Presentation is loading. Please wait.

Presentation is loading. Please wait.

Volume 3, Issue 2, Pages (August 2008)

Similar presentations


Presentation on theme: "Volume 3, Issue 2, Pages (August 2008)"— Presentation transcript:

1 Volume 3, Issue 2, Pages 221-227 (August 2008)
Nf2/Merlin Regulates Hematopoietic Stem Cell Behavior by Altering Microenvironmental Architecture  Jonas Larsson, Masanobu Ohishi, Brian Garrison, Marie Aspling, Viktor Janzen, Gregor B. Adams, Marcello Curto, Andrea I. McClatchey, Ernestina Schipani, David T. Scadden  Cell Stem Cell  Volume 3, Issue 2, Pages (August 2008) DOI: /j.stem Copyright © 2008 Elsevier Inc. Terms and Conditions

2 Figure 1 Rapid Egress of HSCs and Progenitor Cells to the Circulation and Increase in HSC Number over Time following Nf2 Deletion (A) Reduced BM cellularity in Nf2−/− mice 4 weeks after knockout induction (n = 5, p < 0.01). (B) Reduced numbers of primitive Lin−, Sca1+, c-kit+ cells in BM of Nf2-deficient mice (n = 5, p < 0.05). (C) Nf2−/− mice show enhanced numbers of progenitors in peripheral blood as determined by colony-forming cell (CFC) assays (n = 5, p < 0.01). (D) Representative FACS plot showing the content of Lin−, Sca1+, c-kit+ cells in peripheral blood. (E) In vivo competitive repopulation assay to measure the content of stem cells in blood through competition with normal BM cells. Results show increased long-term (16 weeks) repopulating ability of peripheral blood cells from Nf2-deficient mice (n = 4, p = 0.02). (F) Increased BM cellularity in aged (7 months after polyIC induction) mutant mice (n = 4, p < 0.001). (G) Increased numbers of primitive hematopoietic cells determined by FACS (Lin−, Sca1+, c-kit+) in aged mutant mice (n = 4, p < 0.05). (H) Competitive repopulating unit (CRU) assays show an increased frequency of HSCs in the BM of aged mutant mice. Values are mean ± SEM. Cell Stem Cell 2008 3, DOI: ( /j.stem ) Copyright © 2008 Elsevier Inc. Terms and Conditions

3 Figure 2 The HSC Changes Are Non-Cell-Autonomous and Specifically Dependent on Alterations in the Bone Marrow Microenvironment (A and B) BM chimeras were generated by either transplanting Nf2 floxed BM cells into wild-type recipients or wild-type BM into Nf2 floxed recipients. Following hematopoietic reconstitution 8 weeks posttransplantation, the BM chimeras were treated with polyIC to induce gene deletion in Nf2 floxed cells. (A) Nf2-deficient progenitors that have reconstituted a wild-type environment do not show increased egress into the bloodstream as measured by CFC assays 4 weeks after polyIC induction (n = 4, p = NS). (B) Wild-type cells in an Nf2-deficient environment display a similar dramatic retention defect as observed in the Nf2−/− mice (n = 4, p < 0.01). (C and D) Fluorescently labeled wild-type BM cells were intravenously injected to lethally irradiated Nf2−/− or Nf2 flox/flox control mice. Sixteen hours later, mice were sacrificed, and the numbers of homed cells in BM and spleen were determined by FACS. (C) Reduced homing of c-kit + cells to the marrow space in Nf2-deficient mice (n = 3, p = 0.01). (D) No alterations in the homing to spleen could be observed (n = 3, p = NS). (E and F) Lethally irradiated wild-type mice were reconstituted with Nf2 floxed BM cells and after 8 weeks treated with polyIC. Seven months later the BM was analyzed. (E) No alteration in BM cellularity in aged transplant recipients (n = 4, p = NS). (F) Normal numbers of Nf2-deficient HSCs (Lin−, Sca1+, c-kit+, CD34− cells) in a wild-type environment (n = 4, p = NS). Values are mean ± SEM. Cell Stem Cell 2008 3, DOI: ( /j.stem ) Copyright © 2008 Elsevier Inc. Terms and Conditions

4 Figure 3 The Nf2-Deficient Mice Display Increased Vascular Structures and High VEGF Production in the Bone Marrow, which May Account for the Retention and Homing Defect Mice were sacrificed 4 weeks after polyIC induction, and the tibial bones were recovered and processed for histology. (A) Histology of tibias show a dramatic expansion of nonhematopoietic bone marrow stromal cells distal to the cartilage zone in the Nf2 mutants (between arrowheads). (B) Immunohistochemical staining with the endothelial marker CD31 reveals a large increase in the number of vessels in the marrow space of mutant tibias. (C and D) In situ hybridization (C) and immunohistochemistry (D) show markedly increased expression levels of VEGF in tibia sections from the knockout mice, particularly in areas of stromal cell expansion (between arrowheads). Cell Stem Cell 2008 3, DOI: ( /j.stem ) Copyright © 2008 Elsevier Inc. Terms and Conditions

5 Figure 4 Aged Nf2-Deficient Mice Have Increased Trabecular Bone and Osteoblasts Mice were sacrificed 6–7 months after polyIC induction for BM analysis. (A) Tibial sections from the mutant mice showed a marked increase in trabecular bone structures (arrowheads). (B and C) Bone histomorphometry shows a marked increase in the relative bone volume (n = 3, p < 0.05) and in the number of osteoblasts (n = 3, p < 0.05) in the Nf2 mutant animals. (D) Cell cycle distribution of Lin−, Sca1+, c-kit+ cells measured by Hoechst and Pyronin Y staining. Representative FACS plots and graph showing accumulated data (n = 3, p = NS). (E) Apoptosis assay using Annexin V staining on Lin−, Sca1+, c-kit+ cells. The lower right quadrant represents early apoptotic cells that are positive for Annexin V but negative for 7-AAD, which labels dead cells. Representative FACS plots and graph showing accumulated data (n = 3, p = NS). Values are mean ± SEM. Cell Stem Cell 2008 3, DOI: ( /j.stem ) Copyright © 2008 Elsevier Inc. Terms and Conditions


Download ppt "Volume 3, Issue 2, Pages (August 2008)"

Similar presentations


Ads by Google