101
|
Hermans KC, Daskalopoulos EP, Blankesteijn WM. Interventions in Wnt signaling as a novel therapeutic approach to improve myocardial infarct healing. FIBROGENESIS & TISSUE REPAIR 2012; 5:16. [PMID: 22967504 PMCID: PMC3472244 DOI: 10.1186/1755-1536-5-16] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 08/20/2012] [Indexed: 01/12/2023]
Abstract
Following myocardial infarction, wound healing takes place in the infarct area where the non-viable cardiac tissue is replaced by a scar. Inadequate wound healing or insufficient maintenance of the extracellular matrix in the scar can lead to excessive dilatation of the ventricles, one of the hallmarks of congestive heart failure. Therefore, it is important to better understand the wound-healing process in the heart and to develop new therapeutic agents that target the infarct area in order to maintain an adequate cardiac function. One of these potential novel therapeutic targets is Wnt signaling. Wnt signaling plays an important role in embryonic myocardial development but in the adult heart the pathway is thought to be silent. However, there is increasing evidence that components of the Wnt pathway are re-expressed during cardiac repair, implying a regulatory role. Recently, several studies have been published where the effect of interventions in Wnt signaling on infarct healing has been studied. In this review, we will summarize the results of these studies and discuss the effects of these interventions on the different cell types that are involved in the wound healing process.
Collapse
Affiliation(s)
- Kevin Cm Hermans
- Department of Pharmacology, Cardiovascular Research Institute Maastricht, Maastricht University, 50 Universiteitssingel, 6229ER Maastricht, PO Box 616 6200MD, Maastricht, The Netherlands.
| | | | | |
Collapse
|
102
|
Wnt ligands signal in a cooperative manner to promote foregut organogenesis. Proc Natl Acad Sci U S A 2012; 109:15348-53. [PMID: 22949635 DOI: 10.1073/pnas.1201583109] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Endoderm-mesenchyme cross-talk is a central process in the development of foregut-derived organs. How signaling pathways integrate the activity of multiple ligands to guide organ development is poorly understood. We show that two Wnt ligands, Wnt2 and Wnt7b, cooperatively induce Wnt signaling without affecting the stabilization of the Wnt canonical effector β-catenin despite it being necessary for Wnt2-Wnt7b cooperativity. Wnt2-Wnt7b cooperation is specific for mesenchymal cell lineages and the combined loss of Wnt2 and Wnt7b leads to more severe developmental defects in the lung than loss of Wnt2 or Wnt7b alone. High-throughput small-molecule screens and biochemical assays reveal that the Pdgf pathway is required for cooperative Wnt2-Wnt7b signaling. Inhibition of Pdgf signaling in cell culture reduces Wnt2-Wnt7b cooperative signaling. Moreover, inhibition of Pdgf signaling in lung explant cultures results in decreased Wnt signaling and lung smooth-muscle development. These data suggest a model in which Pdgf signaling potentiates Wnt2-Wnt7b signaling to promote high levels of Wnt activity in mesenchymal progenitors that is required for proper development of endoderm-derived organs, such as the lung.
Collapse
|
103
|
Meuten T, Hickey A, Franklin K, Grossi B, Tobias J, Newman DR, Jennings SH, Correa M, Sannes PL. WNT7B in fibroblastic foci of idiopathic pulmonary fibrosis. Respir Res 2012; 13:62. [PMID: 22838404 PMCID: PMC3479038 DOI: 10.1186/1465-9921-13-62] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 07/17/2012] [Indexed: 12/29/2022] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a devastating interstitial pneumonia causing a loss of respiratory surface area due to a proliferative fibrotic response involving hyperplastic, hypertrophic, and metaplastic epithelium, cystic honeycomb change, septal expansion, and variable inflammation. Wnt (wingless) signaling glycoproteins are known to be involved in lung development and tissue repair, and are up-regulated in patients with IPF. Based on previous qRT-PCR data showing increased Wnt7B in lungs of IPF patients, a systematic, quantitative examination of its tissue site distribution was undertaken. Methods Tissue samples from the Lung Tissue Research Consortium (LTRC) of 39 patients diagnosed with mild to severe IPF/usual interstitial pneumonia (UIP) and 19 normal patients were examined for the immunolocalization of Wnt7B. Results In normal lung, moderate Wnt7B reactivity was confined to airway epithelium, smooth muscle of airways and vasculature, and macrophages. IPF lung showed strong Wnt7B reactivity in fibroblastic foci, dysplastic airway and alveolar epithelium, and in highly discrete subepithelial, basement membrane-associated regions. All reactive sites were sized and counted relative to specific microscopic regions. Those in the subepithelial sites were found in significantly greater numbers and larger relative area compared with the others. No reactive sites were present in normal patient controls. Conclusions The results demonstrate Wnt7B to be expressed at high concentrations in regions of active hyperplasia, metaplasia, and fibrotic change in IPF patients. In this context and its previously established biologic activities, Wnt7B would be expected to be of potential importance in the pathogenesis of IPF.
Collapse
Affiliation(s)
- Travis Meuten
- Departments of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
104
|
Mammoto T, Chen J, Jiang E, Jiang A, Smith LE, Ingber DE, Mammoto A. LRP5 regulates development of lung microvessels and alveoli through the angiopoietin-Tie2 pathway. PLoS One 2012; 7:e41596. [PMID: 22848540 PMCID: PMC3404972 DOI: 10.1371/journal.pone.0041596] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 06/27/2012] [Indexed: 01/26/2023] Open
Abstract
Angiogenesis is crucial for lung development. Although there has been considerable exploration, the mechanism by which lung vascular and alveolar formation is controlled is still not completely understood. Here we show that low-density lipoprotein receptor-related protein 5 (LRP5), a component of the Wnt ligand-receptor complex, regulates angiogenesis and alveolar formation in the lung by modulating expression of the angiopoietin (Ang) receptor, Tie2, in vascular endothelial cells (ECs). Vascular development in whole mouse lungs and in cultured ECs is controlled by LRP5 signaling, which is, in turn, governed by a balance between the activities of the antagonistic Tie2 ligands, Ang1 and Ang2. Under physiological conditions when Ang1 is dominant, LRP5 knockdown decreases Tie2 expression and thereby, inhibits vascular and alveolar development in the lung. Conversely, when Ang2 dominates under hyperoxia treatment in neonatal mice, high LRP5 and Tie2 expression suppress angiogenesis and lung development. These findings suggest that the LRP5-Tie2-Ang signaling axis plays a central role in control of both angiogenesis and alveolarization during postnatal lung development, and that deregulation of this signaling mechanism might lead to developmental abnormalities of the lung, such as are observed in bronchopulmonary dysplasia (BPD).
Collapse
Affiliation(s)
- Tadanori Mammoto
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jing Chen
- Department of Ophthalmology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Elisabeth Jiang
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Amanda Jiang
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lois E. Smith
- Department of Ophthalmology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Donald E. Ingber
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts, United States of America
- Harvard School of Engineering and Applied Sciences, Cambridge, Massachusetts, United States of America
| | - Akiko Mammoto
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
105
|
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
|
106
|
Choi HJ, Park H, Lee HW, Kwon YG. The Wnt pathway and the roles for its antagonists, DKKS, in angiogenesis. IUBMB Life 2012; 64:724-31. [PMID: 22807036 DOI: 10.1002/iub.1062] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 05/06/2012] [Accepted: 05/24/2012] [Indexed: 11/07/2022]
Abstract
The Wnt signaling pathway is involved in a wide range of developmental and physiological processes, such as cell fate specification, tissue morphogenesis, and homeostasis. Thus, its dysregulation has been found in multiple diseases, including some cardiovascular disorders. The loss or gain of function of Wnt pathway components results in abnormal vascular development and angiogenesis. Further study has revealed that Wnt signaling in endothelial cells appears to contribute to vascular morphogenesis and endothelial cell specification. Owing to the significance of Wnt signaling in angiogenesis, Wnt antagonists have been considered potential treatments for neovascular disorders. In line with this, members of the Dkk protein family (Dkks), well-known Wnt antagonists, have been recently found to regulate angiogenesis. This review summarizes our present knowledge of the roles of Wnt signaling and Wnt antagonists, particularly Dkks, in angiogenic regulation and explores the therapeutic potential of Wnt antagonists.
Collapse
Affiliation(s)
- Hyun-Jung Choi
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | | | | | | |
Collapse
|
107
|
Jacobs IJ, Ku WY, Que J. Genetic and cellular mechanisms regulating anterior foregut and esophageal development. Dev Biol 2012; 369:54-64. [PMID: 22750256 DOI: 10.1016/j.ydbio.2012.06.016] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2012] [Revised: 06/07/2012] [Accepted: 06/20/2012] [Indexed: 12/22/2022]
Abstract
Separation of the single anterior foregut tube into the esophagus and trachea involves cell proliferation and differentiation, as well as dynamic changes in cell-cell adhesion and migration. These biological processes are regulated and coordinated at multiple levels through the interplay of the epithelium and mesenchyme. Genetic studies and in vitro modeling have shed light on relevant regulatory networks that include a number of transcription factors and signaling pathways. These signaling molecules exhibit unique expression patterns and play specific functions in their respective territories before the separation process occurs. Disruption of regulatory networks inevitably leads to defective separation and malformation of the trachea and esophagus and results in the formation of a relatively common birth defect, esophageal atresia with or without tracheoesophageal fistula (EA/TEF). Significantly, some of the signaling pathways and transcription factors involved in anterior foregut separation continue to play important roles in the morphogenesis of the individual organs. In this review, we will focus on new findings related to these different developmental processes and discuss them in the context of developmental disorders or birth defects commonly seen in clinics.
Collapse
Affiliation(s)
- Ian J Jacobs
- Department of Biology, University of Rochester, Rochester, NY 14642, USA
| | | | | |
Collapse
|
108
|
Cellière G, Menshykau D, Iber D. Simulations demonstrate a simple network to be sufficient to control branch point selection, smooth muscle and vasculature formation during lung branching morphogenesis. Biol Open 2012; 1:775-88. [PMID: 23213471 PMCID: PMC3507219 DOI: 10.1242/bio.20121339] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 05/08/2012] [Indexed: 01/02/2023] Open
Abstract
Proper lung functioning requires not only a correct structure of the conducting airway tree, but also the simultaneous development of smooth muscles and vasculature. Lung branching morphogenesis is strongly stereotyped and involves the recursive use of only three modes of branching. We have previously shown that the experimentally described interactions between Fibroblast growth factor (FGF)10, Sonic hedgehog (SHH) and Patched (Ptc) can give rise to a Turing mechanism that not only reproduces the experimentally observed wildtype branching pattern but also, in part counterintuitive, patterns in mutant mice. Here we show that, even though many proteins affect smooth muscle formation and the expression of Vegfa, an inducer of blood vessel formation, it is sufficient to add FGF9 to the FGF10/SHH/Ptc module to successfully predict simultaneously the emergence of smooth muscles in the clefts between growing lung buds, and Vegfa expression in the distal sub-epithelial mesenchyme. Our model reproduces the phenotype of both wildtype and relevant mutant mice, as well as the results of most culture conditions described in the literature.
Collapse
Affiliation(s)
- Géraldine Cellière
- Department for Biosystems Science and Engineering , ETH Zurich, Mattenstrasse 26, 4058 Basel , Switzerland
| | | | | |
Collapse
|
109
|
Abstract
Wnt signals contribute to melanoma progression by boosting their proliferation and survival. Initially, we expected that activated Wnt signaling also improves their proficiency to recruit blood and lymph vessels. To assess this, we added cell culture supernatants (SNs) of Wnt1(+) and Wnt1(-) melanoma to endothelial spheroids. Whereas SNs of Wnt1(-) melanoma cells induced lymphatic sprouts, those of Wnt1(+) cells were unable to do so and this was restored by vascular endothelial growth factor C (VEGF-C). Subsequent testing of several human melanoma lines revealed that Wnt1 suppressed their VEGF-C expression. This Wnt1 effect did not depend on glycogen synthase kinase-3β (GSK3β), β-catenin, or activator protein-1, but was blocked by cyclosporine A (CsA). To analyze Wnt1 effects in melanoma in vivo, we selected Wnt1(-) melanoma cell lines, overexpressed Wnt1, and injected them subepidermally into severe combined immunodeficient (SCID) mice. We found reduced VEGF-C expression, reduced lymphangiogenesis, and delayed metastasis to sentinel nodes in Wnt1(+) as compared with Wnt1(-) melanoma (P<0.05). Concomitant overexpression of VEGF-C or feeding of animals with CsA restored lymphangiogenesis and metastasis in Wnt1(+) melanoma. In conclusion, Wnt1 is anti-lymphangiogenic by suppressing melanoma-derived VEGF-C expression.
Collapse
|
110
|
Rydell-Törmänen K, Risse PA, Kanabar V, Bagchi R, Czubryt MP, Johnson JR. Smooth muscle in tissue remodeling and hyper-reactivity: airways and arteries. Pulm Pharmacol Ther 2012; 26:13-23. [PMID: 22561160 DOI: 10.1016/j.pupt.2012.04.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 04/20/2012] [Accepted: 04/23/2012] [Indexed: 01/17/2023]
Abstract
Smooth muscle comprises a key functional component of both the airways and their supporting vasculature. Dysfunction of smooth muscle contributes to and exacerbates a host of breathing-associated pathologies such as asthma, chronic obstructive pulmonary disease and pulmonary hypertension. These diseases may be marked by airway and/or vascular smooth muscle hypertrophy, proliferation and hyper-reactivity, and related conditions such as fibrosis and extracellular matrix remodeling. This review will focus on the contribution of airway or vascular smooth dysfunction to common airway diseases.
Collapse
|
111
|
Mill C, George SJ. Wnt signalling in smooth muscle cells and its role in cardiovascular disorders. Cardiovasc Res 2012; 95:233-40. [PMID: 22492675 DOI: 10.1093/cvr/cvs141] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Vascular smooth muscle cells (SMCs) are the major cell type within blood vessels. SMCs exhibit low rates of proliferation, migration, and apoptosis in normal blood vessels. However, increased SMC proliferation, migration, and apoptosis rates radically alter the composition and structure of the blood vessel wall and contribute to cardiovascular diseases, such as atherosclerosis, and restenosis that occur after coronary artery vein grafting and stent implantation. Consequently, therapies that modulate SMC proliferation, migration, and apoptosis may be useful for treating cardiovascular diseases. The family of Wnt proteins, which were first identified in the wingless drosophila, has a well-established role in embryogenesis and development. It is now emerging that Wnt proteins also regulate SMC proliferation, migration, and survival. In this review article, we discuss recently emerging research that has revealed that Wnt proteins are important regulators of SMC behaviour via activation of β-catenin-dependent and β-catenin-independent Wnt signalling pathways.
Collapse
Affiliation(s)
- Carina Mill
- Bristol Heart Institute, School of Clinical Sciences, Research Floor Level 7, Bristol Royal Infirmary, Upper Maudlin St, Bristol BS2 8HW, UK
| | | |
Collapse
|
112
|
Hashimoto S, Chen H, Que J, Brockway BL, Drake JA, Snyder JC, Randell SH, Stripp BR. β-Catenin-SOX2 signaling regulates the fate of developing airway epithelium. J Cell Sci 2012; 125:932-942. [PMID: 22421361 PMCID: PMC3311930 DOI: 10.1242/jcs.092734] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2011] [Indexed: 02/05/2023] Open
Abstract
Wnt-β-catenin signaling regulates cell fate during organ development and postnatal tissue maintenance, but its contribution to specification of distinct lung epithelial lineages is still unclear. To address this question, we used a Cre recombinase (Cre)-LoxP approach to activate canonical Wnt signaling ectopically in developing lung endoderm. We found that persistent activation of canonical Wnt signaling within distal lung endoderm was permissive for normal development of alveolar epithelium, yet led to the loss of developing bronchiolar epithelium and ectasis of distal conducting airways. Activation of canonical Wnt led to ectopic expression of a lymphoid-enhancing factor and a T-cell factor (LEF and TCF, respectively) and absence of SRY (sex-determining region Y)-box 2 (SOX2) and tumor protein p63 (p63) expression in proximal derivatives. Conditional loss of SOX2 in airways phenocopied epithelial differentiation defects observed with ectopic activation of canonical Wnt. Our data suggest that Wnt negatively regulates a SOX2-dependent signaling program required for developmental progression of the bronchiolar lineage.
Collapse
Affiliation(s)
- Shuichi Hashimoto
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Duke University Medical Center, 106 Research Drive, 2075 MSRBII, DUMC Box 103000, Durham, NC, 27710, USA
- Department of Cell Biology, Duke University Medical Center, Box 3709, Durham, NC, 27710, USA
| | - Huaiyong Chen
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Duke University Medical Center, 106 Research Drive, 2075 MSRBII, DUMC Box 103000, Durham, NC, 27710, USA
- Department of Cell Biology, Duke University Medical Center, Box 3709, Durham, NC, 27710, USA
| | - Jianwen Que
- Department of Cell Biology, Duke University Medical Center, Box 3709, Durham, NC, 27710, USA
| | - Brian L. Brockway
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Duke University Medical Center, 106 Research Drive, 2075 MSRBII, DUMC Box 103000, Durham, NC, 27710, USA
- Department of Cell Biology, Duke University Medical Center, Box 3709, Durham, NC, 27710, USA
| | - Jeffrey A. Drake
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Duke University Medical Center, 106 Research Drive, 2075 MSRBII, DUMC Box 103000, Durham, NC, 27710, USA
- Department of Cell Biology, Duke University Medical Center, Box 3709, Durham, NC, 27710, USA
| | - Joshua C. Snyder
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Duke University Medical Center, 106 Research Drive, 2075 MSRBII, DUMC Box 103000, Durham, NC, 27710, USA
- Department of Cell Biology, Duke University Medical Center, Box 3709, Durham, NC, 27710, USA
| | - Scott H. Randell
- Departments of Cell and Molecular Physiology and Medicine, The University of North Carolina at Chapel Hill, 111 Mason Farm Road, 5200 Medical Biomolecular Research Building, CB 7545 Chapel Hill, NC, 27599-7545, USA
| | - Barry R. Stripp
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Duke University Medical Center, 106 Research Drive, 2075 MSRBII, DUMC Box 103000, Durham, NC, 27710, USA
- Department of Cell Biology, Duke University Medical Center, Box 3709, Durham, NC, 27710, USA
| |
Collapse
|
113
|
Ding BS, Nolan DJ, Guo P, Babazadeh AO, Cao Z, Rosenwaks Z, Crystal RG, Simons M, Sato TN, Worgall S, Shido K, Rabbany SY, Rafii S. Endothelial-derived angiocrine signals induce and sustain regenerative lung alveolarization. Cell 2011; 147:539-53. [PMID: 22036563 DOI: 10.1016/j.cell.2011.10.003] [Citation(s) in RCA: 380] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 08/15/2011] [Accepted: 10/05/2011] [Indexed: 12/31/2022]
Abstract
To identify pathways involved in adult lung regeneration, we employ a unilateral pneumonectomy (PNX) model that promotes regenerative alveolarization in the remaining intact lung. We show that PNX stimulates pulmonary capillary endothelial cells (PCECs) to produce angiocrine growth factors that induce proliferation of epithelial progenitor cells supporting alveologenesis. Endothelial cells trigger expansion of cocultured epithelial cells, forming three-dimensional angiospheres reminiscent of alveolar-capillary sacs. After PNX, endothelial-specific inducible genetic ablation of Vegfr2 and Fgfr1 in mice inhibits production of MMP14, impairing alveolarization. MMP14 promotes expansion of epithelial progenitor cells by unmasking cryptic EGF-like ectodomains that activate the EGF receptor (EGFR). Consistent with this, neutralization of MMP14 impairs EGFR-mediated alveolar regeneration, whereas administration of EGF or intravascular transplantation of MMP14(+) PCECs into pneumonectomized Vegfr2/Fgfr1-deficient mice restores alveologenesis and lung inspiratory volume and compliance function. VEGFR2 and FGFR1 activation in PCECs therefore increases MMP14-dependent bioavailability of EGFR ligands to initiate and sustain alveologenesis.
Collapse
Affiliation(s)
- Bi-Sen Ding
- Ansary Stem Cell Institute, Department of Genetic Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Gore AV, Swift MR, Cha YR, Lo B, McKinney MC, Li W, Castranova D, Davis A, Mukouyama YS, Weinstein BM. Rspo1/Wnt signaling promotes angiogenesis via Vegfc/Vegfr3. Development 2011; 138:4875-86. [PMID: 22007135 DOI: 10.1242/dev.068460] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Here, we show that a novel Rspo1-Wnt-Vegfc-Vegfr3 signaling pathway plays an essential role in developmental angiogenesis. A mutation in R-spondin1 (rspo1), a Wnt signaling regulator, was uncovered during a forward-genetic screen for angiogenesis-deficient mutants in the zebrafish. Embryos lacking rspo1 or the proposed rspo1 receptor kremen form primary vessels by vasculogenesis, but are defective in subsequent angiogenesis. Endothelial cell-autonomous inhibition of canonical Wnt signaling also blocks angiogenesis in vivo. The pro-angiogenic effects of Rspo1/Wnt signaling are mediated by Vegfc/Vegfr3(Flt4) signaling. Vegfc expression is dependent on Rspo1 and Wnt, and Vegfc and Vegfr3 are necessary to promote angiogenesis downstream from Rspo1-Wnt. As all of these molecules are expressed by the endothelium during sprouting stages, these results suggest that Rspo1-Wnt-VegfC-Vegfr3 signaling plays a crucial role as an endothelial-autonomous permissive cue for developmental angiogenesis.
Collapse
Affiliation(s)
- Aniket V Gore
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, 6B/3B309, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Yin A, Korzh V, Gong Z. Perturbation of zebrafish swimbladder development by enhancing Wnt signaling in Wif1 morphants. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1823:236-44. [PMID: 22008465 DOI: 10.1016/j.bbamcr.2011.09.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Revised: 09/22/2011] [Accepted: 09/26/2011] [Indexed: 01/05/2023]
Abstract
Wnt signaling plays critical roles in development of both tetrapod lung and fish swimbladder, which are the two evolutionary homologous organs. Our previous data reveal that down-regulation of Wnt signaling leads to defective swimbladder development. However, the effects of up-regulation of Wnt signaling on swimbladder development remain unclear. By knockdown of the Wnt protein inhibitory gene wif1, we demonstrated that up-regulation of Wnt signaling also resulted in perturbed development of the swimbladder. Specifically, the growth of epithelium and mesenchyme was greatly inhibited, the smooth muscle differentiation was abolished, and the organization of mesothelium was disturbed. Furthermore, our data reveal that it is the reduced cell proliferation, but not enhanced apoptosis, that contributes to the disturbance of swimbladder development in wif1 morphants. Blocking Wnt signaling by the Wnt antagonist IWR-1 did not affect wif1 expression in the swimbladder, but complete suppression of Hedgehog signaling in smo-/- mutants abolished wif expression, consistent with our earlier report of a negative feedback regulation of Wnt signaling in the swimbladder by the Hedgehog signaling. Our works established the importance of proper level of Wnt signaling for normal development of swimbladder in zebrafish.
Collapse
Affiliation(s)
- Ao Yin
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | | | | |
Collapse
|
116
|
Yin Y, Wang F, Ornitz DM. Mesothelial- and epithelial-derived FGF9 have distinct functions in the regulation of lung development. Development 2011; 138:3169-77. [PMID: 21750028 DOI: 10.1242/dev.065110] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Fibroblast growth factor (FGF) 9 is a secreted signaling molecule that is expressed in lung mesothelium and epithelium and is required for lung development. Embryos lacking FGF9 show mesenchymal hypoplasia, decreased epithelial branching and, by the end of gestation, hypoplastic lungs that cannot support life. Mesenchymal FGF signaling interacts with β-catenin-mediated WNT signaling in a feed-forward loop that functions to sustain mesenchymal FGF responsiveness and mesenchymal WNT/β-catenin signaling. During pseudoglandular stages of lung development, Wnt2a and Wnt7b are the canonical WNT ligands that activate mesenchymal WNT/β-catenin signaling, whereas FGF9 is the only known ligand that signals to mesenchymal FGF receptors (FGFRs). Here, we demonstrate that mesothelial- and epithelial-derived FGF9, mesenchymal Wnt2a and epithelial Wnt7b have unique functions in lung development in mouse. Mesothelial FGF9 and mesenchymal WNT2A are principally responsible for maintaining mesenchymal FGF-WNT/β-catenin signaling, whereas epithelial FGF9 primarily affects epithelial branching. We show that FGF signaling is primarily responsible for regulating mesenchymal proliferation, whereas β-catenin signaling is a required permissive factor for mesenchymal FGF signaling.
Collapse
Affiliation(s)
- Yongjun Yin
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | |
Collapse
|
117
|
Goss AM, Tian Y, Cheng L, Yang J, Zhou D, Cohen ED, Morrisey EE. Wnt2 signaling is necessary and sufficient to activate the airway smooth muscle program in the lung by regulating myocardin/Mrtf-B and Fgf10 expression. Dev Biol 2011; 356:541-52. [PMID: 21704027 PMCID: PMC3319016 DOI: 10.1016/j.ydbio.2011.06.011] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 06/07/2011] [Accepted: 06/08/2011] [Indexed: 02/08/2023]
Abstract
Smooth muscle in the lung is thought to derive from the developing lung mesenchyme. Smooth muscle formation relies upon coordination of both autocrine and paracrine signaling between the budding epithelium and adjacent mesenchyme to govern its proliferation and differentiation. However, the pathways initiating the earliest aspects of smooth muscle specification and differentiation in the lung are poorly understood. Here, we identify the Wnt2 ligand as a critical regulator of the earliest aspects of lung airway smooth muscle development. Using Wnt2 loss and gain of function models, we show that Wnt2 signaling is necessary and sufficient for activation of a transcriptional and signaling network critical for smooth muscle specification and differentiation including myocardin/Mrtf-B and the signaling factor Fgf10. These studies place Wnt2 high in a hierarchy of signaling molecules that promote the earliest aspects of lung airway smooth muscle development.
Collapse
Affiliation(s)
- Ashley M. Goss
- Department of Medicine University of Pennsylvania Philadelphia, PA 19104
| | - Ying Tian
- Department of Medicine University of Pennsylvania Philadelphia, PA 19104
| | - Lan Cheng
- Department of Medicine University of Pennsylvania Philadelphia, PA 19104
| | | | - Diane Zhou
- Department of Medicine University of Pennsylvania Philadelphia, PA 19104
| | - Ethan David Cohen
- Department of Medicine University of Pennsylvania Philadelphia, PA 19104
| | - Edward E. Morrisey
- Department of Medicine University of Pennsylvania Philadelphia, PA 19104
- Department of Cell and Developmental Biology University of Pennsylvania Philadelphia, PA 19104
- Cardiovascular Institute University of Pennsylvania Philadelphia, PA 19104
- Institute for Regenerative Medicine University of Pennsylvania Philadelphia, PA 19104
| |
Collapse
|
118
|
Miller RK, Canny SGDLT, Jang CW, Cho K, Ji H, Wagner DS, Jones EA, Habas R, McCrea PD. Pronephric tubulogenesis requires Daam1-mediated planar cell polarity signaling. J Am Soc Nephrol 2011; 22:1654-64. [PMID: 21804089 DOI: 10.1681/asn.2010101086] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Canonical β-catenin-mediated Wnt signaling is essential for the induction of nephron development. Noncanonical Wnt/planar cell polarity (PCP) pathways contribute to processes such as cell polarization and cytoskeletal modulation in several tissues. Although PCP components likely establish the plane of polarization in kidney tubulogenesis, whether PCP effectors directly modulate the actin cytoskeleton in tubulogenesis is unknown. Here, we investigated the roles of Wnt PCP components in cytoskeletal assembly during kidney tubule morphogenesis in Xenopus laevis and zebrafish. We found that during tubulogenesis, the developing pronephric anlagen expresses Daam1 and its interacting Rho-GEF (WGEF), which compose one PCP/noncanonical Wnt pathway branch. Knockdown of Daam1 resulted in reduced expression of late pronephric epithelial markers with no apparent effect upon early markers of patterning and determination. Inhibiting various points in the Daam1 signaling pathway significantly reduced pronephric tubulogenesis. These data indicate that pronephric tubulogenesis requires the Daam1/WGEF/Rho PCP pathway.
Collapse
Affiliation(s)
- Rachel K Miller
- Department of Biochemistry and Molecular Biology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
119
|
Reynolds PR, Stogsdill JA, Stogsdill MP, Heimann NB. Up-regulation of receptors for advanced glycation end-products by alveolar epithelium influences cytodifferentiation and causes severe lung hypoplasia. Am J Respir Cell Mol Biol 2011; 45:1195-202. [PMID: 21685154 DOI: 10.1165/rcmb.2011-0170oc] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Receptors for advanced glycation end-products (RAGE) are cell-surface receptors expressed by pulmonary tissue that influence alveolar type (AT) II-ATI transition required for normal alveolar formation. However, the precise contribution of RAGE in interactions between pulmonary epithelium and splanchnic mesenchyme during lung organogenesis remains uncertain. To test the hypothesis that RAGE misexpression adversely affects lung morphogenesis, conditional transgenic mice were generated that overexpress RAGE. Mice that overexpress RAGE throughout embryogenesis experienced 100% mortality and significant lung hypoplasia coincident with large, vacuous areas in the periphery when compared with normal airway and alveolar architecture observed in control mouse lungs. Flow cytometry and immunohistochemistry employing cell-specific markers for distal (forkhead box protein A2) and respiratory (thyroid transcription factor-1) epithelium, ATII cells (pro-surfactant protein-C), and ATI cells (T1-α) demonstrated anomalies in key epithelial cell populations resulting from RAGE up-regulation. These results reveal that precise regulation of RAGE expression is required during lung formation. Furthermore, abundant RAGE results in profound alterations in epithelial cell differentiation that culminate in severe respiratory distress and perinatal lethality.
Collapse
Affiliation(s)
- Paul R Reynolds
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 94602, USA.
| | | | | | | |
Collapse
|
120
|
Hsu YC, Osinski J, Campbell CE, Litwack ED, Wang D, Liu S, Bachurski CJ, Gronostajski RM. Mesenchymal nuclear factor I B regulates cell proliferation and epithelial differentiation during lung maturation. Dev Biol 2011; 354:242-52. [PMID: 21513708 PMCID: PMC3098902 DOI: 10.1016/j.ydbio.2011.04.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 04/04/2011] [Accepted: 04/06/2011] [Indexed: 01/01/2023]
Abstract
The Nuclear factor I (NFI) transcription factor family consists of four genes (Nfia, Nfib, Nfic and Nfix) that regulate the development of multiple organ systems in mice and humans. Nfib is expressed in both lung mesenchyme and epithelium and mice lacking Nfib have severe lung maturation defects and die at birth. Here we continue our analysis of the phenotype of Nfib⁻/⁻ lungs and show that Nfib specifically in lung mesenchyme controls late epithelial and mesenchymal cell proliferation and differentiation. There are more PCNA, BrdU, PHH3 and Ki67 positive cells in Nfib⁻/⁻ lungs than in wild type lungs at E18.5 and this increase in proliferation marker expression is seen in both epithelial and mesenchymal cells. The loss of Nfib in all lung cells decreases the expression of markers for alveolar epithelial cells (Aqp5 and Sftpc), Clara cells (Scgb1a1) and ciliated cells (Foxj1) in E18.5 lungs. To test for a specific role of Nfib in lung mesenchyme we generated and analyzed Nfib(flox/flox), Dermo1-Cre mice. Loss of Nfib only in mesenchyme results in decreased Aqp5, Sftpc and Foxj1 expression, increased cell proliferation, and a defect in sacculation similar to that seen in Nfib⁻/⁻ mice. In contrast, mesenchyme specific loss of Nfib had no effect on the expression of Scgb1a1 in the airway. Microarray and QPCR analyses indicate that the loss of Nfib in lung mesenchyme affects the expression of genes associated with extracellular matrix, cell adhesion and FGF signaling which could affect distal lung maturation. Our data indicate that mesenchymal Nfib regulates both mesenchymal and epithelial cell proliferation through multiple pathways and that mesenchymal NFI-B-mediated signals are essential for the maturation of distal lung epithelium.
Collapse
Affiliation(s)
- Yu-Chih Hsu
- Department of Biochemistry, Developmental Genomics Group, Center of Excellence in Bioinformatics and Life Science, State University of New York at Buffalo, 701 Ellicott St. Buffalo, NY 14203, USA
| | - Jason Osinski
- Department of Biochemistry, Developmental Genomics Group, Center of Excellence in Bioinformatics and Life Science, State University of New York at Buffalo, 701 Ellicott St. Buffalo, NY 14203, USA
| | - Christine E. Campbell
- Department of Biochemistry, Developmental Genomics Group, Center of Excellence in Bioinformatics and Life Science, State University of New York at Buffalo, 701 Ellicott St. Buffalo, NY 14203, USA
| | - E. David Litwack
- Dept. of Anatomy and Neurobiology and the Program in Neuroscience, University of Maryland, Baltimore, School of Medicine, HSF II, S251, 20 Penn St., Baltimore, MD 21201, USA
| | - Dan Wang
- Department of Biostatistics, Center of Excellence in Bioinformatics and Life Science, State University of New York at Buffalo, 701 Ellicott St. Buffalo, NY 14203, USA and Department of Biostatistics, Roswell Park Cancer Institute, Buffalo, NY 14203, USA
| | - Song Liu
- Department of Biostatistics, Center of Excellence in Bioinformatics and Life Science, State University of New York at Buffalo, 701 Ellicott St. Buffalo, NY 14203, USA and Department of Biostatistics, Roswell Park Cancer Institute, Buffalo, NY 14203, USA
| | - Cindy J. Bachurski
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Research Foundation, Cincinnati, OH 45229, USA
| | - Richard M. Gronostajski
- Department of Biochemistry, Developmental Genomics Group, Center of Excellence in Bioinformatics and Life Science, State University of New York at Buffalo, 701 Ellicott St. Buffalo, NY 14203, USA
| |
Collapse
|
121
|
Anokye-Danso F, Trivedi CM, Juhr D, Gupta M, Cui Z, Tian Y, Zhang Y, Yang W, Gruber PJ, Epstein JA, Morrisey EE. Highly efficient miRNA-mediated reprogramming of mouse and human somatic cells to pluripotency. Cell Stem Cell 2011; 8:376-88. [PMID: 21474102 PMCID: PMC3090650 DOI: 10.1016/j.stem.2011.03.001] [Citation(s) in RCA: 894] [Impact Index Per Article: 63.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 02/02/2011] [Accepted: 03/03/2011] [Indexed: 12/14/2022]
Abstract
Transcription factor-based cellular reprogramming has opened the way to converting somatic cells to a pluripotent state, but has faced limitations resulting from the requirement for transcription factors and the relative inefficiency of the process. We show here that expression of the miR302/367 cluster rapidly and efficiently reprograms mouse and human somatic cells to an iPSC state without a requirement for exogenous transcription factors. This miRNA-based reprogramming approach is two orders of magnitude more efficient than standard Oct4/Sox2/Klf4/Myc-mediated methods. Mouse and human miR302/367 iPSCs display similar characteristics to Oct4/Sox2/Klf4/Myc-iPSCs, including pluripotency marker expression, teratoma formation, and, for mouse cells, chimera contribution and germline contribution. We found that miR367 expression is required for miR302/367-mediated reprogramming and activates Oct4 gene expression, and that suppression of Hdac2 is also required. Thus, our data show that miRNA and Hdac-mediated pathways can cooperate in a powerful way to reprogram somatic cells to pluripotency.
Collapse
Affiliation(s)
| | - Chinmay M. Trivedi
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104
| | - Denise Juhr
- The Cardiac Center, Children’s Hospital of Philadelphia, 34 Street and Civic Center Blvd., Philadelphia, PA 19104
| | - Mudit Gupta
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104
| | - Zheng Cui
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Ying Tian
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Yuzhen Zhang
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Wenli Yang
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Peter J. Gruber
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104
- The Cardiac Center, Children’s Hospital of Philadelphia, 34 Street and Civic Center Blvd., Philadelphia, PA 19104
| | - Jonathan A. Epstein
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Edward E. Morrisey
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
122
|
Wang R, Ahmed J, Wang G, Hassan I, Strulovici-Barel Y, Hackett NR, Crystal RG. Down-regulation of the canonical Wnt β-catenin pathway in the airway epithelium of healthy smokers and smokers with COPD. PLoS One 2011; 6:e14793. [PMID: 21490961 PMCID: PMC3072378 DOI: 10.1371/journal.pone.0014793] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2010] [Accepted: 02/14/2011] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND The Wnt pathway mediates differentiation of epithelial tissues; depending on the tissue types, Wnt can either drive or inhibit the differentiation process. We hypothesized that key genes in the Wnt pathway are suppressed in the human airway epithelium under the stress of cigarette smoking, a stress associated with dysregulation of the epithelial differentiated state. METHODOLOGY/PRINCIPAL FINDINGS Microarrays were used to assess the expression of Wnt-related genes in the small airway epithelium (SAE) obtained via bronchoscopy and brushing of healthy nonsmokers, healthy smokers, and smokers with COPD. Thirty-three of 56 known Wnt-related genes were expressed in the SAE. Wnt pathway downstream mediators β-catenin and the transcription factor 7-like 1 were down-regulated in healthy smokers and smokers with COPD, as were many Wnt target genes. Among the extracellular regulators that suppress the Wnt pathway, secreted frizzled-related protein 2 (SFRP2), was up-regulated 4.3-fold in healthy smokers and 4.9-fold in COPD smokers, an observation confirmed by TaqMan Real-time PCR, Western analysis and immunohistochemistry. Finally, cigarette smoke extract mediated up-regulation of SFRP2 and down-regulation of Wnt target genes in airway epithelial cells in vitro. CONCLUSIONS/SIGNIFICANCE Smoking down-regulates the Wnt pathway in the human airway epithelium. In the context that Wnt pathway plays an important role in differentiation of epithelial tissues, the down-regulation of Wnt pathway may contribute to the dysregulation of airway epithelium differentiation observed in smoking-related airway disorders.
Collapse
Affiliation(s)
- Rui Wang
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Joumana Ahmed
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Guoqing Wang
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Ibrahim Hassan
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Yael Strulovici-Barel
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Neil R. Hackett
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Ronald G. Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| |
Collapse
|
123
|
Pacheco-Pinedo EC, Durham AC, Stewart KM, Goss AM, Lu MM, Demayo FJ, Morrisey EE. Wnt/β-catenin signaling accelerates mouse lung tumorigenesis by imposing an embryonic distal progenitor phenotype on lung epithelium. J Clin Invest 2011; 121:1935-45. [PMID: 21490395 DOI: 10.1172/jci44871] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Accepted: 02/02/2011] [Indexed: 01/04/2023] Open
Abstract
Although mutations in Kras are present in 21% of lung tumors, there is a high level of heterogeneity in phenotype and outcome among patients with lung cancer bearing similar mutations, suggesting that other pathways are important. Wnt/β-catenin signaling is a known oncogenic pathway that plays a well-defined role in colon and skin cancer; however, its role in lung cancer is unclear. We have shown here that activation of Wnt/β-catenin in the bronchiolar epithelium of the adult mouse lung does not itself promote tumor development. However, concurrent activation of Wnt/β-catenin signaling and expression of a constitutively active Kras mutant (KrasG12D) led to a dramatic increase in both overall tumor number and size compared with KrasG12D alone. Activation of Wnt/β-catenin signaling altered the KrasG12D tumor phenotype, resulting in a phenotypic switch from bronchiolar epithelium to the highly proliferative distal progenitors found in the embryonic lung. This was associated with decreased E-cadherin expression at the cell surface, which may underlie the increased metastasis of tumors with active Wnt/β-catenin signaling. Together, these data suggest that activation of Wnt/β-catenin signaling can combine with other oncogenic pathways in lung epithelium to produce a more aggressive tumor phenotype by imposing an embryonic distal progenitor phenotype and by decreasing E-cadherin expression.
Collapse
|
124
|
Tian Y, Zhang Y, Hurd L, Hannenhalli S, Liu F, Lu MM, Morrisey EE. Regulation of lung endoderm progenitor cell behavior by miR302/367. Development 2011; 138:1235-45. [PMID: 21350014 PMCID: PMC3050657 DOI: 10.1242/dev.061762] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2011] [Indexed: 11/20/2022]
Abstract
The temporal and spatial control of organ-specific endoderm progenitor development is poorly understood. miRNAs affect cell function by regulating programmatic changes in protein expression levels. We show that the miR302/367 cluster is a target of the transcription factor Gata6 in mouse lung endoderm and regulates multiple aspects of early lung endoderm progenitor development. miR302/367 is expressed at early stages of lung development, but its levels decline rapidly as development proceeds. Gain- and loss-of-function studies show that altering miR302/367 expression disrupts the balance of lung endoderm progenitor proliferation and differentiation, as well as apical-basal polarity. Increased miR302/367 expression results in the formation of an undifferentiated multi-layered lung endoderm, whereas loss of miR302/367 activity results in decreased proliferation and enhanced lung endoderm differentiation. miR302/367 coordinates the balance between proliferation and differentiation, in part, through direct regulation of Rbl2 and Cdkn1a, whereas apical-basal polarity is controlled by regulation of Tiam1 and Lis1. Thus, miR302/367 directs lung endoderm development by coordinating multiple aspects of progenitor cell behavior, including proliferation, differentiation and apical-basal polarity.
Collapse
Affiliation(s)
- Ying Tian
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yuzhen Zhang
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Laura Hurd
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sridhar Hannenhalli
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Feiyan Liu
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Min Min Lu
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward E. Morrisey
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
125
|
Wnt signaling is required for early development of zebrafish swimbladder. PLoS One 2011; 6:e18431. [PMID: 21479192 PMCID: PMC3068184 DOI: 10.1371/journal.pone.0018431] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Accepted: 03/07/2011] [Indexed: 01/15/2023] Open
Abstract
Background Wnt signaling plays critical roles in mammalian lung development. However, Wnt signaling in the development of the zebrafish swimbladder, which is considered as a counterpart of mammalian lungs, have not been explored. To investigate the potential conservation of signaling events in early development of the lung and swimbladder, we wish to address the question whether Wnt signaling plays a role in swimbladder development. Methodology/Principal Findings For analysis of zebrafish swimbladder development, we first identified, by whole-mount in situ hybridization (WISH), has2 as a mesenchymal marker, sox2 as the earliest epithelial marker, as well as hprt1l and elovl1a as the earliest mesothelial markers. We also demonstrated that genes encoding Wnt signaling members Wnt5b, Fz2, Fz7b, Lef1, Tcf3 were expressed in different layers of swimbladder. Then we utilized the heat-shock inducible transgenic lines hs:Dkk1-GFP and hs:ΔTcf-GFP to temporarily block canonical Wnt signaling. Inhibition of canonical Wnt signaling at various time points disturbed precursor cells specification, organization, anterioposterior patterning, and smooth muscle differentiation in all three tissue layers of swimbladder. These observations were also confirmed by using a chemical inhibitor (IWR-1) of Wnt signaling. In addition, we found that Hedgehog (Hh) signaling was activated by canonical Wnt signaling and imposed a negative feedback on the latter. Significance/Conclusion We first provided a new set of gene markers for the three tissue layers of swimbladder in zebrafish and demonstrated the expression of several key genes of Wnt signaling pathway in developing swimbladder. Our functional analysis data indicated that Wnt/β-catenin signaling is required for swimbladder early development and we also provided evidence for the crosstalk between Wnt and Hh signaling in early swimbladder development.
Collapse
|
126
|
Pancratov R, DasGupta R. Postgenomic technologies targeting the Wnt signaling network. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2011; 3:649-65. [PMID: 21381216 DOI: 10.1002/wsbm.140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The recent development of high-throughput sequencing technologies and the availability of whole genome sequences of a variety of living organisms, including that of humans, have led to an enormous push in the quest for a comprehensive inquiry for the function of each and every gene discovered in different model organisms. A major conclusion from the sequencing projects was that while forward genetics had been extremely successful in identifying key genes/components of many biological processes, such as signal transduction cascades, the function(s) of the majority of genes in the genome remains a mystery. In this article, we discuss the use of a variety of high-throughput postgenomic tools, including functional genomics, proteomics, and chemical genetics that are being implemented in an exhaustive molecular dissection of a key evolutionarily conserved signal transduction pathway, namely the Wnt/wingless (wg) pathway and its associated signaling network.
Collapse
Affiliation(s)
- Raluca Pancratov
- Department of Pharmacology, New York University School of Medicine and NYU Cancer Institute, New York, NY, USA
| | | |
Collapse
|
127
|
Abstract
Progress has recently been made in identifying progenitor cell populations in the embryonic lung. Some progenitor cell types have been definitively identified by lineage-tracing studies. However, others are not as well characterized and their existence is inferred on the basis of lung morphology, or mutant phenotypes. Here, I focus on lung development after the specification of the initial lung primordium. The evidence for various lung embryonic progenitor cell types is discussed and future experiments are suggested. The regulation of progenitor proliferation in the embryonic lung, and its coordinate control with morphogenesis, is also discussed. In addition, the relationship between embryonic and adult lung progenitors is considered.
Collapse
Affiliation(s)
- Emma L Rawlins
- Gurdon Institute and Department of Pathology, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
128
|
Beretta CA, Brinkmann I, Carl M. All four zebrafish Wnt7 genes are expressed during early brain development. Gene Expr Patterns 2011; 11:277-84. [PMID: 21300182 DOI: 10.1016/j.gep.2011.01.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Revised: 01/20/2011] [Accepted: 01/30/2011] [Indexed: 01/29/2023]
Abstract
Wnt-signalling is involved in a number of biological processes in the course of embryonic development, cell fate determination, proliferation, stem cell maintenance and oncogenesis. Wnt ligands are secreted glycoproteins and the number of Wnt isoforms varies between five in nematodes and 27 in fish. The highly conserved group of Wnt7 genes has been found to signal via at least three Wnt-signalling pathways dependent on the developmental context. These ligands have been identified as important regulators in a number of processes ranging from formation of bones, lungs, kidneys, reproductive organs and placenta to vasculogenesis and synaptogenesis in the brain. The importance of Wnt7 function is underscored by their implication in disease syndromes in man. Unlike the single Wnt7a and Wnt7b mammalian genes we find that the zebrafish genome contains two paralogues genes for each Wnt7 ligand. Here, we compare these four Wnt7 genes evolutionarily and analyse their expression during the first two days of embryonic development. We find Wnt7 genes mainly expressed in a number of CNS structures at developmental stages at which patterning and neural specification takes place. The timely and spatially overlapping as well as complementary gene expression suggests diverse as well as redundant involvements during brain development.
Collapse
Affiliation(s)
- Carlo A Beretta
- Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology, Ludolf-Krehl-Strasse 13-17, D-68167 Mannheim, Germany
| | | | | |
Collapse
|
129
|
Apparao KBC, Newman DR, Zhang H, Khosla J, Randell SH, Sannes PL. Temporal changes in expression of FoxA1 and Wnt7A in isolated adult human alveolar epithelial cells enhanced by heparin. Anat Rec (Hoboken) 2010; 293:938-46. [PMID: 20503388 DOI: 10.1002/ar.20805] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Pre- and postnatal developmental studies of the lung have provided compelling evidence demonstrating multiple factors that orchestrate alveolar epithelial cell differentiation. The extent to which reactivation of certain developmental pathways in the adult might influence the course of differentiation of alveolar type 2 cells (AT2) into AT1 cells is not known. In this study, we examined selected members of the forkhead (Fox) family of transcription factors and the Wnt (wingless) family of signaling proteins for expression during human alveolar cell differentiation in vitro and determined their potential responses to sulfated components of extracellular matrix (ECM), like those shed from cell surfaces or found in basement membrane and modeled by heparin. Isolated adult human AT2 cells cultured over a 9-day period were used to define the temporal profile of expression of targeted factors during spontaneous differentiation to AT1-like cells. FoxA1 protein was upregulated at early to intermediate time points, where it was strongly elevated by heparin. Gene expression of wnt7A increased dramatically beginning on day 3 and was enhanced even further on days 7 and 9 by heparin, whereas protein expression appeared at days 7 and 9. These temporal changes of expression suggest that sulfated ECMs may act to enhance the increase in FoxA1 at the critical juncture when AT2 cells commence the differentiation process to AT1 cells, in addition to enhancing the increase in wnt7A when the AT1 cell phenotype stabilizes. Collectively, these factors may act to modulate differentiation in the adult human pulmonary alveolus.
Collapse
Affiliation(s)
- K B C Apparao
- Department of Molecular Biomedical Sciences, Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | | | | | | | | | | |
Collapse
|
130
|
Zhang B, Ma JX. Wnt pathway antagonists and angiogenesis. Protein Cell 2010; 1:898-906. [PMID: 21204016 DOI: 10.1007/s13238-010-0112-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2010] [Accepted: 09/27/2010] [Indexed: 11/26/2022] Open
Abstract
Dysregulation of the Wnt pathway has been extensively studied in multiple diseases, including some angiogenic disorders. Wnt signaling activation is a major stimulator in pathological angiogenesis and thus, Wnt antagonists are believed to have therapeutic potential for neovascular disorders. Actually, some Wnt antagonists have been identified directly from the anti-angiogenic factor family. This review summarizes the recent progress toward understanding of the roles of Wnt pathway antagonists in angiogenic regulation and their mechanism of action, and exploring their therapeutic potential.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | |
Collapse
|
131
|
Takayasu H, Murphy P, Sato H, Doi T, Puri P. Embryonic Wnt gene expression in the nitrofen-induced hypoplastic lung using 3-dimensional imaging. J Pediatr Surg 2010; 45:2129-35. [PMID: 21034933 DOI: 10.1016/j.jpedsurg.2010.06.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Revised: 06/24/2010] [Accepted: 06/28/2010] [Indexed: 11/16/2022]
Abstract
PURPOSE Wnts have been reported to play a key role in the lung morphogenesis. We have previously reported that pulmonary gene expression of Wnt2 and Wnt7b is downregulated on day 15 of gestation in the nitrofen-induced congenital diaphragmatic hernia (CDH) model. However, the distribution pattern of gene expression of Wnts in the very early lung development remains unclear. Optical projection tomography (OPT) is a new technique for 3-dimensional imaging of small developing organs and gene distribution combined with whole-mount in situ hybridization. We designed this study to investigate the distribution pattern of Wnts gene expression in lung buds of nitrofen-induced CDH model using OPT. METHODS Embryos from normal and nitrofen-treated dams were harvested on embryonic day 10 (E10), and divided into controls and nitrofen group, respectively. Whole-mount in situ hybridization to detect transcripts of Wnt2 and Wnt7b was performed, analyzed, and reconstructed using OPT. RESULTS The expression of Wnt2 transcripts was detected in the lung bud mesenchyme and markedly diminished in nitrofen group compared to controls, whereas Wnt7b transcripts were expressed in the mesoderm of bronchi and the lung bud with no detectable difference between 2 groups. CONCLUSION We provide evidence for the first time that Wnt2 expression is downregulated at lung bud stage in the nitrofen model. Optical projection tomography is potentially a useful approach to visualize both gene expression and morphology during very early stages of lung development.
Collapse
Affiliation(s)
- Hajime Takayasu
- The Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland
| | | | | | | | | |
Collapse
|
132
|
Abstract
During the development of the pulmonary vasculature in the fetus, many structural and functional changes occur to prepare the lung for the transition to air breathing. The development of the pulmonary circulation is genetically controlled by an array of mitogenic factors in a temporo-spatial order. With advancing gestation, pulmonary vessels acquire increased vasoreactivity. The fetal pulmonary vasculature is exposed to a low oxygen tension environment that promotes high intrinsic myogenic tone and high vasocontractility. At birth, a dramatic reduction in pulmonary arterial pressure and resistance occurs with an increase in oxygen tension and blood flow. The striking hemodynamic differences in the pulmonary circulation of the fetus and newborn are regulated by various factors and vasoactive agents. Among them, nitric oxide, endothelin-1, and prostaglandin I2 are mainly derived from endothelial cells and exert their effects via cGMP, cAMP, and Rho kinase signaling pathways. Alterations in these signaling pathways may lead to vascular remodeling, high vasocontractility, and persistent pulmonary hypertension of the newborn.
Collapse
Affiliation(s)
- Yuansheng Gao
- Department of Physiology and Pathophysiology, Peking University, Health Science Center, Beijing, China; and Department of Pediatrics, University of Illinois, College of Medicine at Chicago, Chicago, Illinois
| | - J. Usha Raj
- Department of Physiology and Pathophysiology, Peking University, Health Science Center, Beijing, China; and Department of Pediatrics, University of Illinois, College of Medicine at Chicago, Chicago, Illinois
| |
Collapse
|
133
|
Kneidinger N, Yildirim AÖ, Callegari J, Takenaka S, Stein MM, Dumitrascu R, Bohla A, Bracke KR, Morty RE, Brusselle GG, Schermuly RT, Eickelberg O, Königshoff M. Activation of the WNT/β-catenin pathway attenuates experimental emphysema. Am J Respir Crit Care Med 2010; 183:723-33. [PMID: 20889911 DOI: 10.1164/rccm.200910-1560oc] [Citation(s) in RCA: 143] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Chronic obstructive pulmonary disease (COPD) is a devastating disease, for which no causal therapy is available. OBJECTIVES To characterize WNT/β-catenin signaling in COPD in humans and elucidate its potential role as a preventive and therapeutic target in experimental emphysema in mice. METHODS The expression, localization, and activity of WNT/β-catenin signaling was assessed in 12 COPD and 12 transplant donor samples using quantitative reverse transcriptase polymerase chain reaction, immunohistochemistry, and Western blotting. The role of WNT/β-catenin signaling was assessed in elastase- and cigarette smoke-induced emphysema and therapeutic modulation thereof in elastase-induced emphysema in TOPGAL reporter and wild-type mice in vivo. MEASUREMENTS AND MAIN RESULTS No differences in the mRNA expression profile of the main WNT/β-catenin signaling components were observed comparing COPD and donor lung homogenates. Immunohistochemical analysis revealed reduced numbers of nuclear β-catenin-positive alveolar epithelial cells in COPD. Similarly, WNT/β-catenin signaling was down-regulated in both experimental emphysema models. Preventive and therapeutic, WNT/β-catenin activation by lithium chloride attenuated experimental emphysema, as assessed by decreased airspace enlargement, improved lung function, reduced collagen content, and elevated expression of alveolar epithelial cell markers. CONCLUSIONS Decreased WNT/β-catenin signaling is involved in parenchymal tissue destruction and impaired repair capacity in emphysema. These data indicate a crucial role of WNT/β-catenin signaling in lung repair mechanisms in vivo, and highlight WNT/β-catenin activation as a future therapeutic approach for emphysema.
Collapse
Affiliation(s)
- Nikolaus Kneidinger
- Department of Medicine, University of Giessen Lung Center, University of Giessen, Giessen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Foronjy R, Imai K, Shiomi T, Mercer B, Sklepkiewicz P, Thankachen J, Bodine P, D'Armiento J. The divergent roles of secreted frizzled related protein-1 (SFRP1) in lung morphogenesis and emphysema. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:598-607. [PMID: 20595636 PMCID: PMC2913334 DOI: 10.2353/ajpath.2010.090803] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/13/2010] [Indexed: 11/20/2022]
Abstract
Developmentally expressed genes are believed to play a central role in tissue repair after injury; however, in lung disease their role has not been established. This study demonstrates that SFRP1, an inhibitor of Wnt signaling normally expressed during lung embryogenesis, is induced in the lungs of emphysema patients and in two murine models of the disease. SFRP1 was found to be essential for alveolar formation as Sfrp1(-/-) mice exhibited aberrant Wnt signaling, mesenchymal proliferation, and impaired alveoli formation. In contrast, SFRP1 activated ERK and up-regulated MMP1 and MMP9 without altering TIMP1 production when expressed in human lung epithelial cells. These findings demonstrate that SFRP1 promotes normal alveolar formation in lung development, although its expression in the adult up-regulates proteins that can cause tissue destruction. Thus, SFRP1 induction during tissue injury is unlikely to contribute to the repair response but rather is a participatory factor in the pathogenesis of emphysema and tissue destruction.
Collapse
Affiliation(s)
- Robert Foronjy
- Department of Medicine, Columbia University, New York, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
135
|
Yin A, Winata CL, Korzh S, Korzh V, Gong Z. Expression of components of Wnt and Hedgehog pathways in different tissue layers during lung development in Xenopus laevis. Gene Expr Patterns 2010; 10:338-44. [PMID: 20682360 DOI: 10.1016/j.gep.2010.07.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 07/21/2010] [Accepted: 07/24/2010] [Indexed: 11/19/2022]
Abstract
Although Wnt and Hedgehog (Hh) signaling pathways play important roles in mouse lung development, these have not been explored in the development of Xenopus lung. This may be due to the lack of specific molecular markers for different layers of tissue in Xenopus lung and/or insufficient knowledge on expression patterns of Wnt and Hh signaling components in Xenopus lung. In this study, we first described the early morphogenesis of Xenopus laevis lung by using surfactant protein C (sftpc) as a marker of lung epithelium and compared it with the expression patterns of several genes of Wnt and Hh pathways in Xenopus lungs. Our data showed that wnt7b was expressed in the entire lung epithelium from stage 37 to stage 45, while two other Wnt signaling components, wnt5a and wif1 (wnt inhibitory factor 1), were expressed in the mesenchyme layer of the entire lungs through stages 39-41. We also found that sonic hedgehog (shh) was expressed at stage 41 only in the anterior, but not in the posterior part of the lungs. These results show the expression of wnt5a, wnt7b, wif1 and shh in different layers of tissue of Xenopus lungs at early developmental stages, which implies different roles of these genes in the early development of Xenopus lungs. Our study for the first time defined specific molecular markers for description of early lung development in Xenopus, as well as provided information about expression of components of Wnt and Hh pathways in early Xenopus lungs, which should be useful for future functional studies.
Collapse
Affiliation(s)
- Ao Yin
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | | | | | | | | |
Collapse
|
136
|
Abstract
Wls/Evi/Srt encoding a multipass transmembrane protein has been identified as a regulator for proper sorting and secretion of Wnt in flies. We have previously demonstrated that Gpr177 is the mouse ortholog required for axis determination. Gpr177 is a transcriptional target of Wnt that is activated to assist its subcellular distribution in a feedback regulatory loop. We, therefore, proposed that reciprocal regulation of Wnt and Gpr177 is essential for the Wnt-dependent developmental and pathogenic processes. Here, we examine the expression pattern of Gpr177 in mouse development. Gpr177 is expressed in a variety of tissues and cell types during organogenesis. Furthermore, Gpr177 is a glycoprotein primarily accumulating in the Golgi apparatus in signal-producing cells. The glycosylation of Gpr177 is necessary for proper transportation in the secretory pathway. Our findings suggest that the Gpr177-mediated regulation of Wnt is crucial for organogenesis in health and disease.
Collapse
Affiliation(s)
- Hsiao-Man Ivy Yu
- Department of Biomedical Genetics, Center for Oral Biology, James Wilmot Cancer Center, University of Rochester Medical Center, 601 Elmwood Avenue, Box 611, Rochester, NY 14642
| | - Ying Jin
- Department of Biomedical Genetics, Center for Oral Biology, James Wilmot Cancer Center, University of Rochester Medical Center, 601 Elmwood Avenue, Box 611, Rochester, NY 14642
| | - Jiang Fu
- Department of Biomedical Genetics, Center for Oral Biology, James Wilmot Cancer Center, University of Rochester Medical Center, 601 Elmwood Avenue, Box 611, Rochester, NY 14642
| | - Wei Hsu
- Department of Biomedical Genetics, Center for Oral Biology, James Wilmot Cancer Center, University of Rochester Medical Center, 601 Elmwood Avenue, Box 611, Rochester, NY 14642
| |
Collapse
|
137
|
Glaw JT, Skalak TC, Peirce SM. Inhibition of canonical Wnt signaling increases microvascular hemorrhaging and venular remodeling in adult rats. Microcirculation 2010; 17:348-57. [PMID: 20618692 PMCID: PMC2904644 DOI: 10.1111/j.1549-8719.2010.00036.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE The canonical Wnt signaling pathway, heavily studied in development and cancer, has recently been implicated in microvascular growth with the use of developmental and in vitro models. To date, however, no study exists showing the effects of perturbing the canonical Wnt pathway in a complete microvascular network undergoing physiological remodeling in vivo. Our objective was to investigate the effects of canonical Wnt inhibition on the microvascular remodeling of adult rats. METHODS Canonical Wnt inhibitor DKK-1, Wnt inhibitor sFRP-1, BSA or saline was superfused onto the exteriorized mesenteric windows of 300 g adult female Sprague-Dawley rats for 20 minutes. Three days following surgery, mesenteric windows were imaged intravitally and harvested for immunofluorescence staining with smooth muscle alpha-actin and BRDU. RESULTS We observed prominent differences in the response of the mesenteric microvasculature amongst the various treatment groups. Significant increases in hemorrhage area, vascular density, and draining vessel diameter were observed in windows treated with Wnt inhibitors as compared to control-treated windows. Additionally, confocal imaging analysis showed significant increases in proliferating cells as well as evidence of proliferating smooth muscle cells along venules. CONCLUSIONS Together, our results suggest that canonical Wnt inhibition plays an important role in microvascular remodeling, specifically venular remodeling.
Collapse
Affiliation(s)
- Jason T Glaw
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908, USA
| | | | | |
Collapse
|
138
|
Weng T, Liu L. The role of pleiotrophin and beta-catenin in fetal lung development. Respir Res 2010; 11:80. [PMID: 20565841 PMCID: PMC2901351 DOI: 10.1186/1465-9921-11-80] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 06/18/2010] [Indexed: 12/21/2022] Open
Abstract
Mammalian lung development is a complex biological process, which is temporally and spatially regulated by growth factors, hormones, and extracellular matrix proteins. Abnormal changes of these molecules often lead to impaired lung development, and thus pulmonary diseases. Epithelial-mesenchymal interactions are crucial for fetal lung development. This paper reviews two interconnected pathways, pleiotrophin and Wnt/β-catenin, which are involved in fibroblast and epithelial cell communication during fetal lung development.
Collapse
Affiliation(s)
- Tingting Weng
- Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma 74078, USA
| | | |
Collapse
|
139
|
Abstract
Cardiac development is comprised of a series of morphological events tightly controlled both spatially and temporally. The molecular pathways controlling early cardiac differentiation are poorly understood, but Wnt signaling is emerging as a critical pathway for multiple aspects of early cardiovascular development. The Wnt pathway plays multiple roles in regulating cellular behavior including proliferation, differentiation, cell migration, and cell polarity. Recent data have demonstrated that Wnt activity is important for early precardiac mesoderm differentiation but must be inhibited in subsequent steps for cardiomyocyte differentiation to proceed. Given the important role that Wnt signaling plays in both the differentiation of cardiomyocytes from pluripotential stem cells and tissue regeneration in general, an increased understanding of this pathway is likely to enhance our knowledge about both cardiovascular development and reparative mechanisms.
Collapse
|
140
|
Abstract
The mammalian respiratory system--the trachea and the lungs--arises from the anterior foregut through a sequence of morphogenetic events involving reciprocal endodermal-mesodermal interactions. The lung itself consists of two highly branched, tree-like systems--the airways and the vasculature--that develop in a coordinated way from the primary bud stage to the generation of millions of alveolar gas exchange units. We are beginning to understand some of the molecular and cellular mechanisms that underlie critical processes such as branching morphogenesis, vascular development, and the differentiation of multipotent progenitor populations. Nevertheless, many gaps remain in our knowledge, the filling of which is essential for understanding respiratory disorders, congenital defects in human neonates, and how the disruption of morphogenetic programs early in lung development can lead to deficiencies that persist throughout life.
Collapse
|
141
|
Tian Y, Yuan L, Goss AM, Wang T, Yang J, Lepore JJ, Zhou D, Schwartz RJ, Patel V, Cohen ED, Morrisey EE. Characterization and in vivo pharmacological rescue of a Wnt2-Gata6 pathway required for cardiac inflow tract development. Dev Cell 2010; 18:275-87. [PMID: 20159597 PMCID: PMC2846539 DOI: 10.1016/j.devcel.2010.01.008] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 12/10/2009] [Accepted: 01/11/2010] [Indexed: 12/17/2022]
Abstract
Little is understood about the molecular mechanisms underlying the morphogenesis of the posterior pole of the heart. Here we show that Wnt2 is expressed specifically in the developing inflow tract mesoderm, which generates portions of the atria and atrio-ventricular canal. Loss of Wnt2 results in defective development of the posterior pole of the heart, resulting in a phenotype resembling the human congenital heart syndrome complete common atrio-ventricular canal. The number and proliferation of posterior second heart field progenitors is reduced in Wnt2(-/-) mutants. Moreover, these defects can be rescued in a temporally restricted manner through pharmacological inhibition of Gsk-3beta. We also show that Wnt2 works in a feedforward transcriptional loop with Gata6 to regulate posterior cardiac development. These data reveal a molecular pathway regulating the posterior cardiac mesoderm and demonstrate that cardiovascular defects caused by loss of Wnt signaling can be rescued pharmacologically in vivo.
Collapse
Affiliation(s)
- Ying Tian
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Lijun Yuan
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Department of Ultrasound Diagnostics, Tangdu Hospital, Forth Military Medical University, Xi'an, China
| | - Ashley M. Goss
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Tao Wang
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Jifu Yang
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - John J. Lepore
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Diane Zhou
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Robert J. Schwartz
- Institute of Biosciences and Technology Texas A & M Health Science Center Houston, Texas 77030, USA
| | - Vickas Patel
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104
| | - Ethan David Cohen
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Edward E. Morrisey
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA 19104
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
142
|
Wang HX, Li TY, Kidder GM. WNT2 regulates DNA synthesis in mouse granulosa cells through beta-catenin. Biol Reprod 2010; 82:865-75. [PMID: 20107203 DOI: 10.1095/biolreprod.109.080903] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
WNTs are secreted extracellular signaling molecules that transduce their signals by binding to G protein-coupled receptors of the frizzled (FZD) family. They control diverse developmental processes, such as cell fate specification, cell proliferation, cell differentiation, and apoptosis. Although WNT signaling has been shown to be essential for development of the ovary, its mechanistic role in folliculogenesis within the adult ovary has not been studied extensively. Therefore, the objective of this study was to investigate the regulation and function of WNT2 signaling in mouse granulosa cells. Immunostaining identified WNT2 as being expressed in granulosa cells throughout folliculogenesis, but with varying signal strength: in sequential sections, WNT2 immunoreactivity was strongest in healthy antral follicles but weak in atretic follicles. Knockdown of WNT2 expression using transfected short interfering RNA decreased DNA synthesis in granulosa cells, whereas WNT2 overexpression using a recombinant viral vector enhanced it. WNT2 knockdown led to accumulation of glycogen synthase kinase-3beta (GSK3B) in the cytoplasm but reduced the expression of beta-catenin. Conversely, WNT2 overexpression reduced the expression of GSK3B in the cytoplasm and induced beta-catenin translocation from the membrane into the nucleus. Beta-catenin knockdown also inhibited DNA synthesis in granulosa cells and neutralized the effect of WNT2 overexpression. WNT2/beta-catenin signaling had a slight effect on the apoptosis of granulosa cells. Taken together, the data indicate that WNT2 regulates beta-catenin localization in granulosa cells, and WNT2/beta-catenin signaling contributes to regulating their proliferation.
Collapse
Affiliation(s)
- Hong-Xing Wang
- Departments of Physiology and Pharmacology, Obstetrics and Gynecology, and Pediatrics, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | | | | |
Collapse
|
143
|
Warburton D, El-Hashash A, Carraro G, Tiozzo C, Sala F, Rogers O, De Langhe S, Kemp PJ, Riccardi D, Torday J, Bellusci S, Shi W, Lubkin SR, Jesudason E. Lung organogenesis. Curr Top Dev Biol 2010; 90:73-158. [PMID: 20691848 PMCID: PMC3340128 DOI: 10.1016/s0070-2153(10)90003-3] [Citation(s) in RCA: 303] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Developmental lung biology is a field that has the potential for significant human impact: lung disease at the extremes of age continues to cause major morbidity and mortality worldwide. Understanding how the lung develops holds the promise that investigators can use this knowledge to aid lung repair and regeneration. In the decade since the "molecular embryology" of the lung was first comprehensively reviewed, new challenges have emerged-and it is on these that we focus the current review. Firstly, there is a critical need to understand the progenitor cell biology of the lung in order to exploit the potential of stem cells for the treatment of lung disease. Secondly, the current familiar descriptions of lung morphogenesis governed by growth and transcription factors need to be elaborated upon with the reinclusion and reconsideration of other factors, such as mechanics, in lung growth. Thirdly, efforts to parse the finer detail of lung bud signaling may need to be combined with broader consideration of overarching mechanisms that may be therapeutically easier to target: in this arena, we advance the proposal that looking at the lung in general (and branching in particular) in terms of clocks may yield unexpected benefits.
Collapse
Affiliation(s)
- David Warburton
- The Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Abstract
Epithelial tubes are crucial to the function of organ systems including the cardiovascular system, pulmonary system, gastrointestinal tract, reproductive organ systems, excretory system, and auditory system. Using a variety of animal model systems, recent studies have substantiated the role of Wnt signaling via the canonical/beta-catenin-mediated trajectory, the non-canonical Wnt trajectories, or both, in forming epithelial tubular tissues. This review focuses on the involvement of the Wnt pathways in the induction, specification, proliferation, and morphogenesis involved in tubulogenesis within tissues including the lungs, kidneys, ears, mammary glands, gut, and heart. The ultimate goal is to describe the developmental processes forming the various tubulogenic organ systems to determine the relationships between these processes.
Collapse
Affiliation(s)
- Rachel K Miller
- Department of Biochemistry and Molecular Biology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA.
| | | |
Collapse
|
145
|
Ringvall M, Kjellén L. Mice deficient in heparan sulfate N-deacetylase/N-sulfotransferase 1. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010; 93:35-58. [PMID: 20807640 DOI: 10.1016/s1877-1173(10)93003-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ndsts (N-deacetylase/N-sulfotransferases) are enzymes responsible for N-sulfation during heparan sulfate (HS) and heparin biosynthesis. In this review, basic features of the Ndst1 enzyme are covered and a brief description of HS biosynthesis and its regulation is presented. Effects of Ndst1 deficiency on embryonic development are described. These include immature lungs, craniofacial dysplasia and eye developmental defects, branching defect during lacrimal gland development, delayed mineralization of the skeleton, and reduced pericyte recruitment during vascular development. A brief account of the effects of Ndst1 deficiency in selective cell types in adult mice is also given.
Collapse
Affiliation(s)
- Maria Ringvall
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
146
|
Alfieri CM, Cheek J, Chakraborty S, Yutzey KE. Wnt signaling in heart valve development and osteogenic gene induction. Dev Biol 2009; 338:127-35. [PMID: 19961844 DOI: 10.1016/j.ydbio.2009.11.030] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Revised: 10/23/2009] [Accepted: 11/13/2009] [Indexed: 11/25/2022]
Abstract
Wnt signaling mediated by beta-catenin has been implicated in early endocardial cushion development, but its roles in later stages of heart valve maturation and homeostasis have not been identified. Multiple Wnt ligands and pathway genes are differentially expressed during heart valve development. At E12.5, Wnt2 is expressed in cushion mesenchyme, whereas Wnt4 and Wnt9b are predominant in overlying endothelial cells. At E17.5, both Wnt3a and Wnt7b are expressed in the remodeling atrioventricular (AV) and semilunar valves. In addition, the TOPGAL Wnt reporter transgene is active throughout the developing AV and semilunar valves at E16.5, with more localized expression in the stratified valve leaflets after birth. In chicken embryo aortic valves, genes characteristic of osteogenic cell lineages including periostin, osteonectin, and Id2 are expressed specifically in the collagen-rich fibrosa layer at E14. Treatment of E14 aortic valve interstitial cells (VICs) in culture with osteogenic media results in increased expression of multiple genes associated with bone formation. Treatment of VIC with Wnt3a leads to nuclear localization of beta-catenin and induction of periostin and matrix gla protein but does not induce genes associated with later stages of osteogenesis. Together, these studies provide evidence for Wnt signaling as a regulator of endocardial cushion maturation as well as valve leaflet stratification, homeostasis, and pathogenesis.
Collapse
Affiliation(s)
- Christina M Alfieri
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Medical Center, ML 7020, 240 Albert Sabin Way, Cincinnati, OH 45229, USA
| | | | | | | |
Collapse
|
147
|
Han W, Wang W, Mohammed KA, Su Y. Alpha-defensins increase lung fibroblast proliferation and collagen synthesis via the beta-catenin signaling pathway. FEBS J 2009; 276:6603-14. [PMID: 19814765 PMCID: PMC2879066 DOI: 10.1111/j.1742-4658.2009.07370.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Alpha-defensins are released from granules of leukocytes and are implicated in inflammatory and fibrotic lung diseases. In the present study, the effects of alpha-defensins on the proliferation and collagen synthesis of lung fibroblasts were examined. We found that alpha-defensin-1 and alpha-defensin-2 induced dose-dependent increases in the incorporation of 5-bromo-2'-deoxy-uridine into newly synthesized DNA in two lines of human lung fibroblasts (HFL-1 and LL-86), suggesting that alpha-defensin-1 and alpha-defensin-2 stimulate the proliferation of lung fibroblasts. alpha-defensin-1 and alpha-defensin-2 also increased collagen-I mRNA (COL1A1) levels and protein contents of collagen-I and active/dephosphorylated beta-catenin without changes in total beta-catenin protein content in lung fibroblasts (HFL-1 and LL-86). Inhibition of the beta-catenin signaling pathway using quercetin prevented increases in cell proliferation and the protein content of collagen-I and active/dephosphorylated beta-catenin in lung fibroblasts, and in COL1A1 mRNA levels and collagen release into culture medium induced by alpha-defensin-1 and alpha-defensin-2. Knocking-down beta-catenin using small interfering RNA technology also prevented alpha-defensin-induced increases in cell proliferation and the protein content of collagen-I and active/dephosphorylated beta-catenin in lung fibroblasts, and in COL1A1 mRNA levels. Moreover, increases in the phosphorylation of glycogen synthase kinase 3beta, accumulation/activation of beta-catenin, and collagen synthesis induced by alpha-defensin-1 and alpha-defensin-2 were prevented by p38 mitogen-activated protein kinase inhibitor SB203580 and phosphoinositide 3-kinase inhibitor LY294002. These results indicate that alpha-defensin-1 and alpha-defensin-2 stimulate proliferation and collagen synthesis of lung fibroblasts. The beta-catenin signaling pathway mediates alpha-defensin-induced increases in cell proliferation and collagen synthesis of lung fibroblasts. alpha-defensin-induced activation of beta-catenin in lung fibroblasts might be caused by phosphorylation/inactivation of glycogen synthase kinase 3beta as a result of the activation of the p38 mitogen-activated protein kinase and phosphoinositide 3-kinase/Akt pathways.
Collapse
Affiliation(s)
- Weihong Han
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta, GA 30912, USA
| | | | | | | |
Collapse
|
148
|
Goss AM, Tian Y, Tsukiyama T, Cohen ED, Zhou D, Lu MM, Yamaguchi TP, Morrisey EE. Wnt2/2b and beta-catenin signaling are necessary and sufficient to specify lung progenitors in the foregut. Dev Cell 2009; 17:290-8. [PMID: 19686689 DOI: 10.1016/j.devcel.2009.06.005] [Citation(s) in RCA: 329] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Revised: 04/14/2009] [Accepted: 06/02/2009] [Indexed: 01/04/2023]
Abstract
Patterning of the primitive foregut promotes appropriate organ specification along its anterior-posterior axis. However, the molecular pathways specifying foregut endoderm progenitors are poorly understood. We show here that Wnt2/2b signaling is required to specify lung endoderm progenitors within the anterior foregut. Embryos lacking Wnt2/2b expression exhibit complete lung agenesis and do not express Nkx2.1, the earliest marker of the lung endoderm. In contrast, other foregut endoderm-derived organs, including the thyroid, liver, and pancreas, are correctly specified. The phenotype observed is recapitulated by an endoderm-restricted deletion of beta-catenin, demonstrating that Wnt2/2b signaling through the canonical Wnt pathway is required to specify lung endoderm progenitors within the foregut. Moreover, activation of canonical Wnt/beta-catenin signaling results in the reprogramming of esophagus and stomach endoderm to a lung endoderm progenitor fate. Together, these data reveal that canonical Wnt2/2b signaling is required for the specification of lung endoderm progenitors in the developing foregut.
Collapse
Affiliation(s)
- Ashley M Goss
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
149
|
Ustiyan V, Wang IC, Ren X, Zhang Y, Snyder J, Xu Y, Wert SE, Lessard JL, Kalin TV, Kalinichenko VV. Forkhead box M1 transcriptional factor is required for smooth muscle cells during embryonic development of blood vessels and esophagus. Dev Biol 2009; 336:266-79. [PMID: 19835856 DOI: 10.1016/j.ydbio.2009.10.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Revised: 09/17/2009] [Accepted: 10/06/2009] [Indexed: 12/16/2022]
Abstract
The forkhead box m1 (Foxm1 or Foxm1b) transcription factor (previously called HFH-11B, Trident, Win, or MPP2) is expressed in a variety of tissues during embryogenesis, including vascular, airway, and intestinal smooth muscle cells (SMCs). Although global deletion of Foxm1 in Foxm1(-/-) mice is lethal in the embryonic period due to multiple abnormalities in the liver, heart, and lung, the specific role of Foxm1 in SMC remains unknown. In the present study, Foxm1 was deleted conditionally in the developing SMC (smFoxm1(-/-) mice). The majority of smFoxm1(-/-) mice died immediately after birth due to severe pulmonary hemorrhage and structural defects in arterial wall and esophagus. Although Foxm1 deletion did not influence SMC differentiation, decreased proliferation of SMC was found in smFoxm1(-/-) blood vessels and esophagus. Depletion of Foxm1 in cultured SMC caused G(2) arrest and decreased numbers of cells undergoing mitosis. Foxm1-deficiency in vitro and in vivo was associated with reduced expression of cell cycle regulatory genes, including cyclin B1, Cdk1-activator Cdc25b phosphatase, Polo-like 1 and JNK1 kinases, and cMyc transcription factor. Foxm1 is critical for proliferation of smooth muscle cells and is required for proper embryonic development of blood vessels and esophagus.
Collapse
Affiliation(s)
- Vladimir Ustiyan
- Divisions of Pulmonary Biology, Perinatal Institute of the Cincinnati Children's Hospital Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Franco CA, Liebner S, Gerhardt H. Vascular morphogenesis: a Wnt for every vessel? Curr Opin Genet Dev 2009; 19:476-83. [DOI: 10.1016/j.gde.2009.09.004] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Accepted: 09/15/2009] [Indexed: 01/24/2023]
|