Your browser doesn't support javascript.
loading
Human endothelial colony-forming cells serve as trophic mediators for mesenchymal stem cell engraftment via paracrine signaling.
Lin, Ruei-Zeng; Moreno-Luna, Rafael; Li, Dan; Jaminet, Shou-Ching; Greene, Arin K; Melero-Martin, Juan M.
Afiliação
  • Lin RZ; Department of Cardiac Surgery, Boston Children's Hospital, and Department of Surgery, Harvard Medical School, Boston, MA 02115;
  • Moreno-Luna R; Department of Cardiac Surgery, Boston Children's Hospital, and Department of Surgery, Harvard Medical School, Boston, MA 02115;
  • Li D; Center for Vascular Biology, Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02215;
  • Jaminet SC; Center for Vascular Biology, Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02215;
  • Greene AK; Department of Plastic and Oral Surgery, Boston Children's Hospital, Boston, MA 02115; and.
  • Melero-Martin JM; Department of Cardiac Surgery, Boston Children's Hospital, and Department of Surgery, Harvard Medical School, Boston, MA 02115;Harvard Stem Cell Institute, Cambridge, MA 02138 juan.meleromartin@childrens.harvard.edu.
Proc Natl Acad Sci U S A ; 111(28): 10137-42, 2014 Jul 15.
Article em En | MEDLINE | ID: mdl-24982174
ABSTRACT
Endothelial colony-forming cells (ECFCs) are endothelial precursors that circulate in peripheral blood. Studies have demonstrated that human ECFCs have robust vasculogenic properties. However, whether ECFCs can exert trophic functions in support of specific stem cells in vivo remains largely unknown. Here, we sought to determine whether human ECFCs can function as paracrine mediators before the establishment of blood perfusion. We used two xenograft models of human mesenchymal stem cell (MSC) transplantation and studied how the presence of ECFCs modulates MSC engraftment and regenerative capacity in vivo. Human MSCs were isolated from white adipose tissue and bone marrow aspirates and were s.c. implanted into immunodeficient mice in the presence or absence of cord blood-derived ECFCs. MSC engraftment was regulated by ECFC-derived paracrine factors via platelet-derived growth factor BB (PDGF-BB)/platelet-derived growth factor receptor (PDGFR)-ß signaling. Cotransplanting ECFCs significantly enhanced MSC engraftment by reducing early apoptosis and preserving stemness-related properties of PDGFR-ß(+) MSCs, including the ability to repopulate secondary grafts. MSC engraftment was negligible in the absence of ECFCs and completely impaired in the presence of Tyrphostin AG1296, an inhibitor of PDGFR kinase. Additionally, transplanted MSCs displayed fate-restricted potential in vivo, with adipose tissue-derived and bone marrow-derived MSCs contributing exclusive differentiation along adipogenic and osteogenic lineages, respectively. This work demonstrates that blood-derived ECFCs can serve as paracrine mediators and regulate the regenerative potential of MSCs via PDGF-BB/PDGFR-ß signaling. Our data suggest the systematic use of ECFCs as a means to improve MSC transplantation.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Comunicação Parácrina / Receptor beta de Fator de Crescimento Derivado de Plaquetas / Proteínas Proto-Oncogênicas c-sis / Células Endoteliais / Células-Tronco Mesenquimais Idioma: En Ano de publicação: 2014 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Comunicação Parácrina / Receptor beta de Fator de Crescimento Derivado de Plaquetas / Proteínas Proto-Oncogênicas c-sis / Células Endoteliais / Células-Tronco Mesenquimais Idioma: En Ano de publicação: 2014 Tipo de documento: Article