Cargando…

Defects in the Acquisition of Tumor-Killing Capability of CD8(+) Cytotoxic T Cells in Streptozotocin-Induced Diabetic Mice

Emerging evidences have shown that diabetes mellitus not only raises risk but also heightens mortality rate of cancer. It is not clear, however, whether antitumor CD8(+) cytotoxic T lymphocyte (CTL) response is down-modulated in diabetic hosts. We investigated the impact of hyperglycemia on CTLs...

Descripción completa

Detalles Bibliográficos
Autores principales: Chen, Shu-Ching, Su, Yu-Chia, Lu, Ya-Ting, Ko, Patrick Chow-In, Chang, Pei-Yu, Lin, Hung-Ju, Ho, Hong-Nerng, Lai, Yo-Ping
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2014
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4229103/
https://www.ncbi.nlm.nih.gov/pubmed/25390652
http://dx.doi.org/10.1371/journal.pone.0109961
_version_ 1782344085319712768
author Chen, Shu-Ching
Su, Yu-Chia
Lu, Ya-Ting
Ko, Patrick Chow-In
Chang, Pei-Yu
Lin, Hung-Ju
Ho, Hong-Nerng
Lai, Yo-Ping
author_facet Chen, Shu-Ching
Su, Yu-Chia
Lu, Ya-Ting
Ko, Patrick Chow-In
Chang, Pei-Yu
Lin, Hung-Ju
Ho, Hong-Nerng
Lai, Yo-Ping
author_sort Chen, Shu-Ching
collection PubMed
description Emerging evidences have shown that diabetes mellitus not only raises risk but also heightens mortality rate of cancer. It is not clear, however, whether antitumor CD8(+) cytotoxic T lymphocyte (CTL) response is down-modulated in diabetic hosts. We investigated the impact of hyperglycemia on CTLs' acquisition of tumor-killing capability by utilizing streptozotocin-induced diabetic (STZ-diabetic) mice. Murine diabetes was induced by intraperitoneal injection of STZ (200 mg/kg) in C57BL/6 mice, 2C-T cell receptor (TCR) transgenic and P14-TCR transgenic mice. The study found that, despite harboring intact proliferative capacity measured with CFSE labeling and MTT assay, STZ-diabetic CD8(+) CTLs displayed impaired effector functions. After stimulation, STZ-diabetic CD8(+) CTLs produced less perforin and TNFα assessed by intracellular staining, as well as expressed less CD103 protein. Furthermore, adoptive transfer of STZ-diabetic P14 CD8(+) effector cells showed an insufficient recruitment to the B16.gp33 melanoma and inadequate production of perforin, granzyme B and TNFα determined by immunohistochemistry in the tumor milieu. As a result, STZ-diabetic CD8(+) effector cells were neither able to eliminate tumor nor to improve survival of tumor-bearing mice. Taken together, our data suggest that CD8(+) CTLs are crippled to infiltrate into tumors and thus fail to acquire tumor-killing capability in STZ-diabetic hosts.
format Online
Article
Text
id pubmed-4229103
institution National Center for Biotechnology Information
language English
publishDate 2014
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-42291032014-11-18 Defects in the Acquisition of Tumor-Killing Capability of CD8(+) Cytotoxic T Cells in Streptozotocin-Induced Diabetic Mice Chen, Shu-Ching Su, Yu-Chia Lu, Ya-Ting Ko, Patrick Chow-In Chang, Pei-Yu Lin, Hung-Ju Ho, Hong-Nerng Lai, Yo-Ping PLoS One Research Article Emerging evidences have shown that diabetes mellitus not only raises risk but also heightens mortality rate of cancer. It is not clear, however, whether antitumor CD8(+) cytotoxic T lymphocyte (CTL) response is down-modulated in diabetic hosts. We investigated the impact of hyperglycemia on CTLs' acquisition of tumor-killing capability by utilizing streptozotocin-induced diabetic (STZ-diabetic) mice. Murine diabetes was induced by intraperitoneal injection of STZ (200 mg/kg) in C57BL/6 mice, 2C-T cell receptor (TCR) transgenic and P14-TCR transgenic mice. The study found that, despite harboring intact proliferative capacity measured with CFSE labeling and MTT assay, STZ-diabetic CD8(+) CTLs displayed impaired effector functions. After stimulation, STZ-diabetic CD8(+) CTLs produced less perforin and TNFα assessed by intracellular staining, as well as expressed less CD103 protein. Furthermore, adoptive transfer of STZ-diabetic P14 CD8(+) effector cells showed an insufficient recruitment to the B16.gp33 melanoma and inadequate production of perforin, granzyme B and TNFα determined by immunohistochemistry in the tumor milieu. As a result, STZ-diabetic CD8(+) effector cells were neither able to eliminate tumor nor to improve survival of tumor-bearing mice. Taken together, our data suggest that CD8(+) CTLs are crippled to infiltrate into tumors and thus fail to acquire tumor-killing capability in STZ-diabetic hosts. Public Library of Science 2014-11-12 /pmc/articles/PMC4229103/ /pubmed/25390652 http://dx.doi.org/10.1371/journal.pone.0109961 Text en © 2014 Chen et al http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.
spellingShingle Research Article
Chen, Shu-Ching
Su, Yu-Chia
Lu, Ya-Ting
Ko, Patrick Chow-In
Chang, Pei-Yu
Lin, Hung-Ju
Ho, Hong-Nerng
Lai, Yo-Ping
Defects in the Acquisition of Tumor-Killing Capability of CD8(+) Cytotoxic T Cells in Streptozotocin-Induced Diabetic Mice
title Defects in the Acquisition of Tumor-Killing Capability of CD8(+) Cytotoxic T Cells in Streptozotocin-Induced Diabetic Mice
title_full Defects in the Acquisition of Tumor-Killing Capability of CD8(+) Cytotoxic T Cells in Streptozotocin-Induced Diabetic Mice
title_fullStr Defects in the Acquisition of Tumor-Killing Capability of CD8(+) Cytotoxic T Cells in Streptozotocin-Induced Diabetic Mice
title_full_unstemmed Defects in the Acquisition of Tumor-Killing Capability of CD8(+) Cytotoxic T Cells in Streptozotocin-Induced Diabetic Mice
title_short Defects in the Acquisition of Tumor-Killing Capability of CD8(+) Cytotoxic T Cells in Streptozotocin-Induced Diabetic Mice
title_sort defects in the acquisition of tumor-killing capability of cd8(+) cytotoxic t cells in streptozotocin-induced diabetic mice
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4229103/
https://www.ncbi.nlm.nih.gov/pubmed/25390652
http://dx.doi.org/10.1371/journal.pone.0109961
work_keys_str_mv AT chenshuching defectsintheacquisitionoftumorkillingcapabilityofcd8cytotoxictcellsinstreptozotocininduceddiabeticmice
AT suyuchia defectsintheacquisitionoftumorkillingcapabilityofcd8cytotoxictcellsinstreptozotocininduceddiabeticmice
AT luyating defectsintheacquisitionoftumorkillingcapabilityofcd8cytotoxictcellsinstreptozotocininduceddiabeticmice
AT kopatrickchowin defectsintheacquisitionoftumorkillingcapabilityofcd8cytotoxictcellsinstreptozotocininduceddiabeticmice
AT changpeiyu defectsintheacquisitionoftumorkillingcapabilityofcd8cytotoxictcellsinstreptozotocininduceddiabeticmice
AT linhungju defectsintheacquisitionoftumorkillingcapabilityofcd8cytotoxictcellsinstreptozotocininduceddiabeticmice
AT hohongnerng defectsintheacquisitionoftumorkillingcapabilityofcd8cytotoxictcellsinstreptozotocininduceddiabeticmice
AT laiyoping defectsintheacquisitionoftumorkillingcapabilityofcd8cytotoxictcellsinstreptozotocininduceddiabeticmice