151
|
Boulet AM, Capecchi MR. Signaling by FGF4 and FGF8 is required for axial elongation of the mouse embryo. Dev Biol 2012; 371:235-45. [PMID: 22954964 DOI: 10.1016/j.ydbio.2012.08.017] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 08/16/2012] [Accepted: 08/21/2012] [Indexed: 11/16/2022]
Abstract
Fibroblast growth factor (FGF) signaling has been shown to play critical roles in vertebrate segmentation and elongation of the embryonic axis. Neither the exact roles of FGF signaling, nor the identity of the FGF ligands involved in these processes, has been conclusively determined. Fgf8 is required for cell migration away from the primitive streak when gastrulation initiates, but previous studies have shown that drastically reducing the level of FGF8 later in gastrulation has no apparent effect on somitogenesis or elongation of the embryo. In this study, we demonstrate that loss of both Fgf8 and Fgf4 expression during late gastrulation resulted in a dramatic skeletal phenotype. Thoracic vertebrae and ribs had abnormal morphology, lumbar and sacral vertebrae were malformed or completely absent, and no tail vertebrae were present. The expression of Wnt3a in the tail and the amount of nascent mesoderm expressing Brachyury were both severely reduced. Expression of genes in the NOTCH signaling pathway involved in segmentation was significantly affected, and somite formation ceased after the production of about 15-20 somites. Defects seen in the mutants appear to result from a failure to produce sufficient paraxial mesoderm, rather than a failure of mesoderm precursors to migrate away from the primitive streak. Although the epiblast prematurely decreases in size, we did not detect evidence of a change in the proliferation rate of cells in the tail region or excessive apoptosis of epiblast or mesoderm cells. We propose that FGF4 and FGF8 are required to maintain a population of progenitor cells in the epiblast that generates mesoderm and contributes to the stem cell population that is incorporated in the tailbud and required for axial elongation of the mouse embryo after gastrulation.
Collapse
Affiliation(s)
- Anne M Boulet
- Howard Hughes Medical Institute, Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| | | |
Collapse
|
152
|
Singh BN, Doyle MJ, Weaver CV, Koyano-Nakagawa N, Garry DJ. Hedgehog and Wnt coordinate signaling in myogenic progenitors and regulate limb regeneration. Dev Biol 2012; 371:23-34. [PMID: 22902898 DOI: 10.1016/j.ydbio.2012.07.033] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 07/28/2012] [Accepted: 07/30/2012] [Indexed: 11/25/2022]
Abstract
Amphibians have a remarkable capacity for limb regeneration. Following a severe injury, there is complete regeneration with restoration of the patterning and cellular architecture of the amputated limb. While studies have focused on the structural anatomical changes during amphibian limb regeneration, the signaling mechanisms that govern cellular dedifferentiation and blastemal progenitors are unknown. Here, we demonstrate the temporal and spatial requirement for hedgehog (Hh) signaling and its hierarchical correlation with respect to Wnt signaling during newt limb regeneration. While the dedifferentiation process of mature lineages does not depend on Hh signaling, the proliferation and the migration of the dedifferentiated cells are dependent on Hh signaling. Temporally controlled chemical inactivation of the Hh pathway indicates that Hh-mediated antero-posterior (AP) specification occurs early during limb regeneration and that Hh is subsequently required for expansion of the blastemal progenitors. Inhibition of Hh signaling results in G0/G1 arrest with a concomitant reduction in S-phase and G2/M population in myogenic progenitors. Furthermore, Hh inhibition leads to reduced Pax7-positive cells and fewer regenerating fibers relative to control tissue. We demonstrate that activation of Wnt signaling rescues the inhibition of Hh pathway mainly by enhancing proliferative signals, possibly mediated through TCF4 activity. Collectively, our results demonstrate coordinated signaling of Hh and Wnt activities in regulating blastemal progenitors and their hierarchical positioning during limb regeneration.
Collapse
Affiliation(s)
- Bhairab N Singh
- Lillehei Heart Institute, University of Minnesota, 420 Delaware Street, SE. MMC508, Minneapolis, MN 55455, USA
| | | | | | | | | |
Collapse
|
153
|
van der Velden YU, Wang L, van Lohuizen M, Haramis APG. The Polycomb group protein Ring1b is essential for pectoral fin development. Development 2012; 139:2210-20. [PMID: 22619390 DOI: 10.1242/dev.077156] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Polycomb group (PcG) proteins are transcriptional repressors that mediate epigenetic gene silencing by chromatin modification. PcG-mediated gene repression is implicated in development, cell differentiation, stem-cell fate maintenance and cancer. However, analysis of the roles of PcG proteins in orchestrating vertebrate developmental programs in vivo has been hampered by the early embryonic lethality of several PcG gene knockouts in mice. Here, we demonstrate that zebrafish Ring1b, the E3 ligase in Polycomb Repressive Complex 1 (PRC1), is essential for pectoral fin development. We show that differentiation of lateral plate mesoderm (LPM) cells into presumptive pectoral fin precursors is initiated normally in ring1b mutants, but fin bud outgrowth is impaired. Fgf signaling, which is essential for migration, proliferation and cell-fate maintenance during fin development, is not sufficiently activated in ring1b mutants. Exogenous application of FGF4, as well as enhanced stimulation of Fgf signaling by overactivated Wnt signaling in apc mutants, partially restores the fin developmental program. These results reveal that, in the absence of functional Ring1b, fin bud cells fail to execute the pectoral fin developmental program. Together, our results demonstrate that PcG-mediated gene regulation is essential for sustained Fgf signaling in vertebrate limb development.
Collapse
Affiliation(s)
- Yme U van der Velden
- Department of Molecular Genetics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
154
|
Hirschberg RM, Saleh M, Kaiser S, Lierz M, Hafez HM, Bragulla HH. Polymelous Layer Chick Displaying Additional Malformations of the Hind Gut: Case Report and In-Depth Review of Related Literature. Anat Histol Embryol 2012; 41:262-73. [DOI: 10.1111/j.1439-0264.2011.01130.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 12/01/2011] [Indexed: 11/30/2022]
Affiliation(s)
- R. M. Hirschberg
- Institute of Veterinary Anatomy, Faculty of Veterinary Medicine; Freie Universität Berlin; Koserstr. 20; D-14195; Berlin; Germany
| | - M. Saleh
- Institute of Poultry Diseases, Faculty of Veterinary Medicine; Freie Universität Berlin; Berlin; Germany
| | - S. Kaiser
- Fachtierarztpraxis am Erzberg; Braunschweig; Germany
| | - M. Lierz
- Institute of Poultry Diseases, Faculty of Veterinary Medicine; Freie Universität Berlin; Berlin; Germany
| | - H. M. Hafez
- Institute of Poultry Diseases, Faculty of Veterinary Medicine; Freie Universität Berlin; Berlin; Germany
| | - H. H. Bragulla
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine; Louisiana State University; Baton Rouge; LA; USA
| |
Collapse
|
155
|
Trowe MO, Airik R, Weiss AC, Farin HF, Foik AB, Bettenhausen E, Schuster-Gossler K, Taketo MM, Kispert A. Canonical Wnt signaling regulates smooth muscle precursor development in the mouse ureter. Development 2012; 139:3099-108. [PMID: 22833126 DOI: 10.1242/dev.077388] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Smooth muscle cells (SMCs) are a key component of many visceral organs, including the ureter, yet the molecular pathways that regulate their development from mesenchymal precursors are insufficiently understood. Here, we identified epithelial Wnt7b and Wnt9b as possible ligands of Fzd1-mediated β-catenin (Ctnnb1)-dependent (canonical) Wnt signaling in the adjacent undifferentiated ureteric mesenchyme. Mice with a conditional deletion of Ctnnb1 in the ureteric mesenchyme exhibited hydroureter and hydronephrosis at newborn stages due to functional obstruction of the ureter. Histological analysis revealed that the layer of undifferentiated mesenchymal cells directly adjacent to the ureteric epithelium did not undergo characteristic cell shape changes, exhibited reduced proliferation and failed to differentiate into SMCs. Molecular markers for prospective SMCs were lost, whereas markers of the outer layer of the ureteric mesenchyme fated to become adventitial fibroblasts were expanded to the inner layer. Conditional misexpression of a stabilized form of Ctnnb1 in the prospective ureteric mesenchyme resulted in the formation of a large domain of cells that exhibited histological and molecular features of prospective SMCs and differentiated along this lineage. Our analysis suggests that Wnt signals from the ureteric epithelium pattern the ureteric mesenchyme in a radial fashion by suppressing adventitial fibroblast differentiation and initiating smooth muscle precursor development in the innermost layer of mesenchymal cells.
Collapse
Affiliation(s)
- Mark-Oliver Trowe
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
156
|
Staines KA, Macrae VE, Farquharson C. Cartilage development and degeneration: a Wnt Wnt situation. Cell Biochem Funct 2012; 30:633-42. [PMID: 22714865 DOI: 10.1002/cbf.2852] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 04/25/2012] [Accepted: 05/20/2012] [Indexed: 12/27/2022]
Abstract
The Wnt signaling pathway plays a crucial role in the development and homeostasis of a variety of adult tissues and, as such, is emerging as an important therapeutic target for numerous diseases. Factors involved in the Wnt pathway are expressed throughout limb development and chondrogenesis and have been shown to be critical in joint homeostasis and endochondral ossification. Therefore, in this review, we discuss Wnt regulation of chondrogenic differentiation, hypertrophy and cartilage function. Moreover, we detail the role of the Wnt signaling pathway in cartilage degeneration and its potential to act as a target for therapy in osteoarthritis.
Collapse
Affiliation(s)
- Katherine Ann Staines
- The Roslin Institute and Royal-Dick School of Veterinary Studies, The University of Edinburgh, Easter Bush, Midlothian, Scotland.
| | | | | |
Collapse
|
157
|
Rabinowitz AH, Vokes SA. Integration of the transcriptional networks regulating limb morphogenesis. Dev Biol 2012; 368:165-80. [PMID: 22683377 DOI: 10.1016/j.ydbio.2012.05.035] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 05/29/2012] [Accepted: 05/29/2012] [Indexed: 12/29/2022]
Abstract
The developing limb is one of the best described vertebrate systems for understanding how coordinated gene expression during embryogenesis leads to the structures present in the mature organism. This knowledge, derived from decades of research, is largely based upon gain- and loss-of-function experiments. These studies have provided limited information about how the key signaling pathways interact with each other and the downstream effectors of these pathways. We summarize our current understanding of known genetic interactions in the context of three temporally defined gene regulatory networks. These networks crystallize our current knowledge, depicting a dynamic process involving multiple feedback loops between the ectoderm and mesoderm. At the same time, they highlight the fact that many essential processes are still largely undescribed. Much of the dynamic transcriptional activity occurring during development is regulated by distal cis-regulatory elements. Modern genomic tools have provided new approaches for studying the function of cis-regulatory elements and we discuss the results of these studies in regard to understanding limb development. Ultimately, these genomic techniques will allow scientists to understand how multiple signaling pathways are integrated in space and time to drive gene expression and regulate the formation of the limb.
Collapse
Affiliation(s)
- Adam H Rabinowitz
- Section of Molecular Cell & Developmental Biology, Institute for Cellular and Molecular Biology, One University Station A4800, Austin, TX 78712, USA
| | | |
Collapse
|
158
|
Coschi CH, Dick FA. Chromosome instability and deregulated proliferation: an unavoidable duo. Cell Mol Life Sci 2012; 69:2009-24. [PMID: 22223110 PMCID: PMC11114883 DOI: 10.1007/s00018-011-0910-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Revised: 12/15/2011] [Accepted: 12/19/2011] [Indexed: 12/14/2022]
Abstract
The concept that aneuploidy is a characteristic of malignant cells has long been known; however, the idea that aneuploidy is an active contributor to tumorigenesis, as opposed to being an associated phenotype, is more recent in its evolution. At the same time, we are seeing the emergence of novel roles for tumor suppressor genes and oncogenes in genome stability. These include the adenomatous polyposis coli gene (APC), p53, the retinoblastoma susceptibility gene (RB1), and Ras. Originally, many of these genes were thought to be tumor suppressive or oncogenic solely because of their role in proliferative control. Because of the frequency with which they are disrupted in cancer, chromosome instability caused by their dysfunction may be more central to tumorigenesis than previously thought. Therefore, this review will highlight how the proper function of cell cycle regulatory genes contributes to the maintenance of genome stability, and how their mutation in cancer obligatorily connects proliferation and chromosome instability.
Collapse
Affiliation(s)
- Courtney H. Coschi
- London Regional Cancer Program, University of Western Ontario, London, ON Canada
- Department of Biochemistry, University of Western Ontario, London, ON Canada
| | - Frederick A. Dick
- London Regional Cancer Program, University of Western Ontario, London, ON Canada
- Children’s Health Research Institute, University of Western Ontario, London, ON Canada
- Department of Biochemistry, University of Western Ontario, London, ON Canada
- Cancer Research Laboratories, 790 Commissioners Road East, London, ON N6A 4L6 Canada
| |
Collapse
|
159
|
Abstract
In this article, development of articular cartilage and endochondral ossification is reviewed, from the perspective of both morphologic aspects of histogenesis and molecular biology, particularly with respect to key signaling molecules and extracellular matrix components most active in cartilage development. The current understanding of the roles of transforming growth factor β and associated signaling molecules, bone morphogenic proteins, and molecules of the Wnt-β catenin system in chondrogenesis are described. Articular cartilage development is a highly conserved complex biological process that is dynamic and robust in nature, which proceeds well without incident or failure in all joints of most young growing individuals.
Collapse
|
160
|
Lopez-Rios J, Speziale D, Robay D, Scotti M, Osterwalder M, Nusspaumer G, Galli A, Holländer GA, Kmita M, Zeller R. GLI3 constrains digit number by controlling both progenitor proliferation and BMP-dependent exit to chondrogenesis. Dev Cell 2012; 22:837-48. [PMID: 22465667 DOI: 10.1016/j.devcel.2012.01.006] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 11/23/2011] [Accepted: 01/11/2012] [Indexed: 12/11/2022]
Abstract
Inactivation of Gli3, a key component of Hedgehog signaling in vertebrates, results in formation of additional digits (polydactyly) during limb bud development. The analysis of mouse embryos constitutively lacking Gli3 has revealed the essential GLI3 functions in specifying the anteroposterior (AP) limb axis and digit identities. We conditionally inactivated Gli3 during mouse hand plate development, which uncoupled the resulting preaxial polydactyly from known GLI3 functions in establishing AP and digit identities. Our analysis revealed that GLI3 directly restricts the expression of regulators of the G(1)-S cell-cycle transition such as Cdk6 and constrains S phase entry of digit progenitors in the anterior hand plate. Furthermore, GLI3 promotes the exit of proliferating progenitors toward BMP-dependent chondrogenic differentiation by spatiotemporally restricting and terminating the expression of the BMP antagonist Gremlin1. Thus, Gli3 is a negative regulator of the proliferative expansion of digit progenitors and acts as a gatekeeper for the exit to chondrogenic differentiation.
Collapse
Affiliation(s)
- Javier Lopez-Rios
- Developmental Genetics, Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
161
|
Itou J, Kawakami H, Quach T, Osterwalder M, Evans SM, Zeller R, Kawakami Y. Islet1 regulates establishment of the posterior hindlimb field upstream of the Hand2-Shh morphoregulatory gene network in mouse embryos. Development 2012; 139:1620-9. [PMID: 22438573 DOI: 10.1242/dev.073056] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
How divergent genetic systems regulate a common pathway during the development of two serial structures, forelimbs and hindlimbs, is not well understood. Specifically, HAND2 has been shown to regulate Shh directly to initiate its expression in the posterior margin of the limb mesenchyme. Although the Hand2-Shh morphoregulatory system operates in both the forelimb and hindlimb bud, a recent analysis suggested that its upstream regulation is different in the forelimb and hindlimb bud. A combination of all four Hox9 genes is required for Hand2 expression in the forelimb-forming region; however, it remains elusive what genetic system regulates the Hand2-Shh pathway in the hindlimb-forming region. By conditional inactivation of Islet1 in the hindlimb-forming region using the Hoxb6Cre transgene, we show that Islet1 is required for establishing the posterior hindlimb field, but not the forelimb field, upstream of the Hand2-Shh pathway. Inactivation of Islet1 caused the loss of posterior structures in the distal and proximal regions, specifically in the hindlimb. We found that Hand2 expression was downregulated in the hindlimb field and that Shh expression was severely impaired in the hindlimb bud. In the Hoxb6Cre; Islet1 mutant pelvis, the proximal element that is formed in a Shh-independent manner, displayed complementary defects in comparison with Pitx1(-/-) hindlimbs. This suggests that Islet1 and Pitx1 function in parallel during girdle development in hindlimbs, which is in contrast with the known requirement for Tbx5 in girdle development in forelimbs. Our studies have identified a role for Islet1 in hindlimb-specific development and have revealed Islet1 functions in two distinct processes: regulation upstream of the Hand2-Shh pathway and contributions to girdle development.
Collapse
Affiliation(s)
- Junji Itou
- Department of Genetics, Cell Biology and Development, University of Minnesota, 321 Church St. SE. Minneapolis, MN 55455, USA
| | | | | | | | | | | | | |
Collapse
|
162
|
Zhu X, Zhu H, Zhang L, Huang S, Cao J, Ma G, Feng G, He L, Yang Y, Guo X. Wls-mediated Wnts differentially regulate distal limb patterning and tissue morphogenesis. Dev Biol 2012; 365:328-38. [PMID: 22377357 DOI: 10.1016/j.ydbio.2012.02.019] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 02/06/2012] [Accepted: 02/14/2012] [Indexed: 10/28/2022]
Abstract
Wnt proteins are diffusible morphogens that play multiple roles during vertebrate limb development. However, the complexity of Wnt signaling cascades and their overlapping expression prevent us from dissecting their function in limb patterning and tissue morphogenesis. Depletion of the Wntless (Wls) gene, which is required for the secretion of various Wnts, makes it possible to genetically dissect the overall effect of Wnts in limb development. In this study, the Wls gene was conditionally depleted in limb mesenchyme and ectoderm. The loss of mesenchymal Wls prevented the differentiation of distal mesenchyme and arrested limb outgrowth, most likely by affecting Wnt5a function. Meanwhile, the deletion of ectodermal Wls resulted in agenesis of distal limb tissue and premature regression of the distal mesenchyme. These observations suggested that Wnts from the two germ layers differentially regulate the pool of undifferentiated distal limb mesenchyme cells. Cellular behavior analysis revealed that ectodermal Wnts sustain mesenchymal cell proliferation and survival in a manner distinct from Fgf. Ectodermal Wnts were also shown for the first time to be essential for distal tendon/ligament induction, myoblast migration and dermis formation in the limb. These findings provide a comprehensive view of the role of Wnts in limb patterning and tissue morphogenesis.
Collapse
Affiliation(s)
- Xuming Zhu
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
163
|
Dexheimer V, Frank S, Richter W. Proliferation as a requirement for in vitro chondrogenesis of human mesenchymal stem cells. Stem Cells Dev 2012; 21:2160-9. [PMID: 22229819 DOI: 10.1089/scd.2011.0670] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
During embryonic cartilage development, proliferation and differentiation are tightly linked with a transient cell cycle arrest observed during determination and before main extracellular matrix production. Aim of this study was to address whether these steps are imitated during in vitro differentiation of mesenchymal stem cells (MSCs) and are crucial for a proper chondrogenesis. Human MSCs were expanded in distinct media and subjected to pellet culture in chondrogenic medium. Cells were labeled with 5-iodo-2'-deoxyuridin (IdU) or treated with mitomycin C at various time points during culture. Apoptosis was detected by cleaved caspase 3. Proliferation rate of expanded MSCs at start of pellet culture showed a positive correlation with chondrogenesis according to DNA content, proteoglycan deposition, collagen type II content, and final pellet size. Evenly distributed IdU signals at day 1 diminished and became restricted primarily to the periphery by day 3. Between days 10 and 21, IdU-positive cells were detected throughout coinciding with collagen type II positivity. Little IdU incorporation occurred after day 21 and in areas of strong matrix deposition. DNA content decreased and apoptosis was detected up to day 14. Irreversible growth arrest by mitomycin C fully blocked chondrogenic differentiation and seemed to arrest differentiation at the stage reached at treatment. In conclusion, chondrogenesis involved a transient proliferation phase appearing simultaneously with start of collagen type II deposition and growth was crucial for proper chondrogenesis. Growth and differentiation steps, thus, seemed closely coordinated and resembled, with respect to proliferation, stages known from embryonic cartilage development. Stimulation of proliferation and prevention of early apoptosis are attractive goals to further improve MSC chondrogenesis.
Collapse
Affiliation(s)
- Verena Dexheimer
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | | | | |
Collapse
|
164
|
Miclea RL, Siebelt M, Finos L, Goeman JJ, Löwik CWGM, Oostdijk W, Weinans H, Wit JM, Robanus-Maandag EC, Karperien M. Inhibition of Gsk3β in cartilage induces osteoarthritic features through activation of the canonical Wnt signaling pathway. Osteoarthritis Cartilage 2011; 19:1363-72. [PMID: 21911068 DOI: 10.1016/j.joca.2011.07.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2010] [Revised: 07/24/2011] [Accepted: 07/29/2011] [Indexed: 02/02/2023]
Abstract
OBJECTIVE In the past years, the canonical Wnt/β-catenin signaling pathway has emerged as a critical regulator of cartilage development and homeostasis. In this pathway, glycogen synthase kinase-3β (GSK3β) down-regulates transduction of the canonical Wnt signal by promoting degradation of β-catenin. In this study we wanted to further investigate the role of Gsk3β in cartilage maintenance. DESIGN Therefore, we have treated chondrocytes ex vivo and in vivo with GIN, a selective GSK3β inhibitor. RESULTS In E17.5 fetal mouse metatarsals, GIN treatment resulted in loss of expression of cartilage markers and decreased chondrocyte proliferation from day 1 onward. Late (3 days) effects of GIN included cartilage matrix degradation and increased apoptosis. Prolonged (7 days) GIN treatment resulted in resorption of the metatarsal. These changes were confirmed by microarray analysis showing a decrease in expression of typical chondrocyte markers and induction of expression of proteinases involved in cartilage matrix degradation. An intra-articular injection of GIN in rat knee joints induced nuclear accumulation of β-catenin in chondrocytes 72 h later. Three intra-articular GIN injections with a 2 days interval were associated with surface fibrillation, a decrease in glycosaminoglycan expression and chondrocyte hypocellularity 6 weeks later. CONCLUSIONS These results suggest that, by down-regulating β-catenin, Gsk3β preserves the chondrocytic phenotype, and is involved in maintenance of the cartilage extracellular matrix. Short term β-catenin up-regulation in cartilage secondary to Gsk3β inhibition may be sufficient to induce osteoarthritis-like features in vivo.
Collapse
Affiliation(s)
- R L Miclea
- Department of Pediatrics, Leiden University Medical Centre, Leiden, Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
165
|
Stamper BD, Park SS, Beyer RP, Bammler TK, Farin FM, Mecham B, Cunningham ML. Differential expression of extracellular matrix-mediated pathways in single-suture craniosynostosis. PLoS One 2011; 6:e26557. [PMID: 22028906 PMCID: PMC3197523 DOI: 10.1371/journal.pone.0026557] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 09/28/2011] [Indexed: 01/17/2023] Open
Abstract
Craniosynostosis is a disease defined by premature fusion of one or more cranial sutures. The mechanistic pathology of single-suture craniosynostosis is complex and while a number of genetic biomarkers and environmental predispositions have been identified, in many cases the causes remain controversial and inconclusive. In this study, gene expression data from 199 patients with isolated sagittal (n = 100), unilateral coronal (n = 50), and metopic (n = 49) synostosis are compared against both a control population (n = 50), as well as each other. After controlling for variables contributing to potential bias, FGF7, SFRP4, and VCAM1 emerged as genes associated with single-suture craniosynostosis due to their significantly large changes in gene expression compared to the control population. Pathway analysis implicated focal adhesion and extracellular matrix (ECM)-receptor interaction as differentially regulated gene networks when comparing all cases of single-suture synostosis and controls. Lastly, overall gene expression was found to be highly conserved between coronal and metopic cases, as evidenced by the fact that WNT2 and IGFBP2 were the only genes differentially regulated to a significantly large extent in a direct comparison. The identification of genes and gene networks associated with Fgf/Igf/Wnt signaling and ECM-mediated focal adhesion not only support the involvement of biomarkers previously reported to be related to craniosynostosis, but also introduce novel transcripts and pathways that may play critical roles in its pathogenesis.
Collapse
Affiliation(s)
- Brendan D Stamper
- Center for Tissue and Cell Sciences, Seattle Children's Research Institute, Seattle, Washington, United States of America.
| | | | | | | | | | | | | |
Collapse
|
166
|
Nelson ER, Levi B, Sorkin M, James AW, Liu KJ, Quarto N, Longaker MT. Role of GSK-3β in the osteogenic differentiation of palatal mesenchyme. PLoS One 2011; 6:e25847. [PMID: 22022457 PMCID: PMC3194817 DOI: 10.1371/journal.pone.0025847] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 09/12/2011] [Indexed: 01/01/2023] Open
Abstract
Introduction The function of Glycogen Synthase Kinases 3β (GSK-3β) has previously been shown to be necessary for normal secondary palate development. Using GSK-3ß null mouse embryos, we examine the potential coordinate roles of Wnt and Hedgehog signaling on palatal ossification. Methods Palates were harvested from GSK-3β, embryonic days 15.0–18.5 (e15.0–e18.5), and e15.5 Indian Hedgehog (Ihh) null embryos, and their wild-type littermates. The phenotype of GSK-3β null embryos was analyzed with skeletal whole mount and pentachrome stains. Spatiotemporal regulation of osteogenic gene expression, in addition to Wnt and Hedgehog signaling activity, were examined in vivo on GSK-3β and Ihh +/+ and −/− e15.5 embryos using in situ hybridization and immunohistochemistry. To corroborate these results, expression of the same molecular targets were assessed by qRT-PCR of e15.5 palates, or e13.5 palate cultures treated with both Wnt and Hedgehog agonists and anatagonists. Results GSK-3β null embryos displayed a 48 percent decrease (*p<0.05) in palatine bone formation compared to wild-type littermates. GSK-3β null embryos also exhibited decreased osteogenic gene expression that was associated with increased Wnt and decreased Hedgehog signaling. e13.5 palate culture studies demonstrated that Wnt signaling negatively regulates both osteogenic gene expression and Hedgehog signaling activity, while inhibition of Wnt signaling augments both osteogenic gene expression and Hedgehog signaling activity. In addition, no differences in Wnt signaling activity were noted in Ihh null embryos, suggesting that canonical Wnt may be upstream of Hedgehog in secondary palate development. Lastly, we found that GSK-3β −/− palate cultures were “rescued” with the Wnt inhibitor, Dkk-1. Conclusions Here, we identify a critical role for GSK-3β in palatogenesis through its direct regulation of canonical Wnt signaling. These findings shed light on critical developmental pathways involved in palatogenesis and may lead to novel molecular targets to prevent cleft palate formation.
Collapse
Affiliation(s)
- Emily R. Nelson
- Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Benjamin Levi
- Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Michael Sorkin
- Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Aaron W. James
- Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Karen J. Liu
- Department of Craniofacial Development, King's College London, London, United Kingdom
| | - Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Dipartimento di Scienze Chirurgiche, Anestesiologiche-Rianimatorie e dell ‘Emergenza “Giuseppe Zannini,” Universita’ degli Studi di Napoli Federico II, Napoli, Italy
| | - Michael T. Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
167
|
Pedersen L, Jensen MH, Krishna S. Dickkopf1--a new player in modelling the Wnt pathway. PLoS One 2011; 6:e25550. [PMID: 22022411 PMCID: PMC3192063 DOI: 10.1371/journal.pone.0025550] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Accepted: 09/07/2011] [Indexed: 01/22/2023] Open
Abstract
The Wnt signaling pathway transducing the stabilization of β-catenin is essential for metazoan embryo development and is misregulated in many diseases such as cancers. In recent years models have been proposed for the Wnt signaling pathway during the segmentation process in developing embryos. Many of these include negative feedback loops where Axin2 plays a key role. However, Axin2 null mice show no segmentation phenotype. We therefore propose a new model where the negative feedback involves Dkk1 rather than Axin2. We show that this model can exhibit the same type of oscillations as the previous models with Axin2 and as observed in experiments. We show that a spatial Wnt gradient can consistently convert this temporal periodicity into the spatial periodicity of somites, provided the oscillations in new cells arising in the presomitic mesoderm are synchronized with the oscillations of older cells. We further investigate the hypothesis that a change in the Wnt level in the tail bud during the later stages of somitogenesis can lengthen the time period of the oscillations and hence the size and separation of the later somites.
Collapse
Affiliation(s)
- Lykke Pedersen
- Center for Models of Life, Niels Bohr Institute, Copenhagen, Denmark.
| | | | | |
Collapse
|
168
|
Kapeli K, Hurlin PJ. Differential regulation of N-Myc and c-Myc synthesis, degradation, and transcriptional activity by the Ras/mitogen-activated protein kinase pathway. J Biol Chem 2011; 286:38498-38508. [PMID: 21908617 DOI: 10.1074/jbc.m111.276675] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Myc transcription factors are important regulators of proliferation and can promote oncogenesis when deregulated. Deregulated Myc expression in cancers can result from MYC gene amplification and translocation but also from alterations in mitogenic signaling pathways that affect Myc levels through both transcriptional and post-transcription mechanisms. For example, mutations in Ras family GTPase proteins that cause their constitutive activation can increase cellular levels of c-Myc by interfering with its rapid proteasomal degradation. Although enhanced protein stability is generally thought to be applicable to other Myc family members, here we show that c-Myc and its paralog N-Myc respond to oncogenic H-Ras (H-Ras(G12V)) in very different ways. H-Ras(G12V) promotes accumulation of both c-Myc and N-Myc, but although c-Myc accumulation is achieved by enhanced protein stability, N-Myc accumulation is associated with an accelerated rate of translation that overcomes a surprising H-Ras(G12V)-mediated destabilization of N-Myc. We show that H-Ras(G12V)-mediated degradation of N-Myc functions independently of key phosphorylation sites in the highly conserved Myc homology box I region that controls c-Myc protein stability by oncogenic Ras. Finally, we found that N-Myc and c-Myc transcriptional activity is associated with their proteasomal degradation but that N-Myc may be uniquely dependent on Ras-stimulated proteolysis for target gene expression. Taken together, these studies provide mechanistic insight into how oncogenic Ras augments N-Myc levels in cells and suggest that enhanced N-Myc translation and degradation-coupled transactivation may contribute to oncogenesis.
Collapse
Affiliation(s)
- Katannya Kapeli
- Department of Cell and Developmental Biology, Oregon Health and Science University, Portland, Oregon 97239
| | - Peter J Hurlin
- Department of Cell and Developmental Biology, Oregon Health and Science University, Portland, Oregon 97239; Shriners Hospital for Children, Portland, Oregon 97239.
| |
Collapse
|
169
|
Abstract
Wnt signaling pathways control lineage specification in vertebrate embryos and regulate pluripotency in embryonic stem (ES) cells, but how the balance between progenitor self-renewal and differentiation is achieved during axis specification and tissue patterning remains highly controversial. The context- and stage-specific effects of the different Wnt pathways produce complex and sometimes opposite outcomes that help to generate embryonic cell diversity. Although the results of recent studies of the Wnt/β-catenin pathway in ES cells appear to be surprising and controversial, they converge on the same conserved mechanism that leads to the inactivation of TCF3-mediated repression.
Collapse
Affiliation(s)
- Sergei Y Sokol
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA.
| |
Collapse
|
170
|
Chimal-Monroy J, Abarca-Buis RF, Cuervo R, Díaz-Hernández M, Bustamante M, Rios-Flores JA, Romero-Suárez S, Farrera-Hernández A. Molecular control of cell differentiation and programmed cell death during digit development. IUBMB Life 2011; 63:922-9. [PMID: 21901820 DOI: 10.1002/iub.563] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 07/18/2011] [Indexed: 12/14/2022]
Abstract
During the hand plate development, the processes of cell differentiation and control of cell death are relevant to ensure a correct shape of the limb. The progenitor cell pool that later will differentiate into cartilage to form the digits arises from undifferentiated mesenchymal cells beneath the apical ectodermal ridge (AER). Once these cells abandon the area of influence of signals from AER and ectoderm, some cells are committed to chondrocyte lineage forming the digital rays. However, if the cells are not committed to chondrocyte lineage, they will form the prospective interdigits that in species with free digits will subsequently die. In this work, we provide the overview of the molecular interactions between different signaling pathways responsible for the formation of digit and interdigit regions. In addition, we briefly describe some experiments concerning the most important signals responsible for promoting cell death. Finally, on the basis that the interdigital tissue has chondrogenic potential, we discuss the hypothesis that apoptotic-promoting signals might also act as antichondrogenic factors and chondrogenic factors might operate as anti-apoptotic factors.
Collapse
Affiliation(s)
- Jesús Chimal-Monroy
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México Ciudad Universitaria. Apartado Postal 70228. México.
| | | | | | | | | | | | | | | |
Collapse
|
171
|
MiR-140 is co-expressed with Wwp2-C transcript and activated by Sox9 to target Sp1 in maintaining the chondrocyte proliferation. FEBS Lett 2011; 585:2992-7. [PMID: 21872590 DOI: 10.1016/j.febslet.2011.08.013] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Revised: 08/08/2011] [Accepted: 08/09/2011] [Indexed: 12/21/2022]
Abstract
MiR-140 is a microRNA specially involved in chondrogenesis and osteoarthritis pathogenesis. However, its transcriptional regulation and target genes in cartilage development are not fully understood. Here we detected that miR-140 was uniquely expressed in chondrocyte and suppressed by Wnt/β-catenin signalling. The miR-140 primary transcript was an intron-retained RNA co-expressed with Wwp2-C isoform, which was directly induced by Sox9 through binding to the intron 10 of Wwp2 gene. Knockdown of miR-140 in limb bud micromass cultures resulted in arrest of chondrogenic proliferation. Sp1, the activator of the cell cycle regulator p15(INK4b), was identified as a target of miR-140 in maintaining the chondrocyte proliferation. Collectively, our findings expand our understanding of the transcriptional regulation and the chondrogenic role of miR-140 in chondrogenesis.
Collapse
|
172
|
Pluripotency factors in embryonic stem cells regulate differentiation into germ layers. Cell 2011; 145:875-89. [PMID: 21663792 DOI: 10.1016/j.cell.2011.05.017] [Citation(s) in RCA: 419] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 03/03/2011] [Accepted: 05/16/2011] [Indexed: 02/06/2023]
Abstract
Cell fate decisions are fundamental for development, but we do not know how transcriptional networks reorganize during the transition from a pluripotent to a differentiated cell state. Here, we asked how mouse embryonic stem cells (ESCs) leave the pluripotent state and choose between germ layer fates. By analyzing the dynamics of the transcriptional circuit that maintains pluripotency, we found that Oct4 and Sox2, proteins that maintain ESC identity, also orchestrate germ layer fate selection. Oct4 suppresses neural ectodermal differentiation and promotes mesendodermal differentiation; Sox2 inhibits mesendodermal differentiation and promotes neural ectodermal differentiation. Differentiation signals continuously and asymmetrically modulate Oct4 and Sox2 protein levels, altering their binding pattern in the genome, and leading to cell fate choice. The same factors that maintain pluripotency thus also integrate external signals and control lineage selection. Our study provides a framework for understanding how complex transcription factor networks control cell fate decisions in progenitor cells.
Collapse
|
173
|
Zhang J, Tu Q, Bonewald LF, He X, Stein G, Lian J, Chen J. Effects of miR-335-5p in modulating osteogenic differentiation by specifically downregulating Wnt antagonist DKK1. J Bone Miner Res 2011; 26:1953-63. [PMID: 21351149 PMCID: PMC3810406 DOI: 10.1002/jbmr.377] [Citation(s) in RCA: 232] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Dickkopf-related protein 1 (DKK1) is essential to maintain skeletal homeostasis as an inhibitor of Wnt signaling and osteogenic differentiation. The purpose of this study was to investigate the molecular mechanisms underlying the developmental stage-specific regulation of the DKK1 protein level. We performed a series of studies including luciferase reporter assays, micro-RNA microarray, site-specific mutations, and gain- and loss-of-function analyses. We found that the DKK1 protein level was regulated via DKK1 3' UTR by miRNA control, which was restricted to osteoblast-lineage cells. As a result of decreased DKK1 protein level by miR-335-5p, Wnt signaling was enhanced, as indicated by elevated GSK-3β phosphorylation and increased β-catenin transcriptional activity. The effects of miR-335-5p were reversed by anti-miR-335-5p treatment, which downregulated endogenous miR-335-5p. In vivo studies showed high expression levels of miR-335-5p in osteoblasts and hypertrophic chondrocytes of mouse embryos, indicating a pivotal role of miR-335-5p in regulating bone development. In conclusion, miR-335-5p activates Wnt signaling and promotes osteogenic differentiation by downregulating DKK1. This cell- and development-specific regulation is essential and mandatory for the initiation and progression of osteogenic differentiation. miR-335-5p proves to be a potential and useful targeting molecule for promoting bone formation and regeneration.
Collapse
Affiliation(s)
- Jin Zhang
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| | | | | | | | | | | | | |
Collapse
|
174
|
Abstract
Salamander limb regeneration is a classical model of tissue morphogenesis and patterning. Through recent advances in cell labeling and molecular analysis, a more precise, mechanistic understanding of this process has started to emerge. Long-standing questions include to what extent limb regeneration recapitulates the events observed in mammalian limb development and to what extent are adult- or salamander- specific aspects deployed. Historically, researchers studying limb development and limb regeneration have proposed different models of pattern formation. Here we discuss recent data on limb regeneration and limb development to argue that although patterning mechanisms are likely to be similar, cell plasticity and signaling from nerves play regeneration-specific roles.
Collapse
Affiliation(s)
- Eugen Nacu
- DFG-Center for Regenerative Therapies Dresden, Germany.
| | | |
Collapse
|
175
|
Roselló-Díez A, Ros MA, Torres M. Diffusible signals, not autonomous mechanisms, determine the main proximodistal limb subdivision. Science 2011; 332:1086-8. [PMID: 21617076 DOI: 10.1126/science.1199489] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Vertebrate limbs develop three main proximodistal (PD) segments (upper arm, forearm, and hand) in a proximal-to-distal sequence. Despite extensive research into limb development, whether PD specification occurs through nonautonomous or autonomous mechanisms is not resolved. Heterotopic transplantation of intact and recombinant chicken limb buds identifies signals in the embryo trunk that proximalize distal limb cells to generate a complete PD axis. In these transplants, retinoic acid induces proximalization, which is counteracted by fibroblast growth factors from the distal limb bud; these related actions suggest that the first limb-bud PD regionalization results from the balance between proximal and distal signals. The plasticity of limb progenitor cell identity in response to diffusible signals provides a unifying view of PD patterning during vertebrate limb development and regeneration.
Collapse
Affiliation(s)
- Alberto Roselló-Díez
- Departamento de Desarrollo y Reparación Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares, Instituto de Salud Carlos III, c/ Melchor Fernández Almagro, 3, E-28029 Madrid, Spain
| | | | | |
Collapse
|
176
|
Cooper KL, Kuang-Hsien Hu J, Berge DT, Fernandez-Teran M, Ros MA, Tabin CJ. Initiation of proximal-distal patterning in the vertebrate limb by signals and growth. Science 2011; 332:1083-6. [PMID: 21617075 PMCID: PMC3258580 DOI: 10.1126/science.1199499] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Two broad classes of models have been proposed to explain the patterning of the proximal-distal axis of the vertebrate limb (from the shoulder to the digit tips). Differentiating between them, we demonstrate that early limb mesenchyme in the chick is initially maintained in a state capable of generating all limb segments through exposure to a combination of proximal and distal signals. As the limb bud grows, the proximal limb is established through continued exposure to flank-derived signal(s), whereas the developmental program determining the medial and distal segments is initiated in domains that grow beyond proximal influence. In addition, the system we have developed, combining in vitro and in vivo culture, opens the door to a new level of analysis of patterning mechanisms in the limb.
Collapse
Affiliation(s)
- Kimberly L. Cooper
- Harvard Medical School, Department of Genetics, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Jimmy Kuang-Hsien Hu
- Harvard Medical School, Department of Genetics, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Derk ten Berge
- Erasmus Stem Cell Institute, Department of Cell Biology, Erasmus Medical Center, 3000 CA Rotterdam, Netherlands
| | - Marian Fernandez-Teran
- Universidad de Cantabria Instituto de Biomedicina y Biotecnologia (CSIC-UC-IDICAN), C/ Herrera Oria s/n, E-39011 Santander, Spain
| | - Maria A. Ros
- Universidad de Cantabria Instituto de Biomedicina y Biotecnologia (CSIC-UC-IDICAN), C/ Herrera Oria s/n, E-39011 Santander, Spain
| | - Clifford J. Tabin
- Harvard Medical School, Department of Genetics, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| |
Collapse
|
177
|
|
178
|
Guerrini L, Costanzo A, Merlo GR. A symphony of regulations centered on p63 to control development of ectoderm-derived structures. J Biomed Biotechnol 2011; 2011:864904. [PMID: 21716671 PMCID: PMC3118300 DOI: 10.1155/2011/864904] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Revised: 01/25/2011] [Accepted: 03/16/2011] [Indexed: 12/27/2022] Open
Abstract
The p53-related transcription factor p63 is critically important for basic cellular functions during development of the ectoderm and derived structure and tissues, including skin, limb, palate, and hair. On the one side, p63 is required to sustain the proliferation of keratinocyte progenitors, while on the other side it is required for cell stratification, commitment to differentiate, cell adhesion, and epithelial-mesenchymal signaling. Molecules that are components or regulators of the p63 pathway(s) are rapidly being identified, and it comes with no surprise that alterations in the p63 pathway lead to congenital conditions in which the skin and other ectoderm-derived structures are affected. In this paper, we summarize the current knowledge of the molecular and cellular regulations centered on p63, derived from the comprehension of p63-linked human diseases and the corresponding animal models, as well as from cellular models and high-throughput molecular approaches. We point out common themes and features, that allow to speculate on the possible role of p63 downstream events and their potential exploitation in future attempts to correct the congenital defect in preclinical studies.
Collapse
Affiliation(s)
- Luisa Guerrini
- Department of Biomolecular Science and Biotechnology, University of Milan, 20133 Milano, Italy
| | - Antonio Costanzo
- Department of Dermatology, University of Rome “Tor Vergata”, 00133 Rome, Italy
- Rome Oncogenomic Centre, Via Elio Chianesi 53, 00144 Rome, Italy
| | - Giorgio R. Merlo
- Molecular Biotechnology Center, Dulbecco Telethon Institute, University of Torino, 10126 Torino, Italy
| |
Collapse
|
179
|
Southgate L, Machado R, Snape K, Primeau M, Dafou D, Ruddy D, Branney P, Fisher M, Lee G, Simpson M, He Y, Bradshaw T, Blaumeiser B, Winship W, Reardon W, Maher E, FitzPatrick D, Wuyts W, Zenker M, Lamarche-Vane N, Trembath R. Gain-of-function mutations of ARHGAP31, a Cdc42/Rac1 GTPase regulator, cause syndromic cutis aplasia and limb anomalies. Am J Hum Genet 2011; 88:574-85. [PMID: 21565291 PMCID: PMC3146732 DOI: 10.1016/j.ajhg.2011.04.013] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 04/19/2011] [Accepted: 04/20/2011] [Indexed: 12/21/2022] Open
Abstract
Regulation of cell proliferation and motility is essential for normal development. The Rho family of GTPases plays a critical role in the control of cell polarity and migration by effecting the cytoskeleton, membrane trafficking, and cell adhesion. We investigated a recognized developmental disorder, Adams-Oliver syndrome (AOS), characterized by the combination of aplasia cutis congenita (ACC) and terminal transverse limb defects (TTLD). Through a genome-wide linkage analysis, we detected a locus for autosomal-dominant ACC-TTLD on 3q generating a maximum LOD score of 4.93 at marker rs1464311. Candidate-gene- and exome-based sequencing led to the identification of independent premature truncating mutations in the terminal exon of the Rho GTPase-activating protein 31 gene, ARHGAP31, which encodes a Cdc42/Rac1 regulatory protein. Mutant transcripts are stable and increase ARHGAP31 activity in vitro through a gain-of-function mechanism. Constitutively active ARHGAP31 mutations result in a loss of available active Cdc42 and consequently disrupt actin cytoskeletal structures. Arhgap31 expression in the mouse is substantially restricted to the terminal limb buds and craniofacial processes during early development; these locations closely mirror the sites of impaired organogenesis that characterize this syndrome. These data identify the requirement for regulated Cdc42 and/or Rac1 signaling processes during early human development.
Collapse
Affiliation(s)
- Laura Southgate
- Department of Medical and Molecular Genetics, King's College London, School of Medicine, Guy's Hospital, London, London SE1 9RT, UK
| | - Rajiv D. Machado
- Department of Medical and Molecular Genetics, King's College London, School of Medicine, Guy's Hospital, London, London SE1 9RT, UK
| | - Katie M. Snape
- Department of Medical and Molecular Genetics, King's College London, School of Medicine, Guy's Hospital, London, London SE1 9RT, UK
| | - Martin Primeau
- Department of Anatomy and Cell Biology, McGill University, 3640 University Street, Montreal, Quebec H3A 2B2, Canada
| | - Dimitra Dafou
- Department of Medical and Molecular Genetics, King's College London, School of Medicine, Guy's Hospital, London, London SE1 9RT, UK
| | - Deborah M. Ruddy
- Department of Clinical Genetics, Guy's Hospital, London SE1 9RT, UK
| | - Peter A. Branney
- Medical Research Council (MRC) Human Genetics Unit, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Malcolm Fisher
- Medical Research Council (MRC) Human Genetics Unit, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Grace J. Lee
- Department of Medical and Molecular Genetics, King's College London, School of Medicine, Guy's Hospital, London, London SE1 9RT, UK
| | - Michael A. Simpson
- Department of Medical and Molecular Genetics, King's College London, School of Medicine, Guy's Hospital, London, London SE1 9RT, UK
| | - Yi He
- Department of Anatomy and Cell Biology, McGill University, 3640 University Street, Montreal, Quebec H3A 2B2, Canada
| | - Teisha Y. Bradshaw
- Department of Medical and Molecular Genetics, King's College London, School of Medicine, Guy's Hospital, London, London SE1 9RT, UK
| | - Bettina Blaumeiser
- Department of Medical Genetics, University and University Hospital of Antwerp, Prins Boudewijnlaan 43, 2650 Edegem, Belgium
| | - William S. Winship
- Nelson R. Mandela School of Medicine, Faculty of Health Sciences, Department of Paediatrics and Child Health, University of KwaZulu-Natal, Durban 4041, South Africa
| | - Willie Reardon
- National Centre for Medical Genetics, Our Lady's Hospital for Sick Children, Crumlin, Dublin 12, Ireland
| | - Eamonn R. Maher
- Medical and Molecular Genetics, School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, Institute of Biomedical Research, University of Birmingham, Birmingham B15 2TT, UK
- West Midlands Regional Genetics Service, Birmingham Women's Hospital, Birmingham B15 2TG, UK
| | - David R. FitzPatrick
- Medical Research Council (MRC) Human Genetics Unit, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Wim Wuyts
- Department of Medical Genetics, University and University Hospital of Antwerp, Prins Boudewijnlaan 43, 2650 Edegem, Belgium
| | - Martin Zenker
- Institute of Human Genetics, University Hospital Erlangen, University of Erlangen-Nuremberg, Schwabachanlage 10, 91054 Erlangen, Germany
- Institute of Human Genetics, University Hospital Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Nathalie Lamarche-Vane
- Department of Anatomy and Cell Biology, McGill University, 3640 University Street, Montreal, Quebec H3A 2B2, Canada
| | - Richard C. Trembath
- Department of Medical and Molecular Genetics, King's College London, School of Medicine, Guy's Hospital, London, London SE1 9RT, UK
- Department of Clinical Genetics, Guy's Hospital, London SE1 9RT, UK
| |
Collapse
|
180
|
Jelinsky SA, Rodeo SA, Li J, Gulotta LV, Archambault JM, Seeherman HJ. Regulation of gene expression in human tendinopathy. BMC Musculoskelet Disord 2011; 12:86. [PMID: 21539748 PMCID: PMC3095578 DOI: 10.1186/1471-2474-12-86] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 05/03/2011] [Indexed: 11/16/2022] Open
Abstract
Background Chronic tendon injuries, also known as tendinopathies, are common among professional and recreational athletes. These injuries result in a significant amount of morbidity and health care expenditure, yet little is known about the molecular mechanisms leading to tendinopathy. Methods We have used histological evaluation and molecular profiling to determine gene expression changes in 23 human patients undergoing surgical procedures for the treatment of chronic tendinopathy. Results Diseased tendons exhibit altered extracellular matrix, fiber disorientation, increased cellular content and vasculature, and the absence of inflammatory cells. Global gene expression profiling identified 983 transcripts with significantly different expression patterns in the diseased tendons. Global pathway analysis further suggested altered expression of extracellular matrix proteins and the lack of an appreciable inflammatory response. Conclusions Identification of the pathways and genes that are differentially regulated in tendinopathy samples will contribute to our understanding of the disease and the development of novel therapeutics.
Collapse
Affiliation(s)
- Scott A Jelinsky
- Tissue Repair, Pfizer Research, 200 Cambridge Park Drive, Cambridge, MA 02140, USA.
| | | | | | | | | | | |
Collapse
|
181
|
Sequential and coordinated actions of c-Myc and N-Myc control appendicular skeletal development. PLoS One 2011; 6:e18795. [PMID: 21494559 PMCID: PMC3073980 DOI: 10.1371/journal.pone.0018795] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Accepted: 03/18/2011] [Indexed: 12/25/2022] Open
Abstract
Background During limb development, chondrocytes and osteoblasts emerge from
condensations of limb bud mesenchyme. These cells then proliferate and
differentiate in separate but adjacent compartments and function
cooperatively to promote bone growth through the process of endochondral
ossification. While many aspects of limb skeletal formation are understood,
little is known about the mechanisms that link the development of
undifferentiated limb bud mesenchyme with formation of the precartilaginous
condensation and subsequent proliferative expansion of chondrocyte and
osteoblast lineages. The aim of this study was to gain insight into these
processes by examining the roles of c-Myc and N-Myc in morphogenesis of the
limb skeleton. Methodology/Principal Findings To investigate c-Myc function in skeletal development, we characterized mice
in which floxed c-Myc alleles were deleted in undifferentiated limb bud
mesenchyme with Prx1-Cre, in chondro-osteoprogenitors with
Sox9-Cre and in osteoblasts with
Osx1-Cre. We show that c-Myc promotes the proliferative
expansion of both chondrocytes and osteoblasts and as a consequence controls
the process of endochondral growth and ossification and determines bone
size. The control of proliferation by c-Myc was related to its effects on
global gene transcription, as phosphorylation of the C-Terminal Domain
(pCTD) of RNA Polymerase II, a marker of general transcription initiation,
was tightly coupled to cell proliferation of growth plate chondrocytes where
c-Myc is expressed and severely downregulated in the absence of c-Myc.
Finally, we show that combined deletion of N-Myc and
c-Myc in early limb bud mesenchyme gives rise to a
severely hypoplastic limb skeleton that exhibits features characteristic of
individual c-Myc and N-Myc mutants. Conclusions/Significance Our results show that N-Myc and c-Myc act sequentially during limb
development to coordinate the expansion of key progenitor populations
responsible for forming the limb skeleton.
Collapse
|
182
|
Hasson P. "Soft" tissue patterning: muscles and tendons of the limb take their form. Dev Dyn 2011; 240:1100-7. [PMID: 21438070 DOI: 10.1002/dvdy.22608] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2011] [Indexed: 12/18/2022] Open
Abstract
The musculoskeletal system grants our bodies a vast range of movements. Because it is mainly composed of easily identifiable components, it serves as an ideal model to study patterning of the specific tissues that make up the organ. Surprisingly, although critical for the function of the musculoskeletal system, understanding of the embryonic processes that regulate muscle and tendon patterning is very limited. The recent identification of specific markers and the reagents stemming from them has revealed some of the molecular events regulating patterning of these soft tissues. This review will focus on some of the current work, with an emphasis on the roles of the muscle connective tissue, and discuss several key points that addressing them will advance our understanding of these patterning events.
Collapse
Affiliation(s)
- Peleg Hasson
- Department of Anatomy and Cell Biology, The Rappaport Family Institute for Research in the Medical Sciences, Technion-Israel Institute of Technology, Bat Galim, Haifa, Israel.
| |
Collapse
|
183
|
Stricker S, Mundlos S. Mechanisms of digit formation: Human malformation syndromes tell the story. Dev Dyn 2011; 240:990-1004. [PMID: 21337664 DOI: 10.1002/dvdy.22565] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2011] [Indexed: 12/29/2022] Open
Abstract
Identifying the genetic basis of human limb malformation disorders has been instrumental in improving our understanding of limb development. Abnormalities of the hands and/or feet include defects affecting patterning, establishment, elongation, and segmentation of cartilaginous condensations, as well as growth of the individual skeletal elements. While the phenotype of such malformations is highly diverse, the mutations identified to date cluster in genes implicated in a limited number of molecular pathways, namely hedgehog, Wnt, and bone morphogenetic protein. The latter pathway appears to function as a key molecular network regulating different phases of digit and joint development. Studies in animal models not only extended our insight into the pathogenesis of these conditions, but have also contributed to our understanding of the in vivo functions and interactions of these key players. This review is aimed at integrating the current understanding of human digit malformations into the increasing knowledge of the molecular mechanisms of digit development. Developmental Dynamics 240:990-1004, 2011. © 2011 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- Sigmar Stricker
- Development and Disease Group, Max Planck-Institute for Molecular Genetics, Berlin, Germany.
| | | |
Collapse
|
184
|
Antagonistic growth regulation by Dpp and Fat drives uniform cell proliferation. Dev Cell 2011; 20:123-30. [PMID: 21238930 DOI: 10.1016/j.devcel.2010.11.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 09/22/2010] [Accepted: 10/25/2010] [Indexed: 11/21/2022]
Abstract
We use the Dpp morphogen gradient in the Drosophila wing disc as a model to address the fundamental question of how a gradient of a growth factor can produce uniform growth. We first show that proper expression and subcellular localization of components in the Fat tumor-suppressor pathway, which have been argued to depend on Dpp activity differences, are not reliant on the Dpp gradient. We next analyzed cell proliferation in discs with uniformly high Dpp or uniformly low Fat signaling activity and found that these pathways regulate growth in a complementary manner. While the Dpp mediator Brinker inhibits growth in the primordium primarily in the lateral regions, Fat represses growth mostly in the medial region. Together, our results indicate that the activities of both signaling pathways are regulated in a parallel rather than sequential manner and that uniform proliferation is achieved by their complementary action on growth.
Collapse
|
185
|
Armstrong A, Ryu YK, Chieco D, Kuruvilla R. Frizzled3 is required for neurogenesis and target innervation during sympathetic nervous system development. J Neurosci 2011; 31:2371-81. [PMID: 21325504 PMCID: PMC3046637 DOI: 10.1523/jneurosci.4243-10.2011] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Revised: 12/02/2010] [Accepted: 12/16/2010] [Indexed: 01/04/2023] Open
Abstract
The sympathetic nervous system has served as an amenable model system to investigate molecular mechanisms underlying developmental processes in the nervous system. While much attention has been focused on neurotrophic factors controlling survival and connectivity of postmitotic sympathetic neurons, relatively little is known about signaling mechanisms regulating development of sympathetic neuroblasts. Here, we report that Frizzled3 (Fz3), a member of the Wnt receptor family, is essential for maintenance of dividing sympathetic neuroblasts. In Fz3(-/-) mice, sympathetic neuroblasts exhibit decreased proliferation and premature cell cycle exit. Fz3(-/-) sympathetic neuroblasts also undergo enhanced apoptosis, which could not be rescued by eliminating the proapoptotic factor, Bax. These deficits result in reduced generation of sympathetic neurons and pronounced decreases in the size of sympathetic chain ganglia. Furthermore, the axons of sympathetic neurons that persist in Fz3(-/-) ganglia are able to extend out of sympathetic ganglia toward distal targets, but fail to fully innervate final peripheral targets. The cell cycle exit, but not target innervation, defects in Fz3(-/-) mice are phenocopied in mice with conditional ablation of β-catenin, a component of canonical Wnt signaling, in sympathetic precursors. Sympathetic ganglia and innervation of target tissues appeared normal in mice lacking a core planar cell polarity (PCP) component, Vangl2. Together, our results suggest distinct roles for Fz3 during sympathetic neuron development; Fz3 acts at early developmental stages to maintain a pool of dividing sympathetic precursors, likely via activation of β-catenin, and Fz3 functions at later stages to promote innervation of final peripheral targets by postmitotic sympathetic neurons.
Collapse
Affiliation(s)
- Alissa Armstrong
- Department of Biology, Johns Hopkins University, Baltimore, Maryland 21218
| | - Yun Kyoung Ryu
- Department of Biology, Johns Hopkins University, Baltimore, Maryland 21218
| | - Deanna Chieco
- Department of Biology, Johns Hopkins University, Baltimore, Maryland 21218
| | - Rejji Kuruvilla
- Department of Biology, Johns Hopkins University, Baltimore, Maryland 21218
| |
Collapse
|
186
|
Stricker S, Mundlos S. FGF and ROR2 receptor tyrosine kinase signaling in human skeletal development. Curr Top Dev Biol 2011; 97:179-206. [PMID: 22074606 DOI: 10.1016/b978-0-12-385975-4.00013-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Skeletal malformations are among the most frequent developmental disturbances in humans. In the past years, progress has been made in unraveling the molecular mechanisms that govern skeletal development by the use of animal models as well as by the identification of numerous mutations that cause human skeletal syndromes. Receptor tyrosine kinases have critical roles in embryonic development. During formation of the skeletal system, the fibroblast growth factor receptor (FGFR) family plays major roles in the formation of cranial, axial, and appendicular bones. Another player of relevance to skeletal development is the unusual receptor tyrosine kinase ROR2, the function of which is as interesting as it is complex. In this chapter, we review the involvement of FGFR signaling in human skeletal disease and provide an update on the growing knowledge of ROR2.
Collapse
Affiliation(s)
- Sigmar Stricker
- Development and Disease Group, Max Planck-Institute for Molecular Genetics, Berlin, Germany
| | | |
Collapse
|
187
|
Abstract
The various Wnt pathways that are related to upper limb anomalies are reviewed. Abnormalities in the Wnt7a pathway (located in the dorsal ectoderm) produce several clinically relevant conditions such as the palmar duplication syndrome, nail patella syndrome, ulnar ray deficiency, limb hypoplasia, polysyndactyly and the palmar nail syndrome. Abnormalities of the Wnt3/3a pathway (located in the apical ectodermal ridge) include tetra-amelia and loss of the distal phalanges/nails. Abnormalities of the Wnt5/5a pathway (located in the apical ectodermal ridge as well as in the mesoderm) will affect chondrogenesis of the developing limb and experimental Wnt5a(-/-) limbs have terminal adactyly. Chondrogenesis and limb muscle differentiation are both affected by several Wnt pathways and these will be reviewed in details. Abnormalities in LRP 5/6 (a co-receptor for Wnts) lead to congenital bone disease and Wnt4 is specifically involved in joint development. Finally, the relationship between the Wnt pathway and SALL4 (mutations of which cause Okihiro/Duane-radial ray deficiency in humans) are discussed.
Collapse
|
188
|
Tran TH, Jarrell A, Zentner GE, Welsh A, Brownell I, Scacheri PC, Atit R. Role of canonical Wnt signaling/ß-catenin via Dermo1 in cranial dermal cell development. Development 2010; 137:3973-84. [PMID: 20980404 PMCID: PMC2976281 DOI: 10.1242/dev.056473] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2010] [Indexed: 01/15/2023]
Abstract
Cranial dermis develops from cephalic mesoderm and neural crest cells, but what signal(s) specifies the dermal lineage is unclear. Using genetic tools to fate map and manipulate a cranial mesenchymal progenitor population in the supraorbital region, we show that the dermal progenitor cells beneath the surface ectoderm process canonical Wnt signaling at the time of specification. We show that Wnt signaling/β-catenin is absolutely required and sufficient for Dermo1 expression and dermal cell identity in the cranium. The absence of the Wnt signaling cue leads to formation of cartilage in craniofacial and ventral trunk regions at the expense of dermal and bone lineages. Dermo1 can be a direct transcription target and may mediate the functional role of Wnt signaling in dermal precursors. This study reveals a lineage-specific role of canonical Wnt signaling/β-catenin in promoting dermal cell fate in distinct precursor populations.
Collapse
Affiliation(s)
- Thu H. Tran
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Andrew Jarrell
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Gabriel E. Zentner
- Department of Genetics, Case Western Reserve University, Cleveland OH 44106, USA
| | - Adrienne Welsh
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Isaac Brownell
- Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | - Peter C. Scacheri
- Department of Genetics, Case Western Reserve University, Cleveland OH 44106, USA
| | - Radhika Atit
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Genetics, Case Western Reserve University, Cleveland OH 44106, USA
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
189
|
Mitogen-activated protein kinases promote WNT/beta-catenin signaling via phosphorylation of LRP6. Mol Cell Biol 2010; 31:179-89. [PMID: 20974802 DOI: 10.1128/mcb.00550-10] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
LDL-related protein 6 (LRP6) is a coreceptor of WNTs and a key regulator of the WNT/β-catenin pathway. Upon activation, LRP6 is phosphorylated within its intracellular PPPS/TP motifs. These phosphorylated motifs are required to recruit axin and to inhibit glycogen synthase kinase 3 (GSK3), two basic components of the β-catenin destruction complex. On the basis of a kinome-wide small interfering RNA (siRNA) screen and confirmative biochemical analysis, we show that several proline-directed mitogen-activated protein kinases (MAPKs), such as p38, ERK1/2, and JNK1 are sufficient and required for the phosphorylation of PPPS/TP motifs of LRP6. External stimuli, which control the activity of MAPKs, such as phorbol esters and fibroblast growth factor 2 (FGF2) control the choice of the LRP6-PPPS/TP kinase and regulate the amplitude of LRP6 phosphorylation and WNT/β-catenin-dependent transcription. Our findings suggest that cells not only recruit one dedicated LRP6 kinase but rather select their LRP6 kinase depending on cell type and the external stimulus. Moreover, direct phosphorylation of LRP6 by MAPKs provides a unique point for convergence between WNT/β-catenin signaling and mitogenic pathways.
Collapse
|
190
|
Aman A, Nguyen M, Piotrowski T. Wnt/β-catenin dependent cell proliferation underlies segmented lateral line morphogenesis. Dev Biol 2010; 349:470-82. [PMID: 20974120 DOI: 10.1016/j.ydbio.2010.10.022] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Revised: 10/14/2010] [Accepted: 10/16/2010] [Indexed: 11/27/2022]
Abstract
Morphogenesis is a fascinating but complex and incompletely understood developmental process. The sensory lateral line system consists of only a few hundred cells and is experimentally accessible making it an excellent model system to interrogate the cellular and molecular mechanisms underlying segmental morphogenesis. The posterior lateral line primordium periodically deposits prosensory organs as it migrates to the tail tip. We demonstrate that periodic proneuromast deposition is governed by a fundamentally different developmental mechanism than the classical models of developmental periodicity represented by vertebrate somitogenesis and early Drosophila development. Our analysis demonstrates that proneuromast deposition is driven by periodic lengthening of the primordium and a stable Wnt/β-catenin activation domain in the leading region of the primordium. The periodic lengthening of the primordium is controlled by Wnt/β-catenin/Fgf-dependent proliferation. Once proneuromasts are displaced into the trailing Wnt/β-catenin-free zone they are deposited. We have previously shown that Wnt/β-catenin signaling induces Fgf signaling and that interactions between these two pathways regulate primordium migration and prosensory organ formation. Therefore, by coordinating migration, prosensory organ formation and proliferation, localized activation of Wnt/β-catenin signaling in the leading zone of the primordium plays a crucial role in orchestrating lateral line morphogenesis.
Collapse
Affiliation(s)
- Andy Aman
- University of Utah Medical School, Dept. of Neurobiology and Anatomy, MREB 401, Salt Lake City, UT 84132, USA
| | | | | |
Collapse
|
191
|
Wang B, Sinha T, Jiao K, Serra R, Wang J. Disruption of PCP signaling causes limb morphogenesis and skeletal defects and may underlie Robinow syndrome and brachydactyly type B. Hum Mol Genet 2010; 20:271-85. [PMID: 20962035 DOI: 10.1093/hmg/ddq462] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Brachydactyly type B (BDB1) and Robinow syndrome (RRS) are two skeletal disorders caused by mutations in ROR2, a co-receptor of Wnt5a. Wnt5a/Ror2 can activate multiple branches of non-canonical Wnt signaling, but it is unclear which branch(es) mediates Wnt5a/Ror2 function in limb skeletal development. Here, we provide evidence implicating the planar cell polarity (PCP) pathway as the downstream component of Wnt5a in the limb. We show that a mutation in the mouse PCP gene Vangl2 causes digit defects resembling the clinical phenotypes in BDB1, including loss of phalanges. Halving the dosage of Wnt5a in Vangl2 mutants enhances the severity and penetrance of the digit defects and causes long bone defects reminiscent of RRS, suggesting that Wnt5a and Vangl2 function in the same pathway and disruption of PCP signaling may underlie both BDB1 and RRS. Consistent with a role for PCP signaling in tissue morphogenesis, mutation of Vangl2 alters the shape and dimensions of early limb buds: the width and thickness are increased, whereas the length is decreased. The digit pre-chondrogenic condensates also become wider, thicker and shorter. Interestingly, altered limb bud dimensions in Vangl2 mutants also affect limb growth by perturbing the signaling network that regulates the balance between Fgf and Bmp signaling. Halving the dosage of Bmp4 partially suppresses the loss of phalanges in Vangl2 mutants, supporting the hypothesis that an aberrant increase in Bmp signaling is the cause of the brachydactyly defect. These findings provide novel insight into the signaling mechanisms of Wnt5a/Ror2 and the pathogenesis in BDB1 and RRS.
Collapse
Affiliation(s)
- Bing Wang
- Department of Cell Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | |
Collapse
|
192
|
Farrow EG, Summers LJ, Schiavi SC, McCormick JA, Ellison DH, White KE. Altered renal FGF23-mediated activity involving MAPK and Wnt: effects of the Hyp mutation. J Endocrinol 2010; 207:67-75. [PMID: 20675303 PMCID: PMC3050595 DOI: 10.1677/joe-10-0181] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Fibroblast growth factor-23 (FGF23), a hormone central to renal phosphate handling, is elevated in multiple hypophosphatemic disorders. Initial FGF23-dependent Erk1/2 activity in the kidney localizes to the distal convoluted tubule (DCT) with the co-receptor α-Klotho (KL), distinct from Npt2a in proximal tubules (PT). The Hyp mouse model of X-linked hypophosphatemic rickets (XLH) is characterized by hypophosphatemia with increased Fgf23, and patients with XLH elevate FGF23 following combination therapy of phosphate and calcitriol. The molecular signaling underlying renal FGF23 activity, and whether these pathways are altered in hypophosphatemic disorders, is unknown. To examine Npt2a in vivo, mice were injected with FGF23. Initial p-Erk1/2 activity in the DCT occurred within 10 min; however, Npt2a protein was latently reduced in the PT at 30-60 min, and was independent of Npt2a mRNA changes. KL-null mice had no DCT p-Erk1/2 staining following FGF23 delivery. Under basal conditions in Hyp mice, c-Fos and Egr1, markers of renal Fgf23 activity, were increased; however, KL mRNA was reduced 60% (P<0.05). Despite the prevailing hypophosphatemia and elevated Fgf23, FGF23 injections into Hyp mice activated p-Erk1/2 in the DCT. FGF23 injection also resulted in phospho-β-catenin (p-β-cat) co-localization with KL in wild-type mice, and Hyp mice demonstrated strong p-β-cat staining under basal conditions, indicating potential crosstalk between mitogen-activated protein kinase and Wnt signaling. Collectively, these studies refine the mechanisms for FGF23 bioactivity, and demonstrate novel suppression of Wnt signaling in a KL-dependent DCT-PT axis, which is likely altered in XLH. Finally, the current treatment of phosphate and calcitriol for hypophosphatemic disorders may increase FGF23 activity.
Collapse
Affiliation(s)
- Emily G. Farrow
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lelia J. Summers
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | - Kenneth E. White
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
193
|
Abstract
Morphogen gradients play a fundamental role in organ patterning and organ growth. Unlike their role in patterning, their function in regulating the growth and the size of organs is poorly understood. How and why do morphogen gradients exert their mitogenic effects to generate uniform proliferation in developing organs, and by what means can morphogens impinge on the final size of organs? The decapentaplegic (Dpp) gradient in the Drosophila wing imaginal disc has emerged as a suitable and established system to study organ growth. Here, we review models and recent findings that attempt to address how the Dpp morphogen contributes to uniform proliferation of cells, and how it may regulate the final size of wing discs.
Collapse
|
194
|
Popelut A, Rooker SM, Leucht P, Medio M, Brunski JB, Helms JA. The acceleration of implant osseointegration by liposomal Wnt3a. Biomaterials 2010; 31:9173-81. [PMID: 20864159 DOI: 10.1016/j.biomaterials.2010.08.045] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Accepted: 08/19/2010] [Indexed: 11/29/2022]
Abstract
The strength of a Wnt-based strategy for tissue regeneration lies in the central role that Wnts play in healing. Tissue injury triggers local Wnt activation at the site of damage, and this Wnt signal is required for the repair and/or regeneration of almost all tissues including bone, neural tissues, myocardium, and epidermis. We developed a biologically based approach to create a transient elevation in Wnt signaling in peri-implant tissues, and in doing so, accelerated bone formation around the implant. Our subsequent molecular and cellular analyses provide mechanistic insights into the basis for this pro-osteogenic effect. Given the essential role of Wnt signaling in bone formation, this protein-based approach may have widespread application in implant osseointegration.
Collapse
Affiliation(s)
- Antoine Popelut
- Department of Periodontology, Service of Odontology, Hotel-Dieu Hospital, AP-HP, U.F.R. of Odontology, Paris 7 Denis Diderot University, Paris, France
| | | | | | | | | | | |
Collapse
|
195
|
Receptor tyrosine kinase-like orphan receptor 2 (ROR2) and Indian hedgehog regulate digit outgrowth mediated by the phalanx-forming region. Proc Natl Acad Sci U S A 2010; 107:14211-6. [PMID: 20660756 DOI: 10.1073/pnas.1009314107] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Elongation of the digit rays resulting in the formation of a defined number of phalanges is a process poorly understood in mammals, whereas in the chicken distal mesenchymal bone morphogenetic protein (BMP) signaling in the so-called phalanx-forming region (PFR) or digit crescent (DC) seems to be involved. The human brachydactylies (BDs) are inheritable conditions characterized by variable degrees of digit shortening, thus providing an ideal model to analyze the development and elongation of phalanges. We used a mouse model for BDB1 (Ror2(W749X/W749X)) lacking middle phalanges and show that a signaling center corresponding to the chick PFR exists in the mouse, which is diminished in BDB1 mice. This resulted in a strongly impaired elongation of the digit condensations due to reduced chondrogenic commitment of undifferentiated distal mesenchymal cells. We further show that a similar BMP-based mechanism accounts for digit shortening in a mouse model for the closely related condition BDA1 (Ihh(E95K/E95K)), altogether indicating the functional significance of the PFR in mammals. Genetic interaction experiments as well as pathway analysis in BDB1 mice suggest that Indian hedgehog and WNT/beta-catenin signaling, which we show is inhibited by receptor tyrosine kinase-like orphan receptor 2 (ROR2) in distal limb mesenchyme, are acting upstream of BMP signaling in the PFR.
Collapse
|
196
|
Boehm B, Westerberg H, Lesnicar-Pucko G, Raja S, Rautschka M, Cotterell J, Swoger J, Sharpe J. The role of spatially controlled cell proliferation in limb bud morphogenesis. PLoS Biol 2010; 8:e1000420. [PMID: 20644711 PMCID: PMC2903592 DOI: 10.1371/journal.pbio.1000420] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Accepted: 06/03/2010] [Indexed: 11/19/2022] Open
Abstract
Although the vertebrate limb bud has been studied for decades as a model system for spatial pattern formation and cell specification, the cellular basis of its distally oriented elongation has been a relatively neglected topic by comparison. The conventional view is that a gradient of isotropic proliferation exists along the limb, with high proliferation rates at the distal tip and lower rates towards the body, and that this gradient is the driving force behind outgrowth. Here we test this hypothesis by combining quantitative empirical data sets with computer modelling to assess the potential role of spatially controlled proliferation rates in the process of directional limb bud outgrowth. In particular, we generate two new empirical data sets for the mouse hind limb--a numerical description of shape change and a quantitative 3D map of cell cycle times--and combine these with a new 3D finite element model of tissue growth. By developing a parameter optimization approach (which explores spatial patterns of tissue growth) our computer simulations reveal that the observed distribution of proliferation rates plays no significant role in controlling the distally extending limb shape, and suggests that directional cell activities are likely to be the driving force behind limb bud outgrowth. This theoretical prediction prompted us to search for evidence of directional cell orientations in the limb bud mesenchyme, and we thus discovered a striking highly branched and extended cell shape composed of dynamically extending and retracting filopodia, a distally oriented bias in Golgi position, and also a bias in the orientation of cell division. We therefore provide both theoretical and empirical evidence that limb bud elongation is achieved by directional cell activities, rather than a PD gradient of proliferation rates.
Collapse
Affiliation(s)
- Bernd Boehm
- EMBL-CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), UPF, Barcelona, Spain
| | - Henrik Westerberg
- EMBL-CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), UPF, Barcelona, Spain
| | - Gaja Lesnicar-Pucko
- EMBL-CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), UPF, Barcelona, Spain
| | - Sahdia Raja
- EMBL-CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), UPF, Barcelona, Spain
- MRC Human Genetics Unit, Edinburgh, Scotland, United Kingdom
| | - Michael Rautschka
- EMBL-CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), UPF, Barcelona, Spain
| | - James Cotterell
- EMBL-CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), UPF, Barcelona, Spain
- MRC Human Genetics Unit, Edinburgh, Scotland, United Kingdom
| | - Jim Swoger
- EMBL-CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), UPF, Barcelona, Spain
| | - James Sharpe
- EMBL-CRG Systems Biology Research Unit, Centre for Genomic Regulation (CRG), UPF, Barcelona, Spain
- ICREA Professor, Centre for Genomic Regulation (CRG), UPF, Barcelona, Spain
| |
Collapse
|
197
|
Abstract
Although the vertebrate limb bud has been studied for decades as a model system for spatial pattern formation and cell specification, the cellular basis of its distally oriented elongation has been a relatively neglected topic by comparison. The conventional view is that a gradient of isotropic proliferation exists along the limb, with high proliferation rates at the distal tip and lower rates towards the body, and that this gradient is the driving force behind outgrowth. Here we test this hypothesis by combining quantitative empirical data sets with computer modelling to assess the potential role of spatially controlled proliferation rates in the process of directional limb bud outgrowth. In particular, we generate two new empirical data sets for the mouse hind limb--a numerical description of shape change and a quantitative 3D map of cell cycle times--and combine these with a new 3D finite element model of tissue growth. By developing a parameter optimization approach (which explores spatial patterns of tissue growth) our computer simulations reveal that the observed distribution of proliferation rates plays no significant role in controlling the distally extending limb shape, and suggests that directional cell activities are likely to be the driving force behind limb bud outgrowth. This theoretical prediction prompted us to search for evidence of directional cell orientations in the limb bud mesenchyme, and we thus discovered a striking highly branched and extended cell shape composed of dynamically extending and retracting filopodia, a distally oriented bias in Golgi position, and also a bias in the orientation of cell division. We therefore provide both theoretical and empirical evidence that limb bud elongation is achieved by directional cell activities, rather than a PD gradient of proliferation rates.
Collapse
|
198
|
Origin matters: differences in embryonic tissue origin and Wnt signaling determine the osteogenic potential and healing capacity of frontal and parietal calvarial bones. J Bone Miner Res 2010; 25:1680-94. [PMID: 19929441 PMCID: PMC3154006 DOI: 10.1359/jbmr.091116] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Calvarial bones arise from two embryonic tissues, namely, the neural crest and the mesoderm. In this study we have addressed the important question of whether disparate embryonic tissue origins impart variable osteogenic potential and regenerative capacity to calvarial bones, as well as what the underlying molecular mechanism(s). Thus, by performing in vitro and in vivo studies, we have investigated whether differences exist between neural crest-derived frontal and paraxial mesodermal-derived parietal bone. Of interest, our data indicate that calvarial bone osteoblasts of neural crest origin have superior potential for osteogenic differentiation. Furthermore, neural crest-derived frontal bone displays a superior capacity to undergo osseous healing compared with calvarial bone of paraxial mesoderm origin. Our study identified both in vitro and in vivo enhanced endogenous canonical Wnt signaling in frontal bone compared with parietal bone. In addition, we demonstrate that constitutive activation of canonical Wnt signaling in paraxial mesodermal-derived parietal osteoblasts mimics the osteogenic potential of frontal osteoblasts, whereas knockdown of canonical Wnt signaling dramatically impairs the greater osteogenic potential of neural crest-derived frontal osteoblasts. Moreover, fibroblast growth factor 2 (FGF-2) treatment induces phosphorylation of GSK-3beta and increases the nuclear levels of beta-catenin in osteoblasts, suggesting that enhanced activation of Wnt signaling might be mediated by FGF. Taken together, our data provide compelling evidence that indeed embryonic tissue origin makes a difference and that active canonical Wnt signaling plays a major role in contributing to the superior intrinsic osteogenic potential and tissue regeneration observed in neural crest-derived frontal bone.
Collapse
|
199
|
Komatsu DE, Mary MN, Schroeder RJ, Robling AG, Turner CH, Warden SJ. Modulation of Wnt signaling influences fracture repair. J Orthop Res 2010; 28:928-36. [PMID: 20063381 PMCID: PMC3412133 DOI: 10.1002/jor.21078] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
While the importance of Wnt signaling in skeletal development and homeostasis is well documented, little is known regarding its function in fracture repair. We hypothesized that activation and inactivation of Wnt signaling would enhance and impair fracture repair, respectively. Femoral fractures were generated in Lrp5 knockout mice (Lrp5-/-) and wild-type littermates (Lrp5+/+), as well as C57BL/6 mice. Lrp5-/- and Lrp5+/+ mice were untreated, while C57BL/6 mice were treated 2x/week with vehicle or anti-Dkk1 antibodies (Dkk1 Ab) initiated immediately postoperatively (Day 0) or 4 days postoperatively (Day 4). Fractures were radiographed weekly until sacrifice at day 28, followed by DXA, pQCT, and biomechanical analyses. Lrp5-/- mice showed impaired repair compared to Lrp5+/+ mice, as evidenced by reduced callus area, BMC, BMD, and biomechanical properties. The effects of Dkk1 Ab treatment depended on the timing of initiation. Day 0 initiation enhanced repair, with significant gains seen for callus area, BMC, BMD, and biomechanical properties, whereas Day 4 initiation had no effect. These results validated our hypothesis that Wnt signaling influences fracture repair, with prompt activation enhancing repair and inactivation impairing it. Furthermore, these data suggest that activation of Wnt signaling during fracture repair may have clinical utility in facilitating fracture repair.
Collapse
Affiliation(s)
- David E. Komatsu
- InMotion Orthopaedic Research Center, 20 South Dudley, Suite 700, Memphis, Tennessee 38103
| | - Michelle N. Mary
- InMotion Orthopaedic Research Center, 20 South Dudley, Suite 700, Memphis, Tennessee 38103
| | - Robert Jason Schroeder
- InMotion Orthopaedic Research Center, 20 South Dudley, Suite 700, Memphis, Tennessee 38103
| | - Alex G. Robling
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Drive, MS-5035, Indianapolis, Indiana 46202
| | - Charles H. Turner
- Department of Biomedical Engineering, Purdue School of Engineering and Technology, Indiana University—Purdue University Indianapolis, 1120 South Drive, FH-115, Indianapolis, Indiana 46202
| | - Stuart J. Warden
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Drive, MS-5035, Indianapolis, Indiana 46202,Department of Biomedical Engineering, Purdue School of Engineering and Technology, Indiana University—Purdue University Indianapolis, 1120 South Drive, FH-115, Indianapolis, Indiana 46202,Department of Physical Therapy, School of Health and Rehabilitation Sciences, Indiana University, 1140 W. Michigan Street, CF-326, Indianapolis, Indiana 46202
| |
Collapse
|
200
|
Collette NM, Genetos D, Murugesh D, Harland RM, Loots GG. Genetic evidence that SOST inhibits WNT signaling in the limb. Dev Biol 2010; 342:169-79. [PMID: 20359476 PMCID: PMC2896299 DOI: 10.1016/j.ydbio.2010.03.021] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2009] [Revised: 03/18/2010] [Accepted: 03/22/2010] [Indexed: 11/17/2022]
Abstract
SOST is a negative regulator of bone formation, and mutations in human SOST are responsible for sclerosteosis. In addition to high bone mass, sclerosteosis patients occasionally display hand defects, suggesting that SOST may function embryonically. Here we report that overexpression of SOST leads to loss of posterior structures of the zeugopod and autopod by perturbing anterior-posterior and proximal-distal signaling centers in the developing limb. Mutant mice that overexpress SOST in combination with Grem1 and Lrp6 mutations display more severe limb defects than single mutants alone, while Sost(-/-) significantly rescues the Lrp6(-/-) skeletal phenotype, signifying that SOST gain-of-function impairs limb patterning by inhibiting the WNT signaling through LRP5/6.
Collapse
Affiliation(s)
- Nicole M. Collette
- Biology and Biotechnology Division, Lawrence Livermore National Laboratory, 7000 East Avenue, L-452, Livermore, CA 94550
- Department of Molecular and Cell Biology, Division of Genetics, Genomics, and Development, and Center for Integrative Genomics, University of California, Berkeley, CA 94720-3204, USA
| | - Damian Genetos
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Deepa Murugesh
- Biology and Biotechnology Division, Lawrence Livermore National Laboratory, 7000 East Avenue, L-452, Livermore, CA 94550
- Department of Molecular and Cell Biology, Division of Genetics, Genomics, and Development, and Center for Integrative Genomics, University of California, Berkeley, CA 94720-3204, USA
| | - Richard M. Harland
- Department of Molecular and Cell Biology, Division of Genetics, Genomics, and Development, and Center for Integrative Genomics, University of California, Berkeley, CA 94720-3204, USA
| | - Gabriela G. Loots
- Biology and Biotechnology Division, Lawrence Livermore National Laboratory, 7000 East Avenue, L-452, Livermore, CA 94550
- Department of Molecular and Cell Biology, Division of Genetics, Genomics, and Development, and Center for Integrative Genomics, University of California, Berkeley, CA 94720-3204, USA
| |
Collapse
|