51
|
Daoud G, Kempf H, Kumar D, Kozhemyakina E, Holowacz T, Kim DW, Ionescu A, Lassar AB. BMP-mediated induction of GATA4/5/6 blocks somitic responsiveness to SHH. Development 2014; 141:3978-87. [PMID: 25294942 DOI: 10.1242/dev.111906] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The relative timing of SHH and BMP signals controls whether presomitic mesoderm (PSM) cells will adopt either a chondrogenic or lateral plate mesoderm fate. Here we document that SHH-mediated induction of Nkx3.2 maintains the competence of somitic cells to initiate chondrogenesis in response to subsequent BMP signals by repressing BMP-dependent induction of GATA genes. Conversely, administration of BMP signals to PSM or forced expression of GATA family members in chick PSM explants blocks induction of hedgehog-dependent gene expression. We demonstrate that GATA factors can interact with Gli factors and can recruit the transcriptional co-factor FOG1 (ZFPM1) to the regulatory region of the mouse Gli1 gene, repressing the induction of Gli1 by SHH by binding to both GATA and Gli binding sites. Knockdown of FOG1 reverses the ability of GATA factors to repress Gli1 expression. Our findings uncover a novel role for GATA transcription factors as repressors of hedgehog signaling, and document that NKX3.2 maintains the ability of sclerotomal cells to express SHH transcriptional targets in the presence of BMP signals by repressing the induction of Gata4/5/6.
Collapse
Affiliation(s)
- Georges Daoud
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Bldg C-Room 303, 240 Longwood Ave, Boston, MA 02115, USA
| | - Hervé Kempf
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Bldg C-Room 303, 240 Longwood Ave, Boston, MA 02115, USA
| | - Deepak Kumar
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Bldg C-Room 303, 240 Longwood Ave, Boston, MA 02115, USA
| | - Elena Kozhemyakina
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Bldg C-Room 303, 240 Longwood Ave, Boston, MA 02115, USA
| | - Tamara Holowacz
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Bldg C-Room 303, 240 Longwood Ave, Boston, MA 02115, USA
| | - Dae-Won Kim
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Bldg C-Room 303, 240 Longwood Ave, Boston, MA 02115, USA
| | - Andreia Ionescu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Bldg C-Room 303, 240 Longwood Ave, Boston, MA 02115, USA
| | - Andrew B Lassar
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Bldg C-Room 303, 240 Longwood Ave, Boston, MA 02115, USA
| |
Collapse
|
52
|
Chatterjee S, Sivakamasundari V, Yap SP, Kraus P, Kumar V, Xing X, Lim SL, Sng J, Prabhakar S, Lufkin T. In vivo genome-wide analysis of multiple tissues identifies gene regulatory networks, novel functions and downstream regulatory genes for Bapx1 and its co-regulation with Sox9 in the mammalian vertebral column. BMC Genomics 2014; 15:1072. [PMID: 25480362 PMCID: PMC4302147 DOI: 10.1186/1471-2164-15-1072] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 11/27/2014] [Indexed: 12/30/2022] Open
Abstract
Background Vertebrate organogenesis is a highly complex process involving sequential cascades of transcription factor activation or repression. Interestingly a single developmental control gene can occasionally be essential for the morphogenesis and differentiation of tissues and organs arising from vastly disparate embryological lineages. Results Here we elucidated the role of the mammalian homeobox gene Bapx1 during the embryogenesis of five distinct organs at E12.5 - vertebral column, spleen, gut, forelimb and hindlimb - using expression profiling of sorted wildtype and mutant cells combined with genome wide binding site analysis. Furthermore we analyzed the development of the vertebral column at the molecular level by combining transcriptional profiling and genome wide binding data for Bapx1 with similarly generated data sets for Sox9 to assemble a detailed gene regulatory network revealing genes previously not reported to be controlled by either of these two transcription factors. Conclusions The gene regulatory network appears to control cell fate decisions and morphogenesis in the vertebral column along with the prevention of premature chondrocyte differentiation thus providing a detailed molecular view of vertebral column development. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-1072) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Thomas Lufkin
- Department of Biology, Clarkson University, 8 Clarkson Avenue, Potsdam, NY 13699, USA.
| |
Collapse
|
53
|
Zhou ZQ, Ota S, Deng C, Akiyama H, Hurlin PJ. Mutant activated FGFR3 impairs endochondral bone growth by preventing SOX9 downregulation in differentiating chondrocytes. Hum Mol Genet 2014; 24:1764-73. [PMID: 25432534 DOI: 10.1093/hmg/ddu594] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Fibroblast growth factor receptor 3 (FGFR3) plays a critical role in the control of endochondral ossification, and bone growth and mutations that cause hyperactivation of FGFR3 are responsible for a collection of developmental disorders that feature poor endochondral bone growth. FGFR3 is expressed in proliferating chondrocytes of the cartilaginous growth plate but also in chondrocytes that have exited the cell cycle and entered the prehypertrophic phase of chondrocyte differentiation. Achondroplasia disorders feature defects in chondrocyte proliferation and differentiation, and the defects in differentiation have generally been considered to be a secondary manifestation of altered proliferation. By initiating a mutant activated knockin allele of FGFR3 (FGFR3K650E) that causes Thanatophoric Dysplasia Type II (TDII) specifically in prehypertrophic chondrocytes, we show that mutant FGFR3 induces a differentiation block at this stage independent of any changes in proliferation. The differentiation block coincided with persistent expression of SOX9, the master regulator of chondrogenesis, and reducing SOX9 dosage allowed chondrocyte differentiation to proceed and significantly improved endochondral bone growth in TDII. These findings suggest that a proliferation-independent and SOX9-dependent differentiation block is a key driving mechanism responsible for poor endochondral bone growth in achondroplasia disorders caused by mutations in FGFR3.
Collapse
Affiliation(s)
- Zi-Qiang Zhou
- Shriners Hospitals for Children, 3101 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Sara Ota
- Shriners Hospitals for Children, 3101 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Chuxia Deng
- Genetics of Development and Disease Branch, National Institute of Diabetes, Digestive and Kidney Diseases, US National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Haruhiko Akiyama
- Department of Orthopedics, Gifu University, Gifu 501-1194, Japan
| | - Peter J Hurlin
- Shriners Hospitals for Children, 3101 SW Sam Jackson Park Road, Portland, OR 97239, USA, Department of Cell and Developmental Biology and Knight Cancer Institute, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| |
Collapse
|
54
|
Zhao J, Li S, Trilok S, Tanaka M, Jokubaitis-Jameson V, Wang B, Niwa H, Nakayama N. Small molecule-directed specification of sclerotome-like chondroprogenitors and induction of a somitic chondrogenesis program from embryonic stem cells. Development 2014; 141:3848-58. [PMID: 25294938 PMCID: PMC7055718 DOI: 10.1242/dev.105981] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 08/16/2014] [Indexed: 02/03/2023]
Abstract
Pluripotent embryonic stem cells (ESCs) generate rostral paraxial mesoderm-like progeny in 5-6 days of differentiation induced by Wnt3a and Noggin (Nog). We report that canonical Wnt signaling introduced either by forced expression of activated β-catenin, or the small-molecule inhibitor of Gsk3, CHIR99021, satisfied the need for Wnt3a signaling, and that the small-molecule inhibitor of BMP type I receptors, LDN193189, was able to replace Nog. Mesodermal progeny generated using such small molecules were chondrogenic in vitro, and expressed trunk paraxial mesoderm markers such as Tcf15 and Meox1, and somite markers such as Uncx, but failed to express sclerotome markers such as Pax1. Induction of the osteochondrogenically committed sclerotome from somite requires sonic hedgehog and Nog. Consistently, Pax1 and Bapx1 expression was induced when the isolated paraxial mesodermal progeny were treated with SAG1 (a hedgehog receptor agonist) and LDN193189, then Sox9 expression was induced, leading to cartilaginous nodules and particles in the presence of BMP, indicative of chondrogenesis via sclerotome specification. By contrast, treatment with TGFβ also supported chondrogenesis and stimulated Sox9 expression, but failed to induce the expression of Pax1 and Bapx1. On ectopic transplantation to immunocompromised mice, the cartilage particles developed under either condition became similarly mineralized and formed pieces of bone with marrow. Thus, the use of small molecules led to the effective generation from ESCs of paraxial mesodermal progeny, and to their further differentiation in vitro through sclerotome specification into growth plate-like chondrocytes, a mechanism resembling in vivo somitic chondrogenesis that is not recapitulated with TGFβ.
Collapse
Affiliation(s)
- Jiangang Zhao
- Institute of Molecular Medicine, The University of Texas Health Science Center Medical School at Houston, Houston, TX 77030, USA
| | - Songhui Li
- Australian Stem Cell Centre, Monash University, Clayton, Victoria 3800, Australia
| | - Suprita Trilok
- Institute of Molecular Medicine, The University of Texas Health Science Center Medical School at Houston, Houston, TX 77030, USA
| | - Makoto Tanaka
- Australian Stem Cell Centre, Monash University, Clayton, Victoria 3800, Australia
| | | | - Bei Wang
- Australian Stem Cell Centre, Monash University, Clayton, Victoria 3800, Australia
| | - Hitoshi Niwa
- RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Naoki Nakayama
- Institute of Molecular Medicine, The University of Texas Health Science Center Medical School at Houston, Houston, TX 77030, USA Australian Stem Cell Centre, Monash University, Clayton, Victoria 3800, Australia Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
55
|
Tam WL, O DF, Hiramatsu K, Tsumaki N, Luyten FP, Roberts SJ. Sox9 reprogrammed dermal fibroblasts undergo hypertrophic differentiation in vitro and trigger endochondral ossification in vivo. Cell Reprogram 2014; 16:29-39. [PMID: 24459991 DOI: 10.1089/cell.2013.0060] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Strategies for bone regeneration are undergoing a paradigm shift, moving away from the replication of end-stage bone tissue and instead aiming to recapture the initial events of fracture repair. Although this is known to resemble endochondral bone formation, chondrogenic cell types with favorable proliferative and hypertrophic differentiation properties are lacking. Recent advances in cellular reprogramming have allowed the creation of alternative cell populations with specific properties through the forced expression of transcription factors. Herein, we investigated the in vitro hypertrophic differentiation and in vivo tissue formation capacity of induced chondrogenic cells (iChon cells) obtained through direct reprogramming. In vitro hypertrophic differentiation was detected in iChon cells that contained a doxycycline-inducible expression system for Klf4, cMyc, and Sox9. Furthermore, endochondral bone formation was detected after implantation in nude mice. The bone tissue was derived entirely from host origin, whereas cartilage tissue contained cells from both host and donor. The results obtained highlight the promise of cellular reprogramming for the creation of functional skeletal cells that can be used for novel bone healing strategies.
Collapse
Affiliation(s)
- Wai Long Tam
- 1 Laboratory for Developmental and Stem Cell Biology (DSB) , Skeletal Biology and Engineering Research Center (SBE), KU Leuven, 3000, Leuven, Belgium
| | | | | | | | | | | |
Collapse
|
56
|
Abstract
Transcription factor, Nkx3.2, is a member of the NK family of developmental genes and is expressed during embryogenesis in a variety of mammalian model organisms, including chicken and mouse. It was first identified in Drosophila as the Bagpipe (bap) gene, where it has been demonstrated to be essential during formation of the midgut musculature. However, mammalian homolog Nkx3.2 has been shown to play a significant role in axial and limb skeletogenesis; in particular, the human skeletal disease, spondylo-megaepiphyseal-metaphyseal dysplasia (SMMD), is associated with mutations of the Nkx3.2 gene. In this review, we highlight the role of Nkx3.2 during musculoskeletal development, with an emphasis on the factor's role in determining chondrogenic cell fate and its subsequent role in endochondral ossification and chondrocyte survival.
Collapse
|
57
|
Cartilage tissue engineering: molecular control of chondrocyte differentiation for proper cartilage matrix reconstruction. Biochim Biophys Acta Gen Subj 2014; 1840:2414-40. [PMID: 24608030 DOI: 10.1016/j.bbagen.2014.02.030] [Citation(s) in RCA: 177] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 02/06/2014] [Accepted: 02/26/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND Articular cartilage defects are a veritable therapeutic problem because therapeutic options are very scarce. Due to the poor self-regeneration capacity of cartilage, minor cartilage defects often lead to osteoarthritis. Several surgical strategies have been developed to repair damaged cartilage. Autologous chondrocyte implantation (ACI) gives encouraging results, but this cell-based therapy involves a step of chondrocyte expansion in a monolayer, which results in the loss in the differentiated phenotype. Thus, despite improvement in the quality of life for patients, reconstructed cartilage is in fact fibrocartilage. Successful ACI, according to the particular physiology of chondrocytes in vitro, requires active and phenotypically stabilized chondrocytes. SCOPE OF REVIEW This review describes the unique physiology of cartilage, with the factors involved in its formation, stabilization and degradation. Then, we focus on some of the most recent advances in cell therapy and tissue engineering that open up interesting perspectives for maintaining or obtaining the chondrogenic character of cells in order to treat cartilage lesions. MAJOR CONCLUSIONS Current research involves the use of chondrocytes or progenitor stem cells, associated with "smart" biomaterials and growth factors. Other influential factors, such as cell sources, oxygen pressure and mechanical strain are considered, as are recent developments in gene therapy to control the chondrocyte differentiation/dedifferentiation process. GENERAL SIGNIFICANCE This review provides new information on the mechanisms regulating the state of differentiation of chondrocytes and the chondrogenesis of mesenchymal stem cells that will lead to the development of new restorative cell therapy approaches in humans. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
|
58
|
|
59
|
Xing L, Chen D, Boyce BF. Mice Deficient in NF-κB p50 and p52 or RANK Have Defective Growth Plate Formation and Post-natal Dwarfism. Bone Res 2013; 1:336-45. [PMID: 26273511 DOI: 10.4248/br201304004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 09/11/2013] [Indexed: 11/10/2022] Open
Abstract
NF-κBp50/p52 double knockout (dKO) and RANK KO mice have no osteoclasts and develop severe osteopetrosis associated with dwarfism. In contrast, Op/Op mice, which form few osteoclasts, and Src KO mice, which have osteoclasts with defective resorptive function, are osteopetrotic, but they are not dwarfed. Here, we compared the morphologic features of long bones from p50/p52 dKO, RANK KO, Op/Op and Src KO mice to attempt to explain the differences in their long bone lengths. We found that growth plates in p50/p52 dKO and RANK KO mice are significantly thicker than those in WT mice due to a 2-3-fold increase in the hypertrophic chondrocyte zone associated with normal a proliferative chondrocyte zone. This growth plate abnormality disappears when animals become older, but their dwarfism persists. Op/Op or Src KO mice have relatively normal growth plate morphology. In-situ hybridization study of long bones from p50/p52 dKO mice showed marked thickening of the growth plate region containing type 10 collagen-expressing chondrocytes. Treatment of micro-mass chondrocyte cultures with RANKL did not affect expression levels of type 2 collagen and Sox9, markers for proliferative chondrocytes, but RANKL reduced the number of type 10 collagen-expressing hypertrophic chondrocytes. Thus, RANK/NF-κB signaling plays a regulatory role in post-natal endochondral ossification that maintains hypertrophic conversion and prevents dwarfism in normal mice.
Collapse
Affiliation(s)
- Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center , Rochester, NY 14642, USA
| | - Di Chen
- Department of Biochemistry, Rush University Medical Center , Chicago, IL 60612 USA
| | - Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center , Rochester, NY 14642, USA
| |
Collapse
|
60
|
Schmid R, Bosserhoff AK. Redundancy in regulation of chondrogenesis in MIA/CD-RAP-deficient mice. Mech Dev 2013; 131:24-34. [PMID: 24269712 DOI: 10.1016/j.mod.2013.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 10/26/2013] [Accepted: 11/11/2013] [Indexed: 10/26/2022]
Abstract
Recent in vitro analysis of MIA/CD-RAP-deficient (MIA(-/-)) mesenchymal stem cells revealed altered chondrogenic differentiation, characterised by enhanced proliferation and delayed differentiation. However, adult MIA(-/-) mice develop normally and show only ultrastructural defects of the cartilage but no major abnormalities. We therefore focused, in this study, on chondrogenesis in vivo in MIA(-/-) mouse embryos to reveal potential molecular changes during embryogenesis and possible redundant mechanisms, which explain the almost normal phenotype despite MIA/CD-RAP loss. In situ hybridisation analysis revealed larger expression areas of Col2a1 and Sox9 positive, proliferating chondrocytes at day 15.5 and 16.5 of embryogenesis in MIA(-/-) mice. The initially diminished zone of Col10a1-expressing hypertrophic chondrocytes at day 15.5 was compensated at day 16.5 in MIA(-/-) embryos. Supported by in vitro studies using mesenchymal stem cells, we discovered that chondrogenesis in MIA(-/-) mice is modified by enhanced Sox9, Sox6 and AP-2α expression. Finally, we identified reduced AP1 and CRE activity, analysed by reporter gene- and electrophoretic mobility shift assays, important for redundancy mechanism which rescued delayed hypertrophic differentiation and allows normal development of MIA(-/-) mice. In summary, as observed in other knockout models of molecules important for cartilage development and differentiation, viability and functional integrity is reached by remarkable molecular redundancy in MIA/CD-RAP knockout mice.
Collapse
Affiliation(s)
- Rainer Schmid
- University of Regensburg Medical School, Institute of Pathology, D-93053 Regensburg, Germany
| | - Anja-Katrin Bosserhoff
- University of Regensburg Medical School, Institute of Pathology, D-93053 Regensburg, Germany.
| |
Collapse
|
61
|
Pax1 acts as a negative regulator of chondrocyte maturation. Exp Cell Res 2013; 319:3128-39. [PMID: 24080012 DOI: 10.1016/j.yexcr.2013.09.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 09/02/2013] [Accepted: 09/18/2013] [Indexed: 12/27/2022]
Abstract
Paired box gene 1 (Pax1) indirectly promotes the early stages of chondrogenic differentiation through induction and transactivation of Nk3 homeobox 2 (Nkx3.2), a transcriptional repressor. Later in chondrogenic differentiation, Nkx3.2 blocks chondrocyte hypertrophy by repressing Runt-related transcription factor 2 (Runx2). Here we report the inhibitory action of Pax1 on chondrocyte maturation, independently of Nkx3.2. Upon cartilage formation, Pax1 expression in the ventral sclerotome was gradually decreased except for the perichondrial region of the vertebral bodies and the intervertebral region, both of which express SRY-box containing gene 9 (Sox9). Forced expression of Pax1 in the chick forelimb resulted in the formation of shortened skeletal elements with a significant reduction of proteoglycans (PGs) accumulation in cartilage as well as a lack of the cortical bone formation and vascular invasion into the primary ossification center. Pax1-misexpressing chondrocytes exhibited aberrant cell morphology with a marked downregulation of Aggrecan (Agc1). Pax1-misexpressing cultured chondrocytes failed to accumulate cartilaginous PGs and became fibroblastic, in association with downregulation of the expression of Sox9, Nkx3.2, Indian hedgehog (Ihh), type II collagen (Col2a1), Chondromodulin-1 (Chm1), and Agc1. Accumulation of cartilaginous PGs in chondrocytes was also reduced by forced expression of Pax1 and Sox9. Thus, chondrocyte maturation driven by Sox9 is antagonized by Pax1 that is downregulated during chondrogenic differentiation.
Collapse
|
62
|
Caron MMJ, Emans PJ, Cremers A, Surtel DAM, Coolsen MME, van Rhijn LW, Welting TJM. Hypertrophic differentiation during chondrogenic differentiation of progenitor cells is stimulated by BMP-2 but suppressed by BMP-7. Osteoarthritis Cartilage 2013; 21:604-13. [PMID: 23353668 DOI: 10.1016/j.joca.2013.01.009] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 12/06/2012] [Accepted: 01/12/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Bone morphogenic protein (BMP)-2 and BMP-7 are clinically approved and their recombinant proteins are used for bone tissue regenerative purposes and widely evaluated for cartilage regeneration. Previous comparison of the in vitro chondrogenic characteristics of BMP-2 vs BMP-7 did not address hypertrophic differentiation and characterizing their chondrogenic properties with a focus in on chondrocyte hypertrophy was topic of investigation in this study. DESIGN Equimolar concentrations of BMP-2 or BMP-7 were added to chondrogenic differentiating ATDC5, human bone marrow stem cells or rabbit periosteal explants. Expression of Col2a1, Sox9, Acan, Col10a1, Runx2, ALP, Mmp13, Mef2c and Bapx1/Nkx3.2 was determined by reverse transcription-quantitative PCR (RT-qPCR) and immunoblotting. Glycosaminoglycan content, cell proliferation capacity and ALP activity were analysed by colourimetric analyses. Expression of Bapx1/Nkx3.2 and Sox9 was targeted by transfection of target specific siRNA duplexes. RESULTS BMP-2 dose-dependently increased chondrocyte hypertrophy during chondrogenic differentiation of progenitor cells, whereas BMP-7 acted hypertrophy-suppressive and chondro-promotive. Both BMPs did not influence cell proliferation, but they did increase total glycosaminoglycan content. In a candidate approach Bapx1/Nkx3.2 was found to be involved in the BMP-7 mediated suppression of chondrocyte hypertrophy in ATDC5 cells. CONCLUSIONS BMP-2 and BMP-7 display opposing actions on the chondrogenic outcome of differentiating progenitor cells: BMP-2 acts a specific inducer of chondrocyte hypertrophy, while BMP-7 appears to increase or maintain chondrogenic potential and prevent chondrocyte hypertrophy. Our results pave the way for an application-dependent differential use of BMP-2 or BMP-7.
Collapse
Affiliation(s)
- M M J Caron
- Department of Orthopaedic Surgery, Caphri School for Public Health and Primary Care, Maastricht University Medical Center, PO Box 5800, 6202 AZ, Maastricht, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
63
|
Zhang W, Chen J, Zhang S, Ouyang HW. Inhibitory function of parathyroid hormone-related protein on chondrocyte hypertrophy: the implication for articular cartilage repair. Arthritis Res Ther 2012; 14:221. [PMID: 22971952 PMCID: PMC3580589 DOI: 10.1186/ar4025] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cartilage repair tissue is usually accompanied by chondrocyte hypertrophy and osseous overgrowths, and a role for parathyroid hormone-related protein (PTHrP) in inhibiting chondrocytes from hypertrophic differentiation during the process of endochondral ossification has been demonstrated. However, application of PTHrP in cartilage repair has not been extensively considered. This review systemically summarizes for the first time the inhibitory function of PTHrP on chondrocyte hypertrophy in articular cartilage and during the process of endochondral ossification, as well as the process of mesenchymal stem cell chondrogenic differentiation. Based on the literature review, the strategy of using PTHrP for articular cartilage repair is suggested, which is instructive for clinical treatment of cartilage injuries as well as osteoarthritis.
Collapse
|
64
|
Cairns DM, Liu R, Sen M, Canner JP, Schindeler A, Little DG, Zeng L. Interplay of Nkx3.2, Sox9 and Pax3 regulates chondrogenic differentiation of muscle progenitor cells. PLoS One 2012; 7:e39642. [PMID: 22768305 PMCID: PMC3388093 DOI: 10.1371/journal.pone.0039642] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 05/26/2012] [Indexed: 01/04/2023] Open
Abstract
Muscle satellite cells make up a stem cell population that is capable of differentiating into myocytes and contributing to muscle regeneration upon injury. In this work we investigate the mechanism by which these muscle progenitor cells adopt an alternative cell fate, the cartilage fate. We show that chick muscle satellite cells that normally would undergo myogenesis can be converted to express cartilage matrix proteins in vitro when cultured in chondrogenic medium containing TGFß3 or BMP2. In the meantime, the myogenic program is repressed, suggesting that muscle satellite cells have undergone chondrogenic differentiation. Furthermore, ectopic expression of the myogenic factor Pax3 prevents chondrogenesis in these cells, while chondrogenic factors Nkx3.2 and Sox9 act downstream of TGFß or BMP2 to promote this cell fate transition. We found that Nkx3.2 and Sox9 repress the activity of the Pax3 promoter and that Nkx3.2 acts as a transcriptional repressor in this process. Importantly, a reverse function mutant of Nkx3.2 blocks the ability of Sox9 to both inhibit myogenesis and induce chondrogenesis, suggesting that Nkx3.2 is required for Sox9 to promote chondrogenic differentiation in satellite cells. Finally, we found that in an in vivo mouse model of fracture healing where muscle progenitor cells were lineage-traced, Nkx3.2 and Sox9 are significantly upregulated while Pax3 is significantly downregulated in the muscle progenitor cells that give rise to chondrocytes during fracture repair. Thus our in vitro and in vivo analyses suggest that the balance of Pax3, Nkx3.2 and Sox9 may act as a molecular switch during the chondrogenic differentiation of muscle progenitor cells, which may be important for fracture healing.
Collapse
Affiliation(s)
- Dana M. Cairns
- Program in Cellular, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts, United States of America
| | - Renjing Liu
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia
- Faculty of Medicine, University of Sydney, Sydney, Australia
| | - Manpreet Sen
- Building Diversity in Biomedical Research Program (BDBS), Tufts University School of Medicine, Massachusetts, United States of America
- Department of Anatomy and Cellular Biology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - James P. Canner
- Program in Cellular, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts, United States of America
| | - Aaron Schindeler
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia
- Faculty of Medicine, University of Sydney, Sydney, Australia
| | - David G. Little
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Westmead, Australia
- Faculty of Medicine, University of Sydney, Sydney, Australia
| | - Li Zeng
- Program in Cellular, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts, United States of America
- Building Diversity in Biomedical Research Program (BDBS), Tufts University School of Medicine, Massachusetts, United States of America
- Department of Anatomy and Cellular Biology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- Department of Orthopaedic Surgery, Tufts Medical Center, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
65
|
Human chondrogenic paraxial mesoderm, directed specification and prospective isolation from pluripotent stem cells. Sci Rep 2012; 2:455. [PMID: 22701159 PMCID: PMC3374161 DOI: 10.1038/srep00455] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 05/22/2012] [Indexed: 12/22/2022] Open
Abstract
Directed specification and prospective isolation of chondrogenic paraxial mesoderm progeny from human pluripotent stem (PS) cells have not yet been achieved. Here we report the successful generation of KDR−PDGFRα+ progeny expressing paraxial mesoderm genes and the mesendoderm reporter MIXL1-GFP in a chemically defined medium containing the canonical WNT signaling activator, BMP-inhibitor, and the Nodal/Activin/TGFβ signaling controller. Isolated (GFP+)KDR−PDGFRα+ mesoderm cells were sensitive to sequential addition of the three chondrogenic factors PDGF, TGFβ and BMP. Under these conditions, the cells showed robust chondrogenic activity in micromass culture, and generated a hyaline-like translucent cartilage particle in serum-free medium. In contrast, both STRO1+ mesenchymal stem/stromal cells from adult human marrow and mesenchymal cells spontaneously arising from hPS cells showed a relatively weaker chondrogenic response in vitro, and formed more of the fibrotic cartilage particles. Thus, hPS cell-derived KDR−PDGFRα+ paraxial mesoderm-like cells have potential in engineered cartilage formation and cartilage repair.
Collapse
|
66
|
Indian Hedgehog signalling triggers Nkx3.2 protein degradation during chondrocyte maturation. Biochem J 2012; 443:789-98. [PMID: 22507129 DOI: 10.1042/bj20112062] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The Ihh (Indian Hedgehog) pathway plays an essential role in facilitating chondrocyte hypertrophy and bone formation during skeletal development. Nkx3.2 (NK3 homeobox 2) is initially induced in chondrocyte precursor cells, maintained in early-stage chondrocytes and down-regulated in terminal-stage chondrocytes. Consistent with these expression patterns, Nkx3.2 has been shown to enhance chondrocyte differentiation and cell survival, while inhibiting chondrocyte hypertrophy and apoptosis. Thus, in the present study, we investigated whether Nkx3.2, an early-stage chondrogenic factor, can be regulated by Ihh, a key regulator for chondrocyte hypertrophy. We show that Ihh signalling can induce proteasomal degradation of Nkx3.2. In addition, we found that Ihh can suppress levels of Lrp (low-density-lipoprotein-receptor-related protein) (Wnt co-receptor) and Sfrp (secreted frizzled-related protein) (Wnt antagonist) expression, which, in turn, may selectively enhance Lrp-independent non-canonical Wnt pathways in chondrocytes. In agreement with these findings, Ihh-induced Nkx3.2 degradation requires Wnt5a, which is capable of triggering Nkx3.2 degradation. Finally, we found that Nkx3.2 protein levels in chondrocytes are remarkably elevated in mice defective in Ihh signalling by deletion of either Ihh or smoothened. Thus these results suggest that Ihh/Wnt5a signalling may play a role in negative regulation of Nkx3.2 for appropriate progression of chondrocyte hypertrophy during chondrogenesis.
Collapse
|
67
|
Kawato Y, Hirao M, Ebina K, Tamai N, Shi K, Hashimoto J, Yoshikawa H, Myoui A. Nkx3.2-induced suppression of Runx2 is a crucial mediator of hypoxia-dependent maintenance of chondrocyte phenotypes. Biochem Biophys Res Commun 2011; 416:205-10. [PMID: 22093831 DOI: 10.1016/j.bbrc.2011.11.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 11/04/2011] [Indexed: 11/17/2022]
Abstract
Hypoxia is a key factor in the maintenance of chondrocyte identity. However, crucial chondrogenic transcription factors in the Sox families are not activated in this phenomenon, indicating that other pathways are involved. Nkx3.2 is a well-known chondrogenic transcription factor induced by Sonic hedgehog (Shh); it suppresses a key osteogenic transcriptional factor, Runt-related transcription factor 2 (Runx2), to maintain the chondrogenic phenotype in mesenchymal lineages. The purpose of this study was to examine the function of Nkx3.2 in hypoxia-dependent maintenance of chondrocyte identity. C3H10T1/2 pluripotent mesenchymal cells were cultured with rh-BMP2 (300 ng/ml) to induce chondrogenesis under normoxic (20% O(2)) or hypoxic (5% O(2)) conditions. Immunohistological detection of Nkx3.2 in a micromass cell culture system revealed that hypoxia promoted expression of the Nkx3.2 protein. Real-time RT-PCR analysis revealed that hypoxia promoted Nkx3.2 mRNA expression and suppressed Runx2 mRNA expression; however, Sox9 mRNA expression was not altered by oxygen conditions, as previously described. Over-expression of exogenous Nkx3.2 promoted glycosaminoglycan (GAG) production and inhibited Runx2 mRNA expression and, based on a dual luciferase assay, Runx2 promoter activity. Interestingly, downregulation of Nkx3.2 using RNAi abolished hypoxia-dependent GAG production and restored Runx2 mRNA expression and promoter activity. These results demonstrated that Nkx3.2-dependent suppression of Runx2 was a crucial factor in hypoxia-dependent maintenance of chondrocyte identity.
Collapse
Affiliation(s)
- Yoshitaka Kawato
- Department of Orthopaedics, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | |
Collapse
|
68
|
McCanless JD, Jennings LK, Cole JA, Bumgardner JD, Haggard WO. In vitro differentiation and biocompatibility of mesenchymal stem cells on a novel platelet releasate-containing injectable composite. J Biomed Mater Res A 2011; 100:220-9. [DOI: 10.1002/jbm.a.33256] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 09/02/2011] [Accepted: 09/06/2011] [Indexed: 12/30/2022]
|
69
|
Bren-Mattison Y, Hausburg M, Olwin BB. Growth of limb muscle is dependent on skeletal-derived Indian hedgehog. Dev Biol 2011; 356:486-95. [PMID: 21683695 PMCID: PMC3322465 DOI: 10.1016/j.ydbio.2011.06.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Revised: 05/31/2011] [Accepted: 06/01/2011] [Indexed: 10/18/2022]
Abstract
During embryogenesis, muscle and bone develop in close temporal and spatial proximity. We show that Indian Hedgehog, a bone-derived signaling molecule, participates in growth of skeletal muscle. In Ihh(-/-) embryos, skeletal muscle development appears abnormal at embryonic day 14.5 and at later ages through embryonic day 20.5, dramatic losses of hindlimb muscle occur. To further examine the role of Ihh in myogenesis, we manipulated Ihh expression in the developing chick hindlimb. Reduction of Ihh in chicken embryo hindlimbs reduced skeletal muscle mass similar to that seen in Ihh(-/-) mouse embryos. The reduction in muscle mass appears to be a direct effect of Ihh since ectopic expression of Ihh by RCAS retroviral infection of chicken embryo hindlimbs restores muscle mass. These effects are independent of bone length, and occur when Shh is not expressed, suggesting Ihh acts directly on fetal myoblasts to regulate secondary myogenesis. Loss of muscle mass in Ihh null mouse embryos is accompanied by a dramatic increase in myoblast apoptosis by a loss of p21 protein. Our data suggest that Ihh promotes fetal myoblast survival during their differentiation into secondary myofibers by maintaining p21 protein levels.
Collapse
Affiliation(s)
- Yvette Bren-Mattison
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado 80309
| | - Melissa Hausburg
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado 80309
| | - Bradley B. Olwin
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado 80309
| |
Collapse
|
70
|
Seriwatanachai D, Densmore MJ, Sato T, Correa D, Neff L, Baron R, Lanske B. Deletion of Zfp521 rescues the growth plate phenotype in a mouse model of Jansen metaphyseal chondrodysplasia. FASEB J 2011; 25:3057-67. [PMID: 21642473 DOI: 10.1096/fj.11-183277] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Jansen metaphyseal chondrodysplasia (JMC) is caused by a constitutively activating mutation of the parathyroid hormone (PTH)/PTH-related protein (PTHrP) receptor (PTHR1) and is characterized by widening of the metaphyses, reduction of long bone length, and short stature. A transgenic mouse expressing this mutation under the collagen α1(II) promoter has been generated to investigate the mechanisms responsible for this chondrodysplasia. We recently identified zinc finger protein 521 (Zfp521) as a downstream target gene of PTHrP signaling. Interestingly, loss of Zfp521 from chondrocytes leads to reduced cell proliferation and increased differentiation in the growth plate. Thus, we hypothesized that specifically ablating Zfp521 from Jansen chondrocytes could sufficiently rescue the chondrodysplasia phenotype. Our results show that Zfp521 expression is up-regulated in Jansen mouse growth plate chondrocytes and that PTHR1 is required for Zfp521 expression. Its ablation from Jansen chondrocytes restored normal cell differentiation, thus initiating chondrocyte apoptosis at the chondro-osseous junction, leading to partial rescue of endochondral bone formation shown by proper bone length. This study provides the first genetic evidence that Zfp521 is required downstream of PTHR1 signaling to act on chondrocyte proliferation, differentiation, and cell death.
Collapse
Affiliation(s)
- Dutmanee Seriwatanachai
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
71
|
Exogenous signal-independent nuclear IkappaB kinase activation triggered by Nkx3.2 enables constitutive nuclear degradation of IkappaB-alpha in chondrocytes. Mol Cell Biol 2011; 31:2802-16. [PMID: 21606193 DOI: 10.1128/mcb.00253-10] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
NF-κB is a multifunctional transcription factor involved in diverse biological processes. It has been well documented that NF-κB can be activated in response to various stimuli. While signal-inducible NF-κB activation mechanisms have been extensively characterized, exogenous signal-independent intrinsic NF-κB activation processes remain poorly understood. Here we show that IκB kinase β (IKKβ) can be intrinsically activated in the nucleus by a homeobox protein termed Nkx3.2 in the absence of exogenous IKK-activating signals. We found that ubiquitin chain-dependent, but persistent, interactions between Nkx3.2 and NEMO (also known as IKKγ) can give rise to constitutive IKKβ activation in the nucleus. Once the Nkx3.2-NEMO-IKKβ complex is formed in the nucleus, IKKβ-induced Nkx3.2 phosphorylation at Ser148 and Ser168 allows βTrCP to be engaged to cause IκB-α ubiquitination independent of IκB-α phosphorylation at Ser32 and Ser36. Taken together, our results provide a novel molecular explanation as to how an intracellular factor such as Nkx3.2 can accomplish persistent nuclear IKK activation to enable intrinsic and constitutive degradation of IκB in the nucleus in the absence of exogenous NF-κB-activating signals, which, in turn, plays a role in chondrocyte viability maintenance.
Collapse
|
72
|
Gawlitta D, Farrell E, Malda J, Creemers LB, Alblas J, Dhert WJA. Modulating endochondral ossification of multipotent stromal cells for bone regeneration. TISSUE ENGINEERING PART B-REVIEWS 2011; 16:385-95. [PMID: 20131956 DOI: 10.1089/ten.teb.2009.0712] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
For years it has been recognized that engineering of large bone constructs will be feasible only if the hurdle of vascularization is overcome. Attempts to engineer bone tissue have predominantly focused on intramembranous (direct) bone formation. A relatively new and most likely more physiological approach in this line is endochondral bone formation, comprising an intermediate cartilaginous stage. Cartilage in nature is an avascular tissue and its cells are equipped to survive the poor oxygenation and nutritional conditions inherent to implanted tissues. Subsequent terminal differentiation (hypertrophy) of the chondrocytes initiates the formation of a mineralized matrix that will then be converted into bone. Through this mechanism, our long bones grow and most fractures heal through the process of secondary fracture healing. The feasibility of the attractive concept of endochondral bone tissue engineering has already been shown. Most emphasis has gone to the multipotent stromal cells because of their great potential for expansion and differentiation and immunoprivileged nature. This review will focus on the promises and current status of this new field. Further, potent modulators of endochondral bone tissue engineering, including oxygen tension and mechanical stimuli, will be discussed.
Collapse
Affiliation(s)
- Debby Gawlitta
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | |
Collapse
|
73
|
Abstract
The middle ear is a composite organ formed from all three germ layers and the neural crest. It provides the link between the outside world and the inner ear, where sound is transduced and routed to the brain for processing. Extensive classical and modern studies have described the complex morphology and origin of the middle ear. Non-mammalian vertebrates have a single ossicle, the columella. Mammals have three functionally equivalent ossicles, designated the malleus, incus and stapes. In this review, I focus on the role of genes known to function in the middle ear. Genetic studies are beginning to unravel the induction and patterning of the multiple middle ear elements including the tympanum, skeletal elements, the air-filled cavity, and the insertion point into the inner ear oval window. Future studies that elucidate the integrated spatio-temporal signaling mechanisms required to pattern the middle ear organ system are needed. The longer-term translational benefits of understanding normal and abnormal ear development will have a direct impact on human health outcomes.
Collapse
|
74
|
Parathyroid hormone/parathyroid hormone-related protein receptor signaling is required for maintenance of the growth plate in postnatal life. Proc Natl Acad Sci U S A 2010; 108:191-6. [PMID: 21173257 DOI: 10.1073/pnas.1005011108] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Parathyroid hormone (PTH)-related protein (PTHrP), regulated by Indian hedgehog and acting through the PTH/PTHrP receptor (PPR), is crucial for normal cartilage development. These observations suggest a possible role of PPR signaling in the postnatal growth plate; however, the role of PPR signaling in postnatal chondrocytes is unknown. In this study, we have generated tamoxifen-inducible and cartilage-specific PPR KO mice to evaluate the physiological role of PPR signaling in postnatal chondrocytes. We found that inactivation of the PPR in chondrocytes postnatally leads to accelerated differentiation of chondrocytes, followed by disappearance of the growth plate. We also observed an increase of TUNEL-positive cells and activities of caspase-3 and caspase-9 in the growth plate, along with a decrease in phosphorylation of Bad at Ser155 in postnatal PPR KO mice. Administration of a low-phosphate diet, which prevents apoptosis of chondrocytes, prevented the disappearance of the growth plate. Taken together, these observations suggest that the major consequences of PPR activation are similar in both the fetal and postnatal growth plates. Moreover, chondrocyte apoptosis through the activation of a mitochondrial pathway may be involved in the process of premature disappearance of the growth plate by postnatal inactivation of the PPR in chondrocytes.
Collapse
|
75
|
Abstract
Wdr5, a bone morphogenetic protein 2 (BMP-2)-induced protein belonging to the family of the WD repeat proteins, is expressed in proliferating and hypertrophic chondrocytes of the growth plate and in osteoblasts. Although previous studies have provided insight into the mechanisms by which Wdr5 affects chondrocyte and osteoblast differentiation, whether Wdr5 is required in vivo for endochondral bone development has not been addressed. In this study, using an avian replication competent retrovirus (RCAS) system delivering Wdr5 short hairpin (sh) RNA to silence Wdr5 in the developing limb, we report that reduction of Wdr5 levels delays endochondral bone development and consequently results in shortening of the skeletal elements. Shortening of the skeletal elements was due to impaired chondrocyte maturation, evidenced by a significant reduction of Runx2, type X collagen, and osteopontin expression. A decrease in Runx2, type collagen I, and ostepontin expression in osteoblasts and a subsequent defect in mineralized bone was observed as well when Wdr5 levels were reduced. Most important, retroviral misexpression of Runx2 rescued the phenotype induced by Wdr5 shRNA. These findings suggest that during limb development, Wdr5 is required for endochondral bone formation and that Wdr5 influences this process, at least in part, by regulating Runx2 expression.
Collapse
Affiliation(s)
- Shimei Zhu
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | | | | | | |
Collapse
|
76
|
Abstract
Nuclear factor kappaB (NF-kappaB) is a set of multifunctional transcription factors that regulate expression of genes involved in numerous normal cellular activities. They also are activated in many inflammatory and neoplastic conditions in which their expression may be stimulated by proinflammatory cytokines. NF-kappaB, in turn, regulates the expression of cytokines and so can mediate autocrine self-amplifying cycles of cytokine release and NF-kappaB activation, leading to maintenance of inflammatory reactions beyond the initial stimulus, as seen in rheumatoid arthritis and asthma. Since discovery of the requirement of NF-kappaB for basal and cytokine-induced osteoclast formation in the mid-1990s, much has been learned about the role of NF-kappaB in bone. NF-kappaB has roles in skeletal development, endochondral ossification, osteoclast and osteoblast functions, and common bone diseases. NF-kappaB inhibitors have been developed, but none have made it to clinical trials for the treatment of common bone diseases. Here we review the roles for NF-kappaB in bone and in common bone diseases.
Collapse
Affiliation(s)
- Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA.
| | | | | |
Collapse
|
77
|
Hellemans J, Simon M, Dheedene A, Alanay Y, Mihci E, Rifai L, Sefiani A, van Bever Y, Meradji M, Superti-Furga A, Mortier G. Homozygous inactivating mutations in the NKX3-2 gene result in spondylo-megaepiphyseal-metaphyseal dysplasia. Am J Hum Genet 2009; 85:916-22. [PMID: 20004766 DOI: 10.1016/j.ajhg.2009.11.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Revised: 10/10/2009] [Accepted: 11/05/2009] [Indexed: 11/29/2022] Open
Abstract
Spondylo-megaepiphyseal-metaphyseal dysplasia (SMMD) is a rare skeletal dysplasia with only a few cases reported in the literature. Affected individuals have a disproportionate short stature with a short and stiff neck and trunk. The limbs appear relatively long and may show flexion contractures of the distal joints. The most remarkable radiographic features are the delayed and impaired ossification of the vertebral bodies as well as the presence of large epiphyseal ossification centers and wide growth plates in the long tubular bones. Numerous pseudoepiphyses of the short tubular bones in hands and feet are another remarkable feature of the disorder. Genome wide homozygosity mapping followed by a candidate gene approach resulted in the elucidation of the genetic cause in three new consanguineous families with SMMD. Each proband was homozygous for a different inactivating mutation in NKX3-2, a homeobox-containing gene located on chromosome 4p15.33. Striking similarities were found when comparing the vertebral ossification defects in SMMD patients with those observed in the Nkx3-2 null mice. Distinguishing features were the asplenia found in the mutant mice and the radiographic abnormalities in the limbs only observed in SMMD patients. The absence of the latter anomalies in the murine model may be due to the perinatal death of the affected animals. This study illustrates that NKX3-2 plays an important role in endochondral ossification of both the axial and appendicular skeleton in humans. In addition, it defines SMMD as yet another skeletal dysplasia with autosomal-recessive inheritance and a distinct phenotype.
Collapse
Affiliation(s)
- Jan Hellemans
- Center for Medical Genetics, Ghent University Hospital, B-9000 Ghent, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Bellon E, Luyten FP, Tylzanowski P. delta-EF1 is a negative regulator of Ihh in the developing growth plate. ACTA ACUST UNITED AC 2009; 187:685-99. [PMID: 19948490 PMCID: PMC2806579 DOI: 10.1083/jcb.200904034] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Indian hedgehog (Ihh) regulates proliferation and differentiation of chondrocytes in the growth plate. Although the biology of Ihh is currently well documented, its transcriptional regulation is poorly understood. delta-EF1 is a two-handed zinc finger/homeodomain transcriptional repressor. Targeted inactivation of mouse delta-EF1 leads to skeletal abnormalities including disorganized growth plates, shortening of long bones, and joint fusions, which are reminiscent of defects associated with deregulation of Ihh signaling. Here, we show that the absence of delta-EF1 results in delayed hypertrophic differentiation of chondrocytes and increased cell proliferation in the growth plate. Further, we demonstrate that delta-EF1 binds to the putative regulatory elements in intron 1 of Ihh in vitro and in vivo, resulting in down-regulation of Ihh expression. Finally, we show that delta-EF1 haploinsufficiency leads to a postnatal increase in trabecular bone mass associated with enhanced Ihh expression. In summary, we have identified delta-EF1 as an in vivo negative regulator of Ihh expression in the growth plate.
Collapse
Affiliation(s)
- Ellen Bellon
- Laboratory of Skeletal Development and Joint Disorders, Division of Rheumatology, Department of Musculoskeletal Sciences, University of Leuven, Leuven 3000, Belgium
| | | | | |
Collapse
|
79
|
Schibler L, Gibbs L, Benoist-Lasselin C, Decraene C, Martinovic J, Loget P, Delezoide AL, Gonzales M, Munnich A, Jais JP, Legeai-Mallet L. New insight on FGFR3-related chondrodysplasias molecular physiopathology revealed by human chondrocyte gene expression profiling. PLoS One 2009; 4:e7633. [PMID: 19898608 PMCID: PMC2764091 DOI: 10.1371/journal.pone.0007633] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Accepted: 10/03/2009] [Indexed: 11/18/2022] Open
Abstract
Endochondral ossification is the process by which the appendicular skeleton, facial bones, vertebrae and medial clavicles are formed and relies on the tight control of chondrocyte maturation. Fibroblast growth factor receptor (FGFR)3 plays a role in bone development and maintenance and belongs to a family of proteins which differ in their ligand affinities and tissue distribution. Activating mutations of the FGFR3 gene lead to craniosynostosis and multiple types of skeletal dysplasia with varying degrees of severity: thanatophoric dysplasia (TD), achondroplasia and hypochondroplasia. Despite progress in the characterization of FGFR3-mediated regulation of cartilage development, many aspects remain unclear. The aim and the novelty of our study was to examine whole gene expression differences occurring in primary human chondrocytes isolated from normal cartilage or pathological cartilage from TD-affected fetuses, using Affymetrix technology. The phenotype of the primary cells was confirmed by the high expression of chondrocytic markers. Altered expression of genes associated with many cellular processes was observed, including cell growth and proliferation, cell cycle, cell adhesion, cell motility, metabolic pathways, signal transduction, cell cycle process and cell signaling. Most of the cell cycle process genes were down-regulated and consisted of genes involved in cell cycle progression, DNA biosynthesis, spindle dynamics and cytokinesis. About eight percent of all modulated genes were found to impact extracellular matrix (ECM) structure and turnover, especially glycosaminoglycan (GAG) and proteoglycan biosynthesis and sulfation. Altogether, the gene expression analyses provide new insight into the consequences of FGFR3 mutations in cell cycle regulation, onset of pre-hypertrophic differentiation and concomitant metabolism changes. Moreover, impaired motility and ECM properties may also provide clues about growth plate disorganization. These results also suggest that many signaling pathways may be directly or indirectly altered by FGFR3 and confirm the crucial role of FGFR3 in the control of growth plate development.
Collapse
Affiliation(s)
- Laurent Schibler
- Unité U781, Institut National de la Santé et de la Recherche Médicale, Université Paris Descartes-Hôpital Necker, Paris, France
- Unité Mixte de Recherche 1313, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | - Linda Gibbs
- Unité U781, Institut National de la Santé et de la Recherche Médicale, Université Paris Descartes-Hôpital Necker, Paris, France
- 4Clinics, Waterloo, Belgique
| | - Catherine Benoist-Lasselin
- Unité U781, Institut National de la Santé et de la Recherche Médicale, Université Paris Descartes-Hôpital Necker, Paris, France
| | | | - Jelena Martinovic
- Service de Fœtopathologie, Hôpital Necker, Université Paris Descartes, Paris, France
| | - Philippe Loget
- Centre Pluridisciplinaire de Diagnostic Prénatal de Rennes, Hôpital de Rennes, Rennes, France
| | - Anne-Lise Delezoide
- Service de Biologie du développement, Hôpital Robert Debré, Université Paris Diderot, Paris, France
| | - Marie Gonzales
- Service de Génétique et d'Embryologie Médicales, Hôpital Armand Trousseau, Université Pierre et Marie Curie, Paris, France
| | - Arnold Munnich
- Unité U781, Institut National de la Santé et de la Recherche Médicale, Université Paris Descartes-Hôpital Necker, Paris, France
| | - Jean-Philippe Jais
- Service de Biostatistique et Informatique Médicale, Hôpital Necker, Université Paris Descartes, Paris, France
| | - Laurence Legeai-Mallet
- Unité U781, Institut National de la Santé et de la Recherche Médicale, Université Paris Descartes-Hôpital Necker, Paris, France
- * E-mail:
| |
Collapse
|
80
|
Hartmann C. Transcriptional networks controlling skeletal development. Curr Opin Genet Dev 2009; 19:437-43. [DOI: 10.1016/j.gde.2009.09.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Revised: 09/04/2009] [Accepted: 09/10/2009] [Indexed: 12/18/2022]
|
81
|
Parathyroid hormone-related peptide represses chondrocyte hypertrophy through a protein phosphatase 2A/histone deacetylase 4/MEF2 pathway. Mol Cell Biol 2009; 29:5751-62. [PMID: 19704004 DOI: 10.1128/mcb.00415-09] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The maturation of immature chondrocytes to hypertrophic chondrocytes is regulated by parathyroid hormone-related peptide (PTHrP). We demonstrate that PTHrP or forskolin administration can block induction of collagen X-luciferase by exogenous Runx2, MEF2, and Smad1 in transfected chondrocytes. We have found that PTHrP/forskolin administration represses the transcriptional activity of MEF2 and that forced expression of MEF2-VP16 can restore expression of the collagen X reporter in chondrocytes treated with these agents. PTHrP/forskolin induces dephosphorylation of histone deacetylase 4 (HDAC4) phospho-S246, which decreases interaction of HDAC4 with cytoplasmic 14-3-3 proteins and promotes nuclear translocation of HDAC4 and repression of MEF2 transcriptional activity. We have found that forskolin increases the activity of an HDAC4 phospho-S246 phosphatase and that forskolin-induced nuclear translocation of HDAC4 was reversed by the protein phosphatase 2A (PP2A) antagonist, okadaic acid. Finally, we demonstrate that knockdown of PP2A inhibits forskolin-induced nuclear translocation of HDAC4 and attenuates the ability of this signaling molecule to repress collagen X expression in chondrocytes, indicating that PP2A is critical for PTHrP-mediated regulation of chondrocyte hypertrophy.
Collapse
|
82
|
Yamashita S, Andoh M, Ueno-Kudoh H, Sato T, Miyaki S, Asahara H. Sox9 directly promotes Bapx1 gene expression to repress Runx2 in chondrocytes. Exp Cell Res 2009; 315:2231-40. [PMID: 19306868 PMCID: PMC2696577 DOI: 10.1016/j.yexcr.2009.03.008] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2009] [Revised: 03/13/2009] [Accepted: 03/14/2009] [Indexed: 12/31/2022]
Abstract
The transcription factor, Sry-related High Mobility Group (HMG) box containing gene 9 (Sox9), plays a critical role in cartilage development by initiating chondrogenesis and preventing the subsequent maturation process called chondrocyte hypertrophy. This suppression mechanism by Sox9 on late-stage chondrogenesis partially results from the inhibition of Runt-related transcription factor 2 (Runx2), the main activator of hypertrophic chondrocyte differentiation. However, the precise mechanism by which Sox9 regulates late chondrogenesis is poorly understood. In the present study, the transcriptional repressor vertebrate homolog of Drosophila bagpipe (Bapx1) was found to be a direct target of Sox9 for repression of Runx2 expression in chondrocytes. We identified a critical Sox9 responsive region in the Bapx1 promoter via a luciferase reporter assay. Analysis by chromatin immunoprecipitation and electrophoretic mobility shift assays indicated that Sox9 physically bound to this region of the Bapx1 promoter. Consistent with the notion that Bapx1 and Sox9 act as negative regulators of chondrocyte hypertrophy by regulating Runx2 expression, transient knockdown of Sox9 or Bapx1 expression by shRNA in chondrocytes increased Runx2 expression, as well as expression of the late chondrogenesis marker, Col10a1. Furthermore, while over-expression of Sox9 decreased Runx2 and Col10a1 expressions, simultaneous transient knockdown of Bapx1 diminished that Sox9 over-expressing effect. Our findings reveal that the molecular pathway modulated by Bapx1 links two major regulators in chondrogenesis, Sox9 and Runx2, to coordinate skeletal formation.
Collapse
Affiliation(s)
- Satoshi Yamashita
- Department of Systems BioMedicine, National Research Institute of Child Health and Development, Tokyo 157-8535, Japan
| | - Masataka Andoh
- New National IT Strategy Business Promotion Division, NEC Corporation, Tokyo 108-8001, Japan
| | - Hiroe Ueno-Kudoh
- Department of Systems BioMedicine, National Research Institute of Child Health and Development, Tokyo 157-8535, Japan
| | - Tempei Sato
- Department of Systems BioMedicine, National Research Institute of Child Health and Development, Tokyo 157-8535, Japan
- School of Biomedical Science, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Shigeru Miyaki
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Hiroshi Asahara
- Department of Systems BioMedicine, National Research Institute of Child Health and Development, Tokyo 157-8535, Japan
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
83
|
Gordon CT, Rodda FA, Farlie PG. The RCAS retroviral expression system in the study of skeletal development. Dev Dyn 2009; 238:797-811. [DOI: 10.1002/dvdy.21907] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
84
|
Abstract
Major advances in the molecular genetics, paleobiology, and the evolutionary developmental biology of vertebrate skeletogenesis have improved our understanding of the early evolution and development of the vertebrate skeleton. These studies have involved genetic analysis of model organisms, human genetics, comparative developmental studies of basal vertebrates and nonvertebrate chordates, and both cladistic and histological analyses of fossil vertebrates. Integration of these studies has led to renaissance in the area of skeletal development and evolution. Among the major findings that have emerged is the discovery of an unexpectedly deep origin of the gene network that regulates chondrogenesis. In this chapter, we discuss recent progress in each these areas and identify a number of questions that need to be addressed in order to fill key gaps in our knowledge of early skeletal evolution.
Collapse
|
85
|
Kim YJ, Kim HJ, Im GI. PTHrP promotes chondrogenesis and suppresses hypertrophy from both bone marrow-derived and adipose tissue-derived MSCs. Biochem Biophys Res Commun 2008; 373:104-8. [PMID: 18554504 DOI: 10.1016/j.bbrc.2008.05.183] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2008] [Accepted: 05/30/2008] [Indexed: 01/13/2023]
Abstract
During chondrogenesis from mesenchymal stem cells (MSCs), inadequate differentiation and hypertrophic differentiation are two important limitations. The purpose of this study was to test the hypothesis that chondrogenesis is enhanced and unwanted hypertrophic changes are suppressed by treating bone marrow-derived (BMMSCs) and adipose tissue-derived mesenchymal stem cells (ATMSCs) with parathyroid hormone-related peptide (PTHrP). To induce chondrogenesis, in vitro pellet cultures were carried out using 2.5x10(5) MSCs at passage 3 in chondrogenic medium containing 5 ng/ml of TGF-beta(2) for BMMSCs, and 5 ng/ml of TGF-beta(2) and 100 ng/ml of BMP-7 for ATMSCs. From the 14th day of culture, subsets of pellets were treated with PTHrP [0, 10, 100 ng/ml], and after two more weeks of in vitro culture, pellets were harvested for analysis. The addition of PTHrP dose-dependently increased DNA contents in both BMMSCs and ATMSCs. GAG contents also increased after PTHrP treatment. The gene expression of COL1A1 decreased by three-fourths, while the decrease was not evident in ATMSCs after PTHrP treatment. SOX-9 mRNA increased up to four fold in both BMMSCs and ATMSCs, and COL2A1 gene expression sharply increased to sevenfold in BMMSCs and to 4 fold in ATMSCs. COL10A1 gene expression decreased by a third in both cell types, and Runx-2 expression dropped sharply in both cell types after PTHrP treatment. Safranin-O and immunohistochemistry for type I, II, X collagen and Runx-2 generally paralleled qRT-PCR findings with minor variations. In conclusion, PTHrP was found to promote chondrogenesis and suppress hypertrophy during in vitro chondrogenesis from both BMMSCs and ATMSCs, which supports its use for cartilage tissue engineering.
Collapse
Affiliation(s)
- Young-Ju Kim
- Department of Orthopaedics, Dongguk University International Hospital, 814 Siksa-Dong, Goyang 411-773, Republic of Korea
| | | | | |
Collapse
|
86
|
Solomon LA, Bérubé NG, Beier F. Transcriptional regulators of chondrocyte hypertrophy. ACTA ACUST UNITED AC 2008; 84:123-30. [DOI: 10.1002/bdrc.20124] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
87
|
Murdoch AD, Grady LM, Ablett MP, Katopodi T, Meadows RS, Hardingham TE. Chondrogenic Differentiation of Human Bone Marrow Stem Cells in Transwell Cultures: Generation of Scaffold-Free Cartilage. Stem Cells 2007; 25:2786-96. [PMID: 17656642 DOI: 10.1634/stemcells.2007-0374] [Citation(s) in RCA: 206] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Human bone marrow stem cells (hMSCs) have been shown to differentiate in vitro into a number of cell lineages and are a potential autologous cell source for the repair and replacement of damaged and diseased musculoskeletal tissues. hMSC differentiation into chondrocytes has been described in high-density cell pellets cultured with specific growth and differentiation factors. We now describe how culture of hMSCs as a shallow multicellular layer on a permeable membrane over 2-4 weeks resulted in a much more efficient formation of cartilaginous tissue than in established chondrogenic assays. In this format, the hMSCs differentiated in 14 days to produce translucent, flexible discs, 6 mm in diameter by 0.8-1 mm in thickness from 0.5 x 10(6) cells. The discs contained an extensive cartilage-like extracellular matrix (ECM), with more than 50% greater proteoglycan content per cell than control hMSCs differentiated in standard cell pellet cultures. The disc constructs were also enriched in the cartilage-specific collagen II, and this was more homogeneously distributed than in cell pellet cultures. The expression of cartilage matrix genes for collagen type II and aggrecan was enhanced in disc cultures, but improved matrix production was not accompanied by increased expression of the transcription factors SOX9, L-SOX5, and SOX6. The fast continuous growth of cartilage ECM in these cultures up to 4 weeks appeared to result from the geometry of the construct and the efficient delivery of nutrients to the cells. Scaffold-free growth of cartilage in this format will provide a valuable experimental system for both experimental and potential clinical studies.
Collapse
Affiliation(s)
- Alan D Murdoch
- UK Centre for Tissue Engineering and Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Faculty of Life Sciences, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | | | | | | | | | | |
Collapse
|
88
|
Kempf H, Ionescu A, Udager AM, Lassar AB. Prochondrogenic signals induce a competence for Runx2 to activate hypertrophic chondrocyte gene expression. Dev Dyn 2007; 236:1954-62. [PMID: 17576141 DOI: 10.1002/dvdy.21205] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Whereas Runx2 is necessary for bone formation and cartilage hypertrophy, it is unclear why Runx2 induces markers of chondrocyte hypertrophy only in chondrocytes. We document that chondrocytes either contain a cofactor, which can be induced in somitic cells by prochondrogenic signals, that is necessary for Runx2 to induce chondrocyte hypertrophy or, alternatively, lack a repressor of this maturation program. Sequential Shh and bone morphogenetic protein (BMP) signals or forced expression of either Nkx3.2 or Sox9 (plus BMP signals) induces chondrogenesis in presomitic mesoderm and simultaneously induces a competence for Runx2 to activate the chondrocyte maturation program. The ability of either sequential Shh and BMP signals or retrovirus-encoded Nkx3.2 or Sox9 to induce this competence correlates with their ability to activate chondrogenesis in various embryonic tissues. Consistent with these findings in embryonic tissues, we have found that cotransfected Runx2 and Smad1 are able to induce the expression of a reporter construct driven by the collagen X regulatory sequences in chondrocytes but not in fibroblasts. In contrast, both Runx2 and Smad1 are competent to activate reporters driven by either reiterated Runx or Smad binding sites, respectively, in both cell types. As Sox9 and Nkx3.2 have previously been shown to block chondrocyte maturation in vivo, our findings suggest that these transcription factors can, in addition, either induce the expression or activity of a factor in chondrocytes that is required for Runx2 to activate the chondrocyte maturation program, or alternatively that these transcription factors block the expression or activity of a repressor of this maturation program.
Collapse
Affiliation(s)
- Hervé Kempf
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
89
|
Park M, Yong Y, Choi SW, Kim JH, Lee JE, Kim DW. Constitutive RelA activation mediated by Nkx3.2 controls chondrocyte viability. Nat Cell Biol 2007; 9:287-98. [PMID: 17310243 DOI: 10.1038/ncb1538] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2006] [Accepted: 01/12/2007] [Indexed: 02/04/2023]
Abstract
During endochondral ossification, a process that accounts for the majority of bone formation in vertebrates, hypertrophic chondrocytes display a greater susceptibility to apoptosis when compared to proliferating chondrocytes. However, the molecular mechanisms underlying this phenomenon remain unclear. Nkx3.2, a member of the NK class of homeoproteins, is initially expressed in chondrogenic precursor cells, and later, during cartilage maturation, its expression is restricted to proliferating chondrocytes. Here, we show that the nuclear factor kappa B (NF-kappaB) pathway is required for chondrocyte viability and that Nkx3.2 supports chondrocyte survival by constitutively activating RelA. Although signal-dependent NF-kappaB activation has been intensively studied, ligand-independent NF-kappaB activation is poorly understood. The data presented here support a novel ligand-independent mechanism of NF-kappaB activation, whereby Nkx3.2 recruits the RelA-IkappaBalpha heteromeric complex into the nucleus by direct protein-protein interactions and activates RelA through proteasome-dependent IkappaBalpha degradation in the nucleus. Furthermore, we demonstrate that stage-specific NF-kappaB activation, mediated by Nkx3.2, regulates chondrocyte viability during cartilage maturation.
Collapse
Affiliation(s)
- Minsun Park
- Department of Biochemistry, Yonsei University, SEOUL, 120-749, Republic of Korea
| | | | | | | | | | | |
Collapse
|
90
|
Kimmel CB, Walker MB, Miller CT. Morphing the hyomandibular skeleton in development and evolution. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2007; 308:609-24. [PMID: 17358015 DOI: 10.1002/jez.b.21155] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
How might changes in developmental regulatory pathways underlie evolutionary changes in morphology? Here we focus on a particular pathway regulated by a secreted, signaling peptide, Endothelin1 (Edn1). Developmental genetic analyses show the Edn1-pathway to be crucial for hyomandibular patterning, and we discuss our work with zebrafish suggesting how the signal may function in regulating numbers of skeletal elements, their sizes and their shapes. We then review a broader collection of comparative studies that examine morphological evolution of a subset of the same skeletal elements-the opercular-branchiostegal series of bones of the hyoid arch. We find that phenotypic changes in zebrafish mutants copy evolutionary changes that recur along many actinopterygian lineages. Hence the developmental genetic studies are informative for providing candidate pathways for macroevolution of facial morphology, as well as for our understanding of how these pathways work.
Collapse
Affiliation(s)
- Charles B Kimmel
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, USA.
| | | | | |
Collapse
|
91
|
Hassan MQ, Tare RS, Lee SH, Mandeville M, Morasso MI, Javed A, van Wijnen AJ, Stein JL, Stein GS, Lian JB. BMP2 commitment to the osteogenic lineage involves activation of Runx2 by DLX3 and a homeodomain transcriptional network. J Biol Chem 2006; 281:40515-26. [PMID: 17060321 DOI: 10.1074/jbc.m604508200] [Citation(s) in RCA: 170] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Several homeodomain (HD) proteins are critical for skeletal patterning and respond directly to BMP2 as an early step in bone formation. RUNX2, the earliest transcription factor proven essential for commitment to osteoblastogenesis, is also expressed in response to BMP2. However, there is a gap in our knowledge of the regulatory cascade from BMP2 signaling to the onset of osteogenesis. Here we show that BMP2 induces DLX3, a homeodomain protein that activates Runx2 gene transcription. Small interfering RNA knockdown studies in osteoblasts validate that DLX3 is a potent regulator of Runx2. Furthermore in Runx2 null cells, DLX3 forced expression suffices to induce transcription of Runx2, osteocalcin, and alkaline phosphatase genes, thus defining DLX3 as an osteogenic regulator independent of RUNX2. Our studies further show regulation of the Runx2 gene by several homeodomain proteins: MSX2 and CDP/cut repress whereas DLX3 and DLX5 activate endogenous Runx2 expression and promoter activity in non-osseous cells and osteoblasts. These HD proteins exhibit distinct temporal expression profiles during osteoblast differentiation as well as selective association with Runx2 chromatin that is related to Runx2 transcriptional activity and recruitment of RNA polymerase II. Runx2 promoter mutagenesis shows that multiple HD elements control expression of Runx2 in relation to the stages of osteoblast maturation. Our studies establish mechanisms for commitment to the osteogenic lineage directly through BMP2 induction of HD proteins DLX3 and DLX5 that activate Runx2, thus delineating a transcriptional regulatory pathway mediating osteoblast differentiation. We propose that the three homeodomain proteins MSX2, DLX3, and DLX5 provide a key series of molecular switches that regulate expression of Runx2 throughout bone formation.
Collapse
Affiliation(s)
- Mohammad Q Hassan
- Department of Cell Biology and Cancer Center, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Lian JB, Stein GS, Javed A, van Wijnen AJ, Stein JL, Montecino M, Hassan MQ, Gaur T, Lengner CJ, Young DW. Networks and hubs for the transcriptional control of osteoblastogenesis. Rev Endocr Metab Disord 2006; 7:1-16. [PMID: 17051438 DOI: 10.1007/s11154-006-9001-5] [Citation(s) in RCA: 349] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We present an overview of the concepts of tissue-specific transcriptional control mechanisms essential for development of the bone cell phenotype. BMP2 induced transcription factors constitute a network of activities and molecular switches for bone development and osteoblast differentiation. Among these regulators are Runx2 (Cbfa1/AML3), the principal osteogenic master gene for bone formation, as well as homeodomain proteins and osterix. Runx2 has multiple regulatory activities, including activation or repression of gene expression, and integration of biological signals from developmental cues, such as BMP/TGFbeta, Wnt and Src signaling pathways. Runx2 provides a new paradigm for transcriptional control by functioning as a principal scaffolding protein in nuclear microenvironments to control gene expression in response to physiologic signals (growth factors, cytokines and hormones). The protein serves as a hub for the coordination of activities essential for the expansion and differentiation of osteogenic lineage cells through the formation of co-regulatory protein complexes organized in subnuclear domains. Mechanisms by which Runx2 supports commitment to osteogenesis and determines cell fate involve its retention on mitotic chromosomes. Disruption of a unique protein module, the subnuclear targeting signal of Runx2, has profound effects on osteoblast differentiation and metastasis of cancer cells in the bone microenvironment. Runx2 target genes include regulators of cell growth control, components of the bone extracellular matrix, angiogenesis, and signaling proteins for development of the osteoblast phenotype and bone turnover. The specificity of Runx2 regulatory activities provides a basis for novel therapeutic strategies to correct bone disorders.
Collapse
Affiliation(s)
- Jane B Lian
- Department of Cell Biology and Cancer Center, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|