Nestin(+) tissue-resident multipotent stem cells contribute to tumor progression by differentiating into pericytes and smooth muscle cells resulting in blood vessel remodeling

Tumor vessels with resistance to anti-angiogenic therapy are characterized by the normalization of the vascular structures through integration of mature pericytes and smooth muscle cells (SMC) into the vessel wall, a process termed vessel stabilization. Unfortunately, stabilization-associated vascul...

Full description

Bibliographic Details
Main Authors: Diana eKlein, Nicole eMeissner, Veronika eKleff, Holger eJastrow, Masahiro eYamaguchi, Süleyman eErgün, Verena eJendrossek
Format: Article
Language:English
Published: Frontiers Media S.A. 2014-06-01
Series:Frontiers in Oncology
Subjects:
Online Access:http://journal.frontiersin.org/Journal/10.3389/fonc.2014.00169/full
_version_ 1811231225075990528
author Diana eKlein
Nicole eMeissner
Veronika eKleff
Holger eJastrow
Masahiro eYamaguchi
Süleyman eErgün
Verena eJendrossek
author_facet Diana eKlein
Nicole eMeissner
Veronika eKleff
Holger eJastrow
Masahiro eYamaguchi
Süleyman eErgün
Verena eJendrossek
author_sort Diana eKlein
collection DOAJ
description Tumor vessels with resistance to anti-angiogenic therapy are characterized by the normalization of the vascular structures through integration of mature pericytes and smooth muscle cells (SMC) into the vessel wall, a process termed vessel stabilization. Unfortunately, stabilization-associated vascular remodeling can result in reduced sensitivity to subsequent anti-angiogenic therapy. We show here that blockade of VEGF by bevacizumab induces stabilization of angiogenic tumor blood vessels in human tumor specimen by recruiting Nestin-positive cells, whereas mature vessels down-regulated Nestin-expression. Using xenograft tumors growing on bone-marrow (BM) chimera of C57Bl/6 wildtype and Nestin-GFP transgenic mice we show for first time that Nestin(+) cells inducing the maturation of tumor vessels do not originate from the BM but presumably reside within the adventitia of adult blood vessels. Complementary ex vivo experiments using explants of murine aortas revealed that Nestin(+) multipotent stem cells (MPSCs) are mobilized from their niche and differentiated into pericytes and SMC through the influence of tumor-cell secreted factors. We conclude that tissue-resident Nestin(+) cells are more relevant than BM-derived cells for vessel stabilization and therefore have to be considered in future strategies for anti-angiogenic therapy. The identification of proteins mediating recruitment or differentiation of local Nestin(+) cells with potential stem cell character to angiogenic blood vessels may allow the definition of new therapeutic targets to reduce tumor resistance against anti-angiogenic drugs.
first_indexed 2024-04-12T10:40:28Z
format Article
id doaj.art-a29a28de57b9424f8ec6525d21818306
institution Directory Open Access Journal
issn 2234-943X
language English
last_indexed 2024-04-12T10:40:28Z
publishDate 2014-06-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Oncology
spelling doaj.art-a29a28de57b9424f8ec6525d218183062022-12-22T03:36:36ZengFrontiers Media S.A.Frontiers in Oncology2234-943X2014-06-01410.3389/fonc.2014.0016979529Nestin(+) tissue-resident multipotent stem cells contribute to tumor progression by differentiating into pericytes and smooth muscle cells resulting in blood vessel remodelingDiana eKlein0Nicole eMeissner1Veronika eKleff2Holger eJastrow3Masahiro eYamaguchi4Süleyman eErgün5Verena eJendrossek6University of Duisburg-EssenUniversity of Duisburg-EssenUniversity of Duisburg-EssenUniversity of Duisburg-EssenUniversity of TokyoUniversity of WürzburgUniversity of Duisburg-EssenTumor vessels with resistance to anti-angiogenic therapy are characterized by the normalization of the vascular structures through integration of mature pericytes and smooth muscle cells (SMC) into the vessel wall, a process termed vessel stabilization. Unfortunately, stabilization-associated vascular remodeling can result in reduced sensitivity to subsequent anti-angiogenic therapy. We show here that blockade of VEGF by bevacizumab induces stabilization of angiogenic tumor blood vessels in human tumor specimen by recruiting Nestin-positive cells, whereas mature vessels down-regulated Nestin-expression. Using xenograft tumors growing on bone-marrow (BM) chimera of C57Bl/6 wildtype and Nestin-GFP transgenic mice we show for first time that Nestin(+) cells inducing the maturation of tumor vessels do not originate from the BM but presumably reside within the adventitia of adult blood vessels. Complementary ex vivo experiments using explants of murine aortas revealed that Nestin(+) multipotent stem cells (MPSCs) are mobilized from their niche and differentiated into pericytes and SMC through the influence of tumor-cell secreted factors. We conclude that tissue-resident Nestin(+) cells are more relevant than BM-derived cells for vessel stabilization and therefore have to be considered in future strategies for anti-angiogenic therapy. The identification of proteins mediating recruitment or differentiation of local Nestin(+) cells with potential stem cell character to angiogenic blood vessels may allow the definition of new therapeutic targets to reduce tumor resistance against anti-angiogenic drugs.http://journal.frontiersin.org/Journal/10.3389/fonc.2014.00169/fullPericytesmesenchymal stem cellvascular remodelingnestinvessel maturation
spellingShingle Diana eKlein
Nicole eMeissner
Veronika eKleff
Holger eJastrow
Masahiro eYamaguchi
Süleyman eErgün
Verena eJendrossek
Nestin(+) tissue-resident multipotent stem cells contribute to tumor progression by differentiating into pericytes and smooth muscle cells resulting in blood vessel remodeling
Frontiers in Oncology
Pericytes
mesenchymal stem cell
vascular remodeling
nestin
vessel maturation
title Nestin(+) tissue-resident multipotent stem cells contribute to tumor progression by differentiating into pericytes and smooth muscle cells resulting in blood vessel remodeling
title_full Nestin(+) tissue-resident multipotent stem cells contribute to tumor progression by differentiating into pericytes and smooth muscle cells resulting in blood vessel remodeling
title_fullStr Nestin(+) tissue-resident multipotent stem cells contribute to tumor progression by differentiating into pericytes and smooth muscle cells resulting in blood vessel remodeling
title_full_unstemmed Nestin(+) tissue-resident multipotent stem cells contribute to tumor progression by differentiating into pericytes and smooth muscle cells resulting in blood vessel remodeling
title_short Nestin(+) tissue-resident multipotent stem cells contribute to tumor progression by differentiating into pericytes and smooth muscle cells resulting in blood vessel remodeling
title_sort nestin tissue resident multipotent stem cells contribute to tumor progression by differentiating into pericytes and smooth muscle cells resulting in blood vessel remodeling
topic Pericytes
mesenchymal stem cell
vascular remodeling
nestin
vessel maturation
url http://journal.frontiersin.org/Journal/10.3389/fonc.2014.00169/full
work_keys_str_mv AT dianaeklein nestintissueresidentmultipotentstemcellscontributetotumorprogressionbydifferentiatingintopericytesandsmoothmusclecellsresultinginbloodvesselremodeling
AT nicoleemeissner nestintissueresidentmultipotentstemcellscontributetotumorprogressionbydifferentiatingintopericytesandsmoothmusclecellsresultinginbloodvesselremodeling
AT veronikaekleff nestintissueresidentmultipotentstemcellscontributetotumorprogressionbydifferentiatingintopericytesandsmoothmusclecellsresultinginbloodvesselremodeling
AT holgerejastrow nestintissueresidentmultipotentstemcellscontributetotumorprogressionbydifferentiatingintopericytesandsmoothmusclecellsresultinginbloodvesselremodeling
AT masahiroeyamaguchi nestintissueresidentmultipotentstemcellscontributetotumorprogressionbydifferentiatingintopericytesandsmoothmusclecellsresultinginbloodvesselremodeling
AT suleymaneergun nestintissueresidentmultipotentstemcellscontributetotumorprogressionbydifferentiatingintopericytesandsmoothmusclecellsresultinginbloodvesselremodeling
AT verenaejendrossek nestintissueresidentmultipotentstemcellscontributetotumorprogressionbydifferentiatingintopericytesandsmoothmusclecellsresultinginbloodvesselremodeling