Your browser doesn't support javascript.
loading
The Src homology 2 protein Shb promotes cell cycle progression in murine hematopoietic stem cells by regulation of focal adhesion kinase activity.
Gustafsson, Karin; Heffner, Garrett; Wenzel, Pamela L; Curran, Matthew; Grawé, Jan; McKinney-Freeman, Shannon L; Daley, George Q; Welsh, Michael.
Afiliação
  • Gustafsson K; Department of Medical Cell Biology, Uppsala University, Uppsala 751 23, Sweden.
  • Heffner G; HHMI, Children's Hospital Boston, Harvard Medical School, Boston, 02115 MA, USA.
  • Wenzel PL; HHMI, Children's Hospital Boston, Harvard Medical School, Boston, 02115 MA, USA.
  • Curran M; HHMI, Children's Hospital Boston, Harvard Medical School, Boston, 02115 MA, USA.
  • Grawé J; Department of Genetics and Pathology, Uppsala University, Uppsala 75185, Sweden.
  • McKinney-Freeman SL; Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
  • Daley GQ; HHMI, Children's Hospital Boston, Harvard Medical School, Boston, 02115 MA, USA.
  • Welsh M; Department of Medical Cell Biology, Uppsala University, Uppsala 751 23, Sweden. Electronic address: michael.welsh@mcb.uu.se.
Exp Cell Res ; 319(12): 1852-1864, 2013 Jul 15.
Article em En | MEDLINE | ID: mdl-23528453
ABSTRACT
The widely expressed adaptor protein Shb has previously been reported to contribute to T cell function due to its association with the T cell receptor and furthermore, several of Shb's known interaction partners are established regulators of blood cell development and function. In addition, Shb deficient embryonic stem cells displayed reduced blood cell colony formation upon differentiation in vitro. The aim of the current study was therefore to explore hematopoietic stem and progenitor cell function in the Shb knockout mouse. Shb deficient bone marrow contained reduced relative numbers of long-term hematopoietic stem cells (LT-HSCs) that exhibited lower proliferation rates. Despite this, Shb knockout LT-HSCs responded promptly by entering the cell cycle in response to genotoxic stress by 5-fluorouracil treatment. In competitive LT-HSC transplantations, Shb null cells initially engrafted as well as the wild-type cells but provided less myeloid expansion over time. Moreover, Shb knockout bone marrow cells exhibited elevated basal activities of focal adhesion kinase/Rac1/p21-activated kinase signaling and reduced responsiveness to Stem Cell Factor stimulation. Consequently, treatment with a focal adhesion kinase inhibitor increased Shb knockout LT-HSC proliferation. The altered signaling characteristics thus provide a plausible mechanistic explanation for the changes in LT-HSC proliferation since these signaling intermediates have all been shown to participate in LT-HSC cell cycle control. In summary, the loss of Shb dependent signaling in bone marrow cells, resulting in elevated focal adhesion kinase activity and reduced proliferative responses in LT-HSCs under steady state hematopoiesis, confers a disadvantage to the maintenance of LT-HSCs over time.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Células-Tronco Hematopoéticas / Ciclo Celular / Proteínas Proto-Oncogênicas / Proteína-Tirosina Quinases de Adesão Focal Limite: Animals Idioma: En Ano de publicação: 2013 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Células-Tronco Hematopoéticas / Ciclo Celular / Proteínas Proto-Oncogênicas / Proteína-Tirosina Quinases de Adesão Focal Limite: Animals Idioma: En Ano de publicação: 2013 Tipo de documento: Article