Cargando…

RB inactivation in keratin 18 positive thymic epithelial cells promotes non-cell autonomous T cell hyperproliferation in genetically engineered mice

Thymic epithelial cells (TEC), as part of thymic stroma, provide essential growth factors/cytokines and self-antigens to support T cell development and selection. Deletion of Rb family proteins in adult thymic stroma leads to T cell hyperplasia in vivo. To determine whether deletion of Rb specifical...

Descripción completa

Detalles Bibliográficos
Autores principales: Song, Yurong, Sullivan, Teresa, Klarmann, Kimberly, Gilbert, Debra, O’Sullivan, T. Norene, Lu, Lucy, Wang, Sophie, Haines, Diana C., Van Dyke, Terry, Keller, Jonathan R.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5291521/
https://www.ncbi.nlm.nih.gov/pubmed/28158249
http://dx.doi.org/10.1371/journal.pone.0171510
_version_ 1782504793565036544
author Song, Yurong
Sullivan, Teresa
Klarmann, Kimberly
Gilbert, Debra
O’Sullivan, T. Norene
Lu, Lucy
Wang, Sophie
Haines, Diana C.
Van Dyke, Terry
Keller, Jonathan R.
author_facet Song, Yurong
Sullivan, Teresa
Klarmann, Kimberly
Gilbert, Debra
O’Sullivan, T. Norene
Lu, Lucy
Wang, Sophie
Haines, Diana C.
Van Dyke, Terry
Keller, Jonathan R.
author_sort Song, Yurong
collection PubMed
description Thymic epithelial cells (TEC), as part of thymic stroma, provide essential growth factors/cytokines and self-antigens to support T cell development and selection. Deletion of Rb family proteins in adult thymic stroma leads to T cell hyperplasia in vivo. To determine whether deletion of Rb specifically in keratin (K) 18 positive TEC was sufficient for thymocyte hyperplasia, we conditionally inactivated Rb and its family members p107 and p130 in K18+ TEC in genetically engineered mice (TgK18GT(121); K18 mice). We found that thymocyte hyperproliferation was induced in mice with Rb inactivation in K18+ TEC, while normal T cell development was maintained; suggesting that inactivation of Rb specifically in K18+ TEC was sufficient and responsible for the phenotype. Transplantation of wild type bone marrow cells into mice with Rb inactivation in K18+ TEC resulted in donor T lymphocyte hyperplasia confirming the non-cell autonomous requirement for Rb proteins in K18+ TEC in regulating T cell proliferation. Our data suggests that thymic epithelial cells play an important role in regulating lymphoid proliferation and thymus size.
format Online
Article
Text
id pubmed-5291521
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-52915212017-02-17 RB inactivation in keratin 18 positive thymic epithelial cells promotes non-cell autonomous T cell hyperproliferation in genetically engineered mice Song, Yurong Sullivan, Teresa Klarmann, Kimberly Gilbert, Debra O’Sullivan, T. Norene Lu, Lucy Wang, Sophie Haines, Diana C. Van Dyke, Terry Keller, Jonathan R. PLoS One Research Article Thymic epithelial cells (TEC), as part of thymic stroma, provide essential growth factors/cytokines and self-antigens to support T cell development and selection. Deletion of Rb family proteins in adult thymic stroma leads to T cell hyperplasia in vivo. To determine whether deletion of Rb specifically in keratin (K) 18 positive TEC was sufficient for thymocyte hyperplasia, we conditionally inactivated Rb and its family members p107 and p130 in K18+ TEC in genetically engineered mice (TgK18GT(121); K18 mice). We found that thymocyte hyperproliferation was induced in mice with Rb inactivation in K18+ TEC, while normal T cell development was maintained; suggesting that inactivation of Rb specifically in K18+ TEC was sufficient and responsible for the phenotype. Transplantation of wild type bone marrow cells into mice with Rb inactivation in K18+ TEC resulted in donor T lymphocyte hyperplasia confirming the non-cell autonomous requirement for Rb proteins in K18+ TEC in regulating T cell proliferation. Our data suggests that thymic epithelial cells play an important role in regulating lymphoid proliferation and thymus size. Public Library of Science 2017-02-03 /pmc/articles/PMC5291521/ /pubmed/28158249 http://dx.doi.org/10.1371/journal.pone.0171510 Text en https://creativecommons.org/publicdomain/zero/1.0/ This is an open access article, free of all copyright, and may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose. The work is made available under the Creative Commons CC0 (https://creativecommons.org/publicdomain/zero/1.0/) public domain dedication.
spellingShingle Research Article
Song, Yurong
Sullivan, Teresa
Klarmann, Kimberly
Gilbert, Debra
O’Sullivan, T. Norene
Lu, Lucy
Wang, Sophie
Haines, Diana C.
Van Dyke, Terry
Keller, Jonathan R.
RB inactivation in keratin 18 positive thymic epithelial cells promotes non-cell autonomous T cell hyperproliferation in genetically engineered mice
title RB inactivation in keratin 18 positive thymic epithelial cells promotes non-cell autonomous T cell hyperproliferation in genetically engineered mice
title_full RB inactivation in keratin 18 positive thymic epithelial cells promotes non-cell autonomous T cell hyperproliferation in genetically engineered mice
title_fullStr RB inactivation in keratin 18 positive thymic epithelial cells promotes non-cell autonomous T cell hyperproliferation in genetically engineered mice
title_full_unstemmed RB inactivation in keratin 18 positive thymic epithelial cells promotes non-cell autonomous T cell hyperproliferation in genetically engineered mice
title_short RB inactivation in keratin 18 positive thymic epithelial cells promotes non-cell autonomous T cell hyperproliferation in genetically engineered mice
title_sort rb inactivation in keratin 18 positive thymic epithelial cells promotes non-cell autonomous t cell hyperproliferation in genetically engineered mice
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5291521/
https://www.ncbi.nlm.nih.gov/pubmed/28158249
http://dx.doi.org/10.1371/journal.pone.0171510
work_keys_str_mv AT songyurong rbinactivationinkeratin18positivethymicepithelialcellspromotesnoncellautonomoustcellhyperproliferationingeneticallyengineeredmice
AT sullivanteresa rbinactivationinkeratin18positivethymicepithelialcellspromotesnoncellautonomoustcellhyperproliferationingeneticallyengineeredmice
AT klarmannkimberly rbinactivationinkeratin18positivethymicepithelialcellspromotesnoncellautonomoustcellhyperproliferationingeneticallyengineeredmice
AT gilbertdebra rbinactivationinkeratin18positivethymicepithelialcellspromotesnoncellautonomoustcellhyperproliferationingeneticallyengineeredmice
AT osullivantnorene rbinactivationinkeratin18positivethymicepithelialcellspromotesnoncellautonomoustcellhyperproliferationingeneticallyengineeredmice
AT lulucy rbinactivationinkeratin18positivethymicepithelialcellspromotesnoncellautonomoustcellhyperproliferationingeneticallyengineeredmice
AT wangsophie rbinactivationinkeratin18positivethymicepithelialcellspromotesnoncellautonomoustcellhyperproliferationingeneticallyengineeredmice
AT hainesdianac rbinactivationinkeratin18positivethymicepithelialcellspromotesnoncellautonomoustcellhyperproliferationingeneticallyengineeredmice
AT vandyketerry rbinactivationinkeratin18positivethymicepithelialcellspromotesnoncellautonomoustcellhyperproliferationingeneticallyengineeredmice
AT kellerjonathanr rbinactivationinkeratin18positivethymicepithelialcellspromotesnoncellautonomoustcellhyperproliferationingeneticallyengineeredmice