimulation in the potentiation of cytotoxic CD8(+) CTL function against the EL-4 lymphoma cells expressing OVA peptide. The effect was seen using several structurally distinct GSK-3 SMIs and was accompanied by an increase in Lamp-1 and GZMB expression. Conversely, CD28 crosslinking obviated the need for GSK-3α/β inhibition in its enhancement of CTL function. Our findings support a model where GSK-3 is the central cosignal for CD28 priming of CD8(+) CTLs in anti-PD-1 immunotherapy.
format Online
Article
Text
id pubmed-5732207
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-57322072018-01-08 Glycogen Synthase Kinase 3 Inactivation Compensates for the Lack of CD28 in the Priming of CD8(+) Cytotoxic T-Cells: Implications for anti-PD-1 Immunotherapy Taylor, Alison Rudd, Christopher E. Front Immunol Immunology The rescue of exhausted CD8(+) cytolytic T-cells (CTLs) by anti-Programmed Cell Death-1 (anti-PD-1) blockade has been found to require CD28 expression. At the same time, we have shown that the inactivation of the serine/threonine kinase glycogen synthase kinase (GSK)-3α/β with small-interfering RNAs (siRNAs) and small molecule inhibitors (SMIs) specifically down-regulates PD-1 expression for enhanced CD8(+) CTL function and clearance of tumors and viral infections. Despite this, it has been unclear whether the GSK-3α/β pathway accounts for CD28 costimulation of CD8(+) CTL function. In this article, we show that inactivation of GSK-3α/β through siRNA or by SMIs during priming can substitute CD28 co-stimulation in the potentiation of cytotoxic CD8(+) CTL function against the EL-4 lymphoma cells expressing OVA peptide. The effect was seen using several structurally distinct GSK-3 SMIs and was accompanied by an increase in Lamp-1 and GZMB expression. Conversely, CD28 crosslinking obviated the need for GSK-3α/β inhibition in its enhancement of CTL function. Our findings support a model where GSK-3 is the central cosignal for CD28 priming of CD8(+) CTLs in anti-PD-1 immunotherapy. Frontiers Media S.A. 2017-12-11 /pmc/articles/PMC5732207/ /pubmed/29312284 http://dx.doi.org/10.3389/fimmu.2017.01653 Text en Copyright © 2017 Taylor and Rudd. http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Immunology
Taylor, Alison
Rudd, Christopher E.
Glycogen Synthase Kinase 3 Inactivation Compensates for the Lack of CD28 in the Priming of CD8(+) Cytotoxic T-Cells: Implications for anti-PD-1 Immunotherapy
title Glycogen Synthase Kinase 3 Inactivation Compensates for the Lack of CD28 in the Priming of CD8(+) Cytotoxic T-Cells: Implications for anti-PD-1 Immunotherapy
title_full Glycogen Synthase Kinase 3 Inactivation Compensates for the Lack of CD28 in the Priming of CD8(+) Cytotoxic T-Cells: Implications for anti-PD-1 Immunotherapy
title_fullStr Glycogen Synthase Kinase 3 Inactivation Compensates for the Lack of CD28 in the Priming of CD8(+) Cytotoxic T-Cells: Implications for anti-PD-1 Immunotherapy
title_full_unstemmed Glycogen Synthase Kinase 3 Inactivation Compensates for the Lack of CD28 in the Priming of CD8(+) Cytotoxic T-Cells: Implications for anti-PD-1 Immunotherapy
title_short Glycogen Synthase Kinase 3 Inactivation Compensates for the Lack of CD28 in the Priming of CD8(+) Cytotoxic T-Cells: Implications for anti-PD-1 Immunotherapy
title_sort glycogen synthase kinase 3 inactivation compensates for the lack of cd28 in the priming of cd8(+) cytotoxic t-cells: implications for anti-pd-1 immunotherapy
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5732207/
https://www.ncbi.nlm.nih.gov/pubmed/29312284
http://dx.doi.org/10.3389/fimmu.2017.01653
work_keys_str_mv AT tayloralison glycogensynthasekinase3inactivationcompensatesforthelackofcd28intheprimingofcd8cytotoxictcellsimplicationsforantipd1immunotherapy
AT ruddchristophere glycogensynthasekinase3inactivationcompensatesforthelackofcd28intheprimingofcd8cytotoxictcellsimplicationsforantipd1immunotherapy