Your browser doesn't support javascript.
loading
Osteopontin is An Important Regulative Component of the Fetal Bone Marrow Hematopoietic Stem Cell Niche.
Cao, Huimin; Cao, Benjamin; Heazlewood, Chad K; Domingues, Melanie; Sun, Xuan; Debele, Emmanuel; McGregor, Narelle E; Sims, Natalie A; Heazlewood, Shen Y; Nilsson, Susan K.
Afiliação
  • Cao H; Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC 3800, Australia.
  • Cao B; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia.
  • Heazlewood CK; Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC 3800, Australia.
  • Domingues M; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia.
  • Sun X; Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC 3800, Australia.
  • Debele E; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia.
  • McGregor NE; Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC 3800, Australia.
  • Sims NA; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia.
  • Heazlewood SY; Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC 3800, Australia.
  • Nilsson SK; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia.
Cells ; 8(9)2019 08 27.
Article em En | MEDLINE | ID: mdl-31461896
ABSTRACT
Osteopontin (OPN) is an important component in both bone and blood regulation, functioning as a bridge between the two. Previously, thrombin-cleaved osteopontin (trOPN), the dominant form of OPN in adult bone marrow (BM), was demonstrated to be a critical negative regulator of adult hematopoietic stem cells (HSC) via interactions with α4ß1 and α9ß1 integrins. We now demonstrate OPN is also required for fetal hematopoiesis in maintaining the HSC and progenitor pool in fetal BM. Specifically, we showed that trOPN is highly expressed in fetal BM and its receptors, α4ß1 and α9ß1 integrins, are both highly expressed and endogenously activated on fetal BM HSC and progenitors. Notably, the endogenous activation of integrins expressed by HSC was attributed to high concentrations of three divalent metal cations, Ca2+, Mg2+ and Mn2+, which were highly prevalent in developing fetal BM. In contrast, minimal levels of OPN were detected in fetal liver, and α4ß1 and α9ß1 integrins expressed by fetal liver HSC were not in the activated state, thereby permitting the massive expansion of HSC and progenitors required during early fetal hematopoiesis. Consistent with these results, no differences in the number or composition of hematopoietic cells in the liver of fetal OPN-/- mice were detected, but significant increases in the hematopoietic progenitor pool in fetal BM as well as an increase in the BM HSC pool following birth and into adulthood were observed. Together, the data demonstrates OPN is a necessary negative regulator of fetal and neonatal BM progenitors and HSC, and it exhibits preserved regulatory roles during early development, adulthood and ageing.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Medula Óssea / Células-Tronco Hematopoéticas / Osteopontina / Nicho de Células-Tronco / Feto Limite: Animals Idioma: En Revista: Cells Ano de publicação: 2019 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Medula Óssea / Células-Tronco Hematopoéticas / Osteopontina / Nicho de Células-Tronco / Feto Limite: Animals Idioma: En Revista: Cells Ano de publicação: 2019 Tipo de documento: Article