51
|
Min KW, Liggett JL, Silva G, Wu WW, Wang R, Shen RF, Eling TE, Baek SJ. NAG-1/GDF15 accumulates in the nucleus and modulates transcriptional regulation of the Smad pathway. Oncogene 2015; 35:377-88. [PMID: 25893289 PMCID: PMC4613816 DOI: 10.1038/onc.2015.95] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 02/25/2015] [Accepted: 03/03/2015] [Indexed: 02/06/2023]
Abstract
Protein dynamics, modifications, and trafficking are all processes that can modulate protein activity. Accumulating evidence strongly suggests that many proteins play distinctive roles dependent on cellular location. Nonsteroidal anti-inflammatory drug activated gene-1 (NAG-1) is a TGF-β superfamily protein that plays a role in cancer, obesity, and inflammation. NAG-1 is synthesized and cleaved into a mature peptide, which is ultimately secreted into the extracellular matrix (ECM). In this study, we have found that full-length NAG-1 is expressed in not only the cytoplasm and ECM, but also in the nucleus. NAG-1 is dynamically moved to the nucleus, exported into cytoplasm, and further transported into the ECM. We have also found that nuclear NAG-1 contributes to inhibition of the Smad pathway by interrupting the Smad complex. Overall, our study indicates that NAG-1 is localized in the nucleus and provides new evidence that NAG-1 controls transcriptional regulation in the Smad pathway.
Collapse
Affiliation(s)
- K-W Min
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA
| | - J L Liggett
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA
| | - G Silva
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA
| | - W W Wu
- Facility for Biotechnology Resources, CBER, Food and Drug Administration, Bethesda, MD, USA
| | - R Wang
- Facility for Biotechnology Resources, CBER, Food and Drug Administration, Bethesda, MD, USA
| | - R-F Shen
- Facility for Biotechnology Resources, CBER, Food and Drug Administration, Bethesda, MD, USA
| | - T E Eling
- Laboratory of Molecular Carcinogenesis, NIH/NIEHS, Research Triangle Park, NC, USA
| | - S J Baek
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA
| |
Collapse
|
52
|
Rajakylä EK, Viita T, Kyheröinen S, Huet G, Treisman R, Vartiainen MK. RNA export factor Ddx19 is required for nuclear import of the SRF coactivator MKL1. Nat Commun 2015; 6:5978. [PMID: 25585691 PMCID: PMC4309436 DOI: 10.1038/ncomms6978] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 11/27/2014] [Indexed: 12/03/2022] Open
Abstract
Controlled transport of macromolecules between the cytoplasm and nucleus is essential for homeostatic regulation of cellular functions. For instance, gene expression entails coordinated nuclear import of transcriptional regulators to activate transcription and nuclear export of the resulting messenger RNAs for cytoplasmic translation. Here we link these two processes by reporting a novel role for the mRNA export factor Ddx19/Dbp5 in nuclear import of MKL1, the signal-responsive transcriptional activator of SRF. We show that Ddx19 is not a general nuclear import factor, and that its specific effect on MKL1 nuclear import is separate from its role in mRNA export. Both helicase and nuclear pore-binding activities of Ddx19 are dispensable for MKL1 nuclear import, but RNA binding is required. Mechanistically, Ddx19 operates by modulating the conformation of MKL1, which affects its interaction with Importin-β for efficient nuclear import. Thus, Ddx19 participates in mRNA export, translation and nuclear import of a key transcriptional regulator.
Collapse
Affiliation(s)
- Eeva Kaisa Rajakylä
- Program in Cell and Molecular Biology, Institute of Biotechnology, University of Helsinki, Viikinkaari 9, Helsinki 00014, Finland
| | - Tiina Viita
- Program in Cell and Molecular Biology, Institute of Biotechnology, University of Helsinki, Viikinkaari 9, Helsinki 00014, Finland
| | - Salla Kyheröinen
- Program in Cell and Molecular Biology, Institute of Biotechnology, University of Helsinki, Viikinkaari 9, Helsinki 00014, Finland
| | - Guillaume Huet
- Program in Cell and Molecular Biology, Institute of Biotechnology, University of Helsinki, Viikinkaari 9, Helsinki 00014, Finland
| | - Richard Treisman
- Transcription group, Cancer Research UK London Research Institute, London WC2A 3LY, UK
| | - Maria K. Vartiainen
- Program in Cell and Molecular Biology, Institute of Biotechnology, University of Helsinki, Viikinkaari 9, Helsinki 00014, Finland
| |
Collapse
|
53
|
Vizza D, Perri A, Toteda G, Lupinacci S, Leone F, Gigliotti P, Lofaro D, La Russa A, Bonofiglio R. Nerve growth factor exposure promotes tubular epithelial-mesenchymal transition via TGF-β1 signaling activation. Growth Factors 2015; 33:169-80. [PMID: 26066770 DOI: 10.3109/08977194.2015.1054989] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Clinical studies showed that renal expression and serum levels of nerve growth factor (NGF) are increased in renal diseases characterized by progressive fibrosis, a pathologic process in which TGF-β1 mediates most of the key events leading to tubular epithelial-mesenchymal transition (EMT). However, the pathogenic role of high NGF levels has not yet been elucidated. In this study, we found that in tubular renal cells, HK-2, NGF transcriptionally up-regulated TGF-β1 expression and secretion and enhanced cell motility by activating EMT markers via its receptors, TrkA and p75(NTR). Interestingly, we observed that TGF-β1-SMAD pathway activation and the up-regulation of EMT markers NGF-induced were both prevented when knockdown of TGF-β1 gene occurred and that the pretreatment with an antibody anti-NGF reversed the nuclear translocation of pSMAD3/SMAD4 complex. Collectively, our results demonstrated that NGF promotes renal fibrosis via TGF-β1-signaling activation, suggesting that in kidney diseases high NGF serum levels could contribute to worsen renal fibrosis.
Collapse
Affiliation(s)
- Donatella Vizza
- a Department of Nephrology Dialysis and Transplantation, Research Center Kidney and Transplantation , Annunziata Hospital , Cosenza , Italy
| | - Anna Perri
- a Department of Nephrology Dialysis and Transplantation, Research Center Kidney and Transplantation , Annunziata Hospital , Cosenza , Italy
| | - Giuseppina Toteda
- a Department of Nephrology Dialysis and Transplantation, Research Center Kidney and Transplantation , Annunziata Hospital , Cosenza , Italy
| | - Simona Lupinacci
- a Department of Nephrology Dialysis and Transplantation, Research Center Kidney and Transplantation , Annunziata Hospital , Cosenza , Italy
| | - Francesca Leone
- a Department of Nephrology Dialysis and Transplantation, Research Center Kidney and Transplantation , Annunziata Hospital , Cosenza , Italy
| | - Paolo Gigliotti
- a Department of Nephrology Dialysis and Transplantation, Research Center Kidney and Transplantation , Annunziata Hospital , Cosenza , Italy
| | - Danilo Lofaro
- a Department of Nephrology Dialysis and Transplantation, Research Center Kidney and Transplantation , Annunziata Hospital , Cosenza , Italy
| | - Antonella La Russa
- a Department of Nephrology Dialysis and Transplantation, Research Center Kidney and Transplantation , Annunziata Hospital , Cosenza , Italy
| | - Renzo Bonofiglio
- a Department of Nephrology Dialysis and Transplantation, Research Center Kidney and Transplantation , Annunziata Hospital , Cosenza , Italy
| |
Collapse
|
54
|
Xia X, Wu W, Huang C, Cen G, Jiang T, Cao J, Huang K, Qiu Z. SMAD4 and its role in pancreatic cancer. Tumour Biol 2014; 36:111-9. [PMID: 25464861 DOI: 10.1007/s13277-014-2883-z] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 11/19/2014] [Indexed: 12/13/2022] Open
Abstract
Transforming growth factor-β (TGF-β) regulates cell functions and has key roles in pancreatic cancer development. SMAD4, as one of the Smads family of signal transducer from TGF-β, mediates pancreatic cell proliferation and apoptosis and is specifically inactivated in half of advanced pancreatic cancers. In recent years, many advances concerning SMAD4 had tried to unravel the complex signaling mechanisms of TGF-β and its dual role of tumor-suppressive and tumor-promoting efforts in pancreatic cancer initiation and progression through SMAD4-dependent TGF-β signaling and SMAD4-independent TGF-β signaling pathways. Meanwhile, its potential prognostic value based on immunohistochemical expression in surgical sample was variably reported by several studies and short of a systematic analysis. This review aimed to discuss the structure, functions, and regulation of this principal protein and its effects in determining the progression and prognosis of pancreatic cancer.
Collapse
Affiliation(s)
- Xiang Xia
- Department of General Surgery, Shanghai Jiaotong University Affiliated First People's Hospital, 100 Hai Ning Road, Shanghai, 200080, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
55
|
GUO JIA, SONG BO, LI XIANG, HE CHENHUI, YANG GANGLONG, YANG XIAOMIN, GUAN FENG. Downregulation of gangliotetraosylceramide and β1,3-galactosyltransferase-4 gene expression by Smads during transforming growth factor β-induced epithelial-mesenchymal transition. Mol Med Rep 2014; 11:2241-7. [DOI: 10.3892/mmr.2014.2912] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 07/28/2014] [Indexed: 11/05/2022] Open
|
56
|
A novel SMAD family protein, SMAD9 is involved in follicular initiation and changes egg yield of geese via synonymous mutations in exon1 and intron2. Mol Biol Rep 2014; 42:289-302. [DOI: 10.1007/s11033-014-3772-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 09/20/2014] [Indexed: 12/16/2022]
|
57
|
Charni Chaabane S, Coomans de Brachène A, Essaghir A, Velghe A, Lo Re S, Stockis J, Lucas S, Khachigian LM, Huaux F, Demoulin JB. PDGF-D expression is down-regulated by TGFβ in fibroblasts. PLoS One 2014; 9:e108656. [PMID: 25280005 PMCID: PMC4184810 DOI: 10.1371/journal.pone.0108656] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 08/24/2014] [Indexed: 02/07/2023] Open
Abstract
Transforming growth factor-β (TGFβ) is a key mediator of fibrogenesis. TGFβ is overexpressed and activated in fibrotic diseases, regulates fibroblast differentiation into myofibroblasts and induces extracellular matrix deposition. Platelet-derived growth factor (PDGF) is also a regulator of fibrogenesis. Some studies showed a link between TGFβ and PDGF in certain fibrotic diseases. TGFβ induces PDGF receptor alpha expression in scleroderma fibroblasts. PDGF-C and -D are the most recently discovered ligands and also play a role in fibrosis. In this study, we report the first link between TGFβ and PDGF-D and -C ligands. In normal fibroblasts, TGFβ down-regulated PDGF-D expression and up-regulated PDGF-C expression at the mRNA and protein levels. This phenomenon is not limited to TGFβ since other growth factors implicated in fibrosis, such as FGF, EGF and PDGF-B, also regulated PDGF-D and PDGF-C expression. Among different kinase inhibitors, only TGFβ receptor inhibitors and the IκB kinase (IKK) inhibitor BMS-345541 blocked the effect of TGFβ. However, activation of the classical NF-κB pathway was not involved. Interestingly, in a model of lung fibrosis induced by either bleomycin or silica, PDGF-D was down-regulated, which correlates with the production of TGFβ and other fibrotic growth factors. In conclusion, the down-regulation of PDGF-D by TGFβ and other growth factors may serve as a negative feedback in the network of cytokines that control fibrosis.
Collapse
Affiliation(s)
| | | | - Ahmed Essaghir
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Amélie Velghe
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Sandra Lo Re
- Louvain center of Toxicology and Applied Pharmacology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Julie Stockis
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Sophie Lucas
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium
| | - Levon M. Khachigian
- Center of Vascular Research, University of New South Wales, Sydney, Australia
| | - François Huaux
- Louvain center of Toxicology and Applied Pharmacology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | | |
Collapse
|
58
|
Howell DW, Popovic N, Metz RP, Wilson E. Regional changes in elastic fiber organization and transforming growth factor β signaling in aortas from a mouse model of marfan syndrome. Cell Tissue Res 2014; 358:807-19. [PMID: 25238995 DOI: 10.1007/s00441-014-1993-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 08/21/2014] [Indexed: 11/27/2022]
Abstract
In Marfan Syndrome (MFS), development of thoracic aortic aneurysms (TAAs) is characterized by degeneration of the medial layer of the aorta, including fragmentation and loss of elastic fibers, phenotypic changes in the smooth muscle cells, and an increase in the active form of transforming growth factor-β (TGFβ), which is thought to play a major role in development and progression of the aneurysm. We hypothesized that regional difference in elastic fiber fragmentation contributes to TGFβ activation and hence the localization of aneurysm formation. The fibrillin-1-deficient mgR/mgR mouse model of MFS was used to investigate regional changes in elastin fiber fragmentation, TGFβ activation and changes in gene expression as compared to wild-type littermates. Knockdown of Smad 2 and Smad 3 with shRNA was used to determine the role of the specific transcription factors in gene regulation in aortic smooth muscle cells. We show increased elastin fiber fragmentation in the regions associated with aneurysm formation and altered TGFβ signaling in these regions. Differential effects of Smad 2 and Smad 3 were observed in cultured smooth muscle cells by shRNA-mediated knockdown of expression of these transcription factors. Differential signaling through Smad 2 and Smad 3 in regions of active vascular remodeling likely contribute to aneurysm formation in the mgR/mgR model of MFS. Increased elastin fiber fragmentation in these regions is associated with these changes as compared to other regions of the thoracic aorta and may contribute to the changes in TGFβ signaling in these regions.
Collapse
Affiliation(s)
- David W Howell
- Department of Medical Physiology, Texas A&M Health Science Center, College Station, TX, 77843-1114, USA
| | | | | | | |
Collapse
|
59
|
Bellaye PS, Wettstein G, Burgy O, Besnard V, Joannes A, Colas J, Causse S, Marchal-Somme J, Fabre A, Crestani B, Kolb M, Gauldie J, Camus P, Garrido C, Bonniaud P. The small heat-shock protein α
B-crystallin is essential for the nuclear localization of Smad4: impact on pulmonary fibrosis. J Pathol 2014; 232:458-72. [DOI: 10.1002/path.4314] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 11/25/2013] [Accepted: 11/29/2013] [Indexed: 11/07/2022]
Affiliation(s)
- Pierre-Simon Bellaye
- INSERM U866 Équipe Labellisée Ligue Contre le Cancer; Dijon France
- Faculty of Medicine and Pharmacy; University of Burgundy; Dijon France
| | - Guillaume Wettstein
- INSERM U866 Équipe Labellisée Ligue Contre le Cancer; Dijon France
- Faculty of Medicine and Pharmacy; University of Burgundy; Dijon France
| | - Olivier Burgy
- INSERM U866 Équipe Labellisée Ligue Contre le Cancer; Dijon France
- Faculty of Medicine and Pharmacy; University of Burgundy; Dijon France
| | | | | | - Julien Colas
- INSERM U866 Équipe Labellisée Ligue Contre le Cancer; Dijon France
- Faculty of Medicine and Pharmacy; University of Burgundy; Dijon France
| | - Sébastien Causse
- INSERM U866 Équipe Labellisée Ligue Contre le Cancer; Dijon France
- Faculty of Medicine and Pharmacy; University of Burgundy; Dijon France
| | | | | | - Bruno Crestani
- INSERM U700; Paris France
- Université Paris Diderot; PRES Sorbonne Paris Cité Paris France
- Faculté de Médecine Bichat; Paris APHP, Hôpital Bichat, Service de Pneumologie A; Paris France
| | - Martin Kolb
- Center for Gene Therapeutics; McMaster University; Hamilton ON Canada
| | - Jack Gauldie
- Center for Gene Therapeutics; McMaster University; Hamilton ON Canada
| | - Philippe Camus
- INSERM U866 Équipe Labellisée Ligue Contre le Cancer; Dijon France
- Faculty of Medicine and Pharmacy; University of Burgundy; Dijon France
- Service de Pneumologie; CHU Dijon France
| | - Carmen Garrido
- INSERM U866 Équipe Labellisée Ligue Contre le Cancer; Dijon France
- Faculty of Medicine and Pharmacy; University of Burgundy; Dijon France
| | - Philippe Bonniaud
- INSERM U866 Équipe Labellisée Ligue Contre le Cancer; Dijon France
- Faculty of Medicine and Pharmacy; University of Burgundy; Dijon France
- Service de Pneumologie; CHU Dijon France
| |
Collapse
|
60
|
Chatterjee S, Elinson RP. Commitment to nutritional endoderm in Eleutherodactylus coqui involves altered nodal signaling and global transcriptional repression. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2013; 322:27-44. [PMID: 24323742 DOI: 10.1002/jez.b.22543] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 09/11/2013] [Accepted: 09/24/2013] [Indexed: 01/01/2023]
Abstract
The vegetal cells of a Xenopus laevis embryo commit to mesendoderm via the Nodal-signaling pathway. In the direct developing frog Eleutherodactylus coqui, mesendoderm is specified at the marginal zone of the early gastrula, and vegetal core cells transform into nutritional endoderm. Nutritional endoderm, a novel tissue, consists of transient, yolky cells that provide nutrition but remain undifferentiated. We report a dual regulation for the generation of nutritional endoderm. First, differential expressions of the Nodal-signal transducers Smad2 and Smad4 were observed during early gastrulation between the marginal zone and the vegetal core cells. Although EcSmad2 RNA as well as total and activated Smad2 protein were detected in the vegetal core, Smad4 protein was expressed less in vegetal core during early gastrulation. Only 12% and 50% of vegetal core cells were positive for nuclear Smad2 and Smad4 signals respectively compared to 100% of marginal zone cells. These results suggest a signaling disruption in the vegetal core. Second, vegetal core cells were transcriptionally repressed. At the blastula stage, both marginal zone and vegetal core cells were transcriptionally silent, but during early gastrulation, only marginal zone cells became transcriptionally active. This indicates the occurrence of a mid-blastula transition in the marginal zone by early gastrulation, but global transcriptional repression persisted in the vegetal core and its derivative, nutritional endoderm, throughout development. We have described a novel mechanism, which prevents differentiation of the vegetal core through differential Nodal-signaling and global transcriptional repression.
Collapse
Affiliation(s)
- Suman Chatterjee
- Department of Biological Sciences, Duquesne University, Pittsburgh, Pennsylvania
| | | |
Collapse
|
61
|
Vizán P, Miller DSJ, Gori I, Das D, Schmierer B, Hill CS. Controlling long-term signaling: receptor dynamics determine attenuation and refractory behavior of the TGF-β pathway. Sci Signal 2013; 6:ra106. [PMID: 24327760 DOI: 10.1126/scisignal.2004416] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Understanding the complex dynamics of growth factor signaling requires both mechanistic and kinetic information. Although signaling dynamics have been studied for pathways downstream of receptor tyrosine kinases and G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptors, they have not been investigated for the transforming growth factor-β (TGF-β) superfamily pathways. Using an integrative experimental and mathematical modeling approach, we dissected the dynamic behavior of the TGF-β to Smad pathway, which is mediated by type I and type II receptor serine/threonine kinases, in response to acute, chronic, and repeated ligand stimulations. TGF-β exposure produced a transient response that attenuated over time, resulting in desensitized cells that were refractory to further acute stimulation. This loss of signaling competence depended on ligand binding, but not on receptor activity, and was restored only after the ligand had been depleted. Furthermore, TGF-β binding triggered the rapid depletion of signaling-competent receptors from the cell surface, with the type I and type II receptors exhibiting different degradation and trafficking kinetics. A computational model of TGF-β signal transduction from the membrane to the nucleus that incorporates our experimental findings predicts that autocrine signaling, such as that associated with tumorigenesis, severely compromises the TGF-β response, which we confirmed experimentally. Thus, we have shown that the long-term signaling behavior of the TGF-β pathway is determined by receptor dynamics, does not require TGF-β-induced gene expression, and influences context-dependent responses in vivo.
Collapse
Affiliation(s)
- Pedro Vizán
- 1Developmental Signalling Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | | | | | | | | | | |
Collapse
|
62
|
Kristensen SG, Andersen K, Clement CA, Franks S, Hardy K, Andersen CY. Expression of TGF-beta superfamily growth factors, their receptors, the associated SMADs and antagonists in five isolated size-matched populations of pre-antral follicles from normal human ovaries. ACTA ACUST UNITED AC 2013; 20:293-308. [DOI: 10.1093/molehr/gat089] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
|
63
|
Abstract
The post-genomic era has produced a variety of new investigation technologies, techniques and approaches that may offer exciting insights into many long-standing questions of scientific research. The microtubule cytoskeleton is a highly conserved system that shows a high degree of internal complexity, is known to be integral to many cell systems and functions on a fundamental level. After decades of study, much is still unknown about microtubules in vivo from the control of dynamics in living cells to their responses to environmental changes and responses to other cellular processes. In the present article, we examine some outstanding questions in the microtubule field and propose a combination of emerging interdisciplinary approaches, i.e. high-throughput functional genomics techniques, quantitative and super-resolution microscopy, and in silico modelling, that could shed light on the systemic regulation of microtubules in cells by networks of regulatory factors. We propose that such an integrative approach is key to elucidate the function of the microtubule cytoskeleton as a complete responsive integral biological system.
Collapse
|
64
|
Nicklas D, Saiz L. Computational modelling of Smad-mediated negative feedback and crosstalk in the TGF-β superfamily network. J R Soc Interface 2013; 10:20130363. [PMID: 23804438 DOI: 10.1098/rsif.2013.0363] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The transforming growth factor-β (TGF-β) signal transduction pathway controls many cellular processes, including differentiation, proliferation and apoptosis. It plays a fundamental role during development and it is dysregulated in many diseases. The factors that control the dynamics of the pathway, however, are not fully elucidated yet and so far computational approaches have been very limited in capturing the distinct types of behaviour observed under different cellular backgrounds and conditions into a single-model description. Here, we develop a detailed computational model for TGF-β signalling that incorporates elements of previous models together with crosstalking between Smad1/5/8 and Smad2/3 channels through a negative feedback loop dependent on Smad7. The resulting model accurately reproduces the diverse behaviour of experimental datasets for human keratinocytes, bovine aortic endothelial cells and mouse mesenchymal cells, capturing the dynamics of activation and nucleocytoplasmic shuttling of both R-Smad channels. The analysis of the model dynamics and its system properties revealed Smad7-mediated crosstalking between Smad1/5/8 and Smad2/3 channels as a major determinant in shaping the distinct responses to single and multiple ligand stimulation for different cell types.
Collapse
Affiliation(s)
- Daniel Nicklas
- Modeling of Biological Networks Laboratory, Department of Biomedical Engineering, University of California, 451 East Health Sciences Drive, Davis, CA 95616, USA
| | | |
Collapse
|
65
|
Shahni R, Czajka A, Mankoo BS, Guvenel AK, King AJ, Malik AN. Nop-7-associated 2 (NSA2), a candidate gene for diabetic nephropathy, is involved in the TGFβ1 pathway. Int J Biochem Cell Biol 2012; 45:626-35. [PMID: 23220173 DOI: 10.1016/j.biocel.2012.11.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 10/28/2012] [Accepted: 11/28/2012] [Indexed: 01/29/2023]
Abstract
We recently showed that Nop-7-associated 2 (NSA2) originally described in yeast as a nuclear protein involved in ribosomal biogenesis, is a hyperglycemia induced gene involved in diabetic nephropathy [Shahni et al., Elevated levels of renal and circulating Nop-7-associated 2 (NSA2) in rat and mouse models of diabetes, in mesangial cells in vitro and in patients with diabetic nephropathy. Diabetologia 2012;55(March(3)):825-34]. However the function of NSA2 in the cell remains unknown. In the current paper we investigate the possible mechanisms for the involvement of NSA2 in diabetic nephropathy by testing the hypothesis that NSA2 expression is linked to the TGFβ1 pathway. Both TGFβ1 and NSA2 mRNAs were significantly up-regulated in cultured renal mesangial cells in response to high glucose, in mouse kidneys during hyperglycemia, and in developing kidneys of mouse embryos during mesenchymal to epithelial transition. Surprisingly, the previously described nuclear NSA2 protein was predominantly located in the cytosol of cultured renal cells. Exogenous TGFβ1 could elevate NSA2 mRNA/protein levels in cultured mesangial cells and could also affect the cellular localization of NSA2, causing the predominantly cytosolic NSA2 protein to rapidly translocate to the nucleus. Increased NSA2 nuclear staining was seen in diabetic mouse kidneys compared to control kidneys. Knock-down of NSA2 expression using RNA interference resulted in significantly decreased TGFβ1 mRNA/protein, almost abolished TGFβ1 activity, and resulted in significantly reduced mRNA levels of the TGFβ1 downstream gene fibronectin. Our data suggest that NSA2 is acting upstream of the TGFβ1 pathway and that NSA2 is needed for TGFβ1 expression and transcriptional activity. In summary, NSA2, which increases in diabetic nephropathy, may be involved in the actions of TGFβ1 and contribute to the development of diabetic nephropathy.
Collapse
Affiliation(s)
- Rojeen Shahni
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, School of Medicine, King's College London, Hodgkin Building, London Bridge, London SE1 1UL, United Kingdom
| | | | | | | | | | | |
Collapse
|
66
|
Li Y, Wang M, Carra C, Cucinotta FA. Modularized Smad-regulated TGFβ signaling pathway. Math Biosci 2012; 240:187-200. [DOI: 10.1016/j.mbs.2012.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 07/19/2012] [Accepted: 07/20/2012] [Indexed: 01/05/2023]
|
67
|
Alcaraz A, Mrowiec A, Insausti CL, García-Vizcaíno EM, Ruiz-Canada C, López-Martínez MC, Moraleda JM, Nicolás FJ. Autocrine TGF-β induces epithelial to mesenchymal transition in human amniotic epithelial cells. Cell Transplant 2012; 22:1351-67. [PMID: 23031712 DOI: 10.3727/096368912x657387] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Human amniotic epithelial cells (hAECs) have been the object of intense research due to their potential therapeutic use. In this paper, we present molecular evidence of a bona fide epithelial to mesenchymal transition (EMT) undergone by hAECs. Amniotic membrane (AM)-derived hAECs showed the presence of typical epithelial markers such as E-cadherin and cytokeratins. hAECs in culture, however, underwent morphological changes acquiring a mesenchymal shape. Epithelial cell markers were lost and typical mesenchymal markers, such as vimentin and α-SMA, appeared. Several genes associated with EMT, such as SNAI1, MMP9, PAI1, or ACTA2, increased their expression. The expression of the transcription activators KLF4 or MTA3 was consistent with the downregulation of CDH1. We have shown that hAECs undergo EMT due to the autocrine production of TGF-β. Furthermore, the addition of the TGF-β receptor I (ALK5) inhibitor SB-431542 or TGF-β neutralizing antibody to hAECs prevented EMT and preserved the hAECs' epithelial phenotype. Altogether, these results suggest that cultured hAECs undergo EMT through the autocrine production of TGF-β.
Collapse
Affiliation(s)
- Antonia Alcaraz
- Oncología Molecular y TGF-β, Unidad de Investigación, Hospital Universitario Virgen de la Arrixaca, El Palmar, Murcia, Spain
| | | | | | | | | | | | | | | |
Collapse
|
68
|
Eroshkin FM, Baĭramov AV, Martynova NI, Zaraĭskiĭ AG. [Using of the luciferase reporter constructs for investigation of the capacity of Noggin2 protein to inhibit cell signaling pathways in early Xenopus laevis embryos]. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2012; 38:385-8. [PMID: 22997712 DOI: 10.1134/s106816201203003x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Noggin (Noggin1) protein inhibits Smad1-dependent TGF-beta signaling pathway by extracellular binding of BMP proteins. Recently we found two previously unknown representatives of Noggin family - Noggin2 and Noggin4. Here we investigate by using the luciferase reporter constructs the ability of Noggin2 to inhibit BMP-, Activin/Nodal- and Wnt-signaling pathways in the early Xenopus laevis embryos. The effectiveness of this inhibitory activity of Noggin2 is comparing with that of well-known inhibitors of the indicated pathways - the secreted proteins Cerberus and Follistatin. Besides revealing of novel properties of Noggin2, our findings demonstrate that luciferase-reporter assay is as a useful tool for signaling pathways investigation in the model of Xenopus embryos.
Collapse
|
69
|
Cao Q, Zhang X, Lu L, Yang L, Gao J, Gao Y, Ma H, Cao Y. Klf4 is required for germ-layer differentiation and body axis patterning during Xenopus embryogenesis. Development 2012; 139:3950-61. [PMID: 22992953 DOI: 10.1242/dev.082024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Klf4 is a transcription factor of the family of Kruppel-like factors and plays important roles in stem cell biology; however, its function during embryogenesis is unknown. Here, we report the characterization of a Klf4 homologue in Xenopus laevis during embryogenesis. Klf4 is transcribed both maternally and zygotically and the transcript is ubiquitous in embryos during germ-layer formation. Klf4 promotes endoderm differentiation in both Nodal/Activin-dependent and -independent manners. Moreover, Klf4 regulates anteroposterior body axis patterning via activation of a subset of genes in the Spemann organizer, such as Noggin, Dkk1 and Cerberus, which encode Nodal, Wnt and BMP antagonists. Loss of Klf4 function leads to the failure of germ-layer differentiation, the loss of responsiveness of early embryonic cells to inducing signals, e.g. Nodal/Activin, and the loss of transcription of genes involved in axis patterning. We conclude that Klf4 is required for germ-layer differentiation and body axis patterning by means of rendering early embryonic cells competent to differentiation signals.
Collapse
Affiliation(s)
- Qing Cao
- Model Animal Research Center of Nanjing University and MOE Key Laboratory of Model Animals for Disease Study, 12 Xuefu Road, Pukou High-Tech Zone, 210061 Nanjing, China
| | | | | | | | | | | | | | | |
Collapse
|
70
|
Dynamics of TGF-β signaling reveal adaptive and pulsatile behaviors reflected in the nuclear localization of transcription factor Smad4. Proc Natl Acad Sci U S A 2012; 109:E1947-56. [PMID: 22689943 DOI: 10.1073/pnas.1207607109] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The TGF-β pathway plays a vital role in development and disease and regulates transcription through a complex composed of receptor-regulated Smads (R-Smads) and Smad4. Extensive biochemical and genetic studies argue that the pathway is activated through R-Smad phosphorylation; however, the dynamics of signaling remain largely unexplored. We monitored signaling and transcriptional dynamics and found that although R-Smads stably translocate to the nucleus under continuous pathway stimulation, transcription of direct targets is transient. Surprisingly, Smad4 nuclear localization is confined to short pulses that coincide with transcriptional activity. Upon perturbation, the dynamics of transcription correlate with Smad4 nuclear localization rather than with R-Smad activity. In Xenopus embryos, Smad4 shows stereotyped, uncorrelated bursts of nuclear localization, but activated R-Smads are uniform. Thus, R-Smads relay graded information about ligand levels that is integrated with intrinsic temporal control reflected in Smad4 into the active signaling complex.
Collapse
|
71
|
Xu P, Liu J, Derynck R. Post-translational regulation of TGF-β receptor and Smad signaling. FEBS Lett 2012; 586:1871-84. [PMID: 22617150 DOI: 10.1016/j.febslet.2012.05.010] [Citation(s) in RCA: 166] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Revised: 05/06/2012] [Accepted: 05/07/2012] [Indexed: 01/17/2023]
Abstract
TGF-β family signaling through Smads is conceptually a simple and linear signaling pathway, driven by sequential phosphorylation, with type II receptors activating type I receptors, which in turn activate R-Smads. Nevertheless, TGF-β family proteins induce highly complex programs of gene expression responses that are extensively regulated, and depend on the physiological context of the cells. Regulation of TGF-β signaling occurs at multiple levels, including TGF-β activation, formation, activation and destruction of functional TGF-β receptor complexes, activation and degradation of Smads, and formation of Smad transcription complexes at regulatory gene sequences that cooperate with a diverse set of DNA binding transcription factors and coregulators. Here we discuss recent insights into the roles of post-translational modifications and molecular interaction networks in the functions of receptors and Smads in TGF-β signal responses. These layers of regulation demonstrate how a simple signaling system can be coopted to exert exquisitely regulated, complex responses.
Collapse
Affiliation(s)
- Pinglong Xu
- Department of Cell and Tissue Biology, Programs in Cell Biology and Developmental Biology, University of California, San Francisco, CA, USA
| | | | | |
Collapse
|
72
|
Zieba A, Pardali K, Söderberg O, Lindbom L, Nyström E, Moustakas A, Heldin CH, Landegren U. Intercellular variation in signaling through the TGF-β pathway and its relation to cell density and cell cycle phase. Mol Cell Proteomics 2012; 11:M111.013482. [PMID: 22442258 DOI: 10.1074/mcp.m111.013482] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Fundamental open questions in signal transduction remain concerning the sequence and distribution of molecular signaling events among individual cells. In this work, we have characterized the intercellular variability of transforming growth factor β-induced Smad interactions, providing essential information about TGF-β signaling and its dependence on the density of cell populations and the cell cycle phase. By employing the recently developed in situ proximity ligation assay, we investigated the dynamics of interactions and modifications of Smad proteins and their partners under native and physiological conditions. We analyzed the kinetics of assembly of Smad complexes and the influence of cellular environment and relation to mitosis. We report rapid kinetics of formation of Smad complexes, including native Smad2-Smad3-Smad4 trimeric complexes, in a manner influenced by the rate of proteasomal degradation of these proteins, and we found a striking cell to cell variation of signaling complexes. The single-cell analysis of TGF-β signaling in genetically unmodified cells revealed previously unknown aspects of regulation of this pathway, and it provided a basis for analysis of these signaling events to diagnose pathological perturbations in patient samples and to evaluate their susceptibility to drug treatment.
Collapse
Affiliation(s)
- Agata Zieba
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Se-75185 Sweden
| | | | | | | | | | | | | | | |
Collapse
|
73
|
D'Inzeo S, Nicolussi A, Donini CF, Zani M, Mancini P, Nardi F, Coppa A. A novel human Smad4 mutation is involved in papillary thyroid carcinoma progression. Endocr Relat Cancer 2012; 19:39-55. [PMID: 22109972 DOI: 10.1530/erc-11-0233] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Smad proteins are the key effectors of the transforming growth factor β (TGFβ) signaling pathway in mammalian cells. Smad4 plays an important role in human physiology, and its mutations were found with high frequency in wide range of human cancer. In this study, we have functionally characterized Smad4 C324Y mutation, isolated from a nodal metastasis of papillary thyroid carcinoma. We demonstrated that the stable expression of Smad4 C324Y in FRTL-5 cells caused a significant activation of TGFβ signaling, responsible for the acquisition of transformed phenotype and invasive behavior. The coexpression of Smad4 C324Y with Smad4 wild-type determined an increase of homo-oligomerization of Smad4 with receptor-regulated Smads and a lengthening of nuclear localization. FRTL-5 clones overexpressing Smad4 C324Y showed a strong reduction of response to antiproliferative action of TGFβ1, acquired the ability to grow in anchorage-independent conditions, showed a fibroblast-like appearance and a strong reduction of the level of E-cadherin, one crucial event of the epithelial-mesenchymal transition process. The acquisition of a mesenchymal phenotype gave the characteristics of increased cellular motility and a significant reduction in adhesion to substrates such as fibronectin and laminin. Overall, our results demonstrate that the Smad4 C324Y mutation plays an important role in thyroid carcinogenesis and can be considered as a new prognostic and therapeutic target for thyroid cancer.
Collapse
Affiliation(s)
- Sonia D'Inzeo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
74
|
Wegner K, Bachmann A, Schad JU, Lucarelli P, Sahle S, Nickel P, Meyer C, Klingmüller U, Dooley S, Kummer U. Dynamics and feedback loops in the transforming growth factor β signaling pathway. Biophys Chem 2012; 162:22-34. [PMID: 22284904 DOI: 10.1016/j.bpc.2011.12.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 12/19/2011] [Accepted: 12/20/2011] [Indexed: 11/25/2022]
Abstract
Transforming growth factor β (TGF-β) ligands activate a signaling cascade with multiple cell context dependent outcomes. Disruption or disturbance leads to variant clinical disorders. To develop strategies for disease intervention, delineation of the pathway in further detail is required. Current theoretical models of this pathway describe production and degradation of signal mediating proteins and signal transduction from the cell surface into the nucleus, whereas feedback loops have not exhaustively been included. In this study we present a mathematical model to determine the relevance of feedback regulators (Arkadia, Smad7, Smurf1, Smurf2, SnoN and Ski) on TGF-β target gene expression and the potential to initiate stable oscillations within a realistic parameter space. We employed massive sampling of the parameters space to pinpoint crucial players for potential oscillations as well as transcriptional product levels. We identified Smad7 and Smurf2 with the highest impact on the dynamics. Based on these findings, we conducted preliminary time course experiments.
Collapse
Affiliation(s)
- Katja Wegner
- Biological and Neural Computation Group, Science and Technology Research Institute, University of Hertfordshire, College Lane, Hatfield, United Kingdom.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Lomniczi A, Garcia-Rudaz C, Ramakrishnan R, Wilmot B, Khouangsathiene S, Ferguson B, Dissen GA, Ojeda SR. A single-nucleotide polymorphism in the EAP1 gene is associated with amenorrhea/oligomenorrhea in nonhuman primates. Endocrinology 2012; 153:339-49. [PMID: 22128021 PMCID: PMC3249686 DOI: 10.1210/en.2011-1540] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Current evidence suggests that the acquisition of female reproductive capacity and the maintenance of mature reproductive function are related processes transcriptionally regulated by gene networks operating within the neuroendocrine brain. One of these genes, termed enhanced at puberty 1 (EAP1), encodes an upstream regulator of these processes. Selective inhibition of EAP1 expression in discrete regions of the rat and nonhuman primate (NHP) hypothalamus, via targeted delivery of RNA interference, either disrupts (rats) or abolishes (monkeys) reproductive cycles. The striking loss of menstrual cyclicity resulting from knocking down hypothalamic EAP1 expression suggests that diminished EAP1 function may contribute to disorders of the menstrual cycle of neuroendocrine origin. Here we show that a single-nucleotide polymorphism in the 5'-flanking region of EAP1 gene is associated with increased incidence of amenorrhea/oligomenorrhea in NHP. In the presence of the risk allele, binding of the transcription factor mothers against decapentaplegic homolog 3 (SMAD3) to its recognition site contained within the polymorphic sequence in the monkey EAP1 promoter is reduced. The risk allele also diminishes the increase in EAP1 promoter activity elicited by TGFβ1, a peptide that activates a SMAD3/4-mediated signaling pathway to regulate gene transcription. These findings indicate that common genetic variation in the EAP1 locus increases the susceptibility of NHP to loss/disruption of menstrual cyclicity. They also raise the possibility that polymorphisms in EAP1 may increase the risk of functional hypothalamic amenorrhea in humans.
Collapse
Affiliation(s)
- Alejandro Lomniczi
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Neuroscience, 505 NW 185th Avenue, Beaverton, Oregon 97006, USA.
| | | | | | | | | | | | | | | |
Collapse
|
76
|
Mechanism and regulation of nucleocytoplasmic trafficking of smad. Cell Biosci 2011; 1:40. [PMID: 22204445 PMCID: PMC3292837 DOI: 10.1186/2045-3701-1-40] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 12/28/2011] [Indexed: 11/24/2022] Open
Abstract
Smad proteins are the intracellular mediators of transforming growth factor β (TGF-β) signaling. Smads function as transcription factors and their activities require carboxyl-terminal phosphorylation by TGF-β receptor kinases which are embedded in the cell membrane. Therefore, the translocation of activated Smads from the cytoplasm into the nucleus is a rate-limiting step in TGF-β signal transduction into the nucleus. On the other hand, the export of Smads out of the nucleus turns off TGF-β effect. Such spatial control of Smad ensures a tight regulation of TGF-β target genes. Several cross-talk pathways have been shown to affect TGF-β signaling by impairing nuclear translocation of Smad, exemplifying the biological importance of the nuclear transport process. Many laboratories have investigated the underlying molecular mechanism of Smad nucleocytoplasmic translocation, combining genetics, biochemistry and sophisticated live cell imaging approaches. The last few years have witnessed the elucidation of several key players in Smad nuclear transport, most importantly the karyopherins that carry Smads across the nuclear envelope and nuclear pore proteins that facilitate the trans-nuclear envelope movement. The foundation is now set to further elucidate how the nuclear transport process is regulated and exploit such knowledge to manipulate TGF-β signaling. In this review we will discuss the current understanding of the molecular machinery responsible for nuclear import and export of Smads.
Collapse
|
77
|
Lomelí H, Vázquez M. Emerging roles of the SUMO pathway in development. Cell Mol Life Sci 2011; 68:4045-64. [PMID: 21892772 PMCID: PMC11115048 DOI: 10.1007/s00018-011-0792-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 08/02/2011] [Accepted: 08/04/2011] [Indexed: 01/01/2023]
Abstract
Sumoylation is a reversible post-translational modification that targets a variety of proteins mainly within the nucleus, but also in the plasma membrane and cytoplasm of the cell. It controls diverse cellular mechanisms such as subcellular localization, protein-protein interactions, or transcription factor activity. In recent years, the use of several developmental model systems has unraveled many critical functions for the sumoylation system in the early life of diverse species. In particular, detailed analyses of mutant organisms in both the components of the SUMO pathway and their targets have established the importance of the SUMO system in early developmental processes, such as cell division, cell lineage commitment, specification, and/or differentiation. In addition, an increasing number of developmental proteins, including transcription factors and epigenetic regulators, have been identified as sumoylation substrates. Sumoylation acts on these targets through various mechanisms. For example, this modification has been involved in converting a transcription factor from an activator to a repressor or in regulating the localization and/or stability of numerous transcription factors. This review will summarize current information on the function of sumoylation in embryonic development in different species from yeast to mammals.
Collapse
Affiliation(s)
- Hilda Lomelí
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| | | |
Collapse
|
78
|
Bayramov AV, Eroshkin FM, Martynova NY, Ermakova GV, Solovieva EA, Zaraisky AG. Novel functions of Noggin proteins: inhibition of Activin/Nodal and Wnt signaling. Development 2011; 138:5345-56. [PMID: 22071106 DOI: 10.1242/dev.068908] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The secreted protein Noggin1 is an embryonic inducer that can sequester TGFβ cytokines of the BMP family with extremely high affinity. Owing to this function, ectopic Noggin1 can induce formation of the headless secondary body axis in Xenopus embryos. Here, we show that Noggin1 and its homolog Noggin2 can also bind, albeit less effectively, to ActivinB, Nodal/Xnrs and XWnt8, inactivation of which, together with BMP, is essential for the head induction. In support of this, we show that both Noggin proteins, if ectopically produced in sufficient concentrations in Xenopus embryo, can induce a secondary head, including the forebrain. During normal development, however, Noggin1 mRNA is translated in the presumptive forebrain with low efficiency, which provides the sufficient protein concentration for only its BMP-antagonizing function. By contrast, Noggin2, which is produced in cells of the anterior margin of the neural plate at a higher concentration, also protects the developing forebrain from inhibition by ActivinB and XWnt8 signaling. Thus, besides revealing of novel functions of Noggin proteins, our findings demonstrate that specification of the forebrain requires isolation of its cells from BMP, Activin/Nodal and Wnt signaling not only during gastrulation but also at post-gastrulation stages.
Collapse
Affiliation(s)
- Andrey V Bayramov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | | | | | | | | | | |
Collapse
|
79
|
Cellière G, Fengos G, Hervé M, Iber D. Plasticity of TGF-β signaling. BMC SYSTEMS BIOLOGY 2011; 5:184. [PMID: 22051045 PMCID: PMC3227652 DOI: 10.1186/1752-0509-5-184] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Accepted: 11/03/2011] [Indexed: 01/17/2023]
Abstract
Background The family of TGF-β ligands is large and its members are involved in many different signaling processes. These signaling processes strongly differ in type with TGF-β ligands eliciting both sustained or transient responses. Members of the TGF-β family can also act as morphogen and cellular responses would then be expected to provide a direct read-out of the extracellular ligand concentration. A number of different models have been proposed to reconcile these different behaviours. We were interested to define the set of minimal modifications that are required to change the type of signal processing in the TGF-β signaling network. Results To define the key aspects for signaling plasticity we focused on the core of the TGF-β signaling network. With the help of a parameter screen we identified ranges of kinetic parameters and protein concentrations that give rise to transient, sustained, or oscillatory responses to constant stimuli, as well as those parameter ranges that enable a proportional response to time-varying ligand concentrations (as expected in the read-out of morphogens). A combination of a strong negative feedback and fast shuttling to the nucleus biases signaling to a transient rather than a sustained response, while oscillations were obtained if ligand binding to the receptor is weak and the turn-over of the I-Smad is fast. A proportional read-out required inefficient receptor activation in addition to a low affinity of receptor-ligand binding. We find that targeted modification of single parameters suffices to alter the response type. The intensity of a constant signal (i.e. the ligand concentration), on the other hand, affected only the strength but not the type of the response. Conclusions The architecture of the TGF-β pathway enables the observed signaling plasticity. The observed range of signaling outputs to TGF-β ligand in different cell types and under different conditions can be explained with differences in cellular protein concentrations and with changes in effective rate constants due to cross-talk with other signaling pathways. It will be interesting to uncover the exact cellular differences as well as the details of the cross-talks in future work.
Collapse
Affiliation(s)
- Geraldine Cellière
- Department of Biosystems Science and Engineering (D-BSSE), Eidgenöossische Technische Hochschule Zurich (ETHZ), Mattenstrasse 26, 4058 Basel, Switzerland
| | | | | | | |
Collapse
|
80
|
Huang D, Wang Y, Wang L, Zhang F, Deng S, Wang R, Zhang Y, Huang K. Poly(ADP-ribose) polymerase 1 is indispensable for transforming growth factor-β Induced Smad3 activation in vascular smooth muscle cell. PLoS One 2011; 6:e27123. [PMID: 22073128 PMCID: PMC3205050 DOI: 10.1371/journal.pone.0027123] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 10/11/2011] [Indexed: 12/31/2022] Open
Abstract
Background Transforming growth factor type-β (TGF-β)/Smad pathway plays an essential role in vascular fibrosis. Reactive oxygen species (ROS) generation also mediates TGF-β signaling-induced vascular fibrosis, suggesting that some sort of interaction exists between Smad and redox pathways. However, the underlying molecular mechanism is largely unknown. This study aims to investigate the influence of poly(ADP-ribose) polymerase 1 (PARP1), a downstream effector of ROS, on TGF-β signaling transduction through Smad3 pathway in rat vascular smooth muscle cells (VSMCs). Methods and Results TGF-β1 treatment promoted PARP1 activation through induction of ROS generation in rat VSMCs. TGF-β1-induced phosphorylation and nuclear accumulation of Smad3 was prevented by treatment of cells with PARP inhibitor, 3-aminobenzamide (3AB) or N-(6-oxo-5,6-dihydrophenanthridin-2-yl)-2-(N,N-dimethylamino)acetami (PJ34), or PARP1 siRNA. TGF-β1 treatment promoted poly(ADP-ribosy)lation of Smad3 via activation of PARP1 in the nucleus. Poly(ADP-ribosy)lation enhanced Smad-Smad binding element (SBE) complex formation in nuclear extracts and increased DNA binding activity of Smad3. Pretreatment with 3AB, PJ34, or PARP1 siRNA prevented TGF-β1-induced Smad3 transactivation and expression of Smad3 target genes, including collagen Iα1, collagen IIIα1 and tissue inhibitor of metalloproteinase 1, in rat VSMCs. Conclusions PARP1 is indispensable for TGF-β1 induced Smad3 activation in rat VSMCs. Targeting PARP1 may be a promising therapeutic approach against vascular diseases induced by dysregulation of TGF-β/Smad3 pathway.
Collapse
MESH Headings
- Animals
- Benzamides/pharmacology
- Blotting, Southwestern
- Blotting, Western
- Cell Nucleus/drug effects
- Cell Nucleus/metabolism
- Cells, Cultured
- Electrophoretic Mobility Shift Assay
- Enzyme Inhibitors/pharmacology
- Enzyme-Linked Immunosorbent Assay
- Fluorescent Antibody Technique
- Immunoenzyme Techniques
- Immunoprecipitation
- Luciferases/metabolism
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Phenanthrenes/pharmacology
- Phosphorylation/drug effects
- Poly(ADP-ribose) Polymerase Inhibitors
- Poly(ADP-ribose) Polymerases/genetics
- Poly(ADP-ribose) Polymerases/metabolism
- Promoter Regions, Genetic
- Protein Binding
- RNA, Messenger/genetics
- RNA, Small Interfering/pharmacology
- Rats
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Real-Time Polymerase Chain Reaction
- Signal Transduction/drug effects
- Smad3 Protein/genetics
- Smad3 Protein/metabolism
- Trans-Activators
- Transcription, Genetic
- Transfection
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
Collapse
Affiliation(s)
- Dan Huang
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Wang
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Wang
- Central Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengxiao Zhang
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan Deng
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Wang
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China
- * E-mail: (KH); (YZ)
| | - Kai Huang
- Department of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan, China
- * E-mail: (KH); (YZ)
| |
Collapse
|
81
|
Icardi L, Lievens S, Mori R, Piessevaux J, De Cauwer L, De Bosscher K, Tavernier J. Opposed regulation of type I IFN-induced STAT3 and ISGF3 transcriptional activities by histone deacetylases (HDACS) 1 and 2. FASEB J 2011; 26:240-9. [PMID: 21957129 DOI: 10.1096/fj.11-191122] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The antiviral and antiproliferative responses mediated by type I interferons (IFNs) depend on JAK/STAT signaling and ISGF3 (STAT1:STAT2:IRF9)-dependent transcription. In addition, type I IFNs stimulate STAT3 activation in many cell types, an event generally associated with cell cycle progression, survival, and proliferation. To gather more insight into this functionally contradictive phenomenon, we studied the regulation of STAT3 transcriptional activity upon type I IFN treatment. We show that IFNα2 stimulation strongly induces STAT3 phosphorylation, nuclear translocation, and promoter binding, yet the activation of transcription of a STAT3-dependent reporter and endogenous genes, such as SOCS3 and c-FOS, is impaired. Simultaneous treatment with IFNα2 and trichostatin A, as well as combined HDAC1/HDAC2 silencing, restores STAT3-dependent reporter gene and endogenous gene expression, strongly suggesting that HDAC1 and HDAC2 are directly involved in repressing IFNα2-activated STAT3. Of note, single silencing of only one of the two HDACs does not lead to enhanced STAT3 activity, supporting a functional redundancy between these two enzymes. In sharp contrast, HDAC1 and HDAC2 activities are required for ISGF3-dependent gene expression. We conclude that HDAC1 and HDAC2 differentially modulate STAT activity in response to IFNα2: while they are required for the induction of ISGF3-responsive genes, they impair the transcription of STAT3-dependent genes.
Collapse
Affiliation(s)
- Laura Icardi
- Department of Medical Protein Research, Vlaams Instituut voor Biotechnologie, Ghent, Belgium
| | | | | | | | | | | | | |
Collapse
|
82
|
Role of Smads in TGFβ signaling. Cell Tissue Res 2011; 347:21-36. [PMID: 21643690 DOI: 10.1007/s00441-011-1190-x] [Citation(s) in RCA: 275] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Accepted: 05/10/2011] [Indexed: 02/07/2023]
Abstract
Transforming growth factor-β (TGFβ) is the prototype for a large family of pleiotropic factors that signal via heterotetrameric complexes of type I and type II serine/threonine kinase receptors. Important intracellular mediators of TGFβ signaling are members of the Smad family. Smad2 and 3 are activated by C-terminal receptor-mediated phosphorylation, whereafter they form complexes with Smad4 and are translocated to the nucleus where they, in cooperation with other transcription factors, co-activators and co-repressors, regulate the transcription of specific genes. Smads have key roles in exerting TGFβ-induced programs leading to cell growth arrest and epithelial-mesenchymal transition. The activity and stability of Smad molecules are carefully regulated by a plethora of post-translational modifications, including phosphorylation, ubiquitination, sumoylation, acetylation and poly(ADP)-ribosylation. The Smad function has been shown to be perturbed in certain diseases such as cancer.
Collapse
|
83
|
Pang L, Qiu T, Cao X, Wan M. Apoptotic role of TGF-β mediated by Smad4 mitochondria translocation and cytochrome c oxidase subunit II interaction. Exp Cell Res 2011; 317:1608-20. [PMID: 21324314 DOI: 10.1016/j.yexcr.2011.02.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 02/08/2011] [Accepted: 02/09/2011] [Indexed: 11/29/2022]
Abstract
Smad4, originally isolated from the human chromosome 18q21, is a key factor in transducing the signals of the TGF-β superfamily of growth hormones and plays a pivotal role in mediating antimitogenic and proapoptotic effects of TGF-β, but the mechanisms by which Smad4 induces apoptosis are elusive. Here we report that Smad4 directly translocates to the mitochondria of apoptotic cells. Smad4 gene silencing by siRNA inhibits TGF-β-induced apoptosis in Hep3B cells and UV-induced apoptosis in PANC-1 cells. Cell fractionation assays demonstrated that a fraction of Smad4 translocates to mitochondria after long time TGF-β treatment or UV exposure, during which the cells were under apoptosis. Smad4 mitochondria translocation during apoptosis was also confirmed by fluorescence observation of Smad4 colocalization with MitoTracker Red. We searched for mitochondria proteins that have physical interactions with Smad4 using yeast two-hybrid screening approach. DNA sequence analysis identified 34 positive clones, five of which encoded subunits in mitochondria complex IV, i.e., one clone encoded cytochrome c oxidase COXII, three clones encoded COXIII and one clone encoded COXVb. Strong interaction between Smad4 with COXII, an important apoptosis regulator, was verified in yeast by β-gal activity assays and in mammalian cells by immunoprecipitation assays. Further, mitochondrial portion of cells was isolated and the interaction between COXII and Smad4 in mitochondria upon TGF-β treatment or UV exposure was confirmed. Importantly, targeting Smad4 to mitochondria using import leader fusions enhanced TGF-β-induced apoptosis. Collectively, the results suggest that Smad4 promote apoptosis of the cells through its mitochondrial translocation and association with mitochondria protein COXII.
Collapse
Affiliation(s)
- Lijuan Pang
- The Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | |
Collapse
|
84
|
Abstract
The growth inhibition of dividing cells and most of the transcriptional responses upon TGF-beta treatment depend on the Smad2, Smad3, and Smad4 transcription factors. These proteins shuttle continuously between the cytoplasm and the nucleus, transmitting the ligand status of the TGF-beta receptors to the nuclear transcription machinery. In the absence of TGF-beta ligand, Smads 2/3/4 reside predominantly in the cytoplasm. Following ligand binding to the TGF-beta receptors, the dynamic equilibrium of shuttling Smads 2/3/4 shifts toward a predominantly nuclear state, where a high concentration of these transcription factors drives transcriptional activation and repression of genes required for proper cellular response. Here, we describe live cell imaging and immunofluorescence microscopy methods for tracking Smads subcellular localization in response to TGF-beta and leptomycin B treatment. In addition, a method of fractionating nuclear and cytoplasmic proteins used to confirm the imaging results was presented. Our results support the notion that the R-Smad shuttling mechanism is distinct from Co-Smad.
Collapse
|
85
|
Diversity of axin in signaling pathways and its relation to colorectal cancer. Med Oncol 2010; 28 Suppl 1:S259-67. [DOI: 10.1007/s12032-010-9722-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Accepted: 10/12/2010] [Indexed: 12/12/2022]
|
86
|
Nakahiro T, Kurooka H, Mori K, Sano K, Yokota Y. Identification of BMP-responsive elements in the mouse Id2 gene. Biochem Biophys Res Commun 2010; 399:416-21. [PMID: 20674548 DOI: 10.1016/j.bbrc.2010.07.090] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 07/25/2010] [Indexed: 12/28/2022]
Abstract
Inhibitor of DNA binding/differentiation (Id) genes are the targets of bone morphogenetic protein (BMP) signals in various types of cells. We investigated the molecular basis of BMP6-induced gene expression of mouse Id2 in C2C12 myoblasts. BMP6-dependent Id2 expression occurred immediately without de novo protein synthesis and was blocked by an inhibitor of the BMP type I receptors. A reporter assay identified a BMP6-responsive region 3.0kb upstream of the transcription initiation site. The region showed sequence similarity to the mouse Id1 promoter and shared potential Smad binding sites with it, two GGCGCC palindromes and one GTCT element. Mutation analysis demonstrated the involvement of these elements in the BMP response. Gel shift and chromatin immunoprecipitation (ChIP) assays confirmed the physical binding of Smad proteins to these elements. The 3'-positioned GGCGCC palindrome and the GTCT element were separated by 5-bp and conformed to the canonical BMP-responsive sequence. In addition, the 5'-positioned GGCGCC was accompanied by a previously uncharacterized CGCC element, which were separated by a 5-bp space, and this configuration coincided with that of a similar but distinct sequence to which a Drosophila homolog of the Smad complex can bind. Reporter and gel shift assays revealed the importance of this bipartite sequence. Therefore, we have identified the BMP-responsive elements in mouse Id2 and also shown that the CGCC sequence contributes to target recognition by Smad proteins.
Collapse
Affiliation(s)
- Takeshi Nakahiro
- Division of Molecular Genetics, Department of Biochemistry and Bioinformative Sciences, School of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | | | | | | | | |
Collapse
|
87
|
Abstract
Cytoplasm-to-nucleus translocation of Smad is a fundamental step in transforming growth factor beta (TGF-beta) signal transduction. Here we identify a subset of nucleoporins that, in conjunction with Msk (Drosophila Imp7/8), specifically mediate activation-induced nuclear translocation of MAD (Drosophila Smad1) but not the constitutive import of proteins harboring a classic nuclear localization signal (cNLS) or the spontaneous nuclear import of Medea (Drosophila Smad4). Surprisingly, many of these nucleoporins, including Sec13, Nup75, Nup93, and Nup205, are scaffold nucleoporins considered important for the overall integrity of the nuclear pore complex (NPC) but not known to have cargo-specific functions. We demonstrate that the roles of these nucleoporins in supporting Smad nuclear import are separate from their previously assigned functions in NPC assembly. Furthermore, we uncovered novel pathway-specific functions of Sec13 and Nup93; both Sec13 and Nup93 are able to preferentially interact with the phosphorylated/activated form of MAD, and Nup93 acts to recruit the importin Msk to the nuclear periphery. These findings, together with the observation that Sec13 and Nup93 could interact directly with Msk, suggest their direct involvement in the nuclear import of MAD. Thus, we have delineated the nucleoporin requirement of MAD nuclear import, reflecting a unique trans-NPC mechanism.
Collapse
|
88
|
Tao S, Sampath K. Alternative splicing of SMADs in differentiation and tissue homeostasis. Dev Growth Differ 2010; 52:335-42. [PMID: 20148926 DOI: 10.1111/j.1440-169x.2009.01163.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Tissue-specific alternate splicing is an important means of regulating gene expression during development. The effector proteins for the transforming growth factor-beta signaling pathway, the SMADs, encode distinct isoforms generated via alternate splicing, which appear to have distinct tissue-specific expression profiles and functions. Here, we discuss the roles of various SMAD isoforms, and the consequences of mis-regulation of SMAD splicing in development and tissue homeostasis.
Collapse
Affiliation(s)
- Shijie Tao
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore
| | | |
Collapse
|
89
|
Yu J, Pan L, Qin X, Chen H, Xu Y, Chen Y, Tang H. MTMR4 attenuates transforming growth factor beta (TGFbeta) signaling by dephosphorylating R-Smads in endosomes. J Biol Chem 2010; 285:8454-62. [PMID: 20061380 DOI: 10.1074/jbc.m109.075036] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Homeostasis of Smad phosphorylation at its C-terminal SXS motif is essential for transforming growth factor beta (TGFbeta) signaling. Whereas it is known that TGFbeta signaling can be terminated by phosphatases, which dephosphorylate R-Smads in the nucleus, it is unclear whether there are any cytoplasmic phosphatase(s) that can attenuate R-Smad phosphorylation and nuclear translocation. Here we demonstrate that myotubularin-related protein 4 (MTMR4), a FYVE domain-containing dual-specificity protein phosphatase (DSP), attenuates TGFbeta signaling by reducing the phosphorylation level of R-Smads in early endosomes. Co-immunoprecipitation experiments showed that endogenous MTMR4 interacts with phosphorylated R-Smads, and that this interaction is correlated with dephosphorylation of R-Smads. Further analysis showed that overexpression of MTMR4 resulted in the sequestration of activated Smad3 in the early endosomes, thus reducing its nuclear translocation. However, both point mutations at the conserved catalytic site of the phosphatase (MTMR4-C407S) and small interference RNA of endogenous Mtmr4 expression led to sustained Smad3 activation. This work therefore suggests that MTMR4 plays an important role in preventing the overactivation of TGFbeta signaling by dephosphorylating activated R-Smads that have been trafficked to early endosomes.
Collapse
Affiliation(s)
- Junjing Yu
- Key Laboratory of Infection and Immunity of the Chinese Academy of Sciences, Institute of Biophysics, Beijing 100101, China
| | | | | | | | | | | | | |
Collapse
|
90
|
Dai F, Duan X, Liang YY, Lin X, Feng XH. Coupling of dephosphorylation and nuclear export of Smads in TGF-beta signaling. Methods Mol Biol 2010; 647:125-37. [PMID: 20694664 PMCID: PMC3153448 DOI: 10.1007/978-1-60761-738-9_7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In eukaryotes, regulation of signaling mediators/effectors in the nucleus is one of the principal mechanisms that govern duration and strength of signaling. Smads are a family of structurally related intracellular proteins that serve as signaling effectors for transforming growth factor beta (TGF-beta) and TGF-beta-related proteins. Accumulating evidence demonstrates that Smads possess intrinsic nucleocytoplasmic shuttling capacity, which enables them to transmit TGF-beta signals from cell membrane to nucleus. We recently identified two important steps in the termination of nuclear Smad signaling. The first step is initiated by a serine/threonine phosphatase PPM1A that dephosphorylates Smad2/3 in the nucleus, thereby shutting down signaling capacity of phosphorylated Smad2/3. The second step involves nuclear export of dephosphorylated Smad2/3 with the aid of nuclear protein RanBP3 to terminate Smad signaling. This chapter introduces methods for examining nuclear export of Smad2/3 in TGF-beta signaling.
Collapse
Affiliation(s)
- Fangyan Dai
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX77030, USA
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX77030, USA
- The Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX77030, USA
| | - Xueyan Duan
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX77030, USA
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX77030, USA
- The Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX77030, USA
| | - Yao-Yun Liang
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX77030, USA
| | - Xia Lin
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX77030, USA
- The Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX77030, USA
| | - Xin-Hua Feng
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX77030, USA
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX77030, USA
- The Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX77030, USA
| |
Collapse
|
91
|
Daly AC, Vizán P, Hill CS. Smad3 protein levels are modulated by Ras activity and during the cell cycle to dictate transforming growth factor-beta responses. J Biol Chem 2009; 285:6489-97. [PMID: 20037158 DOI: 10.1074/jbc.m109.043877] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Transforming growth factor beta (TGF-beta) regulates many biological processes, and aberrant TGF-beta signaling is implicated in tumor development. Smad3 is a central component of the TGF-beta signaling pathway, and once activated, Smad3 forms complexes with Smad4 or other receptor-regulated Smads, which accumulate in the nucleus to transcriptionally regulate TGF-beta target genes. Because Smad3 plays a significant role in mediating the activities of TGF-beta, we examined its regulation during tumor development using a well characterized tumor model. We demonstrate that Smad3 levels are dramatically reduced in the tumorigenic cell line transformed with activated H-Ras compared with the normal parental epithelial cells. Interestingly, we also observe a cell cycle-dependent regulation of Smad3 in both cell types, with high Smad3 levels in quiescent cells and a significant drop in Smad3 protein levels in proliferating cells. Smad3 is regulated at the mRNA level and at the level of protein stability. In addition, functional analysis indicates that down-regulation of Smad3 levels is required for the tumor cells to proliferate in the presence of TGF-beta, because ectopic expression of Smad3 in the tumorigenic cell line restores the growth inhibitory response to TGF-beta. In contrast, expression of high levels of Smad3 did not interfere with the ability of these cells to undergo epithelial to mesenchymal transition upon TGF-beta stimulation. Altogether, our results suggest that the level of Smad3 protein is an important determinant of the progression of tumorigenesis. High levels of Smad3 are required for the tumor suppressor activities of TGF-beta, whereas lower levels are sufficient for the tumor promoting functions.
Collapse
Affiliation(s)
- Amanda C Daly
- Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, London WC2A 3PX, United Kingdom
| | | | | |
Collapse
|
92
|
Epicatechin induces NF-κB, activator protein-1 (AP-1) and nuclear transcription factor erythroid 2p45-related factor-2 (Nrf2) via phosphatidylinositol-3-kinase/protein kinase B (PI3K/AKT) and extracellular regulated kinase (ERK) signalling in HepG2 cells. Br J Nutr 2009; 103:168-79. [DOI: 10.1017/s0007114509991747] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The dietary flavonoid epicatechin has been reported to exhibit a wide range of biological activities. The objective of the present study was to investigate the time-dependent regulation by epicatechin on the activity of the main transcription factors (NF-κB, activator protein-1 (AP-1) and nuclear transcription factor erythroid 2p45-related factor (Nrf2)) related to antioxidant defence and survival and proliferation pathways in HepG2 cells. Treatment of cells with 10 μm-epicatechin induced the NF-κB pathway in a time-dependent manner characterised by increased levels of IκB kinase (IKK) and phosphorylated inhibitor of κB subunit-α (p-IκBα) and proteolytic degradation of IκB, which was consistent with an up-regulation of the NF-κB-binding activity. Time-dependent activation of the AP-1 pathway, in concert with enhanced c-Jun nuclear levels and induction of Nrf2 translocation and phosphorylation were also demonstrated. Additionally, epicatechin-induced NF-κB and Nrf2 were connected to reactive oxygen species intracellular levels and to the activation of cell survival and proliferation pathways, being phosphatidylinositol-3-kinase/protein kinase B (PI3K/AKT) and extracellular regulated kinase (ERK) associated to Nrf2 modulation and ERK to NF-κB induction. These data suggest that the epicatechin-induced survival effect occurs by the induction of redox-sensitive transcription factors through a tight regulation of survival and proliferation pathways.
Collapse
|
93
|
Kardassis D, Murphy C, Fotsis T, Moustakas A, Stournaras C. Control of transforming growth factor β signal transduction by small GTPases. FEBS J 2009; 276:2947-65. [DOI: 10.1111/j.1742-4658.2009.07031.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
94
|
Windish HP, Lin PL, Mattila JT, Green AM, Onuoha EO, Kane LP, Flynn JL. Aberrant TGF-beta signaling reduces T regulatory cells in ICAM-1-deficient mice, increasing the inflammatory response to Mycobacterium tuberculosis. J Leukoc Biol 2009; 86:713-25. [PMID: 19454651 DOI: 10.1189/jlb.1208740] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Foxp3+ T regulatory cells are required to prevent autoimmune disease, but also prevent clearance of some chronic infections. While natural T regulatory cells are produced in the thymus, TGF-beta1 signaling combined with T-cell receptor signaling induces the expression of Foxp3 in CD4+ T cells in the periphery. We found that ICAM-1-/- mice have fewer T regulatory cells in the periphery than WT controls, due to a role for ICAM-1 in induction of Foxp3 expression in response to TGF-beta1. Further investigation revealed a functional deficiency in the TGF-beta1-induced translocation of phosphorylated Smad3 from the cytoplasmic compartment to the nucleus in ICAM-1-deficient mice. This impairment in the TGF-beta1 signaling pathway is most likely responsible for the decrease in T regulatory cell induction in the absence of ICAM-1. We hypothesized that in the presence of an inflammatory response, reduced production of inducible T regulatory cells would be evident in ICAM-1-/- mice. Indeed, following Mycobacterium tuberculosis infection, ICAM-1-/- mice had a pronounced reduction in T regulatory cells in the lungs compared with control mice. Consequently, the effector T-cell response and inflammation were greater in the lungs of ICAM-1-/- mice, resulting in morbidity due to overwhelming pathology.
Collapse
Affiliation(s)
- Hillarie Plessner Windish
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | |
Collapse
|
95
|
Quantitative modeling and analysis of the transforming growth factor beta signaling pathway. Biophys J 2009; 96:1733-50. [PMID: 19254534 DOI: 10.1016/j.bpj.2008.11.050] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2008] [Accepted: 11/12/2008] [Indexed: 11/20/2022] Open
Abstract
Transforming growth factor beta (TGF-beta) signaling, which regulates multiple cellular processes including proliferation, apoptosis, and differentiation, plays an important but incompletely understood role in normal and cancerous tissues. For instance, although TGF-beta functions as a tumor suppressor in the premalignant stages of tumorigenesis, paradoxically, it also seems to act as a tumor promoter in advanced cancer leading to metastasis. The mechanisms by which TGF-beta elicits such diverse responses during cancer progression are still not entirely clear. As a first step toward understanding TGF-beta signaling quantitatively, we have developed a comprehensive, dynamic model of the canonical TGF-beta pathway via Smad transcription factors. By describing how an extracellular signal of the TGF-beta ligand is sensed by receptors and transmitted into the nucleus through intracellular Smad proteins, the model provides quantitative insight into how TGF-beta-induced responses are modulated and regulated. Subsequent model analysis shows that mechanisms associated with Smad activation by ligand-activated receptor, nuclear complex formation among Smad proteins, and inactivation of ligand-activated Smad (e.g., degradation, dephosphorylation) may be critical for regulating TGF-beta-targeted functional responses. The model was also used to predict dynamic characteristics of the Smad-mediated pathway in abnormal cells, from which we generated four testable hypotheses regarding potential mechanisms by which TGF-beta's tumor-suppressive roles may appear to morph into tumor-promotion during cancer progression.
Collapse
|
96
|
Gramley F, Lorenzen J, Koellensperger E, Kettering K, Weiss C, Munzel T. Atrial fibrosis and atrial fibrillation: the role of the TGF-β1 signaling pathway. Int J Cardiol 2009; 143:405-13. [PMID: 19394095 DOI: 10.1016/j.ijcard.2009.03.110] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Revised: 02/23/2009] [Accepted: 03/24/2009] [Indexed: 10/20/2022]
Abstract
BACKGROUND Atrial fibrosis concurs with chronic atrial fibrillation (AF), a phenomenon that contributes to the resistance to restore and maintain sinus rhythm (SR). Fibrogenesis represents a complex process in which the transforming growth factor-β1 (TGF-β1) pathway may play a major role, e.g. in the setting of myocardial infarction. The present study addresses the potential contribution of the TGF-β1 signaling pathway to atrial fibrosis in patients with AF. METHODS AND RESULTS Right atrial appendages of 163 patients were excised during heart surgery and grouped according to rhythm (SR vs. AF) and AF duration. Five groups were defined: SR, paroxysmal/chronic persistent AF (<6 months), chronic permanent AF (CAF) of 7-24 months, 25-60 months, and >60 months duration. Collagen content of atria, determined morphometrically, revealed a steady and significant increase in patients with SR (14.6±8.9%) up to patients with CAF of >60 months (28.1±7.1%). Likewise, expression of TGF-β1 mRNA and protein, TGF-β-receptor-II protein, profibrotic phospho-Smad-2 and -4 proteins increased. However, the TGF-β(1) effect appeared to decline with increasing AF duration, characterized by a decrease in TGF-β-receptor-I protein, increases of TGF-β inhibiting Smad-7 protein and a reduction of ph-Smad-2. CONCLUSIONS Human atrial fibrogenesis in patients with atrial fibrillation is accompanied by a biphasic response, an early increase and later loss of responsiveness to TGF-β(1). It appears that fibrosis progresses despite compensatory changes in the TGF-β-signaling pathway. The sequential changes in the contribution of different profibrotic processes during the establishment of AF may offer the opportunity to selectively interfere with the atrial remodeling process at different stages.
Collapse
Affiliation(s)
- Felix Gramley
- Department of Cardiology and Vascular Medicine, Mainz University, Germany.
| | | | | | | | | | | |
Collapse
|
97
|
Lin X, Chen Y, Meng A, Feng X. Termination of TGF-beta superfamily signaling through SMAD dephosphorylation--a functional genomic view. J Genet Genomics 2009; 34:1-9. [PMID: 17469772 DOI: 10.1016/s1673-8527(07)60001-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2006] [Accepted: 10/31/2006] [Indexed: 12/11/2022]
Abstract
The transforming growth factor-beta (TGF-beta) and related growth factors activate a broad range of cellular responses in metazoan organisms via autocrine, paracrine, and endocrine modes. They play key roles in the pathogenesis of many diseases especially cancer, fibrotic diseases, autoimmune diseases and cardiovascular diseases. TGF-beta receptor-mediated phosphorylation of R-SMADs represents the most critical step in the TGF-beta signaling pathways that triggers a cascade of intracellular events from SMAD complex assembly in the cytoplasm to transcriptional control in the nucleus. Conversely, dephosphorylation of R-SMADs is a key mechanism for terminating TGF-beta signaling. Our labs have recently taken an integrated approach combining functional genomics, biochemistry and development biology to describe the isolation and functional characterization of protein phosphatase PPM1A in controlling TGF-beta signaling. This article briefly reviews how dynamic phosphorylation and dephosphorylation of SMADs control or fine-tune the signaling strength and duration and ultimately the physiological consequences in TGF-beta signaling.
Collapse
Affiliation(s)
- Xia Lin
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston TX 77030, USA
| | | | | | | |
Collapse
|
98
|
Dai F, Lin X, Chang C, Feng XH. Nuclear export of Smad2 and Smad3 by RanBP3 facilitates termination of TGF-beta signaling. Dev Cell 2009; 16:345-57. [PMID: 19289081 PMCID: PMC2676691 DOI: 10.1016/j.devcel.2009.01.022] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2008] [Revised: 12/31/2008] [Accepted: 01/30/2009] [Indexed: 10/21/2022]
Abstract
Smad2 and Smad3 (Smad2/3) are key intracellular signal transducers for TGF-beta signaling, and their transcriptional activities are controlled through reversible phosphorylation and nucleocytoplasmic shuttling. However, the precise mechanism underlying nuclear export of Smad2/3 remains elusive. Here we report the essential function of RanBP3 in selective nuclear export of Smad2/3 in the TGF-beta pathway. RanBP3 directly recognizes dephosphorylated Smad2/3, which results from the activity of nuclear Smad phosphatases, and mediates nuclear export of Smad2/3 in a Ran-dependent manner. As a result, increased expression of RanBP3 inhibits TGF-beta signaling in mammalian cells and Xenopus embryos. Conversely, depletion of RanBP3 expression or dominant-negative inhibition of RanBP3 enhances TGFbeta-induced antiproliferative and transcriptional responses. In conclusion, our study supports a definitive role for RanBP3 in mediating Smad2/3 nuclear export and terminating TGF-beta signaling.
Collapse
Affiliation(s)
- Fangyan Dai
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX77030, USA
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX77030, USA
- The Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX77030, USA
| | - Xia Lin
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX77030, USA
- The Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX77030, USA
| | - Chenbei Chang
- Department of Cell Biology, University of Alabama, Birmingham, AL35294, USA
| | - Xin-Hua Feng
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX77030, USA
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX77030, USA
- The Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX77030, USA
| |
Collapse
|
99
|
Erickson RA, Liu X. Association of v-ErbA with Smad4 disrupts TGF-beta signaling. Mol Biol Cell 2009; 20:1509-19. [PMID: 19144825 PMCID: PMC2649266 DOI: 10.1091/mbc.e08-08-0836] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Revised: 12/15/2008] [Accepted: 01/05/2009] [Indexed: 11/11/2022] Open
Abstract
Disruption of the transforming growth factor-beta (TGF-beta) pathway is observed in the majority of cancers. To further understand TGF-beta pathway inactivation in cancer, we stably expressed the v-ErbA oncoprotein in TGF-beta responsive cells. v-ErbA participates in erythroleukemic transformation of cells induced by the avian erythroblastosis virus (AEV). Here we demonstrate that expression of v-ErbA was sufficient to antagonize TGF-beta-induced cell growth inhibition and that dysregulation of TGF-beta signaling required that v-ErbA associate with the Smad4 which sequesters Smad4 in the cytoplasm. We also show that AEV-transformed erythroleukemia cells were resistant to TGF-beta-induced growth inhibition and that TGF-beta sensitivity could be recovered by reducing v-ErbA expression. Our results reveal a novel mechanism for oncogenic disruption of TGF-beta signaling and provide a mechanistic explanation of v-ErbA activity in AEV-induced erythroleukemia.
Collapse
Affiliation(s)
- Richard A. Erickson
- Department of Chemistry and Biochemistry, University of Colorado-Boulder, Boulder, CO 80309
| | - Xuedong Liu
- Department of Chemistry and Biochemistry, University of Colorado-Boulder, Boulder, CO 80309
| |
Collapse
|
100
|
Transforming growth factor beta depletion is the primary determinant of Smad signaling kinetics. Mol Cell Biol 2009; 29:2443-55. [PMID: 19223462 DOI: 10.1128/mcb.01443-08] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
A cell's decision to growth arrest, apoptose, or differentiate in response to transforming growth factor beta (TGF-beta) superfamily ligands depends on the ligand concentration. How cells sense the concentration of extracellular bioavailable TGF-beta remains poorly understood. We therefore undertook a systematic quantitative analysis of how TGF-beta ligand concentration is transduced into downstream phospho-Smad2 kinetics, and we found that the rate of TGF-beta ligand depletion is the principal determinant of Smad signal duration. TGF-beta depletion is caused by two mechanisms: (i) cellular uptake of TGF-beta by a TGF-beta type II receptor-dependent mechanism and (ii) reversible binding of TGF-beta to the cell surface. Our results indicate that cells sense TGF-beta dose by depleting TGF-beta via constitutive TGF-beta type II receptor trafficking processes. Our results also have implications for the role of the TGF-beta type II receptor in disease, as tumor cells harboring TGF-beta type II receptor mutations exhibit impaired TGF-beta depletion, which may contribute to the overproduction of TGF-beta and a consequently poor prognosis in cancer.
Collapse
|