101
|
Abstract
Nuclear accumulation of active Smad complexes is crucial for transduction of transforming growth factor beta (TGF-beta)-superfamily signals from transmembrane receptors into the nucleus. It is now clear that the nucleocytoplasmic distributions of Smads, in both the absence and the presence of a TGF-beta-superfamily signal, are not static, but instead the Smads are continuously shuttling between the nucleus and the cytoplasm in both conditions. This article presents the evidence for continuous nucleocytoplasmic shuttling of Smads. It then reviews different mechanisms that have been proposed to mediate Smad nuclear import and export, and discusses how the Smad steady-state distributions in the absence and the presence of a TGF-beta-superfamily signal are established. Finally, the biological relevance of continuous nucleocytoplasmic shuttling for signaling by TGF-beta superfamily members is discussed.
Collapse
Affiliation(s)
- Caroline S Hill
- Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3PX, UK.
| |
Collapse
|
102
|
Schnaper HW, Jandeska S, Runyan CE, Hubchak SC, Basu RK, Curley JF, Smith RD, Hayashida T. TGF-beta signal transduction in chronic kidney disease. Front Biosci (Landmark Ed) 2009; 14:2448-65. [PMID: 19273211 DOI: 10.2741/3389] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Transforming growth factor (TGF)-beta is a central stimulus of the events leading to chronic progressive kidney disease, having been implicated in the regulation of cell proliferation, hypertrophy, apoptosis and fibrogenesis. The fact that it mediates these varied events suggests that multiple mechanisms play a role in determining the outcome of TGF-beta signaling. Regulation begins with the availability and activation of TGF-beta and continues through receptor expression and localization, control of the TGF-beta family-specific Smad signaling proteins, and interaction of the Smads with multiple signaling pathways extending into the nucleus. Studies of these mechanisms in kidney cells and in whole-animal experimental models, reviewed here, are beginning to provide insight into the role of TGF-beta in the pathogenesis of renal dysfunction and its potential treatment.
Collapse
Affiliation(s)
- H William Schnaper
- Division of Kidney Diseases, Department of Pediatrics, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave.; Chicago, IL 60611-3008, USA.
| | | | | | | | | | | | | | | |
Collapse
|
103
|
An N-terminally truncated Smad2 protein can partially compensate for loss of full-length Smad2. Biochem J 2008; 417:205-12. [DOI: 10.1042/bj20080014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
TGFβ (transforming growth factor β) superfamily signalling is critical both for early embryonic development and later for tissue homoeostasis in adult organisms. The use of gene-disruption techniques in mice has been essential to understanding the functional roles of the components of the pathways downstream of TGFβ superfamily ligands, in particular, the receptors and the Smads that transduce signals from the plasma membrane to the nucleus. Smad2 functions downstream of TGFβ, Activin and Nodal, and a number of Smad2 mutant mice have been generated by different laboratories. Although in all cases these Smad2-deficient mice were embryonic lethal, those created by deletion of the first coding exon survived longer than those generated by replacing part of the MH (Mad homology) 1 domain or deleting all or part of the MH2 domain. Moreover, they displayed a less severe phenotype, as they were capable of transiently inducing mesoderm. In the present study, we show that embryonic fibroblasts taken from the Smad2 mutant mice created by deletion of the first coding exon express a small amount of an N-terminally truncated Smad2 protein. We show this protein results from internal initiation at Met241 and encodes the entire MH2 domain and the C-terminal part of the linker. We demonstrate that this protein is incorporated into Smad heteromeric complexes, can interact with DNA-binding transcription factors and thereby can mediate TGFβ-induced transcriptional activation from a number of TGFβ-responsive elements. We propose that this functional truncated Smad2 protein can partially compensate for the loss of full-length Smad2, thereby providing an explanation for the differing phenotypes of Smad2 mutant mice.
Collapse
|
104
|
Miles WO, Jaffray E, Campbell SG, Takeda S, Bayston LJ, Basu SP, Li M, Raftery LA, Ashe MP, Hay RT, Ashe HL. Medea SUMOylation restricts the signaling range of the Dpp morphogen in the Drosophila embryo. Genes Dev 2008; 22:2578-90. [PMID: 18794353 PMCID: PMC2546696 DOI: 10.1101/gad.494808] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Accepted: 07/08/2008] [Indexed: 12/17/2022]
Abstract
Morphogens are secreted signaling molecules that form concentration gradients and control cell fate in developing tissues. During development, it is essential that morphogen range is strictly regulated in order for correct cell type specification to occur. One of the best characterized morphogens is Drosophila Decapentaplegic (Dpp), a BMP signaling molecule that patterns the dorsal ectoderm of the embryo by activating the Mad and Medea (Med) transcription factors. We demonstrate that there is a spatial and temporal expansion of the expression patterns of Dpp target genes in SUMO pathway mutant embryos. We identify Med as the primary SUMOylation target in the Dpp pathway, and show that failure to SUMOylate Med leads to the increased Dpp signaling range observed in the SUMO pathway mutant embryos. Med is SUMO modified in the nucleus, and we provide evidence that SUMOylation triggers Med nuclear export. Hence, Med SUMOylation provides a mechanism by which nuclei can continue to monitor the presence of extracellular Dpp signal to activate target gene expression for an appropriate duration. Overall, our results identify an unusual strategy for regulating morphogen range that, rather than impacting on the morphogen itself, targets an intracellular transducer.
Collapse
Affiliation(s)
- Wayne O. Miles
- Faculty of Life Sciences, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Ellis Jaffray
- School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Susan G. Campbell
- Faculty of Life Sciences, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Shugaku Takeda
- Faculty of Life Sciences, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Laura J. Bayston
- Faculty of Life Sciences, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Sanjay P. Basu
- Faculty of Life Sciences, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Mingfa Li
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02109, USA
| | - Laurel A. Raftery
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02109, USA
| | - Mark P. Ashe
- Faculty of Life Sciences, The University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Ronald T. Hay
- School of Life Sciences, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Hilary L. Ashe
- Faculty of Life Sciences, The University of Manchester, Manchester, M13 9PT, United Kingdom
| |
Collapse
|
105
|
Wang B, Suzuki H, Kato M. Roles of mono-ubiquitinated Smad4 in the formation of Smad transcriptional complexes. Biochem Biophys Res Commun 2008; 376:288-92. [PMID: 18783722 DOI: 10.1016/j.bbrc.2008.08.143] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Accepted: 08/22/2008] [Indexed: 10/21/2022]
Abstract
TGF-beta activates receptor-regulated Smad (R-Smad) through phosphorylation by type I receptors. Activated R-Smad binds to Smad4 and the complex translocates into the nucleus and stimulates the transcription of target genes through association with co-activators including p300. It is not clear, however, how activated Smad complexes are removed from target genes. In this study, we show that TGF-beta enhances the mono-ubiquitination of Smad4. Smad4 mono-ubiquitination was promoted by p300 and suppressed by the c-Ski co-repressor. Smad4 mono-ubiquitination disrupted the interaction with Smad2 in the presence of constitutively active TGF-beta type I receptor. Furthermore, mono-ubiquitinated Smad4 was not found in DNA-binding Smad complexes. A Smad4-Ubiquitin fusion protein, which mimics mono-ubiquitinated Smad4, enhanced localization to the cytoplasm. These results suggest that mono-ubiquitination of Smad4 occurs in the transcriptional activator complex and facilitates the turnover of Smad complexes at target genes.
Collapse
Affiliation(s)
- Bei Wang
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki 305-8575, Japan
| | | | | |
Collapse
|
106
|
Clarke DC, Liu X. Decoding the quantitative nature of TGF-beta/Smad signaling. Trends Cell Biol 2008; 18:430-42. [PMID: 18706811 PMCID: PMC2774497 DOI: 10.1016/j.tcb.2008.06.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2008] [Revised: 06/13/2008] [Accepted: 06/17/2008] [Indexed: 01/02/2023]
Abstract
How transforming growth factor-beta (TGF-beta) signaling elicits diverse cell responses remains elusive, despite the major molecular components of the pathway being known. We contend that understanding TGF-beta biology requires mathematical models to decipher the quantitative nature of TGF-beta/Smad signaling and to account for its complexity. Here, we review mathematical models of TGF-beta superfamily signaling that predict how robustness is achieved in bone-morphogenetic-protein signaling in the Drosophila embryo, how changes in receptor-trafficking dynamics can be exploited by cancer cells and how the basic mechanisms of TGF-beta/Smad signaling conspire to promote Smad accumulation in the nucleus. These studies demonstrate the power of mathematical modeling for understanding TGF-beta biology.
Collapse
Affiliation(s)
- David C Clarke
- Department of Chemistry and Biochemistry, University of Colorado-Boulder, Boulder, CO 80309-0215, USA
| | | |
Collapse
|
107
|
Davies MN, O'Callaghan BL, Towle HC. Glucose activates ChREBP by increasing its rate of nuclear entry and relieving repression of its transcriptional activity. J Biol Chem 2008; 283:24029-38. [PMID: 18591247 DOI: 10.1074/jbc.m801539200] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Carbohydrate response element-binding protein (ChREBP) is a glucose-responsive transcription factor that activates genes involved in de novo lipogenesis in mammals. The current model for glucose activation of ChREBP proposes that increased glucose metabolism triggers a cytoplasmic to nuclear translocation of ChREBP that is critical for activation. However, we find that ChREBP actively shuttles between the cytoplasm and nucleus in both low and high glucose in the glucose-sensitive beta cell-derived line, 832/13. Glucose stimulates a 3-fold increase in the rate of ChREBP nuclear entry, but trapping ChREBP in the nucleus by mutagenesis or with a nuclear export inhibitor does not lead to constitutive activation. In fact, mutational studies targeting the nuclear export signal of ChREBP also identified a distinct function essential for glucose-dependent transcriptional activation. From this, we conclude that an additional event independent of nuclear translocation is required for activation. The N-terminal segment of ChREBP (amino acids 1-298) has previously been shown to repress activity under basal conditions. This segment has five highly conserved regions, Mondo conserved regions 1-5 (MCR1 to -5). Based on activating mutations in MCR2 and MCR5, we propose that these two regions act coordinately to repress ChREBP in low glucose. In addition, other mutations in MCR2 and mutations in MCR3 were found to prevent glucose activation. Hence, we conclude that both relief of repression and adoption of an activating form are required for ChREBP activation.
Collapse
Affiliation(s)
- Michael N Davies
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 321 Church Street SE, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
108
|
Beck IME, Vanden Berghe W, Vermeulen L, Bougarne N, Vander Cruyssen B, Haegeman G, De Bosscher K. Altered subcellular distribution of MSK1 induced by glucocorticoids contributes to NF-kappaB inhibition. EMBO J 2008; 27:1682-93. [PMID: 18511904 PMCID: PMC2435130 DOI: 10.1038/emboj.2008.95] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2007] [Accepted: 04/15/2008] [Indexed: 11/09/2022] Open
Abstract
Glucocorticoids are widely used anti-inflammatory and immunomodulatory agents, of which the action mechanism is mainly based on interference of hormone-activated glucocorticoid receptor (GR) with the activity of transcription factors, such as nuclear factor-kappaB (NF-kappaB). In addition to the well described interaction-based mutual repression mechanism between the GR and NF-kappaB, additional mechanisms are at play, which help to explain the efficacy of glucocorticoid-mediated gene repression. In this respect, we found that glucocorticoids counteract the recruitment of activated Mitogen- and Stress-activated protein Kinase-1 (MSK1) at inflammatory gene promoters resulting in the inhibition of NF-kappaB p65 transactivation and of concurrent histone H3 phosphorylation. Additionally, we observed that activated GR can trigger redistribution of nuclear MSK1 to the cytoplasm through a CRM1-dependent export mechanism, as a result of an interaction between liganded GR and activated MSK1. These findings unveil a novel aspect within the GR-mediated NF-kappaB-targeting anti-inflammatory mechanism.
Collapse
Affiliation(s)
- Ilse M E Beck
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Department of Molecular Biology, Gent University, Gent, Belgium
| | - Wim Vanden Berghe
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Department of Molecular Biology, Gent University, Gent, Belgium
| | - Linda Vermeulen
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Department of Molecular Biology, Gent University, Gent, Belgium
| | - Nadia Bougarne
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Department of Molecular Biology, Gent University, Gent, Belgium
| | | | - Guy Haegeman
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Department of Molecular Biology, Gent University, Gent, Belgium
- Both of these authors share senior authorship
| | - Karolien De Bosscher
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Department of Molecular Biology, Gent University, Gent, Belgium
- Both of these authors share senior authorship
| |
Collapse
|
109
|
Yao X, Chen X, Cottonham C, Xu L. Preferential utilization of Imp7/8 in nuclear import of Smads. J Biol Chem 2008; 283:22867-74. [PMID: 18519565 DOI: 10.1074/jbc.m801320200] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Trafficking of Smad proteins between the cytoplasm and nucleus is a critical component of transforming growth factor beta (TGF-beta) signal transduction. Smad4 translocates into the nucleus either in response to TGF-beta stimulation or when its nuclear export is blocked by leptomycin B (LMB). We demonstrate that both TGF-beta-induced and basal state spontaneous nuclear import of Smad4 require importin 7 and 8 (Imp7,8). Our data suggest that in the nuclear import of Smad4, the role of Imp8 is irreplaceable by Imp7, and that Smads preferentially bind Imp8. Interestingly, in contrast to its mammalian counterpart Smad4, Drosophila Medea appears to utilize different mechanisms for TGF-beta-induced or basal state nuclear accumulation, with the latter independent of Msk (Drosophila Imp7/8) function. In addition, overexpression of Imp8 alone was sufficient to cause an increased concentration of Smad1, 3 and 4 in the nucleus, but had very limited effects on Smad2. These observations suggest selective involvement of Imp8/Msk in nuclear import of different Smads under different conditions.
Collapse
Affiliation(s)
- Xiaohao Yao
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | | | | | |
Collapse
|
110
|
Dewint P, Gossye V, De Bosscher K, Vanden Berghe W, Van Beneden K, Deforce D, Van Calenbergh S, Müller-Ladner U, Vander Cruyssen B, Verbruggen G, Haegeman G, Elewaut D. A plant-derived ligand favoring monomeric glucocorticoid receptor conformation with impaired transactivation potential attenuates collagen-induced arthritis. THE JOURNAL OF IMMUNOLOGY 2008; 180:2608-15. [PMID: 18250472 DOI: 10.4049/jimmunol.180.4.2608] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The glucocorticoid receptor (GR) is a transcription factor regulating its target genes either positively, through direct binding to the promoter of target genes, or negatively by the interference with the activity of transcription factors involved in proinflammatory gene expression. The well-known adverse effects of glucocorticoids are believed to be mainly caused by their GR-mediated gene-activating properties. Although dimerization of GR is thought to be essential for gene-activating properties, no compound has yet been described which selectively imposes GR monomer formation and interference with other transcription factors. In the present study, we report on a GR-binding, plant-derived compound with marked dissociative properties in rheumatoid arthritis fibroblast-like synoviocytes, which are important effector cells in inflammation and matrix degradation in rheumatoid arthritis. In addition, these findings could be extended in vivo in murine collagen-induced arthritis, in which joint inflammation was markedly inhibited without inducing hyperinsulinemia. Therefore, we conclude that GR monomers are sufficient for inhibition of inflammation in vivo.
Collapse
Affiliation(s)
- Pieter Dewint
- Department of Rheumatology, Ghent University Hospital, De Pintelaan 185, Ghent, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
111
|
Moustakas A, Heldin CH. Dynamic control of TGF-β signaling and its links to the cytoskeleton. FEBS Lett 2008; 582:2051-65. [DOI: 10.1016/j.febslet.2008.03.027] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2008] [Accepted: 03/18/2008] [Indexed: 12/22/2022]
|
112
|
Buxton ILO, Duan D. Cyclic GMP/protein kinase G phosphorylation of Smad3 blocks transforming growth factor-beta-induced nuclear Smad translocation: a key antifibrogenic mechanism of atrial natriuretic peptide. Circ Res 2008; 102:151-3. [PMID: 18239144 DOI: 10.1161/circresaha.107.170217] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
113
|
Fleur SS, Fujii H. Cytokine-induced nuclear translocation of signaling proteins and their analysis using the inducible translocation trap system. Cytokine 2008; 41:187-97. [PMID: 18203617 PMCID: PMC2289906 DOI: 10.1016/j.cyto.2007.11.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Revised: 11/06/2007] [Accepted: 11/20/2007] [Indexed: 11/16/2022]
Abstract
Binding of cytokines to their specific receptors induces activation of signal transduction pathways, many of which involve nuclear translocation of signaling proteins. In this review, an overview of cytokine-induced nuclear translocation of signaling proteins is provided. In addition, inducible translocation trap (ITT), a novel reporter-based system to detect nuclear translocation, and its application for identification of nuclear translocating proteins are elaborated. Finally, analysis of "nuclear translocatome", the entire set of proteins that translocate into or out of the nucleus in response to extracellular stimuli, by ITT is discussed.
Collapse
Affiliation(s)
- Shella Saint Fleur
- Department of Pathology, New York University School of Medicine, 550 First Avenue, MSB-126, New York, NY 10016, USA
| | - Hodaka Fujii
- Department of Pathology, New York University School of Medicine, 550 First Avenue, MSB-126, New York, NY 10016, USA
| |
Collapse
|
114
|
Ramos MF, Lamé MW, Segall HJ, Wilson DW. Smad Signaling in the Rat Model of Monocrotaline Pulmonary Hypertension. Toxicol Pathol 2008; 36:311-20. [DOI: 10.1177/0192623307311402] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mutations in the bone morphogenetic protein receptor type II (BMPrII) gene have been implicated in the development of familial pulmonary artery hypertension (PAH). The function of BMP signal transduction within the pulmonary vasculature and the role BMPrII mutations have in the development of PAH are incompletely understood. We used the monocrotaline (MCT) model of PAH to examine alterations in Smad signal transduction pathways in vivo. Lungs harvested from Sprague-Dawley rats treated with a single 60-mg/kg intraperitoneal (IP) injection of MCT were compared to saline-treated controls 2 weeks following treatment. Smad 4 was localized by immunohistochemistry to endothelial nuclei of the intra-acinar vessels undergoing remodeling. Smad 4, common to both BMP and transforming growth factor β (TGFβ) signaling, and BMP-specific Smad 1 were significantly decreased in western blot from whole lungs of treated animals, while no change was found for TGFβ-specific Smad 2. MCT-treated rats also had increased expression of phosphorylated Smad 1 (P-Smad 1) but not phosphorylated Smad 2 (P-Smad 2). There was a decrease in the expression of the full BMPrII protein but not its short form variant in MCT-treated rat lungs. The type I receptor Alk1 had increased expression. Collectively, our data indicate that vascular remodeling in the MCT model is associated with alterations in BMP receptors and persistent endothelial Smad 1 signaling.
Collapse
Affiliation(s)
| | - Michael W. Lamé
- Molecular BioSciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Henry J. Segall
- Molecular BioSciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | | |
Collapse
|
115
|
TGFbeta-SMAD signal transduction: molecular specificity and functional flexibility. Nat Rev Mol Cell Biol 2007; 8:970-82. [PMID: 18000526 DOI: 10.1038/nrm2297] [Citation(s) in RCA: 1006] [Impact Index Per Article: 55.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Ligands of the transforming growth factor-beta (TGFbeta) superfamily of growth factors initiate signal transduction through a bewildering complexity of ligand-receptor interactions. Signalling then converges to nuclear accumulation of transcriptionally active SMAD complexes and gives rise to a plethora of specific functional responses in both embryos and adult organisms. Current research is focused on the mechanisms that regulate SMAD activity to evoke cell-type-specific and context-dependent transcriptional programmes. An equally important challenge is understanding the functional role of signal strength and duration. How are these quantitative aspects of the extracellular signal regulated? How are they then sensed and interpreted, and how do they affect responses?
Collapse
|
116
|
Gromova KV, Friedrich M, Noskov A, Harms GS. Visualizing Smad1/4 signaling response to bone morphogenetic protein-4 activation by FRET biosensors. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1773:1759-73. [PMID: 18006160 DOI: 10.1016/j.bbamcr.2007.09.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2007] [Revised: 09/11/2007] [Accepted: 09/20/2007] [Indexed: 11/18/2022]
Abstract
Smad proteins are the major signal transducers for the Transforming Growth Factor superfamily of cytokines and their serine/threonine kinase receptors. Smads mediate the signal from the membrane into the nucleus. Bone Morphogenetic Protein-4 stimulates phosphorylation of Smad1, which interacts with Smad4. This complex translocates into the nucleus and regulates transcription of target genes. Here, we report our development of cellular fluorescence biosensors for direct visualization of Smad signaling in live mammalian cells. Fluorescence resonance energy transfer between cyan and yellow fluorescent proteins fused to the Smad1 and Smad4 proteins was used to unravel the temporal aspects of BMP/Smad signaling. A rate-limiting delay of 2-5 min occurred between BMP activation and Smad1 activity. A similar delay was observed in the Smad1/Smad4 complexation. Further experimentation indicated that the delay is dependent on the MH1 domain and linker of Smad1. These results give new insights into the dynamics of the BMP receptor -Smad1/4 signaling process and provide a new tool for studying Smads.
Collapse
Affiliation(s)
- Kira V Gromova
- Molecular Microscopy Group, Rudolf-Virchow-Center, University of Würzburg, Versbacher Str. 9, D-97078 Würzburg, Germany
| | | | | | | |
Collapse
|
117
|
Ross S, Hill CS. How the Smads regulate transcription. Int J Biochem Cell Biol 2007; 40:383-408. [PMID: 18061509 DOI: 10.1016/j.biocel.2007.09.006] [Citation(s) in RCA: 291] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2007] [Revised: 09/19/2007] [Accepted: 09/20/2007] [Indexed: 01/01/2023]
Abstract
The primary signalling pathway downstream of ligands of the transforming growth factor beta (TGF-beta) superfamily is the Smad pathway. Activated receptors phosphorylate receptor-regulated Smads, which form homomeric complexes and heteromeric complexes with Smad4. These activated Smad complexes accumulate in the nucleus, where they are directly involved in the regulation of transcription of target genes. This apparently very simple pathway is subject to complex regulation, much of which is at the level of post-translational modifications of pathway components, in particular, the Smads. The enzymes responsible may be constitutively active, may be cell type-specific or may be regulated by other signalling pathways or by the cell cycle. In this way, signals from TGF-beta superfamily ligands are integrated with signals from other growth factors and cytokines, are regulated by the cell cycle and are dependent on cell type. This may go some way to explaining the pleiotropic nature of TGF-beta superfamily responses. In this review we focus on the mechanisms whereby the Smads are modified and regulated. We then go on to discuss how the activated Smad complexes regulate transcription.
Collapse
Affiliation(s)
- Sarah Ross
- Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, Lincoln's Inn Fields Laboratories, 44 Lincoln's Inn Fields, London WC2A 3PX, United Kingdom
| | | |
Collapse
|
118
|
Zi Z, Klipp E. Constraint-based modeling and kinetic analysis of the Smad dependent TGF-beta signaling pathway. PLoS One 2007; 2:e936. [PMID: 17895977 PMCID: PMC1978528 DOI: 10.1371/journal.pone.0000936] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2007] [Accepted: 09/06/2007] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Investigation of dynamics and regulation of the TGF-beta signaling pathway is central to the understanding of complex cellular processes such as growth, apoptosis, and differentiation. In this study, we aim at using systems biology approach to provide dynamic analysis on this pathway. METHODOLOGY/PRINCIPAL FINDINGS We proposed a constraint-based modeling method to build a comprehensive mathematical model for the Smad dependent TGF-beta signaling pathway by fitting the experimental data and incorporating the qualitative constraints from the experimental analysis. The performance of the model generated by constraint-based modeling method is significantly improved compared to the model obtained by only fitting the quantitative data. The model agrees well with the experimental analysis of TGF-beta pathway, such as the time course of nuclear phosphorylated Smad, the subcellular location of Smad and signal response of Smad phosphorylation to different doses of TGF-beta. CONCLUSIONS/SIGNIFICANCE The simulation results indicate that the signal response to TGF-beta is regulated by the balance between clathrin dependent endocytosis and non-clathrin mediated endocytosis. This model is useful to be built upon as new precise experimental data are emerging. The constraint-based modeling method can also be applied to quantitative modeling of other signaling pathways.
Collapse
Affiliation(s)
- Zhike Zi
- Computational Systems Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Edda Klipp
- Computational Systems Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Theoretical Biophysics, Humboldt University Berlin, Institute for Biology, Berlin, Germany
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
119
|
Levy L, Howell M, Das D, Harkin S, Episkopou V, Hill CS. Arkadia activates Smad3/Smad4-dependent transcription by triggering signal-induced SnoN degradation. Mol Cell Biol 2007; 27:6068-83. [PMID: 17591695 PMCID: PMC1952153 DOI: 10.1128/mcb.00664-07] [Citation(s) in RCA: 149] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2007] [Revised: 05/25/2007] [Accepted: 06/14/2007] [Indexed: 01/17/2023] Open
Abstract
E3 ubiquitin ligases play important roles in regulating transforming growth factor beta (TGF-beta)/Smad signaling. Screening of an E3 ubiquitin ligase small interfering RNA library, using TGF-beta induction of a Smad3/Smad4-dependent luciferase reporter as a readout, revealed that Arkadia is an E3 ubiquitin ligase that is absolutely required for this TGF-beta response. Knockdown of Arkadia or overexpression of a dominant-negative mutant completely abolishes transcription from Smad3/Smad4-dependent reporters, but not from Smad1/Smad4-dependent reporters or from reporters driven by Smad2/Smad4/FoxH1 complexes. We show that Arkadia specifically activates transcription via Smad3/Smad4 binding sites by inducing degradation of the transcriptional repressor SnoN. Arkadia is essential for TGF-beta-induced SnoN degradation, but it has little effect on SnoN levels in the absence of signal. Arkadia interacts with SnoN and induces its ubiquitination irrespective of TGF-beta/Activin signaling, but SnoN is efficiently degraded only when it forms a complex with both Arkadia and phosphorylated Smad2 or Smad3. Finally, we describe an esophageal cancer cell line (SEG-1) that we show has lost Arkadia expression and is deficient for SnoN degradation. Reintroduction of wild-type Arkadia restores TGF-beta-induced Smad3/Smad4-dependent transcription and SnoN degradation in these cells, raising the possibility that loss of Arkadia function may be relevant in cancer.
Collapse
Affiliation(s)
- Laurence Levy
- Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, Lincoln's Inn Fields Laboratories, 44 Lincoln's Inn Fields, London WC2A 3PX, United Kingdom
| | | | | | | | | | | |
Collapse
|
120
|
Gao S, Laughon A. Flexible interaction of Drosophila Smad complexes with bipartite binding sites. ACTA ACUST UNITED AC 2007; 1769:484-96. [PMID: 17610966 DOI: 10.1016/j.bbaexp.2007.05.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2006] [Revised: 04/24/2007] [Accepted: 05/29/2007] [Indexed: 01/11/2023]
Abstract
A subset of BMP-responsive enhancer elements are characterized by pairing of a GC-rich Smad1 binding site and an SBE-type Smad4 binding site. Such paired, or bipartite, sites are in some cases just 5 bp apart and thus might be contacted by a single Smad1-Smad4 complex. Other potential pairings are separated as much as 60 bp but it is not known whether such longer distances can be spanned by a Smad1-Smad4 complex, indeed binding of native Smad1-Smad4 complexes to any of these bipartite elements has yet to be reported. Here we report that a complex of the homologous Drosophila Smad proteins, Mad and Medea, is capable of concerted binding to GC-rich and SBE sites separated by as much as 20 bp. The wider the separation, the more severely binding affinity was reduced by shortening of the linker region that tethers the DNA binding domain of Medea. In contrast, length of the Mad linker did not affect the allowed distance between paired sites, rather it contributes specifically to Mad contact with the GC-rich site. Finally, we show that Smad1 and Smad4 can participate in binding to bipartite sites.
Collapse
Affiliation(s)
- Sheng Gao
- Laboratory of Genetics, University of Wisconsin, 425G Henry Mall, Madison, WI 53706, USA
| | | |
Collapse
|
121
|
Brown KA, Pietenpol JA, Moses HL. A tale of two proteins: differential roles and regulation of Smad2 and Smad3 in TGF-beta signaling. J Cell Biochem 2007; 101:9-33. [PMID: 17340614 DOI: 10.1002/jcb.21255] [Citation(s) in RCA: 305] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Transforming growth factor-beta (TGF-beta) is an important growth inhibitor of epithelial cells, and insensitivity to this cytokine results in uncontrolled cell proliferation and can contribute to tumorigenesis. Smad2 and Smad3 are direct mediators of TGF-beta signaling, however little is known about the selective activation of Smad2 versus Smad3. The Smad2 and Smad3 knockout mouse phenotypes and studies comparing Smad2 and Smad3 activation of TGF-beta target genes, suggest that Smad2 and Smad3 have distinct roles in TGF-beta signaling. The observation that TGF-beta inhibits proliferation of Smad3-null mammary gland epithelial cells, whereas Smad3 deficient fibroblasts are only partially growth inhibited, suggests that Smad3 has a different role in epithelial cells and fibroblasts. Herein, the current understanding of Smad2 and Smad3-mediated TGF-beta signaling and their relative roles are discussed, in addition to potential mechanisms for the selective activation of Smad2 versus Smad3. Since alterations in the TGF-beta signaling pathway play an important role in promoting tumorigenesis and cancer progression, methods for therapeutic targeting of the TGF-beta signaling pathway are being pursued. Determining how Smad2 or Smad3 differentially regulate the TGF-beta response may translate into developing more effective strategies for cancer therapy.
Collapse
Affiliation(s)
- Kimberly A Brown
- Department of Cancer Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37232, USA
| | | | | |
Collapse
|
122
|
Zhu S, Wang W, Clarke DC, Liu X. Activation of Mps1 Promotes Transforming Growth Factor-β-independent Smad Signaling. J Biol Chem 2007; 282:18327-18338. [PMID: 17452325 DOI: 10.1074/jbc.m700636200] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The primary intracellular mediators of TGF-beta signaling are the Smad proteins. Phosphorylation of R-Smad at the C-terminal SSXS motif by the activated TGF-beta type I receptor kinase triggers a conformation change in R-Smad and facilitates complex formation between R-Smad and Smad4, which shuttle into the nucleus where they interact with DNA and other transcription factors to regulate gene expression. In an attempt to identify proteins interacting with activated Smad signaling complex, we discovered that Mps1, a protein kinase that plays important roles in normal mitotic progression and mitotic checkpoint signaling, co-purifies with this complex. We demonstrated that Smad2 and Smad3 but not Smad4 are substrates of Mps1 in vitro and in vivo. Mps1 phosphorylates Smad2 and Smad3 at the SSXS motif in their C-terminal regions in vitro and in vivo. Disruption of microtubule networks by nocodazole activates Mps1 and promotes TGF-beta-independent activation of Smad signaling. We found that Mps1 is involved in turning on Smad signaling by phosphorylating R-Smads. Our results reveal a novel functional link between Mps1 and Smads in a non-canonical Smad signaling pathway.
Collapse
Affiliation(s)
- Songcheng Zhu
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, Colorado 80309
| | - Wei Wang
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, Colorado 80309
| | - David C Clarke
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, Colorado 80309
| | - Xuedong Liu
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, Colorado 80309.
| |
Collapse
|
123
|
Ramos M, Lamé MW, Segall HJ, Wilson DW. Monocrotaline pyrrole induces Smad nuclear accumulation and altered signaling expression in human pulmonary arterial endothelial cells. Vascul Pharmacol 2007; 46:439-48. [PMID: 17336165 PMCID: PMC2570208 DOI: 10.1016/j.vph.2007.01.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2006] [Revised: 01/03/2007] [Accepted: 01/22/2007] [Indexed: 12/31/2022]
Abstract
The mechanistic relationship between the widely used monocrotaline model of primary pulmonary hypertension and altered TGFbeta family signaling due to genetic defects in the Bone Morphogenetic Protein type II receptor in affected humans has not been investigated. In this study we use fluorescent microscopy to demonstrate nuclear translocation of Smad 4 in human pulmonary arterial endothelial cell (HPAEC) cultures treated with monocrotaline pyrrole (MCTP), Bone Morphogenetic Protein (BMP) and TGFbeta. While MCTP induced transient nuclear accumulation of phosphorylated Smad 1 (P-Smad 1) and phosphorylated Smad 2 (P-Smad 2), only expression of P-Smad 1 was significantly altered in western blots. P-Smad 1 expression significantly increased 30 min following treatment with MCTP correlating with P-Smad 1 and Smad 4 nuclear translocation. Although a modest, but significant decrease in P-Smad 1 expression occurred 1 h after treatment, expression was significantly increased at 72 h. Evaluation of components of the signal and response pathway at 72 h showed decreased expression of the BMP type II receptor (BMPrII), no change in TGFbeta Activin Receptor-like Kinase 1 (Alk 1), no change in Smad 4 but increase in the inhibitory Smad 6, decrease in the alternate BMP signaling pathway p38(MAPK) but no change in the psmad1 response element ID 1. Our results suggest transient activation of Smad signaling pathways in initial MCTP endothelial cell toxicity, and a persistent dysregulation of BMP signaling. Electron microscopy of cell membrane caveoli revealed a dramatic decrease in these structures after 72 h. Loss of these structural elements, noted for their sequestration and inhibition of receptor activity, may contribute to prolonged alterations in BMP signaling.
Collapse
Affiliation(s)
- M Ramos
- Department of Veterinary Medicine, Pathology, Immunology, Microbiology, University of California, Davis, Davis, California 95616, USA
| | | | | | | |
Collapse
|
124
|
Biondi C, Das D, Howell M, Islam A, Bikoff E, Hill C, Robertson E. Mice develop normally in the absence of Smad4 nucleocytoplasmic shuttling. Biochem J 2007; 404:235-45. [PMID: 17300215 PMCID: PMC1868808 DOI: 10.1042/bj20061830] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2006] [Revised: 02/08/2007] [Accepted: 02/14/2007] [Indexed: 01/02/2023]
Abstract
Smad4 in partnership with R-Smads (receptor-regulated Smads) activates TGF-beta (transforming growth factor-beta)-dependent signalling pathways essential for early mouse development. Smad4 null embryos die shortly after implantation due to severe defects in cell proliferation and visceral endoderm differentiation. In the basal state, Smad4 undergoes continuous shuttling between the cytoplasm and the nucleus due to the combined activities of an N-terminal NLS (nuclear localization signal) and an NES (nuclear export signal) located in its linker region. Cell culture experiments suggest that Smad4 nucleocytoplasmic shuttling plays an important role in TGF-beta signalling. In the present study we have investigated the role of Smad4 shuttling in vivo using gene targeting to engineer two independent mutations designed to eliminate Smad4 nuclear export. As predicted this results in increased levels of Smad4 in the nucleus of homozygous ES cells (embryonic stem cells) and primary keratinocytes, in the presence or absence of ligand. Neither mutation affects Smad4 expression levels nor its ability to mediate transcriptional activation in homozygous cell lines. Remarkably mouse mutants lacking the Smad4 NES develop normally. Smad4 NES mutants carrying one copy of a Smad4 null allele also fail to display developmental defects. The present study clearly demonstrates that Smad4 nucleocytoplasmic shuttling is not required for embryonic development or tissue homoeostasis in normal, healthy adult mice.
Collapse
Key Words
- embryonic
- gene targetting
- nuclear export
- nucleocytoplasmic shuttling
- smad4
- transforming growth factor-β signal
- bmp, bone morphogenetic proteins
- crm1, chromosomal region maintenance 1
- d.p.c., days post-coitum
- es cell, embryonic stem cell
- fbs, foetal bovine serum
- gdf, growth and differentiation factor
- lmb, leptomycin b
- mef, murine embryonic fibroblast
- mh domain, mad homology domain
- nes, nuclear export signal
- nls, nuclear localization signal
- rpa, ribonuclease protection assay
- r-smad, receptor-regulated smad
- snon, ski-related novel protein n
- tgf-β, transforming growth factor-β
Collapse
Affiliation(s)
- Christine A. Biondi
- *The Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, U.K
| | - Debipriya Das
- †Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3PX, U.K
| | - Michael Howell
- †Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3PX, U.K
| | - Ayesha Islam
- *The Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, U.K
| | - Elizabeth K. Bikoff
- *The Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, U.K
| | - Caroline S. Hill
- †Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3PX, U.K
| | - Elizabeth J. Robertson
- *The Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, U.K
| |
Collapse
|
125
|
Tu AW, Luo K. Acetylation of Smad2 by the co-activator p300 regulates activin and transforming growth factor beta response. J Biol Chem 2007; 282:21187-96. [PMID: 17478422 DOI: 10.1074/jbc.m700085200] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Transforming growth factor beta (TGFbeta) signals primarily through the Smad proteins to regulate cell growth, differentiation, and extracellular matrix production. Post-translational modifications, such as phosphorylation, play an important role in the regulation of the Smad proteins. TGFbeta signaling results in the phosphorylation of Smad2 and Smad3 that then oligomerize with Smad4 and translocate into the nucleus to initiate transcription of TGFbeta target genes. The initiation of transcription is significantly enhanced by the direct interaction of the Smad complex with p300/CBP (CREB-binding protein), a co-activator with intrinsic acetyltransferase activity. However, how p300/CBP enhances transcription through this interaction is not entirely understood. In this report, we show that Smad2, but not the highly homologous Smad3, can be acetylated by p300/CBP in a ligand-dependent manner. At least three lysine residues, Lys(19), Lys(20), and Lys(39), are required for efficient acetylation of Smad2, as mutations altering these lysines abolished Smad2 acetylation in vivo. This acetylation event is required for the ability of Smad2 to mediate activin and TGFbeta signaling. Mutation of the three key lysine residues did not alter the stability of Smad2 or the ability of Smad2 to form a complex with Smad4 on promoter DNA, but it prevented nuclear accumulation of Smad2 and subsequent TGFbeta and activin responses. Thus, our studies reveal a novel mechanism of modulating Smad2 activity and localization through protein acetylation.
Collapse
Affiliation(s)
- Andrea W Tu
- Department of Molecular and Cell Biology, University of California, and Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
| | | |
Collapse
|
126
|
Wang LH, Kim SH, Lee JH, Choi YL, Kim YC, Park TS, Hong YC, Wu CF, Shin YK. Inactivation of SMAD4 tumor suppressor gene during gastric carcinoma progression. Clin Cancer Res 2007; 13:102-10. [PMID: 17200344 DOI: 10.1158/1078-0432.ccr-06-1467] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Mothers against decapentaplegic homologue 4 (SMAD4) is a tumor suppressor gene associated with gastrointestinal carcinogenesis. The aim of the present study is to more precisely characterize its role in the development and progression of human gastric carcinoma. EXPERIMENTAL DESIGN The expression of SMAD4 was investigated in 283 gastric adenocarcinomas and related lesions, as well as in 9 gastric carcinoma cell lines. We also analyzed the methylation status of SMAD4 gene by using methylation-specific PCR, examined loss of heterozygosity (LOH) of this gene locus by using a vicinal marker, and detected exon mutation of SMAD4 through exon-by-exon amplification. Moreover, we assessed whether MG132, a proteasome inhibitor, affected the SMAD4 protein level. RESULTS We found loss of SMAD4 protein expression in the cytoplasm (36 of 114, 32%) and in the nucleus (46 of 114, 40%) of gastric cancer cells. The loss of nuclear SMAD4 expression in primary tumors correlated significantly with poor survival, and was an independent prognostic marker in multivariate analysis. We also found a substantial decrease in SMAD4 expression at both the RNA and protein level in several human gastric carcinoma cell lines. In addition, we found that LOH (20 of 70, 29%) and promoter hypermethylation (4 of 73, 5%) were associated with the loss of SMAD4 expression. SMAD4 protein levels were also affected in certain gastric carcinoma cell lines following incubation with MG132. CONCLUSION Taken together, our results indicate that the loss of SMAD4, especially loss of nuclear SMAD4 expression, is involved in gastric cancer progression. The loss of SMAD4 in gastric carcinomas was due to several mechanisms, including LOH, hypermethylation, and proteasome degradation.
Collapse
Affiliation(s)
- Li-Hui Wang
- Research Institute of Pharmaceutical Science, Department of Pharmacy, Seoul National University College of Pharmacy, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Batut J, Howell M, Hill CS. Kinesin-mediated transport of Smad2 is required for signaling in response to TGF-beta ligands. Dev Cell 2007; 12:261-74. [PMID: 17276343 DOI: 10.1016/j.devcel.2007.01.010] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2006] [Revised: 11/30/2006] [Accepted: 01/17/2007] [Indexed: 01/13/2023]
Abstract
During vertebrate development, Activin/Nodal-related ligands signal through Smad2, leading to its activation and accumulation in the nucleus. Here, we demonstrate that Smad2 constantly shuttles between the cytoplasm and nucleus both in early Xenopus embryo explants and in living zebrafish embryos, providing a mechanism whereby the intracellular components of the pathway constantly monitor receptor activity. We have gone on to demonstrate that an intact microtubule network and kinesin ATPase activity are required for Smad2 phosphorylation and nuclear accumulation in response to Activin/Nodal in early vertebrate embryos and TGF-beta in mammalian cells. The kinesin involved is kinesin-1, and Smad2 interacts with the kinesin-1 light chain subunit. Interfering with kinesin activity in Xenopus and zebrafish embryos phenocopies loss of Nodal signaling. Our results reveal that kinesin-mediated transport of Smad2 along microtubules to the receptors is an essential step in ligand-induced Smad2 activation.
Collapse
Affiliation(s)
- Julie Batut
- Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3PX, United Kingdom
| | | | | |
Collapse
|
128
|
Chalmers KA, Love S. Neurofibrillary Tangles May Interfere With Smad 2/3 Signaling in Neurons. J Neuropathol Exp Neurol 2007; 66:158-67. [PMID: 17279001 DOI: 10.1097/nen.0b013e3180303b93] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Transforming growth factor (TGF)-beta is a multifunctional cytokine with anti-inflammatory, reparative and neuroprotective functions. Increased levels of TGFbeta in Alzheimer disease (AD) are associated with perivascular deposition of extracellular matrix, which may impair clearance of beta-amyloid and contribute to the development of cerebral amyloid angiopathy. TGFbeta signaling is transduced by Smad proteins: on TGFbeta receptor activation, Smads 2 and 3 are released from sequestration by microtubules, phosphorylated (forming pSmad2/3), and, together with Smad 4, translocated to the nucleus, where they initiate the transcription of multiple genes. Neuronal microtubule assembly is disturbed in AD when tau, a microtubule-stabilizing protein, is hyperphosphorylated and forms neurofibrillary tangles. We have investigated the relationship between Ser202 phospho-tau and pSmads 2 and 3 in the temporal lobe in AD. Within neurons in control brains, pSmads 2 and 3 were almost exclusively intranuclear. In AD, pSmad 3 bound to phospho-tau (mostly insoluble tau) and accumulated in the cytoplasm of tangle-bearing neurons; this was accompanied by a marked decrease in nuclear pSmad3. pSmads 2 and 3 were also present in neuronal granulovacuolar inclusions. Our findings suggest that neurofibrillary tangles sequester pSmad3, preventing its translocation into the nucleus and the induction of gene transcription. Interference with the Smad signaling may adversely affect survival of tangle-bearing neurons in AD.
Collapse
Affiliation(s)
- Katy A Chalmers
- Dementia Research Group, University of Bristol Institute of Clinical Neurosciences, Department of Clinical Science at North Bristol, Frenchay Hospital, Bristol, UK
| | | |
Collapse
|
129
|
Abstract
Activation of Smad signaling pathways downstream of TGF-beta superfamily ligands via receptor-mediated Smad phosphorylation is well understood, but little is known about the phosphatases that turn off Smad activity. Now in Cell, Feng and colleagues (Lin, X., et al. (2006) Cell 125 , 915-928) report their discovery that PPM1A acts as a Smad phosphatase to terminate TGF-beta signaling.
Collapse
Affiliation(s)
- Caroline S Hill
- Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London, WC2A 3PX, United Kingdom
| |
Collapse
|
130
|
Lim SK, Hoffmann FM. Smad4 cooperates with lymphoid enhancer-binding factor 1/T cell-specific factor to increase c-myc expression in the absence of TGF-beta signaling. Proc Natl Acad Sci U S A 2006; 103:18580-5. [PMID: 17132729 PMCID: PMC1693705 DOI: 10.1073/pnas.0604773103] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The c-myc protooncogene is a key regulator of cell proliferation whose expression is reduced in normal epithelial cells in response to the growth inhibitory cytokine TGF-beta. Smad4 mediates this inhibitory effect of TGF-beta by forming a complex with Smad3, E2F4/5, and p107 at the TGF-beta inhibitory element (TIE) element on the c-myc promoter. In contrast, cell proliferation and c-myc expression are increased in response to Wnt ligands; this effect is mediated through the lymphoid enhancer-binding factor 1/T cell-specific factor (LEF/TCF) family of transcription factors on the c-myc promoter LEF/TCF-binding elements (TBE1 and TBE2). We report that a peptide aptamer designed to inhibit the binding between Smad4 and LEF/TCF reduced c-myc expression and the growth rate of HepG2 cells. Further analysis demonstrated that, in the absence of TGF-beta, Smad4 was bound to the positive regulatory element TBE1 from the c-myc promoter and activated c-myc promoter activity. Smad4 binding to the positive TBE1 c-myc element was reduced by TGF-beta, consistent with Smad4's inhibitory role on c-myc expression in response to TGF-beta. Reduction of Smad4 levels by RNAi knockdown also reduced c-myc expression levels and sensitized hepatocytes to cell death by serum deprivation. Two tumor-derived mutant Smad4 proteins that fail to mediate TGF-beta responses were still competent to cooperate with LEF1 to activate the c-myc promoter. These results support a previously unreported TGF-beta-independent function for Smad4 in cooperating with LEF/TCF to activate c-myc expression.
Collapse
Affiliation(s)
| | - F. Michael Hoffmann
- *McArdle Laboratory for Cancer Research and
- Laboratory of Genetics, University of Wisconsin, Madison, WI 53706
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
131
|
Xu L. Regulation of Smad activities. ACTA ACUST UNITED AC 2006; 1759:503-13. [PMID: 17182123 PMCID: PMC1805629 DOI: 10.1016/j.bbaexp.2006.11.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2006] [Revised: 10/27/2006] [Accepted: 11/07/2006] [Indexed: 01/15/2023]
Abstract
TGF-beta (Transforming Growth Factor-beta) cytokines employ Smad proteins as the intracellular mediator of signaling. Upon TGF-beta stimulation, the cytoplasmic Smads become phosphorylated and consequently accumulate in the nucleus to regulate target gene expression. The cytoplasm-to-nucleus redistribution of Smads, as well as the ability of Smads to activate or repress gene transcription, is under multiple layers of regulation by factors not limited to TGF-beta. With recent advance in the knowledge of regulatory factors impinged on Smads, we are beginning to understand the complexity in cellular responses to TGF-beta.
Collapse
Affiliation(s)
- Lan Xu
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street, Rm. 308, Worcester, MA 01605, USA.
| |
Collapse
|
132
|
Ross S, Cheung E, Petrakis TG, Howell M, Kraus WL, Hill CS. Smads orchestrate specific histone modifications and chromatin remodeling to activate transcription. EMBO J 2006; 25:4490-502. [PMID: 16990801 PMCID: PMC1589990 DOI: 10.1038/sj.emboj.7601332] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2006] [Accepted: 08/11/2006] [Indexed: 11/09/2022] Open
Abstract
Smads are intracellular transducers for TGF-beta superfamily ligands, but little is known about the mechanism by which complexes of receptor-phosphorylated Smad2 and Smad4 regulate transcription. Using an in vitro transcription system, we have discovered that, unlike most transcription factors that are sufficient to recruit the basal transcription machinery and therefore activate transcription on both naked DNA and chromatin templates, the Smads only activate transcription from chromatin templates. We demonstrate that Smad2-mediated transcription requires the histone acetyltransferase, p300. Smad2-recruited p300 exhibits an altered substrate specificity, specifically acetylating nucleosomal histone H3 at lysines 9 and 18, and these modifications are also detected on an endogenous Smad2-dependent promoter in a ligand-induced manner. Furthermore, we show that endogenous Smad2 interacts with the SWI/SNF ATPase, Brg1, in a TGF-beta-dependent manner, and demonstrate that Brg1 is recruited to Smad2-dependent promoters and is specifically required for TGF-beta-induced expression of endogenous Smad2 target genes. Our data indicate that the Smads define a new class of transcription factors that absolutely require chromatin to assemble the basal transcription machinery and activate transcription.
Collapse
Affiliation(s)
- Sarah Ross
- Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, London, UK
| | - Edwin Cheung
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
- Genome Institute of Singapore, Genome, Singapore
| | - Thodoris G Petrakis
- Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, London, UK
| | - Michael Howell
- Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, London, UK
| | - W Lee Kraus
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Caroline S Hill
- Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, London, UK
| |
Collapse
|
133
|
Abstract
In this issue of Cell, Lin et al. (2006) answer one of the long-standing questions in the TGFbeta field by identifying a phosphatase, PPM1A, that directly dephosphorylates Smad2 and Smad3 to limit their activation.
Collapse
Affiliation(s)
- Stephen H Schilling
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | |
Collapse
|
134
|
Luo Q, Nieves E, Kzhyshkowska J, Angeletti RH. Endogenous transforming growth factor-beta receptor-mediated Smad signaling complexes analyzed by mass spectrometry. Mol Cell Proteomics 2006; 5:1245-60. [PMID: 16582422 DOI: 10.1074/mcp.m600065-mcp200] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
ASmad proteins are the central feature of the transforming growth factor-beta (TGF-beta) intracellular signaling cascade. They function by carrying signals from the cell surface to the nucleus through the formation of a series of signaling complexes. Changes in Smad proteins and their complexes upon treatment with TGF-beta were studied in mink lung epithelial (Mv1Lu) cell cultures. A time course of incubation with TGF-beta was carried out to determine the peak of appearance of phosphorylated Smad2. Immobilized monoclonal antibody against Smad2 was then used to isolate the naturally occurring complexes. Three strategies were used to identify changes in proteins partnering with Smad2: separation by one-dimensional SDS-PAGE followed by MALDI peptide mass fingerprinting, cleavable ICAT labeling of the protein mixtures analyzed by LC-MS/MS, and nano-LC followed by MALDI MS TOF/TOF. Smad2 forms complexes with many other polypeptides both in the presence and absence of TGF-beta. Some of the classes of proteins identified include: transcription regulators, proteins of the cytoskeletal scaffold and other tethering proteins, motility proteins, proteins involved in transport between the cytoplasm and nucleus, and a group of membrane adaptor proteins. Although some of these have been reported in the literature, most have not been reported previously. This work expands the repertoire of proteins known to participate in the TGF-beta signal transduction processes.
Collapse
Affiliation(s)
- Qilie Luo
- Laboratory for Macromolecular Analysis and Proteomics, Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | |
Collapse
|
135
|
Kim SH, Rowe J, Fujii H, Jones R, Schmierer B, Kong BW, Kuchler K, Foster D, Ish-Horowicz D, Peters G. Upregulation of chicken p15INK4b at senescence and in the developing brain. J Cell Sci 2006; 119:2435-43. [PMID: 16720639 DOI: 10.1242/jcs.02989] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In mammalian cells, products of the INK4a-ARF locus play major roles in senescence and tumour suppression in different contexts, whereas the adjacent INK4b gene is more generally associated with transforming growth factor β (TGF-β)-mediated growth arrest. As the chicken genome does not encode an equivalent of INK4a, we asked whether INK4b and/or ARF contribute to replicative senescence in chicken cells. In chicken embryo fibroblasts (CEFs), INK4b levels increase substantially at senescence and the gene is transcriptionally silenced in two spontaneously immortalised chicken cell lines. By contrast, ARF levels are unaffected by prolonged culture or immortalisation. These expression patterns resemble the behaviour of INK4a and ARF in human fibroblasts. However, short-hairpin RNA (shRNA)-mediated knockdown of chicken INK4b or ARF provides only modest lifespan extension, suggesting that other factors contribute to senescence in CEFs. As well as underscoring the importance of the INK4b-ARF-INK4a locus in senescence, these findings imply that the encoded products have assumed different roles in different evolutionary niches. Although ARF RNA is not detectable in early chicken embryos, the INK4b transcript is expressed in the roof-plate of the developing hind-brain, consistent with a role in limiting cell proliferation.
Collapse
Affiliation(s)
- Soo-Hyun Kim
- Molecular Oncology, Cancer Research UK London Research Institute, Lincoln's Inn Fields, London, WC2A 3PX, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Chen YG, Wang Q, Lin SL, Chang CD, Chuang J, Chung J, Ying SY. Activin signaling and its role in regulation of cell proliferation, apoptosis, and carcinogenesis. Exp Biol Med (Maywood) 2006; 231:534-44. [PMID: 16636301 DOI: 10.1177/153537020623100507] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Activins, cytokine members of the transforming growth factor-beta superfamily, have various effects on many physiological processes, including cell proliferation, cell death, metabolism, homeostasis, differentiation, immune responses endocrine function, etc. Activins interact with two structurally related serine/threonine kinase receptors, type I and type II, and initiate downstream signaling via Smads to regulate gene expression. Understanding how activin signaling is controlled extracellularly and intracellularly would not only lead to more complete understanding of cell growth and apoptosis, but would also provide the basis for therapeutic strategies to treat cancer and other related diseases. This review focuses on the recent progress on activin-receptor interactions, regulations of activin signaling by ligand-binding proteins, receptor-binding proteins, and nucleocytoplasmic shuttling of Smad proteins.
Collapse
Affiliation(s)
- Ye-Guang Chen
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Department of Biological Sciences and Biotechnology, Tsinghua University, Beijing 100084, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
137
|
Natarajan E, Omobono JD, Guo Z, Hopkinson S, Lazar AJF, Brenn T, Jones JC, Rheinwald JG. A keratinocyte hypermotility/growth-arrest response involving laminin 5 and p16INK4A activated in wound healing and senescence. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 168:1821-37. [PMID: 16723698 PMCID: PMC1606631 DOI: 10.2353/ajpath.2006.051027] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/15/2006] [Indexed: 11/20/2022]
Abstract
Keratinocytes become migratory to heal wounds, during early neoplastic invasion, and when undergoing telomere-unrelated senescence in culture. All three settings are associated with expression of the cell cycle inhibitor p16INK4A (p16) and of the basement membrane protein laminin 5 (LN5). We have investigated cause-and-effect relationships among laminin 5, p16, hypermotility, and growth arrest. Plating primary human keratinocytes on the gamma2 precursor form of laminin 5 (LN5') immediately induced directional hypermotility at approximately 125 microm/hour, followed by p16 expression and growth arrest. Cells deficient in p16 and either p14ARF or p53 became hypermotile in response to LN5' but did not arrest growth. Plating on LN5' triggered smad nuclear translocation, and all LN5' effects were blocked by a transforming growth factor (TGF) beta receptor I (TGFbetaRI) kinase inhibitor. In contrast, plating cells on collagen I triggered a TGFbetaRI kinase-independent hypermotility unaccompanied by smad translocation or growth arrest. Plating on control surfaces with TGFbeta induced hypermotility after a 1-day lag time and growth arrest by a p16-independent mechanism. Keratinocytes serially cultured with TGFbetaRI kinase inhibitor exhibited an extended lifespan, and immortalization was facilitated following transduction to express the catalytic subunit of telomerase (TERT). These results reveal fundamental features of a keratinocyte hyper-motility/growth-arrest response that is activated in wound healing, tumor suppression, and during serial culture.
Collapse
Affiliation(s)
- Easwar Natarajan
- Department of Dermatology, Brigham and Women's Hospital and Harvard Skin Disease Research Center, Harvard Institutes of Medicine, Room 664, 77 Ave. Louis Pasteur, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
138
|
Kaivo-oja N, Jeffery LA, Ritvos O, Mottershead DG. Smad signalling in the ovary. Reprod Biol Endocrinol 2006; 4:21. [PMID: 16611366 PMCID: PMC1459162 DOI: 10.1186/1477-7827-4-21] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2005] [Accepted: 04/12/2006] [Indexed: 02/08/2023] Open
Abstract
It has now been a decade since the first discovery of the intracellular Smad proteins, the downstream signalling molecules of one of the most important growth factor families in the animal kingdom, the transforming growth factor beta (TGF-beta) superfamily. In the ovary, several TGF-beta superfamily members are expressed by the oocyte, granulosa and thecal cells at different stages of folliculogenesis, and they signal mainly through two different Smad pathways in an autocrine/paracrine manner. Defects in the upstream signalling cascade molecules, the ligands and receptors, are known to have adverse effects on ovarian organogenesis and folliculogenesis, but the role of the individual Smad proteins in the proper function of the ovary is just beginning to be understood for example through the use of Smad knockout models. Although most of the different Smad knockouts are embryonic lethal, it is known, however, that in Smad1 and Smad5 knockout mice primordial germ cell development is impaired and that Smad3 deficient mice harbouring a deletion in exon 8 exhibit impaired folliculogenesis and reduced fertility. In this minireview we discuss the role of Smad structure and function in the ovarian context.
Collapse
Affiliation(s)
- Noora Kaivo-oja
- Programme for Developmental and Reproductive Biology, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland and Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Luke A Jeffery
- Programme for Developmental and Reproductive Biology, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland and Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Olli Ritvos
- Programme for Developmental and Reproductive Biology, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland and Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - David G Mottershead
- Programme for Developmental and Reproductive Biology, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland and Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland
| |
Collapse
|
139
|
Kurisaki A, Kurisaki K, Kowanetz M, Sugino H, Yoneda Y, Heldin CH, Moustakas A. The mechanism of nuclear export of Smad3 involves exportin 4 and Ran. Mol Cell Biol 2006; 26:1318-32. [PMID: 16449645 PMCID: PMC1367208 DOI: 10.1128/mcb.26.4.1318-1332.2006] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Transforming growth factor beta (TGF-beta) receptors phosphorylate Smad3 and induce its nuclear import so it can regulate gene transcription. Smad3 can return to the cytoplasm to propagate further cycles of signal transduction or to be degraded. We demonstrate that Smad3 is exported by a constitutive mechanism that is insensitive to leptomycin B. The Mad homology 2 (MH2) domain is responsible for Smad3 export, which requires the GTPase Ran. Inactive, GDP-locked RanT24N or nuclear microinjection of Ran GTPase activating protein 1 blocked Smad3 export. Inactivation of the Ran guanine nucleotide exchange factor RCC1 inhibited Smad3 export and led to nuclear accumulation of phosphorylated Smad3. A screen for importin/exportin family members that associate with Smad3 identified exportin 4, which binds a conserved peptide sequence in the MH2 domain of Smad3 in a Ran-dependent manner. Exportin 4 is sufficient for carrying the in vitro nuclear export of Smad3 in cooperation with Ran. Knockdown of endogenous exportin 4 completely abrogates the export of endogenous Smad3. A short peptide representing the minimal interaction domain in Smad3 effectively competes with Smad3 association to exportin 4 and blocks nuclear export of Smad3 in vivo. We thus delineate a novel nuclear export pathway for Smad3.
Collapse
Affiliation(s)
- Akira Kurisaki
- Ludwig Institute for Cancer Research, Box 595 Biomedical Center, SE-751 24 Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
140
|
Kumar S, Saradhi M, Chaturvedi NK, Tyagi RK. Intracellular localization and nucleocytoplasmic trafficking of steroid receptors: an overview. Mol Cell Endocrinol 2006; 246:147-56. [PMID: 16388893 DOI: 10.1016/j.mce.2005.11.028] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Subcellular compartmentalization and dynamic movements of steroid receptors are major steps in executing their transcription regulatory function. Though significant progress has been made in understanding the mechanisms underlying nuclear import of NLS-bearing proteins, our general and mechanistic understanding about the nuclear export processes has begun to emerge only recently. The discovery of most commonly utilized CRM1/exportin1 dependent nuclear export pathway is attributed to a potent nuclear export inhibitor leptomycin B that helped dissecting this and other nuclear export pathways. Simultaneously, utilization of green fluorescent protein (GFP)-tagged intracellular steroid receptors has contributed to not only resolving controversial issue of subcellular localization of unliganded hormone receptors but also provided further insight into finer details of receptor dynamics in living cells. With judicious use of leptomycin B and expression of GFP-tagged receptors in living cells, existence of exportin1/CRM1 independent pathway(s), nuclear export signals and receptors for bi-directional translocation that are unique to steroid receptor trafficking have been specified. Currently, we appear to be arriving at a consensus that steroid/nuclear receptors follow dynamic nucleocytoplasmic processes that deviate from the ones commonly utilized by majority of other proteins.
Collapse
Affiliation(s)
- Sanjay Kumar
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | | | | | | |
Collapse
|
141
|
Abstract
Smad transcription factors lie at the core of one of the most versatile cytokine signaling pathways in metazoan biology-the transforming growth factor-beta (TGFbeta) pathway. Recent progress has shed light into the processes of Smad activation and deactivation, nucleocytoplasmic dynamics, and assembly of transcriptional complexes. A rich repertoire of regulatory devices exerts control over each step of the Smad pathway. This knowledge is enabling work on more complex questions about the organization, integration, and modulation of Smad-dependent transcriptional programs. We are beginning to uncover self-enabled gene response cascades, graded Smad response mechanisms, and Smad-dependent synexpression groups. Our growing understanding of TGFbeta signaling through the Smad pathway provides general principles for how animal cells translate complex inputs into concrete behavior.
Collapse
Affiliation(s)
- Joan Massagué
- Cancer Biology and Genetics Program, Howard Hughes Medical Institute, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA.
| | | | | |
Collapse
|
142
|
Alabert C, Rogers L, Kahn L, Niellez S, Fafet P, Cerulis S, Blanchard JM, Hipskind RA, Vignais ML. Cell type-dependent control of NF-Y activity by TGF-beta. Oncogene 2006; 25:3387-96. [PMID: 16434965 DOI: 10.1038/sj.onc.1209385] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Transforming growth factor beta (TGF-beta) is a pluripotent cytokine that regulates cell growth and differentiation in a cell type-dependent fashion. TGF-beta exerts its effects through the activation of several signaling pathways. One involves membrane proximal events that lead to nuclear translocation of members of the Smad family of transcriptional regulators. TGF-beta can also activate MAPK cascades. Here, we show that TGF-beta induces nuclear translocation of the NF-YA subunit of the transcription factor NF-Y by a process that requires activation of the ERK cascade. This results in increased binding of endogenous NF-Y to chromatin and TGF-beta-dependent transcriptional regulation of the NF-Y target gene cyclin A2. Interestingly, the kinetics of NF-YA relocalization differs between epithelial cells and fibroblasts. NIH3T3 fibroblasts show an elevated basal level of phosphorylated p38 and delayed nuclear accumulation of NF-YA after TGF-beta treatment. In contrast, MDCK cells show low basal p38 activation, higher basal ERK phosphorylation and more rapid localization of NF-YA after induction. Thus, NF-Y activation by TGF-beta1 involves ERK1/2 and potentially an interplay between MAPK pathways, thereby opening the possibility for finely tuned transcriptional regulation.
Collapse
Affiliation(s)
- C Alabert
- Institut de Génétique Moléculaire de Montpellier, CNRS-UMR5535-IFR122, Montpellier, France
| | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Lee HG, Ueda M, Zhu X, Perry G, Smith MA. Ectopic expression of phospho-Smad2 in Alzheimer's disease: Uncoupling of the transforming growth factor-β pathway? J Neurosci Res 2006; 84:1856-61. [PMID: 16998902 DOI: 10.1002/jnr.21072] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Transforming growth factor-beta (TGF-beta), a multifunctional cytokine, has been widely suggested to play a role in the pathogenesis of Alzheimer's disease. Supporting this, levels of TGF-beta are elevated in the cerebrospinal fluid, sera, and brain of patients with Alzheimer's disease. Since TGF-beta is neuroprotective, whereas Alzheimer's disease is typified by neurodegeneration, we speculated that defects in TGF-beta signaling might abrogate its neuroprotective properties. Consistently with an increase in TGF-beta in Alzheimer's disease, we found significant increases in phospho-Smad2, a major downstream signaling molecule of TGF-beta, in hippocampal neurons of Alzheimer's disease compared with age-matched control patients. However, in contrast to an expected nuclear localization, phosphorylated Smad2 in Alzheimer's disease was predominantly, and ectopically, found in the neuronal cytoplasm, specifically colocalized with neurofibrillary tangles and granulovacuolar degeneration. Given that a nuclear localization is required to regulate the transcription of TGF-beta target genes to afford neuroprotection, the ectopic localization of phosphorylated Smad2 suggests a defect in the Smad-mediated signaling pathway of TGF-beta in Alzheimer's disease and consequent loss of neuroprotective function.
Collapse
Affiliation(s)
- Hyoung-gon Lee
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | | | |
Collapse
|
144
|
Schmierer B, Hill CS. Kinetic analysis of Smad nucleocytoplasmic shuttling reveals a mechanism for transforming growth factor beta-dependent nuclear accumulation of Smads. Mol Cell Biol 2005; 25:9845-58. [PMID: 16260601 PMCID: PMC1280270 DOI: 10.1128/mcb.25.22.9845-9858.2005] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Upon transforming growth factor beta (TGF-beta) stimulation, Smads accumulate in the nucleus, where they regulate gene expression. Using fluorescence perturbation experiments on Smad2 and Smad4 fused to either enhanced green fluorescent protein or photoactivatable green fluorescent protein, we have studied the kinetics of Smad nucleocytoplasmic shuttling in a quantitative manner in vivo. We have obtained rate constants for import and export of Smad2 and show that the cytoplasmic localization of Smad2 in uninduced cells reflects its nuclear export being more rapid than import. We find that TGF-beta-induced nuclear accumulation of Smad2 is caused by a pronounced drop in the export rate of Smad2 from the nucleus, which is associated with a strong decrease in nuclear mobility of Smad2 and Smad4. TGF-beta-induced nuclear accumulation involves neither a release from cytoplasmic retention nor an increase in Smad2 import rate. Hence, TGF-beta-dependent nuclear accumulation of Smad2 is caused exclusively by selective nuclear trapping of phosphorylated, complexed Smad2. The proposed mechanism reconciles signal-dependent nuclear accumulation of Smad2 with its continuous nucleocytoplasmic cycling properties.
Collapse
Affiliation(s)
- Bernhard Schmierer
- Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3PX, United Kingdom
| | | |
Collapse
|
145
|
Abstract
The establishment of human embryonic stem (ES) cells has opened possibilities for cell replacement therapy to treat diseases such as diabetes, Parkinson's disease and cardiac myopathies. Self-renewal is one of the essential defining characteristics of stem cells. If stem cells are to have widespread therapeutic applications, it is essential to identify the extrinsic and intrinsic factors maintaining self-renewal, particularly in culture. Insight into the regulation of known self-renewal transcription factors and cross-talk between their upstream signalling pathways is important for a better understanding of how stem cell self-renewal and differentiation are related to downstream target genes. This may lead to the establishment of protocols for obtaining a large supply of ES cells. Here, we review the role that TGFbeta superfamily members are thought to play in self-renewal and differentiation of human and mouse ES cells. We focus on the prototype TGFbeta, TGFbeta1, activin A, nodal and bone morphogenetic proteins and their expression, activity and function in embryonic stem cells.
Collapse
Affiliation(s)
- Gudrun Valdimarsdottir
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | | |
Collapse
|
146
|
De Bosscher K, Vanden Berghe W, Beck IME, Van Molle W, Hennuyer N, Hapgood J, Libert C, Staels B, Louw A, Haegeman G. A fully dissociated compound of plant origin for inflammatory gene repression. Proc Natl Acad Sci U S A 2005; 102:15827-32. [PMID: 16243974 PMCID: PMC1276063 DOI: 10.1073/pnas.0505554102] [Citation(s) in RCA: 209] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The identification of selective glucocorticoid receptor (GR) modifiers, which separate transactivation and transrepression properties, represents an important research goal for steroid pharmacology. Although the gene-activating properties of GR are mainly associated with undesirable side effects, its negative interference with the activity of transcription factors, such as NF-kappaB, greatly contributes to its antiinflammatory and immune-suppressive capacities. In the present study, we found that Compound A (CpdA), a plant-derived phenyl aziridine precursor, although not belonging to the steroidal class of GR-binding ligands, does mediate gene-inhibitory effects by activating GR. We demonstrate that CpdA exerts an antiinflammatory potential by down-modulating TNF-induced proinflammatory gene expression, such as IL-6 and E-selectin, but, interestingly, does not at all enhance glucocorticoid response element-driven genes or induce GR binding to glucocorticoid response element-dependent genes in vivo. We further show that the specific gene-repressive effect of CpdA depends on the presence of functional GR, displaying a differential phosphorylation status with CpdA as compared with dexamethasone treatment. The antiinflammatory mechanism involves both a reduction of the in vivo DNA-binding activity of p65 as well as an interference with the transactivation potential of NF-kappaB. Finally, we present evidence that CpdA is as effective as dexamethasone in counteracting acute inflammation in vivo and does not cause a hyperglycemic side effect. Taken together, this compound may be a lead compound of a class of antiinflammatory agents with fully dissociated properties and might thus hold great potential for therapeutic use.
Collapse
Affiliation(s)
- Karolien De Bosscher
- Laboratory of Eukaryotic Gene Expression and Signal Transduction, Department of Molecular Biology, Ghent University, K. L. Ledeganckstraat 35, B-9000 Ghent, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Smad4 dependency defines two classes of transforming growth factor {beta} (TGF-{beta}) target genes and distinguishes TGF-{beta}-induced epithelial-mesenchymal transition from its antiproliferative and migratory responses. Mol Cell Biol 2005. [PMID: 16135802 DOI: 10.1128/mcb.25.18.8108-8125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
In response to transforming growth factor beta (TGF-beta), Smad4 forms complexes with activated Smad2 and Smad3, which accumulate in the nucleus, where they both positively and negatively regulate TGF-beta target genes. Mutation or deletion of Smad4 is found in about 50% of pancreatic tumors and in about 15% of colorectal tumors. As Smad4 is a central component of the TGF-beta/Smad pathway, we have determined whether Smad4 is absolutely required for all TGF-beta responses, to evaluate the effect of its loss during human tumor development. We have generated cell lines from the immortalized human keratinocyte cell line HaCaT or the pancreatic tumor cell line Colo-357, which stably express a tetracyline-inducible small interfering RNA targeted against Smad4. In response to tetracycline, Smad4 expression is effectively silenced. Large-scale microarray analysis identifies two populations of TGF-beta target genes that are distinguished by their dependency on Smad4. Some genes absolutely require Smad4 for their regulation, while others do not. Functional analysis also indicates a differential Smad4 requirement for TGF-beta-induced functions; TGF-beta-induced cell cycle arrest and migration, but not epithelial-mesenchymal transition, are abolished after silencing of Smad4. Altogether our results suggest that loss of Smad4 might promote TGF-beta-mediated tumorigenesis by abolishing tumor-suppressive functions of TGF-beta while maintaining some tumor-promoting TGF-beta responses.
Collapse
|
148
|
Levy L, Hill CS. Smad4 dependency defines two classes of transforming growth factor {beta} (TGF-{beta}) target genes and distinguishes TGF-{beta}-induced epithelial-mesenchymal transition from its antiproliferative and migratory responses. Mol Cell Biol 2005; 25:8108-25. [PMID: 16135802 PMCID: PMC1234333 DOI: 10.1128/mcb.25.18.8108-8125.2005] [Citation(s) in RCA: 275] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
In response to transforming growth factor beta (TGF-beta), Smad4 forms complexes with activated Smad2 and Smad3, which accumulate in the nucleus, where they both positively and negatively regulate TGF-beta target genes. Mutation or deletion of Smad4 is found in about 50% of pancreatic tumors and in about 15% of colorectal tumors. As Smad4 is a central component of the TGF-beta/Smad pathway, we have determined whether Smad4 is absolutely required for all TGF-beta responses, to evaluate the effect of its loss during human tumor development. We have generated cell lines from the immortalized human keratinocyte cell line HaCaT or the pancreatic tumor cell line Colo-357, which stably express a tetracyline-inducible small interfering RNA targeted against Smad4. In response to tetracycline, Smad4 expression is effectively silenced. Large-scale microarray analysis identifies two populations of TGF-beta target genes that are distinguished by their dependency on Smad4. Some genes absolutely require Smad4 for their regulation, while others do not. Functional analysis also indicates a differential Smad4 requirement for TGF-beta-induced functions; TGF-beta-induced cell cycle arrest and migration, but not epithelial-mesenchymal transition, are abolished after silencing of Smad4. Altogether our results suggest that loss of Smad4 might promote TGF-beta-mediated tumorigenesis by abolishing tumor-suppressive functions of TGF-beta while maintaining some tumor-promoting TGF-beta responses.
Collapse
Affiliation(s)
- Laurence Levy
- Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3PX, United Kingdom
| | | |
Collapse
|
149
|
Davies M, Robinson M, Smith E, Huntley S, Prime S, Paterson I. Induction of an epithelial to mesenchymal transition in human immortal and malignant keratinocytes by TGF-beta1 involves MAPK, Smad and AP-1 signalling pathways. J Cell Biochem 2005; 95:918-31. [PMID: 15861394 DOI: 10.1002/jcb.20458] [Citation(s) in RCA: 192] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Recent data indicate that transforming growth factor-beta1 (TGF-beta1) can act to promote tumour progression in the late stages of carcinogenesis. The mechanism by which this occurs is unknown although a ligand-induced epithelial-mesenchymal transition (EMT) is thought to be important. In this study, we demonstrate that active Ras is required for TGF-beta1-induced EMT in human keratinocytes and that epidermal growth factor (EGF) can substitute for mutant Ras. EMT was reversed by the removal of TGF-beta1. Under conditions of TGF-beta1-induced EMT, cells were growth inhibited by the ligand resulting in G1 arrest. In cells containing normal Ras, TGF-beta1-activated ERK and p38 mitogen-activated protein kinases (MAPKs), and levels of activation were further increased by co-treatment with EGF. Inhibition of MAPK pathways and Smad2/3 signalling blocked the induction of EMT by TGF-beta1. Further, inhibition of the AP-1 transcriptional complex by [6]-Gingerol, or by the ectopic expression of JDP2, blocked TGF-beta1-induced EMT and conversely, stimulation of AP-1 by 12-O-tetradecanoylphorbol 13-acetate (TPA) substituted for EGF in the induction of EMT by TGF-beta1 in cells containing normal Ras. The presence of oncogenic Ras, the treatment of cells with EGF, or the treatment of cells with TPA to activate AP-1, potentiated TGF-beta1-induced Smad-dependent transcription, an effect that was attenuated by the inhibition of MAPKs and AP-1. The results demonstrate that active Ras and TGF-beta1 co-operate to reversibly induce EMT in human keratinocytes by mechanisms that involve MAPKs, Smad2/3 and AP-1. Further we demonstrate that MAPK/AP-1 signalling enhances Smad transcriptional activity under conditions associated with TGF-beta1-induced EMT.
Collapse
Affiliation(s)
- Maria Davies
- Department of Oral and Dental Science, University of Bristol, Bristol BS1 2LY, United Kingdom.
| | | | | | | | | | | |
Collapse
|
150
|
Lazzereschi D, Nardi F, Turco A, Ottini L, D'Amico C, Mariani-Costantini R, Gulino A, Coppa A. A complex pattern of mutations and abnormal splicing of Smad4 is present in thyroid tumours. Oncogene 2005; 24:5344-54. [PMID: 15940269 DOI: 10.1038/sj.onc.1208603] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Sensitivity to transforming growth factor-beta is impaired in thyroid tumours. Similar to Mad -- Mother Against Decapentaplegic-(Smad)4 is frequently altered in cancers, but its involvement in this system is unknown. We analysed 56 thyroid tumours of various histotypes for Smad4 mutations by PCR-SSCP and sequencing, linking them to Smad4 reactivity as examined by immunohistochemistry (IHC), and 29 of them also for abnormalities in RNA expression due to alternative splicing. In all, 15/56 cases (27%), both benign and malignant lesions, harbour alterations of Smad4 coding sequence. We found several novel intragenic mutations (13 missense, two silent, one frameshift and one large insertion-deletion), with high incidence in the linker region. A subset of mutated tumours failed to express Smad4 protein by IHC. We have also detected four alternatively spliced tumour-associated Smad4 isoforms, lacking portions of the linker region, and three more due to unreported internal exon-exon rearrangements. Smad4 is both frequently mutated and deregulated by aberrant splicing in thyroid tumours and these alterations may contribute as an early event to thyroid tumorigenesis.
Collapse
Affiliation(s)
- Davide Lazzereschi
- Department of Experimental Medicine and Pathology, I Faculty of Medicine, University of Rome 'La Sapienza', V.le Regina Elena 324, Rome 00161, Italy.
| | | | | | | | | | | | | | | |
Collapse
|