51
|
Milgrom-Hoffman M, Michailovici I, Ferrara N, Zelzer E, Tzahor E. Endothelial cells regulate neural crest and second heart field morphogenesis. Biol Open 2014; 3:679-88. [PMID: 24996922 PMCID: PMC4133721 DOI: 10.1242/bio.20148078] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cardiac and craniofacial developmental programs are intricately linked during early embryogenesis, which is also reflected by a high frequency of birth defects affecting both regions. The molecular nature of the crosstalk between mesoderm and neural crest progenitors and the involvement of endothelial cells within the cardio–craniofacial field are largely unclear. Here we show in the mouse that genetic ablation of vascular endothelial growth factor receptor 2 (Flk1) in the mesoderm results in early embryonic lethality, severe deformation of the cardio–craniofacial field, lack of endothelial cells and a poorly formed vascular system. We provide evidence that endothelial cells are required for migration and survival of cranial neural crest cells and consequently for the deployment of second heart field progenitors into the cardiac outflow tract. Insights into the molecular mechanisms reveal marked reduction in Transforming growth factor beta 1 (Tgfb1) along with changes in the extracellular matrix (ECM) composition. Our collective findings in both mouse and avian models suggest that endothelial cells coordinate cardio–craniofacial morphogenesis, in part via a conserved signaling circuit regulating ECM remodeling by Tgfb1.
Collapse
Affiliation(s)
- Michal Milgrom-Hoffman
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Inbal Michailovici
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Napoleone Ferrara
- Department of Pathology, University of California at San Diego, La Jolla, CA 92093, USA
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Eldad Tzahor
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
52
|
Guerrero L, Villar P, Martínez L, Badia-Careaga C, Arredondo JJ, Cervera M. In vivo cell tracking of mouse embryonic myoblasts and fast fibers during development. Genesis 2014; 52:793-808. [PMID: 24895317 DOI: 10.1002/dvg.22796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 05/30/2014] [Accepted: 05/31/2014] [Indexed: 11/05/2022]
Abstract
Fast and slow TnI are co-expressed in E11.5 embryos, and fast TnI is present from the very beginning of myogenesis. A novel green fluorescent protein (GFP) reporter mouse lines (FastTnI/GFP lines) that carry the primary and secondary enhancer elements of the mouse fast troponin I (fast TnI), in which reporter expression correlates precisely with distribution of the endogenous fTnI protein was generated. Using the FastTnI/GFP mouse model, we characterized the early myogenic events in mice, analyzing the migration of GFP+ myoblasts, and the formation of primary and secondary myotubes in transgenic embryos. Interestingly, we found that the two contractile fast and slow isoforms of TnI are expressed during the migration of myoblasts from the somites to the limbs and body wall, suggesting that both participate in these events. Since no sarcomeres are present in myoblasts, we speculate that the function of fast TnI in early myogenesis is, like Myosin and Tropomyosin, to participate in cell movement during the initial myogenic stages. genesis
Collapse
Affiliation(s)
- Lucia Guerrero
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid, Instituto de Investigaciones Biomédicas Alberto Sols, C.S.I.C., Madrid, Spain
| | | | | | | | | | | |
Collapse
|
53
|
Gámez B, Rodriguez-Carballo E, Ventura F. MicroRNAs and post-transcriptional regulation of skeletal development. J Mol Endocrinol 2014; 52:R179-97. [PMID: 24523514 DOI: 10.1530/jme-13-0294] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
MicroRNAs (miRNAs) have become integral nodes of post-transcriptional control of genes that confer cellular identity and regulate differentiation. Cell-specific signaling and transcriptional regulation in skeletal biology are extremely dynamic processes that are highly reliant on dose-dependent responses. As such, skeletal cell-determining genes are ideal targets for quantitative regulation by miRNAs. So far, large amounts of evidence have revealed a characteristic temporal miRNA signature in skeletal cell differentiation and confirmed the essential roles that numerous miRNAs play in bone development and homeostasis. In addition, microarray expression data have provided evidence for their role in several skeletal pathologies. Mouse models in which their expression is altered have provided evidence of causal links between miRNAs and bone abnormalities. Thus, a detailed understanding of the function of miRNAs and their tight relationship with bone diseases would constitute a powerful tool for early diagnosis and future therapeutic approaches.
Collapse
Affiliation(s)
- Beatriz Gámez
- Departament de Ciències Fisiològiques II, Universitat de Barcelona, IDIBELL, C/Feixa Llarga s/n, E-08907 L'Hospitalet de Llobregat, Spain
| | | | | |
Collapse
|
54
|
Garcez RC, Le Douarin NM, Creuzet SE. Combinatorial activity of Six1-2-4 genes in cephalic neural crest cells controls craniofacial and brain development. Cell Mol Life Sci 2014; 71:2149-64. [PMID: 24061537 PMCID: PMC11113736 DOI: 10.1007/s00018-013-1477-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 08/16/2013] [Accepted: 09/11/2013] [Indexed: 12/21/2022]
Abstract
The combinatorial expression of Hox genes is an evolutionarily ancient program underlying body axis patterning in all Bilateria. In the head, the neural crest (NC)--a vertebrate innovation that contributes to evolutionarily novel skeletal and neural features--develops as a structure free of Hox-gene expression. The activation of Hoxa2 in the Hox-free facial NC (FNC) leads to severe craniofacial and brain defects. Here, we show that this condition unveils the requirement of three Six genes, Six1, Six2, and Six4, for brain development and morphogenesis of the maxillo-mandibular and nasofrontal skeleton. Inactivation of each of these Six genes in FNC generates diverse brain defects, ranging from plexus agenesis to mild or severe holoprosencephaly, and entails facial hypoplasia or truncation of the craniofacial skeleton. The triple silencing of these genes reveals their complementary role in face and brain morphogenesis. Furthermore, we show that the perturbation of the intrinsic genetic FNC program, by either Hoxa2 expression or Six gene inactivation, affects Bmp signaling through the downregulation of Bmp antagonists in the FNC cells. When upregulated in the FNC, Bmp antagonists suppress the adverse skeletal and cerebral effects of Hoxa2 expression. These results demonstrate that the combinatorial expression of Six1, Six2, and Six4 is required for the molecular programs governing craniofacial and cerebral development. These genes are crucial for the signaling system of FNC origin, which regulates normal growth and patterning of the cephalic neuroepithelium. Our results strongly suggest that several congenital craniofacial and cerebral malformations could be attributed to Six genes' misregulation.
Collapse
Affiliation(s)
- Ricardo C. Garcez
- Institut de Neurobiologie, Laboratoire Neurobiologie et Développement, CNRS-UPR3294, avenue de la Terrasse, 91198 Gif-sur-Yvette, France
- Present Address: Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC 88040-900 Brazil
| | | | - Sophie E. Creuzet
- Institut de Neurobiologie, Laboratoire Neurobiologie et Développement, CNRS-UPR3294, avenue de la Terrasse, 91198 Gif-sur-Yvette, France
| |
Collapse
|
55
|
Horvath JE, Ramachandran GL, Fedrigo O, Nielsen WJ, Babbitt CC, St Clair EM, Pfefferle LW, Jernvall J, Wray GA, Wall CE. Genetic comparisons yield insight into the evolution of enamel thickness during human evolution. J Hum Evol 2014; 73:75-87. [PMID: 24810709 DOI: 10.1016/j.jhevol.2014.01.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 10/29/2013] [Accepted: 01/09/2014] [Indexed: 12/29/2022]
Abstract
Enamel thickness varies substantially among extant hominoids and is a key trait with significance for interpreting dietary adaptation, life history trajectory, and phylogenetic relationships. There is a strong link in humans between enamel formation and mutations in the exons of the four genes that code for the enamel matrix proteins and the associated protease. The evolution of thick enamel in humans may have included changes in the regulation of these genes during tooth development. The cis-regulatory region in the 5' flank (upstream non-coding region) of MMP20, which codes for enamelysin, the predominant protease active during enamel secretion, has previously been shown to be under strong positive selection in the lineages leading to both humans and chimpanzees. Here we examine evidence for positive selection in the 5' flank and 3' flank of AMELX, AMBN, ENAM, and MMP20. We contrast the human sequence changes with other hominoids (chimpanzees, gorillas, orangutans, gibbons) and rhesus macaques (outgroup), a sample comprising a range of enamel thickness. We find no evidence for positive selection in the protein-coding regions of any of these genes. In contrast, we find strong evidence for positive selection in the 5' flank region of MMP20 and ENAM along the lineage leading to humans, and in both the 5' flank and 3' flank regions of MMP20 along the lineage leading to chimpanzees. We also identify putative transcription factor binding sites overlapping some of the species-specific nucleotide sites and we refine which sections of the up- and downstream putative regulatory regions are most likely to harbor important changes. These non-coding changes and their potential for differential regulation by transcription factors known to regulate tooth development may offer insight into the mechanisms that allow for rapid evolutionary changes in enamel thickness across closely-related species, and contribute to our understanding of the enamel phenotype in hominoids.
Collapse
Affiliation(s)
- Julie E Horvath
- North Carolina Museum of Natural Sciences, Nature Research Center, Raleigh, NC 27601, USA; Department of Biology, North Carolina Central University, Durham, NC 27707, USA; Department of Evolutionary Anthropology, Duke University, Durham, NC 27708, USA; Duke Institute for Genome Sciences and Policy, Duke University, Durham, NC 27708, USA
| | | | - Olivier Fedrigo
- Duke Institute for Genome Sciences and Policy, Duke University, Durham, NC 27708, USA
| | | | - Courtney C Babbitt
- Duke Institute for Genome Sciences and Policy, Duke University, Durham, NC 27708, USA; Department of Biology, Duke University, Durham, NC 27708, USA
| | | | | | - Jukka Jernvall
- Institute for Biotechnology, University of Helsinki, Helsinki, Finland
| | - Gregory A Wray
- Department of Evolutionary Anthropology, Duke University, Durham, NC 27708, USA; Duke Institute for Genome Sciences and Policy, Duke University, Durham, NC 27708, USA; Department of Biology, Duke University, Durham, NC 27708, USA
| | - Christine E Wall
- Department of Evolutionary Anthropology, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
56
|
Schwarz D, Varum S, Zemke M, Schöler A, Baggiolini A, Draganova K, Koseki H, Schübeler D, Sommer L. Ezh2 is required for neural crest-derived cartilage and bone formation. Development 2014; 141:867-77. [PMID: 24496623 DOI: 10.1242/dev.094342] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The emergence of craniofacial skeletal elements, and of the jaw in particular, was a crucial step in the evolution of higher vertebrates. Most facial bones and cartilage are generated during embryonic development by cranial neural crest cells, while an osteochondrogenic fate is suppressed in more posterior neural crest cells. Key players in this process are Hox genes, which suppress osteochondrogenesis in posterior neural crest derivatives. How this specific pattern of osteochondrogenic competence is achieved remains to be elucidated. Here we demonstrate that Hox gene expression and osteochondrogenesis are controlled by epigenetic mechanisms. Ezh2, which is a component of polycomb repressive complex 2 (PRC2), catalyzes trimethylation of lysine 27 in histone 3 (H3K27me3), thereby functioning as transcriptional repressor of target genes. Conditional inactivation of Ezh2 does not interfere with localization of neural crest cells to their target structures, neural development, cell cycle progression or cell survival. However, loss of Ezh2 results in massive derepression of Hox genes in neural crest cells that are usually devoid of Hox gene expression. Accordingly, craniofacial bone and cartilage formation is fully prevented in Ezh2 conditional knockout mice. Our data indicate that craniofacial skeleton formation in higher vertebrates is crucially dependent on epigenetic regulation that keeps in check inhibitors of an osteochondrogenic differentiation program.
Collapse
Affiliation(s)
- Daniel Schwarz
- Cell and Developmental Biology, Institute of Anatomy, University of Zurich, CH-8057 Zurich, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
57
|
Kokabu S, Sato T, Ohte S, Enoki Y, Okubo M, Hayashi N, Nojima J, Tsukamoto S, Fukushima Y, Sakata Y, Katagiri T, Rosen V, Yoda T. Expression of TLE3 by bone marrow stromal cells is regulated by canonical Wnt signaling. FEBS Lett 2014; 588:614-9. [PMID: 24444608 DOI: 10.1016/j.febslet.2013.12.031] [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] [Received: 10/24/2013] [Revised: 12/19/2013] [Accepted: 12/20/2013] [Indexed: 11/17/2022]
Abstract
Transducing-like enhancer of split 3 (TLE3), one of the Groucho/TLE family members, targets Runx2 transcription and suppresses osteoblast differentiation in bone marrow stromal cells (BMSCs). Here, we identify Wnt responsive elements of the TLE3 promoter region through comparative genomic and functional analyses and show that expression of TLE3 is increased by Wnt signaling, which is important for osteoblast differentiation. We also demonstrated that TLE3 is able to suppress canonical Wnt signaling in BMSCs. Taken together, our data suggest that induction of TLE3 by Wnt signaling is part of a negative feedback loop active during osteoblast differentiation.
Collapse
Affiliation(s)
- Shoichiro Kokabu
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-machi, Saitama 350-0495, Japan; Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, USA.
| | - Tsuyoshi Sato
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-machi, Saitama 350-0495, Japan
| | - Satoshi Ohte
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, USA
| | - Yuichiro Enoki
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-machi, Saitama 350-0495, Japan
| | - Masahiko Okubo
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-machi, Saitama 350-0495, Japan
| | - Naoki Hayashi
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-machi, Saitama 350-0495, Japan
| | - Junya Nojima
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-machi, Saitama 350-0495, Japan
| | - Sho Tsukamoto
- Division of Pathophysiology, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama 350-1241, Japan
| | - Yosuke Fukushima
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-machi, Saitama 350-0495, Japan
| | - Yasuaki Sakata
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-machi, Saitama 350-0495, Japan
| | - Takenobu Katagiri
- Division of Pathophysiology, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka-shi, Saitama 350-1241, Japan
| | - Vicki Rosen
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA 02115, USA
| | - Tetsuya Yoda
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama-machi, Saitama 350-0495, Japan
| |
Collapse
|
58
|
Coelho-Aguiar JM, Le Douarin NM, Dupin E. Environmental factors unveil dormant developmental capacities in multipotent progenitors of the trunk neural crest. Dev Biol 2013; 384:13-25. [DOI: 10.1016/j.ydbio.2013.09.030] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 09/27/2013] [Accepted: 09/28/2013] [Indexed: 12/13/2022]
|
59
|
Bergiers I, Bridoux L, Nguyen N, Twizere JC, Rezsöhazy R. The homeodomain transcription factor Hoxa2 interacts with and promotes the proteasomal degradation of the E3 ubiquitin protein ligase RCHY1. PLoS One 2013; 8:e80387. [PMID: 24244684 PMCID: PMC3820564 DOI: 10.1371/journal.pone.0080387] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 10/02/2013] [Indexed: 12/19/2022] Open
Abstract
Hox proteins are conserved homeodomain transcription factors known to be crucial regulators of animal development. As transcription factors, the functions and modes of action (co-factors, target genes) of Hox proteins have been very well studied in a multitude of animal models. However, a handful of reports established that Hox proteins may display molecular activities distinct from gene transcription regulation. Here, we reveal that Hoxa2 interacts with 20S proteasome subunits and RCHY1 (also known as PIRH2), an E3 ubiquitin ligase that targets p53 for degradation. We further show that Hoxa2 promotes proteasome-dependent degradation of RCHY1 in an ubiquitin-independent manner. Correlatively, Hoxa2 alters the RCHY1-mediated ubiquitination of p53 and promotes p53 stabilization. Together, our data establish that Hoxa2 can regulate the proteasomal degradation of RCHY1 and stabilization of p53.
Collapse
Affiliation(s)
- Isabelle Bergiers
- Molecular and Cellular Animal Embryology Group, Life Sciences Institute, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Laure Bridoux
- Molecular and Cellular Animal Embryology Group, Life Sciences Institute, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Nathan Nguyen
- Molecular and Cellular Animal Embryology Group, Life Sciences Institute, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Jean-Claude Twizere
- Laboratory of Signaling and Protein Interactions, GIGA-R, University of Liege, Liège, Belgium
| | - René Rezsöhazy
- Molecular and Cellular Animal Embryology Group, Life Sciences Institute, Université catholique de Louvain, Louvain-la-Neuve, Belgium
- * E-mail:
| |
Collapse
|
60
|
Deprez PML, Nichane MG, Lengelé BG, Rezsöhazy R, Nyssen-Behets C. Molecular study of a Hoxa2 gain-of-function in chondrogenesis: a model of idiopathic proportionate short stature. Int J Mol Sci 2013; 14:20386-98. [PMID: 24129174 PMCID: PMC3821620 DOI: 10.3390/ijms141020386] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 09/16/2013] [Accepted: 09/16/2013] [Indexed: 12/31/2022] Open
Abstract
In a previous study using transgenic mice ectopically expressing Hoxa2 during chondrogenesis, we associated the animal phenotype to human idiopathic proportionate short stature. Our analysis showed that this overall size reduction was correlated with a negative influence of Hoxa2 at the first step of endochondral ossification. However, the molecular pathways leading to such phenotype are still unknown. Using protein immunodetection and histological techniques comparing transgenic mice to controls, we show here that the persistent expression of Hoxa2 in chondrogenic territories provokes a general down-regulation of the main factors controlling the differentiation cascade, such as Bapx1, Bmp7, Bmpr1a, Ihh, Msx1, Pax9, Sox6, Sox9 and Wnt5a. These data confirm the impairment of chondrogenic differentiation by Hoxa2 overexpression. They also show a selective effect of Hoxa2 on endochondral ossification processes since Gdf5 and Gdf10, and Bmp4 or PthrP were up-regulated and unmodified, respectively. Since Hoxa2 deregulation in mice induces a proportionate short stature phenotype mimicking human idiopathic conditions, our results give an insight into understanding proportionate short stature pathogenesis by highlighting molecular factors whose combined deregulation may be involved in such a disease.
Collapse
Affiliation(s)
- Pierre M. L. Deprez
- Ecole de Kinésiologie et Récréologie, Faculté des Sciences de la Santé et Services Communautaires, Université de Moncton, Moncton, NB E1A 3E9, Canada; E-Mail:
| | - Miloud G. Nichane
- Embryologie Moléculaire et Cellulaire Animale, Institut des Sciences de la Vie, Université catholique de Louvain, Louvain-la-Neuve 1348, Belgium; E-Mails: (M.G.N.); (R.R.)
| | - Benoît G. Lengelé
- Pôle de Morphologie, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels 1200, Belgium; E-Mail:
| | - René Rezsöhazy
- Embryologie Moléculaire et Cellulaire Animale, Institut des Sciences de la Vie, Université catholique de Louvain, Louvain-la-Neuve 1348, Belgium; E-Mails: (M.G.N.); (R.R.)
| | - Catherine Nyssen-Behets
- Pôle de Morphologie, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels 1200, Belgium; E-Mail:
| |
Collapse
|
61
|
Switching axial progenitors from producing trunk to tail tissues in vertebrate embryos. Dev Cell 2013; 25:451-62. [PMID: 23763947 DOI: 10.1016/j.devcel.2013.05.009] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 04/30/2013] [Accepted: 05/10/2013] [Indexed: 11/21/2022]
Abstract
The vertebrate body is made by progressive addition of new tissue from progenitors at the posterior embryonic end. Axial extension involves different mechanisms that produce internal organs in the trunk but not in the tail. We show that Gdf11 signaling is a major coordinator of the trunk-to-tail transition. Without Gdf11 signaling, the switch from trunk to tail is significantly delayed, and its premature activation brings the hindlimbs and cloaca next to the forelimbs, leaving extremely short trunks. Gdf11 activity includes activation of Isl1 to promote formation of the hindlimbs and cloaca-associated mesoderm as the most posterior derivatives of lateral mesoderm progenitors. Gdf11 also coordinates reallocation of bipotent neuromesodermal progenitors from the anterior primitive streak to the tail bud, in part by reducing the retinoic acid available to the progenitors. Our findings provide a perspective to understand the evolution of the vertebrate body plan.
Collapse
|
62
|
Differential distribution of the Ca (2+) regulator Pcp4 in the branchial arches is regulated by Hoxa2. PLoS One 2013; 8:e63160. [PMID: 23671666 PMCID: PMC3650044 DOI: 10.1371/journal.pone.0063160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 03/29/2013] [Indexed: 12/31/2022] Open
Abstract
Branchial arches are externally visible tissue bands in the head region of all vertebrate embryos. Although initially formed from similar components, each arch will give rise to different head and neck structures. In a screen designed to characterize the molecular control of branchial arch identity in mouse, we identified Pcp4 as a second branchial arch-specific molecular signature. We further show that the transcription factor Hoxa2 binds to Pcp4 chromatin and regulates Pcp4 expression in the second arch. Hoxa2 is also sufficient to induce Pcp4 expression in anterior first arch cells, which are Pcp4-negative.
Collapse
|
63
|
Münch J, González-Rajal A, de la Pompa JL. Notch regulates blastema proliferation and prevents differentiation during adult zebrafish fin regeneration. Development 2013; 140:1402-11. [DOI: 10.1242/dev.087346] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Zebrafish have the capacity to regenerate several organs, including the heart and fins. Fin regeneration is epimorphic, involving the formation at the amputation plane of a mass of undifferentiated, proliferating mesenchymal progenitor-like cells, called blastema. This tissue provides all the cell types that form the fin, so that after damage or amputation the fin pattern and structure are fully restored. How blastema cells remain in this progenitor-like state is poorly understood. Here, we show that the Notch pathway plays an essential role during fin regeneration. Notch signalling is activated during blastema formation and remains active throughout the regeneration process. Chemical inhibition or morpholino-mediated knockdown of Notch signalling impairs fin regeneration via decreased proliferation accompanied by reduced expression of Notch target genes in the blastema. Conversely, overexpression of a constitutively active form of the Notch1 receptor (N1ICD) in the regenerating fin leads to increased proliferation and to the expansion of the blastema cell markers msxe and msxb, as well as increased expression of the proliferation regulator aldh1a2. This blastema expansion prevents regenerative fin outgrowth, as indicated by the reduction in differentiating osteoblasts and the inhibition of bone regeneration. We conclude that Notch signalling maintains blastema cells in a plastic, undifferentiated and proliferative state, an essential requirement for fin regeneration.
Collapse
Affiliation(s)
- Juliane Münch
- Program of Cardiovascular Developmental Biology, Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Alvaro González-Rajal
- Program of Cardiovascular Developmental Biology, Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - José Luis de la Pompa
- Program of Cardiovascular Developmental Biology, Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| |
Collapse
|
64
|
Chen JW, Zahid S, Shilts MH, Weaver SJ, Leskowitz RM, Habbsa S, Aronowitz D, Rokins KP, Chang Y, Pinnella Z, Holloway L, Mansfield JH. Hoxa-5 acts in segmented somites to regulate cervical vertebral morphology. Mech Dev 2013; 130:226-40. [DOI: 10.1016/j.mod.2013.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 02/09/2013] [Accepted: 02/12/2013] [Indexed: 01/25/2023]
|
65
|
Luxán G, Casanova JC, Martínez-Poveda B, Prados B, D'Amato G, MacGrogan D, Gonzalez-Rajal A, Dobarro D, Torroja C, Martinez F, Izquierdo-García JL, Fernández-Friera L, Sabater-Molina M, Kong YY, Pizarro G, Ibañez B, Medrano C, García-Pavía P, Gimeno JR, Monserrat L, Jiménez-Borreguero LJ, de la Pompa JL. Mutations in the NOTCH pathway regulator MIB1 cause left ventricular noncompaction cardiomyopathy. Nat Med 2013; 19:193-201. [PMID: 23314057 DOI: 10.1038/nm.3046] [Citation(s) in RCA: 267] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 11/26/2012] [Indexed: 02/07/2023]
Abstract
Left ventricular noncompaction (LVNC) causes prominent ventricular trabeculations and reduces cardiac systolic function. The clinical presentation of LVNC ranges from asymptomatic to heart failure. We show that germline mutations in human MIB1 (mindbomb homolog 1), which encodes an E3 ubiquitin ligase that promotes endocytosis of the NOTCH ligands DELTA and JAGGED, cause LVNC in autosomal-dominant pedigrees, with affected individuals showing reduced NOTCH1 activity and reduced expression of target genes. Functional studies in cells and zebrafish embryos and in silico modeling indicate that MIB1 functions as a dimer, which is disrupted by the human mutations. Targeted inactivation of Mib1 in mouse myocardium causes LVNC, a phenotype mimicked by inactivation of myocardial Jagged1 or endocardial Notch1. Myocardial Mib1 mutants show reduced ventricular Notch1 activity, expansion of compact myocardium to proliferative, immature trabeculae and abnormal expression of cardiac development and disease genes. These results implicate NOTCH signaling in LVNC and indicate that MIB1 mutations arrest chamber myocardium development, preventing trabecular maturation and compaction.
Collapse
Affiliation(s)
- Guillermo Luxán
- Program of Cardiovascular Developmental Biology, Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Casanova JC, Travisano S, de la Pompa JL. Epithelial-to-mesenchymal transition in epicardium is independent of Snail1. Genesis 2012; 51:32-40. [PMID: 23097346 DOI: 10.1002/dvg.22353] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 10/15/2012] [Indexed: 11/07/2022]
Abstract
The epicardium is the outer epithelial covering the heart. This tissue undergoes an epithelial-to-mesenchymal transition (EMT) to generate mesenchymal epicardial-derived cells (EPDCs) that populate the extracellular matrix of the subepicardium and contribute to the development of the coronary vessels and cardiac interstitial cells. Although epicardial EMT plays a crucial role in heart development, the molecular regulation of this process is incompletely understood. Here we examined the possible role of the EMT regulator Snail1 in this process. Snail1 is expressed in the epicardium and EPDCs during mouse cardiac development. To determine the function of Snail1 in epicardial EMT, we deleted Snail1 in the epicardium using Wt1- and Tbx18-Cre drivers. Unexpectedly, epicardial-specific Snail1 mutants are viable and fertile and do not display any obvious morphological or functional cardiac abnormalities. Molecular analysis of these mice reveals that epicardial EMT occurs normally, and epicardial derivatives are established in these mutants. We conclude that Snail1 is not required for the initiation and progression of embryonic epicardial EMT.
Collapse
|
67
|
Two origins of blastemal progenitors define blastemal regeneration of zebrafish lower jaw. PLoS One 2012; 7:e45380. [PMID: 23028974 PMCID: PMC3448660 DOI: 10.1371/journal.pone.0045380] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 08/17/2012] [Indexed: 12/16/2022] Open
Abstract
Zebrafish possess a remarkable ability to regenerate complicated structures by formation of a mass of undifferentiated mesenchymal cells called blastema. To understand how the blastema retains the original structural form, we investigate cellular transitions and transcriptional characteristics of cell identity genes during all stages of regeneration of an amputated lower jaw. We find that mesenchymal blastema originates from multiple sources including nucleated blood cells, fibroblasts, damaged muscle cells and pigment cells. These cells are transformed into two populations of blastemal progenitors: foxi1-expression and isl1-expression, before giving rise to cartilage, bone, and muscle. Time point- based transcriptomal analysis of 45 annotated Hox genes reveal that five 3′-end Hox genes and an equal number of 5′-end Hox genes are activated largely at the stage of blastema reformation. RNA in situ hybridization shows that foxi1 and pax3a are respectively expressed in the presumptive mandible skeletal region and regenerating muscle at 5 dpa. In contrast, hoxa2b and hoxa11b are widely expressed with different domain in chondrogenic blastema and blastema mesenchyme. Knockdown foxi1 changes the expression patterns of sox9a and hoxa2b in chondrogenic blastema. From these results we propose that two origins of blastemal progenitors define blastema skeleton and muscle respecifications through distinct signaling pathways. Meanwhile, the positional identity of blastema reformation is implicated in mesenchymal segmentation and characteristic expression pattern of Hox genes.
Collapse
|
68
|
Zhang P, Men J, Fu Y, Shan T, Ye J, Wu Y, Tao Z, Liu L, Jiang H. Contribution of SATB2 to the stronger osteogenic potential of bone marrow stromal cells from craniofacial bones. Cell Tissue Res 2012; 350:425-37. [DOI: 10.1007/s00441-012-1487-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2011] [Accepted: 08/15/2012] [Indexed: 12/01/2022]
|
69
|
Thompson H, Ohazama A, Sharpe PT, Tucker AS. The origin of the stapes and relationship to the otic capsule and oval window. Dev Dyn 2012; 241:1396-404. [PMID: 22778034 DOI: 10.1002/dvdy.23831] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2012] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND The stapes, an ossicle found within the middle ear, is involved in transmitting sound waves to the inner ear by means of the oval window. There are several developmental problems associated with this ossicle and the oval window, which cause hearing loss. The developmental origin of these tissues has not been fully elucidated. RESULTS Using transgenic reporter mice, we have shown that the stapes is of dual origin with the stapedial footplate being composed of cells of both neural crest and mesodermal origin. Wnt1cre/Dicer mice fail to develop neural crest-derived cartilages, therefore, have no middle ear ossicles. We have shown in these mice the mesodermal stapedial footplate fails to form and the oval window is induced but underdeveloped. CONCLUSIONS If the neural crest part of the stapes fails to form the mesodermal part does not develop, indicating that the two parts are interdependent. The stapes develops tightly associated with the otic capsule, however, it is not essential for the positioning of the oval window, suggesting that other tissues, perhaps within the inner ear are needed for oval window placement.
Collapse
Affiliation(s)
- Hannah Thompson
- Department of Craniofacial Development and Stem Cell Biology, Kings College London, Guy's Tower, Guy's Hospital, London Bridge, London, UK
| | | | | | | |
Collapse
|
70
|
Barradas AM, Lachmann K, Hlawacek G, Frielink C, Truckenmoller R, Boerman OC, van Gastel R, Garritsen H, Thomas M, Moroni L, van Blitterswijk C, de Boer J. Surface modifications by gas plasma control osteogenic differentiation of MC3T3-E1 cells. Acta Biomater 2012; 8:2969-77. [PMID: 22522130 DOI: 10.1016/j.actbio.2012.04.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 03/16/2012] [Accepted: 04/11/2012] [Indexed: 10/28/2022]
Abstract
Numerous studies have shown that the physicochemical properties of biomaterials can control cell activity. Cell adhesion, proliferation, differentiation as well as tissue formation in vivo can be tuned by properties such as the porosity, surface micro- and nanoscale topography and chemical composition of biomaterials. This concept is very appealing for tissue engineering since instructive properties in bioactive materials can be more economical and time efficient than traditional strategies of cell pre-differentiation in vitro prior to implantation. The biomaterial surface, which is easy to modify due to its accessibility, may provide the necessary signals to elicit a certain cellular behavior. Here, we used gas plasma technology at atmospheric pressure to modify the physicochemical properties of polylactic acid and analyzed how this influenced pre-osteoblast proliferation and differentiation. Tetramethylsilane and 3-aminopropyl-trimethoxysilane with helium as a carrier gas or a mixture of nitrogen and hydrogen were discharged to polylactic acid discs to create different surface chemical compositions, hydrophobicity and microscale topographies. Such modifications influenced protein adsorption and pre-osteoblast cell adhesion, proliferation and osteogenic differentiation. Furthermore polylactic acid treated with tetramethylsilane enhanced osteogenic differentiation compared to the other surfaces. This promising surface modification could be further explored for potential development of bone graft substitutes.
Collapse
|
71
|
Wei J, Shi Y, Zheng L, Zhou B, Inose H, Wang J, Guo XE, Grosschedl R, Karsenty G. miR-34s inhibit osteoblast proliferation and differentiation in the mouse by targeting SATB2. ACTA ACUST UNITED AC 2012; 197:509-21. [PMID: 22564414 PMCID: PMC3352956 DOI: 10.1083/jcb.201201057] [Citation(s) in RCA: 195] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
miR-34b and -c inhibit osteoblast proliferation and differentiation by decreasing the levels of cell cycle proteins and of the nuclear matrix protein SATB2. A screen of microRNAs preferentially expressed in osteoblasts identified members of the miR-34 family as regulators of osteoblast proliferation and/or differentiation. Osteoblast-specific gain- and loss-of-function experiments performed in vivo revealed that miR-34b and -c affected skeletogenesis during embryonic development, as well as bone mass accrual after birth, through two complementary cellular and molecular mechanisms. First, they inhibited osteoblast proliferation by suppressing Cyclin D1, CDK4, and CDK6 accumulation. Second, they inhibited terminal differentiation of osteoblasts, at least in part through the inhibition of SATB2, a nuclear matrix protein that is a critical determinant of osteoblast differentiation. Genetic evidence obtained in the mouse confirmed the importance of SATB2 regulation by miR-34b/c. These results are the first to identify a family of microRNAs involved in bone formation in vivo and to identify a specific genetic pathway by which these microRNAs regulate osteoblast differentiation.
Collapse
Affiliation(s)
- Jianwen Wei
- Department of Genetics and Development, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Donaldson IJ, Amin S, Hensman JJ, Kutejova E, Rattray M, Lawrence N, Hayes A, Ward CM, Bobola N. Genome-wide occupancy links Hoxa2 to Wnt-β-catenin signaling in mouse embryonic development. Nucleic Acids Res 2012; 40:3990-4001. [PMID: 22223247 PMCID: PMC3351182 DOI: 10.1093/nar/gkr1240] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The regulation of gene expression is central to developmental programs and largely depends on the binding of sequence-specific transcription factors with cis-regulatory elements in the genome. Hox transcription factors specify the spatial coordinates of the body axis in all animals with bilateral symmetry, but a detailed knowledge of their molecular function in instructing cell fates is lacking. Here, we used chromatin immunoprecipitation with massively parallel sequencing (ChIP-seq) to identify Hoxa2 genomic locations in a time and space when it is actively instructing embryonic development in mouse. Our data reveals that Hoxa2 has large genome coverage and potentially regulates thousands of genes. Sequence analysis of Hoxa2-bound regions identifies high occurrence of two main classes of motifs, corresponding to Hox and Pbx-Hox recognition sequences. Examination of the binding targets of Hoxa2 faithfully captures the processes regulated by Hoxa2 during embryonic development; in addition, it uncovers a large cluster of potential targets involved in the Wnt-signaling pathway. In vivo examination of canonical Wnt-β-catenin signaling reveals activity specifically in Hoxa2 domain of expression, and this is undetectable in Hoxa2 mutant embryos. The comprehensive mapping of Hoxa2-binding sites provides a framework to study Hox regulatory networks in vertebrate developmental processes.
Collapse
Affiliation(s)
- Ian J Donaldson
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Building strong bones: molecular regulation of the osteoblast lineage. Nat Rev Mol Cell Biol 2011; 13:27-38. [DOI: 10.1038/nrm3254] [Citation(s) in RCA: 823] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
74
|
Deprez PML, Nichane MG, Rousseaux P, Devogelaer JP, Chappard D, Lengelé BG, Rezsöhazy R, Nyssen-Behets C. Postnatal growth defect in mice upon persistent Hoxa2 expression in the chondrogenic cell lineage. Differentiation 2011; 83:158-67. [PMID: 22093256 DOI: 10.1016/j.diff.2011.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 09/07/2011] [Accepted: 10/07/2011] [Indexed: 12/18/2022]
Abstract
Hoxa2 is a homeotic transcription factor, which is downregulated once chondrogenic differentiation is initiated. We previously generated a transgenic mouse model, which turns Hoxa2 on in cells expressing Collagen II A1, i.e. in cells entering chondrogenesis. As a consequence, mice display a general embryonic delay of ossification and then a postnatal growth defect. Col2a1-Cre mice were crossed with an inducible β-actin driven Hoxa2 transgene. Spines, vertebrae and limbs were measured and skeletal elements were studied by X-ray, microCT, pQCT, TEM, western-blotting, histomorphometry and immunohistochemistry. Mice expressing Hoxa2 in chondrogenic cells feature a proportionate short stature phenotype with a severe lordosis, which appeared significant from postnatal day 4. Analysis of both cartilage and bone development in affected embryos and mice from birth till P35 did not reveal any major defect in histogenesis, except a reduced number of chondrocytes in the vertebral anlage at E13.5. In conclusion, the sustained expression of Hoxa2 in the chondrocyte lineage is characterized by a proportionate short stature resulting from skeletal growth defect. The indepth analysis of cartilage and bone histogenesis points towards an initial deficit in cell mobilization to enter chondrogenesis.
Collapse
Affiliation(s)
- Pierre Marcel Louis Deprez
- Pôle de Morphologie, Institut de recherche expérimentale et clinique, Université catholique de Louvain, Brussels, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
75
|
Ye JH, Xu YJ, Gao J, Yan SG, Zhao J, Tu Q, Zhang J, Duan XJ, Sommer CA, Mostoslavsky G, Kaplan D, Wu YN, Zhang CP, Wang L, Chen J. Critical-size calvarial bone defects healing in a mouse model with silk scaffolds and SATB2-modified iPSCs. Biomaterials 2011; 32:5065-76. [PMID: 21492931 PMCID: PMC3100415 DOI: 10.1016/j.biomaterials.2011.03.053] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Accepted: 03/21/2011] [Indexed: 12/16/2022]
Abstract
Induced pluripotent stem cells (iPSCs) can differentiate into mineralizing cells and thus have a great potential in application in engineered bone substitutes with bioactive scaffolds in regeneration medicine. In the current study we characterized and demonstrated the pluripotency and osteogenic differentiation of mouse iPSCs. To enhance the osteogenic differentiation of iPSCs, we then transduced the iPSCs with the potent transcription factor, nuclear matrix protein SATB2. We observed that in SATB2-overexpressing iPSCs there were increased mineral nodule formation and elevated mRNA levels of key osteogenic genes, osterix (OSX), Runx2, bone sialoprotein (BSP) and osteocalcin (OCN). Moreover, the mRNA levels of HoxA2 was reduced after SATB2 overexpression in iPSCs. The SATB2-overexpressing iPSCs were then combined with silk scaffolds and transplanted into critical-size calvarial bone defects created in nude mice. Five weeks post-surgery, radiological and micro-CT analysis revealed enhanced new bone formation in calvarial defects in SATB2 group. Histological analysis also showed increased new bone formation and mineralization in the SATB2 group. In conclusion, the results demonstrate that SATB2 facilitates the differentiation of iPSCs towards osteoblast-lineage cells by repressing HoxA2 and augmenting the functions of the osteoblast determinants Runx2, BSP and OCN.
Collapse
Affiliation(s)
- Jin-Hai Ye
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
- Institute of Stomatology, School of Stomatology, Nanjing Medical University, Nanjing 210029, China
- Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Yuan-Jin Xu
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
- Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Jun Gao
- Model Animal Research Center, MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, 210061, China
| | - Shi-Guo Yan
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
- School of Stomatology, Shandong University, Jinan 250012, China
| | - Jun Zhao
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
- Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Qisheng Tu
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| | - Jin Zhang
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
- School of Stomatology, Shandong University, Jinan 250012, China
| | - Xue-Jing Duan
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
- School of Stomatology, Shandong University, Jinan 250012, China
| | - Cesar A. Sommer
- Section of Gastroenterology, Department of Medicine, and Center for Regenerative Medicine (CReM), Boston University School of Medicine, Boston, MA 02118, USA
| | - Gustavo Mostoslavsky
- Section of Gastroenterology, Department of Medicine, and Center for Regenerative Medicine (CReM), Boston University School of Medicine, Boston, MA 02118, USA
| | - David Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Yu-Nong Wu
- Institute of Stomatology, School of Stomatology, Nanjing Medical University, Nanjing 210029, China
| | - Chen-Ping Zhang
- Ninth People’s Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Lin Wang
- Institute of Stomatology, School of Stomatology, Nanjing Medical University, Nanjing 210029, China
| | - Jake Chen
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| |
Collapse
|
76
|
Mansfield JH, Abzhanov A. Hox expression in the American alligator and evolution of archosaurian axial patterning. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2011; 314:629-44. [PMID: 20623505 DOI: 10.1002/jez.b.21364] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The avian body plan has undergone many modifications, most associated with adaptation to flight and bipedal walking. Some of these modifications may be owing to avian-specific changes in the embryonic Hox expression code. Here, we have examined Hox expression in alligator, the closest living relative of birds, and an archosaur with a more conservative body plan. Two differences in Hox expression between chick, alligator, and other tetrapods correlate with aspects of alligator or bird-specific skeletal morphology. First, absence of a thoracic subdomain of Hoxc-8 expression in alligator correlates with morphological adaptations in crocodilian thoracic segments. Second, Hoxa-5, a gene required to pattern the cervical-thoracic transition, shows unique patterns of expression in chick, alligator, and mouse, correlating with species-specific morphological patterning of this region. Given that cervical vertebral morphologies evolved independently in the bird and mammalian lineages, the underlying developmental mechanisms, including refinement of Hox expression domains, may be distinct.
Collapse
Affiliation(s)
- Jennifer H Mansfield
- Department of Biological Sciences, Barnard College, Columbia University, 3009 Broadway, New York, NY 10027, USA.
| | | |
Collapse
|
77
|
Kirilenko P, He G, Mankoo BS, Mallo M, Jones R, Bobola N. Transient activation of meox1 is an early component of the gene regulatory network downstream of hoxa2. Mol Cell Biol 2011; 31:1301-8. [PMID: 21245383 PMCID: PMC3067911 DOI: 10.1128/mcb.00705-10] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Revised: 08/10/2010] [Accepted: 01/02/2011] [Indexed: 11/20/2022] Open
Abstract
Hox genes encode transcription factors that regulate morphogenesis in all animals with bilateral symmetry. Although Hox genes have been extensively studied, their molecular function is not clear in vertebrates, and only a limited number of genes regulated by Hox transcription factors have been identified. Hoxa2 is required for correct development of the second branchial arch, its major domain of expression. We now show that Meox1 is genetically downstream from Hoxa2 and is a direct target. Meox1 expression is downregulated in the second arch of Hoxa2 mouse mutant embryos. In chromatin immunoprecipitation (ChIP), Hoxa2 binds to the Meox1 proximal promoter. Two highly conserved binding sites contained in this sequence are required for Hoxa2-dependent activation of the Meox1 promoter. Remarkably, in the absence of Meox1 and its close homolog Meox2, the second branchial arch develops abnormally and two of the three skeletal elements patterned by Hoxa2 are malformed. Finally, we show that Meox1 can specifically bind the DNA sequences recognized by Hoxa2 on its functional target genes. These results provide new insight into the Hoxa2 regulatory network that controls branchial arch identity.
Collapse
Affiliation(s)
- Pavel Kirilenko
- School of Dentistry, Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom, Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom, Instituto Gulbenkian de Ciência, Oeiras, Portugal, Genetic Medicine, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - Guiyuan He
- School of Dentistry, Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom, Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom, Instituto Gulbenkian de Ciência, Oeiras, Portugal, Genetic Medicine, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - Baljinder S. Mankoo
- School of Dentistry, Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom, Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom, Instituto Gulbenkian de Ciência, Oeiras, Portugal, Genetic Medicine, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - Moises Mallo
- School of Dentistry, Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom, Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom, Instituto Gulbenkian de Ciência, Oeiras, Portugal, Genetic Medicine, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - Richard Jones
- School of Dentistry, Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom, Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom, Instituto Gulbenkian de Ciência, Oeiras, Portugal, Genetic Medicine, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - Nicoletta Bobola
- School of Dentistry, Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom, Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom, Instituto Gulbenkian de Ciência, Oeiras, Portugal, Genetic Medicine, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
78
|
Hu R, Liu W, Li H, Yang L, Chen C, Xia ZY, Guo LJ, Xie H, Zhou HD, Wu XP, Luo XH. A Runx2/miR-3960/miR-2861 regulatory feedback loop during mouse osteoblast differentiation. J Biol Chem 2011; 286:12328-39. [PMID: 21324897 DOI: 10.1074/jbc.m110.176099] [Citation(s) in RCA: 189] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Our recent study showed that miR-2861 promotes osteoblast differentiation by targeting histone deacetylase 5, resulting in increased runt-related transcription factor 2 (Runx2) protein production. Here we identified another new microRNA (miRNA) (miR-3960) that played a regulatory role in osteoblast differentiation through a regulatory feedback loop with miR-2861. miR-3960 and miR-2861 were found clustered at the same loci. miR-3960 was transcribed during bone morphogenic protein 2 (BMP2)-induced osteogenesis of ST2 stromal cells. Overexpression of miR-3960 promoted BMP2-induced osteoblastogenesis. However, the inhibition of miR-3960 expression attenuated the osteoblastogenesis. Homeobox A2 (Hoxa2), a repressor of Runx2 expression, was confirmed to be a target of miR-3960. Electrophoretic mobility shift assay and chromatin immunoprecipitation experiments confirmed that Runx2 bound to the promoter of the miR-3960/miR-2861 cluster. Furthermore, overexpression of Runx2 induced miR-3960/miR-2861 transcription, and block of Runx2 expression attenuated BMP2-induced miR-3960/miR-2861 transcription. Here we report that miR-3960 and miR-2861, transcribed together from the same miRNA polycistron, both function in osteoblast differentiation through a novel Runx2/miR-3960/miR-2861 regulatory feedback loop. Our findings provide new insights into the roles of miRNAs in osteoblast differentiation.
Collapse
Affiliation(s)
- Rong Hu
- Institute of Endocrinology and Metabolism, The Second Xiangya Hospital of Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Lourenço R, Lopes SS, Saúde L. Left-right function of dmrt2 genes is not conserved between zebrafish and mouse. PLoS One 2010; 5:e14438. [PMID: 21203428 PMCID: PMC3010978 DOI: 10.1371/journal.pone.0014438] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Accepted: 12/07/2010] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Members of the Dmrt family, generally associated with sex determination, were shown to be involved in several other functions during embryonic development. Dmrt2 has been studied in the context of zebrafish development where, due to a duplication event, two paralog genes dmrt2a and dmrt2b are present. Both zebrafish dmrt2a/terra and dmrt2b are important to regulate left-right patterning in the lateral plate mesoderm. In addition, dmrt2a/terra is necessary for symmetric somite formation while dmrt2b regulates somite differentiation impacting on slow muscle development. One dmrt2 gene is also expressed in the mouse embryo, where it is necessary for somite differentiation but with an impact on axial skeleton development. However, nothing was known about its role during left-right patterning in the lateral plate mesoderm or in the symmetric synchronization of somite formation. METHODOLOGY/PRINCIPAL FINDINGS Using a dmrt2 mutant mouse line, we show that this gene is not involved in symmetric somite formation and does not regulate the laterality pathway that controls left-right asymmetric organ positioning. We reveal that dmrt2a/terra is present in the zebrafish laterality organ, the Kupffer's vesicle, while its homologue is excluded from the mouse equivalent structure, the node. On the basis of evolutionary sub-functionalization and neo-functionalization theories we discuss this absence of functional conservation. CONCLUSIONS/SIGNIFICANCE Our results show that the role of dmrt2 gene is not conserved during zebrafish and mouse embryonic development.
Collapse
Affiliation(s)
- Raquel Lourenço
- Instituto de Medicina Molecular e Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Susana S. Lopes
- Instituto de Medicina Molecular e Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Leonor Saúde
- Instituto de Medicina Molecular e Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
- * E-mail:
| |
Collapse
|
80
|
Minoux M, Rijli FM. Molecular mechanisms of cranial neural crest cell migration and patterning in craniofacial development. Development 2010; 137:2605-21. [DOI: 10.1242/dev.040048] [Citation(s) in RCA: 329] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
During vertebrate craniofacial development, neural crest cells (NCCs) contribute much of the cartilage, bone and connective tissue that make up the developing head. Although the initial patterns of NCC segmentation and migration are conserved between species, the variety of vertebrate facial morphologies that exist indicates that a complex interplay occurs between intrinsic genetic NCC programs and extrinsic environmental signals during morphogenesis. Here, we review recent work that has begun to shed light on the molecular mechanisms that govern the spatiotemporal patterning of NCC-derived skeletal structures – advances that are central to understanding craniofacial development and its evolution.
Collapse
Affiliation(s)
- Maryline Minoux
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland
- Faculté de Chirurgie Dentaire, 1, Place de l'Hôpital, 67000 Strasbourg, France
| | - Filippo M. Rijli
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland
| |
Collapse
|
81
|
Mao XY, Tang SJ. Effects of phenytoin on Satb2 and Hoxa2 gene expressions in mouse embryonic craniofacial tissue. Biochem Cell Biol 2010; 88:731-5. [PMID: 20651846 DOI: 10.1139/o10-013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Cleft lip and cleft palate are common congenital craniofacial birth defects in humans. Phenytoin (PHT) is a risk factor of cleft palate formation; however, the molecular mechanisms by which phenytoin exerts its teratogenic effects resulting in cleft palate remain unknown. The Satb2 gene mutation is associated with cleft palate. Satb2-deficient mice exhibit cleft palate deformity and an up-regulation of Hoxa2 in the fronto-nasal region. In this study, phenytoin was administered intraperitoneally to pregnant C57BL/6 mice on the 10th day of gestation. Real-time PCR results showed that the expressions of Satb2 and Hoxa2 in craniofacial tissues of mouse embryos were obviously different at different time points. The Satb2 gene was down-regulated and the Hoxa2 gene was up-regulated in phenytoin-treated mouse embryonic craniofacial tissue. We conclude that phenytoin may regulate the expression of these two genes in C57BL/6 mice and it may also be involved in the formation of cleft palate.
Collapse
Affiliation(s)
- Xiao Yan Mao
- Cleft Lip and Palate Treatment Center, the Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | | |
Collapse
|
82
|
Trowe MO, Shah S, Petry M, Airik R, Schuster-Gossler K, Kist R, Kispert A. Loss of Sox9 in the periotic mesenchyme affects mesenchymal expansion and differentiation, and epithelial morphogenesis during cochlea development in the mouse. Dev Biol 2010; 342:51-62. [DOI: 10.1016/j.ydbio.2010.03.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Revised: 02/24/2010] [Accepted: 03/16/2010] [Indexed: 10/19/2022]
|
83
|
Inactivation of Six2 in mouse identifies a novel genetic mechanism controlling development and growth of the cranial base. Dev Biol 2010; 344:720-30. [PMID: 20515681 DOI: 10.1016/j.ydbio.2010.05.509] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2009] [Revised: 05/03/2010] [Accepted: 05/24/2010] [Indexed: 12/17/2022]
Abstract
The cranial base is essential for integrated craniofacial development and growth. It develops as a cartilaginous template that is replaced by bone through the process of endochondral ossification. Here, we describe a novel and specific role for the homeoprotein Six2 in the growth and elongation of the cranial base. Six2-null newborn mice display premature fusion of the bones in the cranial base. Chondrocyte differentiation is abnormal in the Six2-null cranial base, with reduced proliferation and increased terminal differentiation. Gain-of-function experiments indicate that Six2 promotes cartilage development and growth in other body areas and appears therefore to control general regulators of chondrocyte differentiation. Our data indicate that the main factors restricting Six2 function to the cranial base are tissue-specific transcription of the gene and compensatory effects of other Six family members. The comparable expression during human embryogenesis and the high protein conservation from mouse to human implicate SIX2 loss-of-function as a potential congenital cause of anterior cranial base defects in humans.
Collapse
|
84
|
Evidence for a myotomal Hox/Myf cascade governing nonautonomous control of rib specification within global vertebral domains. Dev Cell 2010; 18:655-61. [PMID: 20412779 DOI: 10.1016/j.devcel.2010.02.011] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Revised: 12/28/2009] [Accepted: 02/18/2010] [Indexed: 11/24/2022]
Abstract
Hox genes are essential for the patterning of the axial skeleton. Hox group 10 has been shown to specify the lumbar domain by setting a rib-inhibiting program in the presomitic mesoderm (PSM). We have now produced mice with ribs in every vertebra by ectopically expressing Hox group 6 in the PSM, indicating that Hox genes are also able to specify the thoracic domain. We show that the information provided by Hox genes to specify rib-containing and rib-less areas is first interpreted in the myotome through the regional-specific control of Myf5 and Myf6 expression. This information is then transmitted to the sclerotome by a system that includes FGF and PDGF signaling to produce vertebrae with or without ribs at different axial levels. Our findings offer a new perspective of how Hox genes produce global patterns in the axial skeleton and support a redundant nonmyogenic role of Myf5 and Myf6 in rib formation.
Collapse
|
85
|
Hoshiba T, Kawazoe N, Tateishi T, Chen G. Development of stepwise osteogenesis-mimicking matrices for the regulation of mesenchymal stem cell functions. J Biol Chem 2009; 284:31164-73. [PMID: 19762920 DOI: 10.1074/jbc.m109.054676] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
An extracellular microenvironment, including an extracellular matrix (ECM), is an important factor in regulating stem cell differentiation. During tissue development, the ECM is dynamically remodeled to regulate stem cell functions. Here, we developed matrices mimicking ECM remodeling during the osteogenesis of mesenchymal stem cells (MSCs). The matrices were prepared from cultured MSCs controlled at different stages of osteogenesis and referred to as "stepwise osteogenesis-mimicking matrices." The matrices supported the adhesion and proliferation of MSCs and showed different effects on the osteogenesis of MSCs. On the matrices mimicking the early stage of osteogenesis (early stage matrices), the osteogenesis occurred more rapidly than did that on the matrices mimicking undifferentiated stem cells (stem cell matrices) and the late stage of osteogenesis (late stage matrices). RUNX2 was similarly expressed when MSCs were cultured on both the early stage and late stage matrices but decreased on the stem cell matrices. PPARG expression in the MSCs cultured on the late stage matrices was higher than for those cultured on the stem cell and early stage matrices. This increase of PPARG expression was caused by the suppression of the amount of beta-catenin and downstream signal transduction. These results demonstrate that the osteogenesis-mimicking matrices had different effects on the osteogenesis of MSCs, and the early stage matrices provided a favorable microenvironment for the osteogenesis.
Collapse
Affiliation(s)
- Takashi Hoshiba
- Biomaterials Center, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | | | | | | |
Collapse
|
86
|
Leach SM, Tipney H, Feng W, Baumgartner WA, Kasliwal P, Schuyler RP, Williams T, Spritz RA, Hunter L. Biomedical discovery acceleration, with applications to craniofacial development. PLoS Comput Biol 2009; 5:e1000215. [PMID: 19325874 PMCID: PMC2653649 DOI: 10.1371/journal.pcbi.1000215] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2008] [Accepted: 02/12/2009] [Indexed: 01/17/2023] Open
Abstract
The profusion of high-throughput instruments and the explosion of new results in the scientific literature, particularly in molecular biomedicine, is both a blessing and a curse to the bench researcher. Even knowledgeable and experienced scientists can benefit from computational tools that help navigate this vast and rapidly evolving terrain. In this paper, we describe a novel computational approach to this challenge, a knowledge-based system that combines reading, reasoning, and reporting methods to facilitate analysis of experimental data. Reading methods extract information from external resources, either by parsing structured data or using biomedical language processing to extract information from unstructured data, and track knowledge provenance. Reasoning methods enrich the knowledge that results from reading by, for example, noting two genes that are annotated to the same ontology term or database entry. Reasoning is also used to combine all sources into a knowledge network that represents the integration of all sorts of relationships between a pair of genes, and to calculate a combined reliability score. Reporting methods combine the knowledge network with a congruent network constructed from experimental data and visualize the combined network in a tool that facilitates the knowledge-based analysis of that data. An implementation of this approach, called the Hanalyzer, is demonstrated on a large-scale gene expression array dataset relevant to craniofacial development. The use of the tool was critical in the creation of hypotheses regarding the roles of four genes never previously characterized as involved in craniofacial development; each of these hypotheses was validated by further experimental work.
Collapse
Affiliation(s)
- Sonia M. Leach
- Center for Computational Pharmacology, University of Colorado at Denver, Denver, Colorado, United States of America
| | - Hannah Tipney
- Center for Computational Pharmacology, University of Colorado at Denver, Denver, Colorado, United States of America
| | - Weiguo Feng
- Department of Craniofacial Biology, University of Colorado at Denver, Denver, Colorado, United States of America
| | - William A. Baumgartner
- Center for Computational Pharmacology, University of Colorado at Denver, Denver, Colorado, United States of America
| | - Priyanka Kasliwal
- Center for Computational Pharmacology, University of Colorado at Denver, Denver, Colorado, United States of America
| | - Ronald P. Schuyler
- Center for Computational Pharmacology, University of Colorado at Denver, Denver, Colorado, United States of America
| | - Trevor Williams
- Department of Craniofacial Biology, University of Colorado at Denver, Denver, Colorado, United States of America
| | - Richard A. Spritz
- Human Medical Genetics Program, University of Colorado at Denver, Denver, Colorado, United States of America
| | - Lawrence Hunter
- Center for Computational Pharmacology, University of Colorado at Denver, Denver, Colorado, United States of America
- * E-mail:
| |
Collapse
|
87
|
Rose C. Generating, growing and transforming skeletal shape: insights from amphibian pharyngeal arch cartilages. Bioessays 2009; 31:287-99. [DOI: 10.1002/bies.200800059] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
88
|
Tümpel S, Wiedemann LM, Krumlauf R. Hox genes and segmentation of the vertebrate hindbrain. Curr Top Dev Biol 2009; 88:103-37. [PMID: 19651303 DOI: 10.1016/s0070-2153(09)88004-6] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the vertebrate central nervous system, the hindbrain is an important center for coordinating motor activity, posture, equilibrium, sleep patterns, and essential unconscious functions, such as breathing rhythms and blood circulation. During development, the vertebrate hindbrain depends upon the process of segmentation or compartmentalization to create and organize regional properties essential for orchestrating its highly conserved functional roles. The process of segmentation in the hindbrain differs from that which functions in the paraxial mesoderm to generate somites and the axial skeleton. In the prospective hindbrain, cells in the neural epithelia transiently alter their ability to interact with their neighbors, resulting in the formation of seven lineage-restricted cellular compartments. These different segments or rhombomeres each go on to adopt unique characters in response to environmental signals. The Hox family of transcription factors is coupled to this process. Overlapping or nested patterns of Hox gene expression correlate with segmental domains and provide a combinatorial code and molecular framework for specifying the unique identities of hindbrain segments. The segmental organization and patterns of Hox expression and function are highly conserved among vertebrates and, as a consequence, comparative studies between different species have greatly enhanced our ability to build a picture of the regulatory cascades that control early hindbrain development. The purpose of this chapter is to review what is known about the regulatory mechanisms which establish and maintain Hox gene expression and function in hindbrain development.
Collapse
Affiliation(s)
- Stefan Tümpel
- Stowers Institute for Medical Research, Kansas City, Missouri, USA
| | | | | |
Collapse
|
89
|
Chae SW, Jee BK, Lee JY, Han CW, Jeon YW, Lim Y, Lee KH, Rha HK, Chae GT. HOX gene analysis in the osteogenic differentiation of human mesenchymal stem cells. Genet Mol Biol 2008. [DOI: 10.1590/s1415-47572008005000019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Song Wha Chae
- Neuroscience Genome Research Center, The Catholic University of Korea, Republic of Korea
| | - Bo Keun Jee
- Neuroscience Genome Research Center, The Catholic University of Korea, Republic of Korea
| | - Joo Yong Lee
- Neuroscience Genome Research Center, The Catholic University of Korea, Republic of Korea
| | - Chang Whan Han
- Department of Orthopedic Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Republic of Korea
| | - Yang-Whan Jeon
- Department of Psychiatry, Our Lady of Mercy Hospital, The Catholic University of Korea, Republic of Korea
| | - Young Lim
- Department of Occupational and Environmental Medicine, St. Mary's Hospital, The Catholic University of Korea, Republic of Korea
| | - Kweon-Haeng Lee
- Neuroscience Genome Research Center, The Catholic University of Korea, Republic of Korea
| | - Hyoung Kyun Rha
- Neuroscience Genome Research Center, The Catholic University of Korea, Republic of Korea
| | - Gue-Tae Chae
- Institute of Hansen's Disease, The Catholic University of Korea, Republic of Korea
| |
Collapse
|
90
|
Abstract
The skeleton contains three specific cell types: chondrocytes in cartilage and osteoblasts and osteoclasts in bone. Our understanding of the transcriptional mechanisms that lead to cell differentiation along these three lineages has increased considerably in the past ten years. In the case of chondrocytes and osteoblasts advances have been made possible largely through the molecular elucidation of human skeletal dysplasias. This review discusses the key transcription factors that regulate skeletogenesis and highlights their function, mode of action, and regulation by other factors, with a special emphasis on how human genetics has contributed to this knowledge.
Collapse
Affiliation(s)
- Gerard Karsenty
- Department of Genetics and Development, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA.
| |
Collapse
|
91
|
Bobola N, Engist B. IGFBP5 is a potential regulator of craniofacial skeletogenesis. Genesis 2008; 46:52-9. [DOI: 10.1002/dvg.20360] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
92
|
Matis C, Oury F, Remacle S, Lampe X, Gofflot F, Picard JJ, Rijli FM, Rezsohazy R. Identification of Lmo1 as part of a Hox-dependent regulatory network for hindbrain patterning. Dev Dyn 2007; 236:2675-84. [PMID: 17676642 DOI: 10.1002/dvdy.21266] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The embryonic functions of Hox proteins have been extensively investigated in several animal phyla. These transcription factors act as selectors of developmental programmes, to govern the morphogenesis of multiple structures and organs. However, despite the variety of morphogenetic processes Hox proteins are involved in, only a limited set of their target genes has been identified so far. To find additional targets, we used a strategy based upon the simultaneous overexpression of Hoxa2 and its cofactors Pbx1 and Prep in a cellular model. Among genes whose expression was upregulated, we identified LMO1, which codes for an intertwining LIM-only factor involved in protein-DNA oligomeric complexes. By analysing its expression in Hox knockout mice, we show that Lmo1 is differentially regulated by Hoxa2 and Hoxb2, in specific columns of hindbrain neuronal progenitors. These results suggest that Lmo1 takes part in a Hox paralogue 2-dependent network regulating anteroposterior and dorsoventral hindbrain patterning.
Collapse
Affiliation(s)
- Christelle Matis
- Unit of Developmental Genetics, Université Catholique de Louvain, Brussels, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
93
|
Massip L, Ectors F, Deprez P, Maleki M, Behets C, Lengelé B, Delahaut P, Picard J, Rezsöhazy R. Expression of Hoxa2 in cells entering chondrogenesis impairs overall cartilage development. Differentiation 2007; 75:256-67. [PMID: 17359301 DOI: 10.1111/j.1432-0436.2006.00132.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Vertebrate Hox genes act as developmental architects by patterning embryonic structures like axial skeletal elements, limbs, brainstem territories, or neural crest derivatives. While active during the patterning steps of development, these genes turn out to be down-regulated in specific differentiation programs like that leading to chondrogenesis. To investigate why chondrocyte differentiation is correlated to the silencing of a Hox gene, we generated transgenic mice allowing Cre-mediated conditional misexpression of Hoxa2 and induced this gene in Collagen 2 alpha 1-expressing cells committed to enter chondrogenesis. Persistent Hoxa2 expression in chondrogenic cells resulted in overall chondrodysplasia with delayed cartilage hypertrophy, mineralization, and ossification but without proliferation defects. The absence of skeletal patterning anomaly and the regular migration of precursor cells indicated that the condensation step of chondrogenesis was normal. In contrast, closer examination at the differentiation step showed severely impaired chondrocyte differentiation. In addition, this inhibition affected structures independently of their embryonic origin. In conclusion, for the first time here, by a cell-type specific misexpression, we precisely uncoupled the patterning function of Hoxa2 from its involvement in regulating differentiation programs per se and demonstrate that Hoxa2 displays an anti-chondrogenic activity that is distinct from its patterning function.
Collapse
Affiliation(s)
- Laurent Massip
- Developmental Genetics Unit, Université catholique de Louvain, Brussels, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Flores MV, Lam EYN, Crosier P, Crosier K. A hierarchy of Runx transcription factors modulate the onset of chondrogenesis in craniofacial endochondral bones in zebrafish. Dev Dyn 2007; 235:3166-76. [PMID: 17013873 DOI: 10.1002/dvdy.20957] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The Runx (runt-related) family of transcription factors are important regulators of cell fate decisions in early embryonic development, and in differentiation of tissues including blood, neurons, and bone. During skeletal development in mammals, while only Runx2 is essential for osteoblast differentiation, all family members seem to be involved in chondrogenesis. Runx2 and Runx3 control chondrocyte maturation. Both Runx1 and Runx2 are expressed early in mesenchymal condensations, but how they contribute to the initial stages of chondrocyte differentiation is unclear. Here we show that a hierarchy of Runx transcriptional regulation promotes the early program of chondrocyte differentiation from pre-cartilage mesenchyme in the zebrafish head skeleton. We have previously characterized the zebrafish orthologs for all Runx genes. Zebrafish runx2 is duplicated, but not runx1 or runx3. In the work presented here, we determined the early expression pattern of the runx genes in the craniofacial region. The earliest expression detected was that of runx3 in the pharyngeal endoderm, then runx2a and b in mesenchymal condensations, and later runx1 in the epithelium. Using antisense morpholino knockdown analysis, we examined their respective activities in early chondrogenesis. Depletion of runx2b (but not runx2a) and runx3 severely compromised craniofacial cartilage formation. Because runx2b expression was abolished in Runx3 morphants, we propose that endodermal Runx3 has a role in influencing signaling activities from the endoderm to promote chondrocyte differentiation. We also show that, in contrast to data from mouse studies, zebrafish Runx1 is not required in the initial steps of chondrogenesis leading to endochondral bone formation.
Collapse
Affiliation(s)
- Maria Vega Flores
- Department of Molecular Medicine and Pathology, School of Medical Sciences, The University of Auckland, Auckland, New Zealand
| | | | | | | |
Collapse
|
95
|
Correia AC, Costa M, Moraes F, Bom J, Nóvoa A, Mallo M. Bmp2 is required for migration but not for induction of neural crest cells in the mouse. Dev Dyn 2007; 236:2493-501. [PMID: 17676634 DOI: 10.1002/dvdy.21256] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Bone morphogenetic protein (BMP) signaling is essential for neural crest development in several vertebrates. Genetic experiments in the mouse have shown that Bmp2 is essential for the genesis of migratory neural crest cells. Using several markers and a transgenic reporter approach, we now show that neural crest cells are induced in Bmp2 null mutant embryos, but that these cells fail to migrate out of the neural tube. The absence of migratory neural crest cells in these mutants is not due to their elimination by cell death. The neuroectoderm of Bmp2-/- embryos fail to close and create abnormal folds both along the anterior-posterior and medio-lateral axes, which are associated with an apparent medio-lateral expansion of the neural tube. Finally, our data suggest that the molecular cascade downstream of BMP signaling in early neural crest development may be different in mouse and avian embryos.
Collapse
|
96
|
Abstract
Bone morphogenetic proteins (BMPs) are members of the transforming growth factor (TGF)-beta superfamily of signal molecules that mediate many diverse biological processes ranging from early embryonic tissue patterning to postnatal tissue homeostasis. BMPs trigger cell responses mainly through the canonical signaling pathway where intracellular Smads play central roles in delivering the extracellular signals to the nucleus. While the same Smads are used by BMPs in all types of cells, different transcription factors account in part for the functional diversity of BMPs. These transcription factors are recruited by Smads to regulate the expression of specific subsets of target genes depending on the cell types. Among the transcription factors are Hox proteins. Experimental gain and loss-of-function studies as well as naturally occurring mutations in Hox genes demonstrate their central roles in embryonic skeletal patterning. In addition to the interactions with Smads observed for several Hox proteins, there is also evidence that the expression of a number of Hox genes is regulated by BMPs. It is suggested that Hox proteins play an important role in the BMP pathway.
Collapse
Affiliation(s)
- Xuelin Li
- Department of Pathology, University of Alabama at Birmingham, 1670 University Blvd., VHG003, Birmingham, AL 35294, USA
| | | |
Collapse
|
97
|
Dobreva G, Chahrour M, Dautzenberg M, Chirivella L, Kanzler B, Fariñas I, Karsenty G, Grosschedl R. SATB2 is a multifunctional determinant of craniofacial patterning and osteoblast differentiation. Cell 2006; 125:971-86. [PMID: 16751105 DOI: 10.1016/j.cell.2006.05.012] [Citation(s) in RCA: 408] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2005] [Revised: 03/13/2006] [Accepted: 05/02/2006] [Indexed: 01/04/2023]
Abstract
Vertebrate skeletogenesis involves two processes, skeletal patterning and osteoblast differentiation. Here, we show that Satb2, encoding a nuclear matrix protein, is expressed in branchial arches and in cells of the osteoblast lineage. Satb2-/- mice exhibit both craniofacial abnormalities that resemble those observed in humans carrying a translocation in SATB2 and defects in osteoblast differentiation and function. Multiple osteoblast-specific genes were identified as targets positively regulated by SATB2. In addition, SATB2 was found to repress the expression of several Hox genes including Hoxa2, an inhibitor of bone formation and regulator of branchial arch patterning. Molecular analysis revealed that SATB2 directly interacts with and enhances the activity of both Runx2 and ATF4, transcription factors that regulate osteoblast differentiation. This synergy was genetically confirmed by bone formation defects in Satb2/Runx2 and Satb2/Atf4 double heterozygous mice. Thus, SATB2 acts as a molecular node in a transcriptional network regulating skeletal development and osteoblast differentiation.
Collapse
Affiliation(s)
- Gergana Dobreva
- Max-Planck Institute of Immunobiology, Department of Cellular and Molecular Immunology, 79108 Freiburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
98
|
Baltzinger M, Ori M, Pasqualetti M, Nardi I, Rijli FM. Hoxa2 knockdown in Xenopus results in hyoid to mandibular homeosis. Dev Dyn 2006; 234:858-67. [PMID: 16222714 DOI: 10.1002/dvdy.20567] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The skeletal structures of the face and throat are derived from cranial neural crest cells (NCCs) that migrate from the embryonic neural tube into a series of branchial arches (BAs). The first arch (BA1) gives rise to the upper and lower jaw cartilages, whereas hyoid structures are generated from the second arch (BA2). The Hox paralogue group 2 (PG2) genes, Hoxa2 and Hoxb2, show distinct roles for hyoid patterning in tetrapods and fishes. In the mouse, Hoxa2 acts as a selector of hyoid identity, while its paralogue Hoxb2 is not required. On the contrary, in zebrafish Hoxa2 and Hoxb2 are functionally redundant for hyoid arch patterning. Here, we show that in Xenopus embryos morpholino-induced functional knockdown of Hoxa2 is sufficient to induce homeotic changes of the second arch cartilage. Moreover, Hoxb2 is downregulated in the BA2 of Xenopus embryos, even though initially expressed in second arch NCCs, similar to mouse and unlike in zebrafish. Finally, Xbap, a gene involved in jaw joint formation, is selectively upregulated in the BA2 of Hoxa2 knocked-down frog embryos, supporting a hyoid to mandibular change of NCC identity. Thus, in Xenopus Hoxa2 does not act redundantly with Hoxb2 for BA2 patterning, similar to mouse and unlike in fish. These data bring novel insights into the regulation of Hox PG2 genes and hyoid patterning in vertebrate evolution and suggest that Hoxa2 function is required at late stages of BA2 development.
Collapse
Affiliation(s)
- Mireille Baltzinger
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP, Strasbourg, France
| | | | | | | | | |
Collapse
|
99
|
Abstract
In this issue of Cell, Grosschedl and colleagues (Dobreva et al., 2006) report that the nuclear matrix protein Satb2 represses Hoxa2 expression and acts with other regulatory proteins to promote osteoblast differentiation. This work suggests a molecular mechanism that enables the integration of patterning and differentiation during bone formation.
Collapse
Affiliation(s)
- Debra L Ellies
- Stowers Institute for Medical Research, 1000 50(th) Street, Kansas City, MO 64110, USA
| | | |
Collapse
|
100
|
Perinpanayagam H, Martin T, Mithal V, Dahman M, Marzec N, Lampasso J, Dziak R. Alveolar bone osteoblast differentiation and Runx2/Cbfa1 expression. Arch Oral Biol 2006; 51:406-15. [PMID: 16253204 DOI: 10.1016/j.archoralbio.2005.09.004] [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] [Received: 07/10/2005] [Revised: 09/06/2005] [Accepted: 09/19/2005] [Indexed: 11/19/2022]
Abstract
Alveolar bone cells have a unique origin and functionality, but may resemble skeletal osteoblasts. Osteoblast differentiation and gene expression are regulated by the Runx2/Cbfa1 transcription factor. However, most studies on Runx2/Cbfa1 expression have been on rodent cells and the few studies on human osteoblasts have had differing results. The purpose of this study was to characterize Runx2/Cbfa1 expression in primary cell cultures derived from human alveolar bone. An alveolar bone chip was incubated in alpha-minimum essential medium (alpha-MEM) supplemented with fetal calf serum (10% FCS). Explant cultures were harvested after 3-4 weeks of outgrowth and grown in alpha-MEM with FCS. This media was supplemented with ascorbate, beta-glycerophosphate and dexamethasone to promote osteoblast differentiation over 14 days. RT-PCR analysis and Western blots showed a rapid increase in Runx2/Cbfa1 mRNA (2.1-fold) and protein (2.3-fold) levels in 3 days, followed by a slight decline. There was also a rapid increase in bone sialoprotein expression (2.9-fold) in 3 days, followed by a further increase (3.6-fold) at 14 days. There was a slower increase in alkaline phosphatase expression (1.6-fold) and activity (3.1-fold) over 7 days, followed by a gradual decline. In contrast, collagen mRNA levels showed little change over 14 days. These findings attest to the osteogenic potential of primary cell cultures derived from human alveolar bone. Osteoblastic differentiation in human alveolar bone involves an increase in Runx2/Cbfa1 expression that may be an important component of the differentiation process.
Collapse
Affiliation(s)
- Hiran Perinpanayagam
- School of Dental Medicine, University at Buffalo, 135 Foster Hall, Buffalo, NY 14214-8031, USA.
| | | | | | | | | | | | | |
Collapse
|