Cargando…

Human multipotent adult progenitor cells effectively reduce graft‐vs‐host disease while preserving graft‐vs‐leukemia activity

Graft‐vs‐host disease (GvHD) limits successful outcomes following allogeneic blood and marrow transplantation (allo‐BMT). We examined whether the administration of human, bone marrow‐derived, multipotent adult progenitor cells (MAPCs™) could regulate experimental GvHD. The immunoregulatory capacity...

Descripción completa

Detalles Bibliográficos
Autores principales: Metheny, Leland, Eid, Saada, Wuttisarnwattana, Patiwet, Auletta, Jeffery J., Liu, Chen, Van Dervort, Alana, Paez, Conner, Lee, ZhengHong, Wilson, David, Lazarus, Hillard M., Deans, Robert, Vant Hof, Wouter, Ktena, Yiouli, Cooke, Kenneth R.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley & Sons, Inc. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8596993/
https://www.ncbi.nlm.nih.gov/pubmed/34255899
http://dx.doi.org/10.1002/stem.3434
_version_ 1784600511635259392
author Metheny, Leland
Eid, Saada
Wuttisarnwattana, Patiwet
Auletta, Jeffery J.
Liu, Chen
Van Dervort, Alana
Paez, Conner
Lee, ZhengHong
Wilson, David
Lazarus, Hillard M.
Deans, Robert
Vant Hof, Wouter
Ktena, Yiouli
Cooke, Kenneth R.
author_facet Metheny, Leland
Eid, Saada
Wuttisarnwattana, Patiwet
Auletta, Jeffery J.
Liu, Chen
Van Dervort, Alana
Paez, Conner
Lee, ZhengHong
Wilson, David
Lazarus, Hillard M.
Deans, Robert
Vant Hof, Wouter
Ktena, Yiouli
Cooke, Kenneth R.
author_sort Metheny, Leland
collection PubMed
description Graft‐vs‐host disease (GvHD) limits successful outcomes following allogeneic blood and marrow transplantation (allo‐BMT). We examined whether the administration of human, bone marrow‐derived, multipotent adult progenitor cells (MAPCs™) could regulate experimental GvHD. The immunoregulatory capacity of MAPC cells was evaluated in vivo using established murine GvHD models. Injection of MAPC cells on day +1 (D1) and +4 (D4) significantly reduced T‐cell expansion and the numbers of donor‐derived, Tumor Necrosis Factor Alpha (TNFα) and Interferon Gamma (IFNγ)‐producing, CD4+ and CD8+ cells by D10 compared with untreated controls. These findings were associated with reductions in serum levels of TNFα and IFNγ, intestinal and hepatic inflammation and systemic GvHD as measured by survival and clinical score. Biodistribution studies showed that MAPC cells tracked from the lung and to the liver, spleen, and mesenteric nodes within 24 hours after injection. MAPC cells inhibited mouse T‐cell proliferation in vitro and this effect was associated with reduced T‐cell activation and inflammatory cytokine secretion and robust increases in the concentrations of Prostaglandin E2 (PGE2) and Transforming Growth Factor Beta (TGFβ). Indomethacin and E‐prostanoid 2 (EP2) receptor antagonism both reversed while EP2 agonism restored MAPC cell‐mediated in vitro T‐cell suppression, confirming the role for PGE2. Furthermore, cyclo‐oxygenase inhibition following allo‐BMT abrogated the protective effects of MAPC cells. Importantly, MAPC cells had no effect on the generation cytotoxic T lymphocyte activity in vitro, and the administration of MAPC cells in the setting of leukemic challenge resulted in superior leukemia‐free survival. Collectively, these data provide valuable information regarding the biodistribution and regulatory capacity of MAPC cells, which may inform future clinical trial design.
format Online
Article
Text
id pubmed-8596993
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher John Wiley & Sons, Inc.
record_format MEDLINE/PubMed
spelling pubmed-85969932021-11-22 Human multipotent adult progenitor cells effectively reduce graft‐vs‐host disease while preserving graft‐vs‐leukemia activity Metheny, Leland Eid, Saada Wuttisarnwattana, Patiwet Auletta, Jeffery J. Liu, Chen Van Dervort, Alana Paez, Conner Lee, ZhengHong Wilson, David Lazarus, Hillard M. Deans, Robert Vant Hof, Wouter Ktena, Yiouli Cooke, Kenneth R. Stem Cells Tissue‐specific Stem Cells Graft‐vs‐host disease (GvHD) limits successful outcomes following allogeneic blood and marrow transplantation (allo‐BMT). We examined whether the administration of human, bone marrow‐derived, multipotent adult progenitor cells (MAPCs™) could regulate experimental GvHD. The immunoregulatory capacity of MAPC cells was evaluated in vivo using established murine GvHD models. Injection of MAPC cells on day +1 (D1) and +4 (D4) significantly reduced T‐cell expansion and the numbers of donor‐derived, Tumor Necrosis Factor Alpha (TNFα) and Interferon Gamma (IFNγ)‐producing, CD4+ and CD8+ cells by D10 compared with untreated controls. These findings were associated with reductions in serum levels of TNFα and IFNγ, intestinal and hepatic inflammation and systemic GvHD as measured by survival and clinical score. Biodistribution studies showed that MAPC cells tracked from the lung and to the liver, spleen, and mesenteric nodes within 24 hours after injection. MAPC cells inhibited mouse T‐cell proliferation in vitro and this effect was associated with reduced T‐cell activation and inflammatory cytokine secretion and robust increases in the concentrations of Prostaglandin E2 (PGE2) and Transforming Growth Factor Beta (TGFβ). Indomethacin and E‐prostanoid 2 (EP2) receptor antagonism both reversed while EP2 agonism restored MAPC cell‐mediated in vitro T‐cell suppression, confirming the role for PGE2. Furthermore, cyclo‐oxygenase inhibition following allo‐BMT abrogated the protective effects of MAPC cells. Importantly, MAPC cells had no effect on the generation cytotoxic T lymphocyte activity in vitro, and the administration of MAPC cells in the setting of leukemic challenge resulted in superior leukemia‐free survival. Collectively, these data provide valuable information regarding the biodistribution and regulatory capacity of MAPC cells, which may inform future clinical trial design. John Wiley & Sons, Inc. 2021-07-28 2021-11 /pmc/articles/PMC8596993/ /pubmed/34255899 http://dx.doi.org/10.1002/stem.3434 Text en © 2021 The Authors. STEM CELLS published by Wiley Periodicals LLC on behalf of AlphaMed Press. https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc-nd/4.0/ (https://creativecommons.org/licenses/by-nc-nd/4.0/) License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made.
spellingShingle Tissue‐specific Stem Cells
Metheny, Leland
Eid, Saada
Wuttisarnwattana, Patiwet
Auletta, Jeffery J.
Liu, Chen
Van Dervort, Alana
Paez, Conner
Lee, ZhengHong
Wilson, David
Lazarus, Hillard M.
Deans, Robert
Vant Hof, Wouter
Ktena, Yiouli
Cooke, Kenneth R.
Human multipotent adult progenitor cells effectively reduce graft‐vs‐host disease while preserving graft‐vs‐leukemia activity
title Human multipotent adult progenitor cells effectively reduce graft‐vs‐host disease while preserving graft‐vs‐leukemia activity
title_full Human multipotent adult progenitor cells effectively reduce graft‐vs‐host disease while preserving graft‐vs‐leukemia activity
title_fullStr Human multipotent adult progenitor cells effectively reduce graft‐vs‐host disease while preserving graft‐vs‐leukemia activity
title_full_unstemmed Human multipotent adult progenitor cells effectively reduce graft‐vs‐host disease while preserving graft‐vs‐leukemia activity
title_short Human multipotent adult progenitor cells effectively reduce graft‐vs‐host disease while preserving graft‐vs‐leukemia activity
title_sort human multipotent adult progenitor cells effectively reduce graft‐vs‐host disease while preserving graft‐vs‐leukemia activity
topic Tissue‐specific Stem Cells
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8596993/
https://www.ncbi.nlm.nih.gov/pubmed/34255899
http://dx.doi.org/10.1002/stem.3434
work_keys_str_mv AT methenyleland humanmultipotentadultprogenitorcellseffectivelyreducegraftvshostdiseasewhilepreservinggraftvsleukemiaactivity
AT eidsaada humanmultipotentadultprogenitorcellseffectivelyreducegraftvshostdiseasewhilepreservinggraftvsleukemiaactivity
AT wuttisarnwattanapatiwet humanmultipotentadultprogenitorcellseffectivelyreducegraftvshostdiseasewhilepreservinggraftvsleukemiaactivity
AT aulettajefferyj humanmultipotentadultprogenitorcellseffectivelyreducegraftvshostdiseasewhilepreservinggraftvsleukemiaactivity
AT liuchen humanmultipotentadultprogenitorcellseffectivelyreducegraftvshostdiseasewhilepreservinggraftvsleukemiaactivity
AT vandervortalana humanmultipotentadultprogenitorcellseffectivelyreducegraftvshostdiseasewhilepreservinggraftvsleukemiaactivity
AT paezconner humanmultipotentadultprogenitorcellseffectivelyreducegraftvshostdiseasewhilepreservinggraftvsleukemiaactivity
AT leezhenghong humanmultipotentadultprogenitorcellseffectivelyreducegraftvshostdiseasewhilepreservinggraftvsleukemiaactivity
AT wilsondavid humanmultipotentadultprogenitorcellseffectivelyreducegraftvshostdiseasewhilepreservinggraftvsleukemiaactivity
AT lazarushillardm humanmultipotentadultprogenitorcellseffectivelyreducegraftvshostdiseasewhilepreservinggraftvsleukemiaactivity
AT deansrobert humanmultipotentadultprogenitorcellseffectivelyreducegraftvshostdiseasewhilepreservinggraftvsleukemiaactivity
AT vanthofwouter humanmultipotentadultprogenitorcellseffectivelyreducegraftvshostdiseasewhilepreservinggraftvsleukemiaactivity
AT ktenayiouli humanmultipotentadultprogenitorcellseffectivelyreducegraftvshostdiseasewhilepreservinggraftvsleukemiaactivity
AT cookekennethr humanmultipotentadultprogenitorcellseffectivelyreducegraftvshostdiseasewhilepreservinggraftvsleukemiaactivity