Volume 8, Issue 5, Pages (May 2005)

Slides:



Advertisements
Similar presentations
Journal of Nutritional Biochemistry
Advertisements

CXC chemokine ligand 12a enhances chondrocyte proliferation and maturation during endochondral bone formation  G.-W. Kim, M.-S. Han, H.-R. Park, E.-J.
Heparanase Stimulates Chondrogenesis and Is Up-Regulated in Human Ectopic Cartilage  Julianne Huegel, Motomi Enomoto-Iwamoto, Federica Sgariglia, Eiki.
MicroRNA-30b is a multifunctional regulator of aortic valve interstitial cells  Mi Zhang, MD, Xiaohong Liu, MD, Xiwu Zhang, MD, Zhigang Song, MD, Lin Han,
CXC chemokine ligand 12a enhances chondrocyte proliferation and maturation during endochondral bone formation  G.-W. Kim, M.-S. Han, H.-R. Park, E.-J.
Volume 17, Issue 4, Pages (October 2009)
Hypertrophic differentiation during chondrogenic differentiation of progenitor cells is stimulated by BMP-2 but suppressed by BMP-7  M.M.J. Caron, P.J.
M. Wang, H. Jin, D. Tang, S. Huang, M.J. Zuscik, D. Chen 
Visualization of Cartilage Formation: Insight into Cellular Properties of Skeletal Progenitors and Chondrodysplasia Syndromes  Maria Barna, Lee Niswander 
M. Wang, H. Jin, D. Tang, S. Huang, M.J. Zuscik, D. Chen 
Muscle Contraction Is Necessary to Maintain Joint Progenitor Cell Fate
Nan Gao, Peter White, Klaus H. Kaestner  Developmental Cell 
HIF-1α as a Regulator of BMP2-Induced Chondrogenic Differentiation, Osteogenic Differentiation, and Endochondral Ossification in Stem Cells Cell Physiol.
Volume 6, Issue 4, Pages (April 2016)
Volume 4, Issue 4, Pages (April 2009)
Volume 118, Issue 4, Pages (August 2004)
Exaggerated inflammatory environment decreases BMP-2/ACS-induced ectopic bone mass in a rat model: implications for clinical use of BMP-2  R.-L. Huang,
CaMKII inhibition in human primary and pluripotent stem cell-derived chondrocytes modulates effects of TGFβ and BMP through SMAD signaling  B. Saitta,
Volume 19, Issue 2, Pages (August 2010)
Volume 6, Issue 3, Pages (March 2004)
Volume 14, Issue 4, Pages (April 2008)
Makoto Takeo, Christopher S. Hale, Mayumi Ito 
Volume 29, Issue 2, Pages (April 2014)
Volume 53, Issue 1, Pages (January 2014)
Volume 43, Issue 5, Pages e3 (December 2017)
Radhika Atit, Ronald A. Conlon, Lee Niswander  Developmental Cell 
Volume 29, Issue 3, Pages (May 2014)
Volume 105, Issue 5, Pages (June 2001)
Gufa Lin, Ying Chen, Jonathan M.W. Slack  Developmental Cell 
Lydia Koziel, Melanie Kunath, Olivia G Kelly, Andrea Vortkamp 
All Mouse Ventral Spinal Cord Patterning by Hedgehog Is Gli Dependent and Involves an Activator Function of Gli3  C.Brian Bai, Daniel Stephen, Alexandra.
M.M.-G. Sun, F. Beier  Osteoarthritis and Cartilage 
Matthew P. Harris, Sean M. Hasso, Mark W.J. Ferguson, John F. Fallon 
Hoxd13 Contribution to the Evolution of Vertebrate Appendages
Volume 8, Issue 5, Pages (May 2005)
Kaoru Sugimoto, Yuling Jiao, Elliot M. Meyerowitz  Developmental Cell 
Volume 26, Issue 5, Pages (September 2013)
SoxE Factors Function Equivalently during Neural Crest and Inner Ear Development and Their Activity Is Regulated by SUMOylation  Kimberly M. Taylor, Carole.
Volume 9, Issue 3, Pages (September 2017)
Volume 13, Issue 6, Pages (December 2007)
Jianjun Sun, Wu-Min Deng  Developmental Cell 
Wnt/β-Catenin Signaling in Mesenchymal Progenitors Controls Osteoblast and Chondrocyte Differentiation during Vertebrate Skeletogenesis  Timothy F. Day,
Volume 17, Issue 1, Pages (July 2009)
Kimberly M. McDermott, Bob Y. Liu, Thea D. Tlsty, Gregory J. Pazour 
Volume 27, Issue 5, Pages (December 2013)
Volume 3, Issue 3, Pages (September 2002)
Volume 24, Issue 2, Pages (February 2016)
Tbx5 and Tbx4 Are Not Sufficient to Determine Limb-Specific Morphologies but Have Common Roles in Initiating Limb Outgrowth  Carolina Minguillon, Jo Del.
Early Lineage Segregation between Epiblast and Primitive Endoderm in Mouse Blastocysts through the Grb2-MAPK Pathway  Claire Chazaud, Yojiro Yamanaka,
Volume 23, Issue 1, Pages (July 2012)
Promotion Effects of miR-375 on the Osteogenic Differentiation of Human Adipose- Derived Mesenchymal Stem Cells  Si Chen, Yunfei Zheng, Shan Zhang, Lingfei.
Thomas Andl, Seshamma T. Reddy, Trivikram Gaddapara, Sarah E. Millar 
Volume 18, Issue 4, Pages (April 2010)
Lydia Koziel, Melanie Kunath, Olivia G Kelly, Andrea Vortkamp 
Volume 14, Issue 5, Pages (May 2008)
Volume 11, Issue 1, Pages (July 2018)
Volume 108, Issue 1, Pages (January 2002)
Volume 22, Issue 5, Pages (May 2012)
The Chick Transcriptional Repressor Nkx3
Volume 12, Issue 1, Pages (January 2007)
Jonathan M. Lehman, Essam Laag, Edward J. Michaud, Bradley K. Yoder 
In Vivo Human Somitogenesis Guides Somite Development from hPSCs
EBF2 Regulates Osteoblast-Dependent Differentiation of Osteoclasts
Volume 10, Issue 5, Pages (May 2006)
Volume 15, Issue 10, Pages (October 2007)
Visualization of Cartilage Formation: Insight into Cellular Properties of Skeletal Progenitors and Chondrodysplasia Syndromes  Maria Barna, Lee Niswander 
Volume 8, Issue 6, Pages (June 2017)
Wnt-14 Plays a Pivotal Role in Inducing Synovial Joint Formation in the Developing Appendicular Skeleton  Christine Hartmann, Clifford J Tabin  Cell 
Volume 22, Issue 5, Pages (May 2012)
Zhen Zhang, Jamie M. Verheyden, John A. Hassell, Xin Sun 
Presentation transcript:

Volume 8, Issue 5, Pages 727-738 (May 2005) Canonical Wnt/β-Catenin Signaling Prevents Osteoblasts from Differentiating into Chondrocytes  Theo P. Hill, Daniela Später, Makoto M. Taketo, Walter Birchmeier, Christine Hartmann  Developmental Cell  Volume 8, Issue 5, Pages 727-738 (May 2005) DOI: 10.1016/j.devcel.2005.02.013 Copyright © 2005 Elsevier Inc. Terms and Conditions

Figure 1 Endogenous β-Catenin Expression in Skeletal Tissue and Prx1-Cre Activity (A–A″) Immunohistochemical staining showing higher levels of β-catenin protein in cells of the (A) periarticular region of the joint, in the (A′ and star in A″) nucleus of flattened proliferating and prehypertrophic chondrocytes, and in the (A″) periosteum at E13.5. (B and C) Alkaline phosphatase staining of ZAP-reporter/Prx1-Cre embryos at (B) E9.5 and (C) E11. (D) Coronal section through the head at the level of the white line shown in (C). (E and E′) In situ hybridization showing β-catenin expression in a (E) wild-type hindlimb and in a (E′) β-catΔPrx/− hindlimb at E11.5. Arrows indicate the expression in cells that are probably migrating myoblasts. Developmental Cell 2005 8, 727-738DOI: (10.1016/j.devcel.2005.02.013) Copyright © 2005 Elsevier Inc. Terms and Conditions

Figure 2 Skeletal Alterations in β-Catenin Mutant Animals at Late Stages of Development (A–C) Skeletal preparations of wild-type (WT) and β-catΔPrx/− (ΔPrx/−) mutant P0 embryos showing the whole skeleton, the hindlimbs, and the head skeleton. (D–G) Wild-type and mutant femurs at PO. (D) Hematoxylin/eosin staining. (E) Van Kossa staining of bone collar in the wild-type (arrow), which is absent (star) in the mutant. (F) TRAP staining, demonstrating that multinucleated osteoclasts (inlet) are present in the mutant. High-magnification (20×) hematoxylin/eosin-stained sections, showing that eosin-positive red blood cells and hematopoietic cells are present (E). Developmental Cell 2005 8, 727-738DOI: (10.1016/j.devcel.2005.02.013) Copyright © 2005 Elsevier Inc. Terms and Conditions

Figure 3 Absence of Mature Osteoblast Markers in β-Catenin Mutant Limbs and Mineralization Defects of Femora Cultures (A–I) Nonradioactive section in situ hybridizations on wild-type and β-catΔPrx/− (ΔPrx/−) mutant femora at (A–C) E14.5, (D–G) E16.5, and (H and I) E18.5, showing expression of (A) Ihh, (B and E) Runx2, (C and F) Osx, (D) Ptc-1, (G) Tcf-1, (H) Col1a1, and (I) Osc. Note that the folds within the skeletal elements are due to sectioning and do not reflect increased expression. (J) Alizarin red-stained mineralization assay of cultures from β-catenin heterozygous and mutant ΔPrx/− femora at days (d) 15 and 21 of differentiation, without (w/o) addition of exogenous factors and in the presence of exogenous recombinant Shh and BMP2 protein; this panel shows decreased mineralization potential of mutant cultures. (K) Relative expression of Osteocalcin (Osc), Alkaline phosphatase (ALP), and bone sialoprotein (Bsp) in heterozygous (blue) and mutant cultures (red) at day 6, without the addition of factors (solid bars) and with the addition of Shh and BMP2 (stippled bars). Error bars represent the standard deviation of the mean of the results (n = 3). Developmental Cell 2005 8, 727-738DOI: (10.1016/j.devcel.2005.02.013) Copyright © 2005 Elsevier Inc. Terms and Conditions

Figure 4 Differentiation of Perichondrial/Periosteal Cells into Chondrocytes in β-Catenin Mutants (A–C) Sections through wild-type and β-catΔPrx/− (ΔPrx/−) mutant femora at E18.5 stained with van Kossa/alcian blue at low magnification. High magnification showing alcian blue-positive cells within the mutant perichondrium/periosteum (arrow in [A′]), which express Col2a1 (arrow in [B]) and Sox9 (arrow in [C]). (D–F) (D) Ihh and (E) Col10a1 are only expressed in cells along the wedge-shaped mutant perichondrium, which can be also seen in the (F) hematoxylin/eosin-stained section. Col2a1, Sox9, Ihh, and Col10a1 expression are visualized by in situ hybridization. Developmental Cell 2005 8, 727-738DOI: (10.1016/j.devcel.2005.02.013) Copyright © 2005 Elsevier Inc. Terms and Conditions

Figure 5 Loss of β-Catenin in Skull Mesenchyme and Calvarial Osteoblast Progenitors Leads to Ectopic Cartilage Formation (A) Coronal sections through the skull of wild-type and β-catΔPrx/− (ΔPrx/−) mutants at P0 stained with van Kossa alcian blue (blue), showing the presence of mineralized bone (arrow) in the wild-type, which is absent in the mutant and replaced by ectopic cartilage. (B–D) In situ hybridizations on coronal sections showing a dorsal expansion of chondrogenic markers (B) Sox 9 and (C) Col2 in the mutants (see arrows) at E13.5 and (D) loss of Osx expression at E16.5. (E) Whole-mount in situ hybridizations on E16.5 wild-type and β-catΔPrx/− mutant heads, showing lack of Osc expression (arrow) in the mutant skull. (F) Primary osteoblast cultures from β-catenin fl/fl E16.5 calvariae treated with AdGfp (left panel) and AdCre (right panel) showing different morphology at (Fa) day 15 and formation of alcian blue-positive nodules at (Fb) day 21. In situ hybridization for (Fc) Col2a1, (Fd) Sox9, and (Fe) Col1a1. (G) Relative expression levels of chondrogenic markers Sox9 and Col2a1 and of the marker Col1a1 at days 5, 10, and 15 of culture, analyzed by real-time PCR. Error bars represent the standard deviation of the mean of the results (n = 2). Developmental Cell 2005 8, 727-738DOI: (10.1016/j.devcel.2005.02.013) Copyright © 2005 Elsevier Inc. Terms and Conditions

Figure 6 Alterations of Prechondrogenic Marker Expression in β-Catenin Gain- and Loss-of-Function Mutant Limb Buds (A–J) Transverse sections through the forelimb of (A–D) wild-type, (E–G) β-catenin gain-of-function (Δex3Prx/+) mutants, and (H–J) β-catenin loss-of-function (ΔPrx/−) mutants at E11.0. (A) Hematoxylin/eosin staining visualizing the condensed mesenchyme in the core of a wild-type limb. (B) In situ hybridization showing Sox9 expression in the condensed core. (C) Immunohistochemical staining showing higher levels of β-catenin protein in the mesenchyme underneath the ectoderm, and lower levels in the central core. (D) In situ hybridization showing Runx2 expression in the scapula region. (E) No condensation is visible in β-catΔex3Prx/+ mutant limb buds. (F) Sox9 expression is downregulated in β-catΔex3Prx/+ mutant limbs. (G) Ectopic stabilization of β-catenin protein in β-catΔex3Prx/+ limbs. Mosaic activity of the Prx1-Cre line is reflected by the presence of a few clusters of cells, which do not show ectopic stabilization of β-catenin. (H) No condensation is visible in β-catΔPrx/− mutants, and (I) Sox9 expression is expanded in the distal half of the limb bud. (J) Expanded expression of Runx2 in β-catΔPrx/− mutant limb bud. Developmental Cell 2005 8, 727-738DOI: (10.1016/j.devcel.2005.02.013) Copyright © 2005 Elsevier Inc. Terms and Conditions

Figure 7 Alteration of β-Catenin Levels in Micromass Cultures Affects Sox9 and Runx2 Expression Levels (A) Western blot showing reduction of β-catenin levels in AdCre-treated cultures from 24 hr onward. (B) Time course of real-time PCR analysis showing upregulation of Sox9 starting at 24 hr in AdCre-treated β-catenin fl/fl cultures (n = 2). (C) Alcian blue-stained micromass cultures at days 4 and 6 from β-catenin fl/fl limbs infected with AdenoGfp or AdenoCre virus with and without (w/o) addition of rhBmp4, showing accelerated cartilage nodule formation upon deletion of β-catenin. (D) Western blot showing the appearance of the N-terminal-deleted β-catenin in ex3fl/ex3fl AdCre-treated cultures at 10 hr. (E) Real-time PCR analysis showing downregulation of Sox9 from 10 hr onward in AdCre-treated β-catenin ex3fl/ex3fl cultures (n = 3). (F) Real-time PCR analysis showing relative expression levels of Runx2 at 14 hr going up in AdCre-treated β-catenin fl/fl cultures and going down in AdCre-treated β-catenin ex3fl/ex3fl cultures. Error bars represent the standard deviation of the mean of the results (n = 3). (G) Semiquantitative RT-PCR analysis of Lef-1 expression, showing an upregulation of Lef-1 in AdCre-treated β-catenin ex3fl/ex3fl at 14 hr of culturing. Developmental Cell 2005 8, 727-738DOI: (10.1016/j.devcel.2005.02.013) Copyright © 2005 Elsevier Inc. Terms and Conditions