Human Teratoma-Derived Hematopoiesis Is a Highly Polyclonal Process Supported by Human Umbilical Vein Endothelial Cells

Summary: Hematopoietic stem cells (HSCs) ensure a life-long regeneration of the blood system and are therefore an important source for transplantation and gene therapy. The teratoma environment supports the complex development of functional HSCs from human pluripotent stem cells, which is difficult...

Full description

Bibliographic Details
Main Authors: Friederike Philipp, Anton Selich, Michael Rothe, Dirk Hoffmann, Susanne Rittinghausen, Michael A. Morgan, Denise Klatt, Silke Glage, Stefan Lienenklaus, Vanessa Neuhaus, Katherina Sewald, Armin Braun, Axel Schambach
Format: Article
Language:English
Published: Elsevier 2018-11-01
Series:Stem Cell Reports
Online Access:http://www.sciencedirect.com/science/article/pii/S2213671118303953
id doaj-8106813214384656a4a7ca4fb6cb1c6a
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Friederike Philipp
Anton Selich
Michael Rothe
Dirk Hoffmann
Susanne Rittinghausen
Michael A. Morgan
Denise Klatt
Silke Glage
Stefan Lienenklaus
Vanessa Neuhaus
Katherina Sewald
Armin Braun
Axel Schambach
spellingShingle Friederike Philipp
Anton Selich
Michael Rothe
Dirk Hoffmann
Susanne Rittinghausen
Michael A. Morgan
Denise Klatt
Silke Glage
Stefan Lienenklaus
Vanessa Neuhaus
Katherina Sewald
Armin Braun
Axel Schambach
Human Teratoma-Derived Hematopoiesis Is a Highly Polyclonal Process Supported by Human Umbilical Vein Endothelial Cells
Stem Cell Reports
author_facet Friederike Philipp
Anton Selich
Michael Rothe
Dirk Hoffmann
Susanne Rittinghausen
Michael A. Morgan
Denise Klatt
Silke Glage
Stefan Lienenklaus
Vanessa Neuhaus
Katherina Sewald
Armin Braun
Axel Schambach
author_sort Friederike Philipp
title Human Teratoma-Derived Hematopoiesis Is a Highly Polyclonal Process Supported by Human Umbilical Vein Endothelial Cells
title_short Human Teratoma-Derived Hematopoiesis Is a Highly Polyclonal Process Supported by Human Umbilical Vein Endothelial Cells
title_full Human Teratoma-Derived Hematopoiesis Is a Highly Polyclonal Process Supported by Human Umbilical Vein Endothelial Cells
title_fullStr Human Teratoma-Derived Hematopoiesis Is a Highly Polyclonal Process Supported by Human Umbilical Vein Endothelial Cells
title_full_unstemmed Human Teratoma-Derived Hematopoiesis Is a Highly Polyclonal Process Supported by Human Umbilical Vein Endothelial Cells
title_sort human teratoma-derived hematopoiesis is a highly polyclonal process supported by human umbilical vein endothelial cells
publisher Elsevier
series Stem Cell Reports
issn 2213-6711
publishDate 2018-11-01
description Summary: Hematopoietic stem cells (HSCs) ensure a life-long regeneration of the blood system and are therefore an important source for transplantation and gene therapy. The teratoma environment supports the complex development of functional HSCs from human pluripotent stem cells, which is difficult to recapitulate in culture. This model mimics various aspects of early hematopoiesis, but is restricted by the low spontaneous hematopoiesis rate. In this study, a feasible protocol for robust hematopoiesis has been elaborated. We achieved a significant increase of the teratoma-derived hematopoietic population when teratomas were generated in the NSGS mouse, which provides human cytokines, together with co-injection of human umbilical vein endothelial cells. Since little is known about hematopoiesis in teratomas, we addressed localization and clonality of the hematopoietic lineage. Our results indicate that early human hematopoiesis is closely reflected in teratoma formation, and thus highlight the value of this model. : Schambach and colleagues show evidence that hematopoiesis in hiPSC-derived teratomas occurs by endothelial-to-hematopoietic transition in a highly polyclonal manner. This process was supported by the presence of HUVECs in the NSGS mouse, which provides human SCF, IL-3, and GM-CSF. The established hematopoiesis model can potentially be used for disease modeling and improvement of hematopoietic differentiation protocols. Keywords: hematopoiesis, teratoma, hiPSC, EHT, genetic barcoding, HUVECs, embryogenesis
url http://www.sciencedirect.com/science/article/pii/S2213671118303953
work_keys_str_mv AT friederikephilipp humanteratomaderivedhematopoiesisisahighlypolyclonalprocesssupportedbyhumanumbilicalveinendothelialcells
AT antonselich humanteratomaderivedhematopoiesisisahighlypolyclonalprocesssupportedbyhumanumbilicalveinendothelialcells
AT michaelrothe humanteratomaderivedhematopoiesisisahighlypolyclonalprocesssupportedbyhumanumbilicalveinendothelialcells
AT dirkhoffmann humanteratomaderivedhematopoiesisisahighlypolyclonalprocesssupportedbyhumanumbilicalveinendothelialcells
AT susannerittinghausen humanteratomaderivedhematopoiesisisahighlypolyclonalprocesssupportedbyhumanumbilicalveinendothelialcells
AT michaelamorgan humanteratomaderivedhematopoiesisisahighlypolyclonalprocesssupportedbyhumanumbilicalveinendothelialcells
AT deniseklatt humanteratomaderivedhematopoiesisisahighlypolyclonalprocesssupportedbyhumanumbilicalveinendothelialcells
AT silkeglage humanteratomaderivedhematopoiesisisahighlypolyclonalprocesssupportedbyhumanumbilicalveinendothelialcells
AT stefanlienenklaus humanteratomaderivedhematopoiesisisahighlypolyclonalprocesssupportedbyhumanumbilicalveinendothelialcells
AT vanessaneuhaus humanteratomaderivedhematopoiesisisahighlypolyclonalprocesssupportedbyhumanumbilicalveinendothelialcells
AT katherinasewald humanteratomaderivedhematopoiesisisahighlypolyclonalprocesssupportedbyhumanumbilicalveinendothelialcells
AT arminbraun humanteratomaderivedhematopoiesisisahighlypolyclonalprocesssupportedbyhumanumbilicalveinendothelialcells
AT axelschambach humanteratomaderivedhematopoiesisisahighlypolyclonalprocesssupportedbyhumanumbilicalveinendothelialcells
_version_ 1725891966042898432
spelling doaj-8106813214384656a4a7ca4fb6cb1c6a2020-11-24T21:48:27ZengElsevierStem Cell Reports2213-67112018-11-0111510511060Human Teratoma-Derived Hematopoiesis Is a Highly Polyclonal Process Supported by Human Umbilical Vein Endothelial CellsFriederike Philipp0Anton Selich1Michael Rothe2Dirk Hoffmann3Susanne Rittinghausen4Michael A. Morgan5Denise Klatt6Silke Glage7Stefan Lienenklaus8Vanessa Neuhaus9Katherina Sewald10Armin Braun11Axel Schambach12Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Straße1, 30625 Hannover, Germany; Fraunhofer Institute for Toxicology and Experimental Medicine, 30625 Hannover, Germany; REBIRTH Cluster of Excellence, Hannover Medical School, 30625 Hannover, GermanyInstitute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Straße1, 30625 Hannover, GermanyInstitute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Straße1, 30625 Hannover, Germany; REBIRTH Cluster of Excellence, Hannover Medical School, 30625 Hannover, GermanyInstitute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Straße1, 30625 Hannover, Germany; REBIRTH Cluster of Excellence, Hannover Medical School, 30625 Hannover, GermanyFraunhofer Institute for Toxicology and Experimental Medicine, 30625 Hannover, Germany; REBIRTH Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; German Centre for Lung Research DZL, Hannover, GermanyInstitute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Straße1, 30625 Hannover, Germany; REBIRTH Cluster of Excellence, Hannover Medical School, 30625 Hannover, GermanyInstitute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Straße1, 30625 Hannover, Germany; REBIRTH Cluster of Excellence, Hannover Medical School, 30625 Hannover, GermanyInstitute for Laboratory Animal Science, Hannover Medical School, 30625 Hannover, Germany; REBIRTH Cluster of Excellence, Hannover Medical School, 30625 Hannover, GermanyInstitute for Laboratory Animal Science, Hannover Medical School, 30625 Hannover, GermanyFraunhofer Institute for Toxicology and Experimental Medicine, 30625 Hannover, Germany; REBIRTH Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; German Centre for Lung Research DZL, Hannover, GermanyFraunhofer Institute for Toxicology and Experimental Medicine, 30625 Hannover, Germany; REBIRTH Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; German Centre for Lung Research DZL, Hannover, GermanyFraunhofer Institute for Toxicology and Experimental Medicine, 30625 Hannover, Germany; REBIRTH Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; German Centre for Lung Research DZL, Hannover, GermanyInstitute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Straße1, 30625 Hannover, Germany; REBIRTH Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany; Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Corresponding authorSummary: Hematopoietic stem cells (HSCs) ensure a life-long regeneration of the blood system and are therefore an important source for transplantation and gene therapy. The teratoma environment supports the complex development of functional HSCs from human pluripotent stem cells, which is difficult to recapitulate in culture. This model mimics various aspects of early hematopoiesis, but is restricted by the low spontaneous hematopoiesis rate. In this study, a feasible protocol for robust hematopoiesis has been elaborated. We achieved a significant increase of the teratoma-derived hematopoietic population when teratomas were generated in the NSGS mouse, which provides human cytokines, together with co-injection of human umbilical vein endothelial cells. Since little is known about hematopoiesis in teratomas, we addressed localization and clonality of the hematopoietic lineage. Our results indicate that early human hematopoiesis is closely reflected in teratoma formation, and thus highlight the value of this model. : Schambach and colleagues show evidence that hematopoiesis in hiPSC-derived teratomas occurs by endothelial-to-hematopoietic transition in a highly polyclonal manner. This process was supported by the presence of HUVECs in the NSGS mouse, which provides human SCF, IL-3, and GM-CSF. The established hematopoiesis model can potentially be used for disease modeling and improvement of hematopoietic differentiation protocols. Keywords: hematopoiesis, teratoma, hiPSC, EHT, genetic barcoding, HUVECs, embryogenesishttp://www.sciencedirect.com/science/article/pii/S2213671118303953