301
|
Boucher J, Gridley T, Liaw L. Molecular pathways of notch signaling in vascular smooth muscle cells. Front Physiol 2012; 3:81. [PMID: 22509166 PMCID: PMC3321637 DOI: 10.3389/fphys.2012.00081] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2012] [Accepted: 03/19/2012] [Indexed: 11/20/2022] Open
Abstract
Notch signaling in the cardiovascular system is important during embryonic development, vascular repair of injury, and vascular pathology in humans. The vascular smooth muscle cell (VSMC) expresses multiple Notch receptors throughout its life cycle, and responds to Notch ligands as a regulatory mechanism of differentiation, recruitment to growing vessels, and maturation. The goal of this review is to provide an overview of the current understanding of the molecular basis for Notch regulation of VSMC phenotype. Further, we will explore Notch interaction with other signaling pathways important in VSMC.
Collapse
Affiliation(s)
- Joshua Boucher
- Center for Molecular Medicine, Maine Medical Center Research Institute Scarborough, ME, USA
| | | | | |
Collapse
|
302
|
Ma YC, Shi C, Zhang YN, Wang LG, Liu H, Jia HT, Zhang YX, Sarkar FH, Wang ZS. The tyrosine kinase c-Src directly mediates growth factor-induced Notch-1 and Furin interaction and Notch-1 activation in pancreatic cancer cells. PLoS One 2012; 7:e33414. [PMID: 22479394 PMCID: PMC3316571 DOI: 10.1371/journal.pone.0033414] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 02/08/2012] [Indexed: 01/03/2023] Open
Abstract
The proteolytic activity of Furin responsible for processing full length Notch-1 (p300) plays a critical role in Notch signaling. The amplitude and duration of Notch activity can be regulated at various points in the pathway, but there has been no report regarding regulation of the Notch-1-Furin interaction, despite its importance. In the present study, we found that the Notch-1-Furin interaction is regulated by the non-receptor tyrosine kinase, c-Src. c-Src and Notch-1 are physically associated, and this association is responsible for Notch-1 processing and activation. We also found that growth factor TGF-α, an EGFR ligand, and PDGF-BB, a PDGFR ligand, induce the Notch-1-Furin interaction mediated by c-Src. Our results support three new and provocative conclusions: (1) The association between Notch-1 and Furin is a well-regulated process; (2) Extracellular growth factor signals regulate this interaction, which is mediated by c-Src; (3) There is cross-talk between the plasma growth factor receptor-c-Src and Notch pathways. Co-localization of Notch-1 and c-Src was confirmed in xenograft tumor tissues and in the tissues of pancreatic cancer patients. Our findings have implications for the mechanism by which the Notch and growth factor receptor-c-Src signaling pathways regulate carcinogenesis and cancer cell growth.
Collapse
MESH Headings
- Animals
- Becaplermin
- Blotting, Western
- Cell Line, Tumor
- Female
- Furin/genetics
- Furin/metabolism
- HeLa Cells
- Humans
- Intercellular Signaling Peptides and Proteins/pharmacology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Microscopy, Confocal
- Mutation
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Protein Binding/drug effects
- Proto-Oncogene Proteins c-sis/pharmacology
- Pyrimidines/pharmacology
- Receptor, Notch1/genetics
- Receptor, Notch1/metabolism
- Transforming Growth Factor alpha/pharmacology
- Transplantation, Heterologous
- Tumor Burden/drug effects
- src-Family Kinases/antagonists & inhibitors
- src-Family Kinases/genetics
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- Yong-Chao Ma
- Department of Biochemistry and Molecular Biology, Cancer Institute, Capital Medical University, Beijing, China
| | - Chong Shi
- Department of Biochemistry and Molecular Biology, Cancer Institute, Capital Medical University, Beijing, China
| | - Yao-Nan Zhang
- Department of Biochemistry and Molecular Biology, Cancer Institute, Capital Medical University, Beijing, China
| | - Lan-Ge Wang
- Department of Biochemistry and Molecular Biology, Cancer Institute, Capital Medical University, Beijing, China
| | - Hao Liu
- Department of Biochemistry and Molecular Biology, Cancer Institute, Capital Medical University, Beijing, China
| | - Hong-Ti Jia
- Department of Biochemistry and Molecular Biology, Cancer Institute, Capital Medical University, Beijing, China
| | - Yu-Xiang Zhang
- Department of Biochemistry and Molecular Biology, Cancer Institute, Capital Medical University, Beijing, China
- * E-mail: (YXZ); (ZSW)
| | - Fazlul H. Sarkar
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, United States of America
| | - Ze-Sheng Wang
- Department of Biochemistry and Molecular Biology, Cancer Institute, Capital Medical University, Beijing, China
- * E-mail: (YXZ); (ZSW)
| |
Collapse
|
303
|
Abstract
Temporal changes in transcription programs are coupled to control of cell growth and division. We here report that Mediator, a conserved coregulator of eukaryotic transcription, is part of a regulatory pathway that controls mitotic entry in fission yeast. The Mediator subunit cyclin-dependent kinase 8 (Cdk8) phosphorylates the forkhead 2 (Fkh2) protein in a periodic manner that coincides with gene activation during mitosis. Phosphorylation prevents degradation of the Fkh2 transcription factor by the proteasome, thus ensuring cell cycle-dependent variations in Fkh2 levels. Interestingly, Cdk8-dependent phosphorylation of Fkh2 controls mitotic entry, and mitotic entry is delayed by inactivation of the Cdk8 kinase activity or mutations replacing the phosphorylated serine residues of Fkh2. In addition, mutations in Fkh2, which mimic protein phosphorylation, lead to premature mitotic entry. Therefore, Fkh2 regulates not only the onset of mitotic transcription but also the correct timing of mitotic entry via effects on the Wee1 kinase. Our findings thus establish a new pathway linking the Mediator complex to control of mitotic transcription and regulation of mitotic entry in fission yeast.
Collapse
|
304
|
Galbraith MD, Donner AJ, Espinosa JM. CDK8: a positive regulator of transcription. Transcription 2012; 1:4-12. [PMID: 21327159 DOI: 10.4161/trns.1.1.12373] [Citation(s) in RCA: 165] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 05/12/2010] [Accepted: 05/13/2010] [Indexed: 01/09/2023] Open
Abstract
CDK8 belongs to a group of cyclin-dependent kinases involved in transcriptional regulation from yeast to mammals. CDK8 associates with the Mediator complex, but functions outside of Mediator are also likely. Historically, CDK8 has been described mostly as a transcriptional repressor, but a growing body of research provides unequivocal evidence for various roles of CDK8 in gene activation. Several transcriptional programs of biomedical importance employ CDK8 as a co-activator, including the p53 network, the Wnt/β-catenin pathway, the serum response network, and those governed by SMADs and the thyroid hormone receptor, thus highlighting the importance of further investigation into this enigmatic transcriptional regulator.
Collapse
|
305
|
Pathways involved in Drosophila and human cancer development: the Notch, Hedgehog, Wingless, Runt, and Trithorax pathway. Ann Hematol 2012; 91:645-669. [PMID: 22418742 DOI: 10.1007/s00277-012-1435-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 02/19/2012] [Indexed: 12/15/2022]
Abstract
Animal models are established tools to study basic questions of biology in a systematic way. They have greatly facilitated our understanding of the mechanisms by which nature forms and maintains organisms. Much of the knowledge on molecular changes underlying the development of organisms originates from research in the fruit fly model Drosophila melanogaster. Vertebrate models including the mouse and zebrafish model, but also other animal models coming from different corners of the animal kingdom have shown that much of the basic machinery of development is essentially identical, not just in all vertebrates but in all major phyla of invertebrates too. Moreover, key elements of this machinery have been demonstrated to be involved in recurrent molecular abnormalities detected in tumor-tissue from patients, indicating their implication in the genesis of human cancer. Thus, research in this field has become a common topic for both biologists and hemato-oncologists. In this review, we summarize current knowledge on some of these key elements and molecular pathways such as Notch, Hedgehog, Wingless, Runt, and Trithorax that have been originally described and studied in animal models and which seem to play a major role in the pathophysiology and targeted management of human cancer.
Collapse
|
306
|
Blazsó P, Sinkó I, Praznovszky T, Hadlaczky G, Katona RL. 3.6-KB mouse cyclin C promoter fragment is predominantly active in the testis. ACTA BIOLOGICA HUNGARICA 2012; 63:26-37. [PMID: 22453798 DOI: 10.1556/abiol.63.2012.1.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cyclin C is a highly conserved protein that regulates cell-cycle, messenger RNA transcription and cell adhesion. Recently published studies demonstrate that this protein is an essential player during early embryonic development of multicellular eukaryotes as well. In order to understand better its complex function at the level of tissues or organs, spatial expression characteristics of cyclin C and regulatory components of its expression are needed to be determined. In vitro studies on human cells suggested that approximately the first 3 kilobases of the cyclin C promoter might contain all the regulatory elements that might mimic transcription of cyclin C. To test the hypothesis, we generated reporter transgenic lines where the first 3.6-kilobase region of mouse cyclin C promoter fragment drives the transcription of a marker gene. Messenger RNA levels of the marker gene and cyclin C isoforms were measured in nine organs with reverse transcription coupled quantitative realtime polymerase chain reaction and their expression patterns were compared. The marker gene is predominantly transcribed in testes and does not follow the transcriptional regulation of the examined cyclin C isoforms. Thus, the isolated promoter fragment alone is not sufficient for the complete physiological modulation of cyclin C RNA levels, however, it is capable of enhancing testicular transcription which can be exploited in future applications.
Collapse
Affiliation(s)
- P Blazsó
- Institute of Genetics, Biological Research Center, Hungarian Academy of Sciences, Temesvári krt. 62 H-6726 Szeged Hungary.
| | | | | | | | | |
Collapse
|
307
|
Gasimli L, Linhardt RJ, Dordick JS. Proteoglycans in stem cells. Biotechnol Appl Biochem 2012; 59:65-76. [PMID: 23586787 DOI: 10.1002/bab.1002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2011] [Accepted: 01/18/2012] [Indexed: 12/18/2022]
Abstract
The remarkable promise of pluripotent and multipotent stem cells (SCs) imparts tremendous optimism for advancement of regenerative medicine, developmental biology, and drug discovery. Perhaps the greatest challenge is to finely direct, control, and command their differentiation. As those processes are managed on many levels, including genomic, transcriptomic, and epigenomic, examination of all of these components will yield powerful tools for manipulation of SCs. Carbohydrates surround all cells, including SCs as a glycocalyx. Of particular interest is the class of carbohydrates known as proteoglycans (PGs), which are a diverse group of glycoconjugates consisting of core protein with one or more glycosaminoglycan (GAG) chains attached. They are primarily located in the extracellular matrix as well as at cell surfaces, where they are bound or anchored to the membrane through their core proteins. GAG chains are linear, anionic, and highly heterogeneous carbohydrates consisting of repeating disaccharides. PGs facilitate interaction of cells with the extracellular environment by interacting with chemokines, growth factors, and other signaling molecules. Core proteins are involved in many signaling pathways, both individually, as well as through attached proteins via GAG-mediated interactions. These essential and accessible functions make PGs an excellent target for manipulating SCs and guiding their fate. Studying the role of PGs in cell development will yield valuable insight into the mechanism of SC differentiation and suggest approaches toward directing those pathways. Such studies may also help identify valuable markers for distinguishing between various cell populations during differentiation.
Collapse
Affiliation(s)
- Leyla Gasimli
- Department of Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | | | | |
Collapse
|
308
|
Adler AS, McCleland ML, Truong T, Lau S, Modrusan Z, Soukup TM, Roose-Girma M, Blackwood EM, Firestein R. CDK8 maintains tumor dedifferentiation and embryonic stem cell pluripotency. Cancer Res 2012; 72:2129-39. [PMID: 22345154 DOI: 10.1158/0008-5472.can-11-3886] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CDK8 is a cyclin-dependent kinase that mediates transcriptional control of pathways linked to both cancer and stem cells. In this study, we show that CDK8 is required for both tumor growth and maintenance of tumor dedifferentiation in vivo and uncover a common role for CDK8 in controlling cancer and stem cell function. Acute CDK8 loss in vivo strongly inhibited tumor growth and promoted differentiation. Transcriptional profiling identified a set of embryonic stem cell-related genes that are activated by CDK8 in cancer. Consistent with this, we found that CDK8 expression correlated to the embryonic stem cell pluripotency state and loss of CDK8 caused embryonic stem cells to differentiate. This effect was, at least partially, mediated by the ability of CDK8 to regulate MYC protein and downstream MYC target gene expression. Similar regulation of MYC target genes by CDK8 was observed in colon tumor cells, and increased expression of a CDK8-regulated, embryonic stem cell MYC target gene signature was associated with loss of differentiation and poor outcome in primary human colon cancers. Together, these observations reveal that CDK8 acts, at least in part, through MYC to maintain both tumors and embryonic stem cells in an undifferentiated state. This raises the intriguing possibility that targeting CDK8 therapeutically may specifically inhibit the stem-like properties of cancer cells.
Collapse
Affiliation(s)
- Adam S Adler
- Department of Pathology, Genentech, Inc., South San Francisco, California 94080, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
309
|
The molecular basis of Notch signaling: a brief overview. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 727:1-14. [PMID: 22399335 DOI: 10.1007/978-1-4614-0899-4_1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The Notch signaling pathway is evolutionarily conserved and has been associated with numerous developmental processes, including stem cell maintenance and adult tissue homeostasis. Notably, both abnormal increases and deficiencies of Notch signaling result in human developmental anomalies and cancer development implying that the precise regulation of the intensity and duration of Notch signals is imperative. Numerous studies have demonstrated that the aberrant gain or loss of Notch signaling pathway components is critically linked to multiple human diseases. In this chapter, we will briefly summarize the molecular basis of Notch signaling, focusing on the modulation of Notch signals, and its developmental outcomes including vessel formation and the onset of cancer.
Collapse
|
310
|
Notch and the p53 clan of transcription factors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 727:223-40. [PMID: 22399351 DOI: 10.1007/978-1-4614-0899-4_17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Notch 1 to 4 and the p53 clan, comprising p53, p63 and p73 plus numerous isoforms thereof, are gene transcription regulators that are critically involved in various aspects of cell differentiation, stem cell maintenance and tumour suppression. It is thus perhaps no surprise that extensive crosstalk between the Notch and p53 pathways is implemented during these processes. Typically, Notch together with p53 and even more so with transactivation competent p63 or p73, drives differentiation, whereas Notch combined with transactivation impaired p63 or p73 helps maintain undifferentiated stem cell compartments. With regard to cancer, it seems that Notch acts as a tumour suppressor in cellular contexts where Notch signalling supports p53 activation and both together can bring on its way an anti-proliferative programme of differentiation, senescence or apoptosis. In contrast, Notch often acts as an oncoprotein in contexts where it suppresses p53 activation and activity and where differentiation is unwanted. It is no accident that the latter pathways-the inhibition by Notch of p53 and differentiation-are operative in somatic stem cells as well as in tumour cells.
Collapse
|
311
|
Shah DK, Zúñiga-Pflücker JC. Notch receptor-ligand interactions during T cell development, a ligand endocytosis-driven mechanism. Curr Top Microbiol Immunol 2012; 360:19-46. [PMID: 22581027 DOI: 10.1007/82_2012_225] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Notch signaling plays an important role during the development of different cell types and tissues. The role of Notch signaling in lymphocyte development, in particular in the development and commitment to the T cell lineage, has been the focus of research for many years. Notch signaling is absolutely required during the commitment and early stages of T cell development. Activation of the Notch signaling pathway is initiated by ligand-receptor interactions and appears to require active endocytosis of Notch ligands. Studies addressing the mechanism underlying endocytosis of Notch ligands have helped to identify the main players important and necessary for this process. Here, we review the Notch ligands, and the proposed models of Notch activation by Notch ligand endocytosis, highlighting key molecules involved. In particular, we discuss recent studies on Notch ligands involved in T cell development, current studies aimed at elucidating the relevance of Notch ligand endocytosis during T cell development and the identification of key players necessary for ligand endocytosis in the thymus and during T cell development.
Collapse
Affiliation(s)
- Divya K Shah
- Department of Immunology, Sunnybrook Research Institute, University of Toronto, 2075 Bayview Avenue, Toronto, ON, M4 N 3M5, Canada.
| | | |
Collapse
|
312
|
Reedijk M. Notch Signaling and Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 727:241-57. [DOI: 10.1007/978-1-4614-0899-4_18] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
313
|
Nusser-Stein S, Beyer A, Rimann I, Adamczyk M, Piterman N, Hajnal A, Fisher J. Cell-cycle regulation of NOTCH signaling during C. elegans vulval development. Mol Syst Biol 2012; 8:618. [PMID: 23047528 PMCID: PMC3501274 DOI: 10.1038/msb.2012.51] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 09/04/2012] [Indexed: 01/05/2023] Open
Abstract
C. elegans vulval development is one of the best-characterized systems to study cell fate specification during organogenesis. The detailed knowledge of the signaling pathways determining vulval precursor cell (VPC) fates permitted us to create a computational model based on the antagonistic interactions between the epidermal growth factor receptor (EGFR)/RAS/MAPK and the NOTCH pathways that specify the primary and secondary fates, respectively. A key notion of our model is called bounded asynchrony, which predicts that a limited degree of asynchrony in the progression of the VPCs is necessary to break their equivalence. While searching for a molecular mechanism underlying bounded asynchrony, we discovered that the termination of NOTCH signaling is tightly linked to cell-cycle progression. When single VPCs were arrested in the G1 phase, intracellular NOTCH failed to be degraded, resulting in a mixed primary/secondary cell fate. Moreover, the G1 cyclins CYD-1 and CYE-1 stabilize NOTCH, while the G2 cyclin CYB-3 promotes NOTCH degradation. Our findings reveal a synchronization mechanism that coordinates NOTCH signaling with cell-cycle progression and thus permits the formation of a stable cell fate pattern.
Collapse
Affiliation(s)
- Stefanie Nusser-Stein
- Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
- Molecular Life Sciences PhD program, Uni ETH Zürich, Switzerland
| | - Antje Beyer
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Ivo Rimann
- Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
| | - Magdalene Adamczyk
- Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
- Molecular Life Sciences PhD program, Uni ETH Zürich, Switzerland
| | - Nir Piterman
- Department of Computer Science, University of Leicester, Leicester, UK
| | - Alex Hajnal
- Institute of Molecular Life Sciences, University of Zürich, Zürich, Switzerland
| | | |
Collapse
|
314
|
Tzoneva G, Ferrando AA. Recent advances on NOTCH signaling in T-ALL. Curr Top Microbiol Immunol 2012; 360:163-82. [PMID: 22673746 DOI: 10.1007/82_2012_232] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
NOTCH1 receptor signaling plays a central role in T-cell lineage specification and in supporting the growth and proliferation of immature T-cell progenitors in the thymus during lymphoid development. In T-cell acute lymphoblastic leukemia (T-ALL), a tumor resulting from the malignant transformation of T-cell progenitors, aberrant and constitutively active NOTCH1 signaling triggered by activating mutations in the NOTCH1 gene contributes to oncogenic transformation and is a hallmark of this disease. Most notably, small molecule γ-secretase inhibitors (GSIs) can effectively block NOTCH1 signaling in T-ALL, and could be exploited as a targeted therapy in this disease. In addition, a number of emerging anti-NOTCH therapeutic strategies including anti-NOTCH1 inhibitory antibodies, small peptide inhibitors of NOTCH signaling and combination therapies with GSIs and glucocorticoids, have recently been proposed. Finally, the identification of NOTCH1 mutations in solid tumors and chronic lymphocytic leukemias has increased even further the clinical relevance of NOTCH signaling as a therapeutic target in human cancer. Here we review our current understanding of NOTCH1-induced transformation, the mechanisms of action of oncogenic NOTCH1 in T-ALL and the therapeutic and prognostic implications of NOTCH1 mutations in T-ALL.
Collapse
Affiliation(s)
- Gannie Tzoneva
- Institute for Cancer Genetics and Graduate Program in Pathobiology and Molecular Medicine, Columbia University Medical Center, New York 10032, USA
| | | |
Collapse
|
315
|
Jo HS, Kang KH, Joe CO, Kim JW. Pten coordinates retinal neurogenesis by regulating Notch signalling. EMBO J 2011; 31:817-28. [PMID: 22258620 DOI: 10.1038/emboj.2011.443] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 11/08/2011] [Indexed: 11/09/2022] Open
Abstract
Development of nervous tissue is a coordinated process of neural progenitor cell (NPC) proliferation and neuronal differentiation. Intracellular signalling events that regulate the balance between NPC proliferation and neuronal differentiation, therefore, determine the size and composition of nervous tissues. Here, we demonstrate that negative regulation of phosphoinosite 3-kinase (PI3K)-Akt signalling by phosphatase tensin homologue (Pten) is essential for maintaining NPC population in mouse retina. We found that mouse retinal progenitor cells (RPCs) lacking the Pten gene complete neurogenesis earlier than their normal developmental schedule, resulting in their premature depletion in the mature retina. We further discover that Notch intracellular domain (NICD) fails to form transcription activator complex in Pten-deficient RPCs, and thereby unable to support RPC maintenance. Taken together, our results suggest that Pten plays a pivotal role in retinal neurogenesis by supporting Notch-driven RPC maintenance against neurogenic PI3K-Akt signalling.
Collapse
Affiliation(s)
- Hong Seok Jo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | | | | | | |
Collapse
|
316
|
Cdk8 regulates stability of the transcription factor Phd1 to control pseudohyphal differentiation of Saccharomyces cerevisiae. Mol Cell Biol 2011; 32:664-74. [PMID: 22124158 DOI: 10.1128/mcb.05420-11] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The yeast Saccharomyces differentiates into filamentous pseudohyphae when exposed to a poor source of nitrogen in a process involving a collection of transcription factors regulated by nutrient signaling pathways. Phd1 is important for this process in that it regulates expression of most other transcription factors involved in differentiation and can induce filamentation on its own when overproduced. In this article, we show that Phd1 is an unstable protein whose degradation is initiated through phosphorylation by Cdk8 of the RNA polymerase II mediator subcomplex. Phd1 is stabilized by cdk8 disruption, and the naturally filamenting Σ1278b strain was found to have a sequence polymorphism that eliminates a Cdk8 phosphorylation site, which both stabilizes the protein and contributes to enhanced differentiation. In nitrogen-starved cells, PHD1 expression is upregulated and the Phd1 protein becomes stabilized, which causes its accumulation during differentiation. PHD1 expression is partially dependent upon Ste12, which was also previously shown to be destabilized by Cdk8-dependent phosphorylations, but to a significantly smaller extent than Phd1. These observations demonstrate the central role that Cdk8 plays in initiation of differentiation. Cdk8 activity is inhibited in cells shifted to limiting nutrient conditions, and we argue that this effect drives the initiation of differentiation through stabilization of multiple transcription factors, including Phd1, that cause activation of genes necessary for filamentous response.
Collapse
|
317
|
He SB, Yuan Y, Wang L, Yu MJ, Zhu YB, Zhu XG. Effects of cyclin-dependent kinase 8 specific siRNA on the proliferation and apoptosis of colon cancer cells. J Exp Clin Cancer Res 2011; 30:109. [PMID: 22104393 PMCID: PMC3271993 DOI: 10.1186/1756-9966-30-109] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 11/22/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To investigate the expression of cyclin-dependent kinase 8 (CDK8) and β-catenin in colon cancer and evaluate the role of CDK8 in the proliferation, apoptosis and cell cycle progression of colon cancer cells, especially in HCT116 cell line. METHODS Colon cancer cell line HCT116 was transfected with small interfering RNA (siRNA) targeting on CDK8. After CDK8-siRNA transfection, mRNA and protein expression levels of CDK8 and β-catenin were determined by reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blot assay in HCT116 cells. Cell proliferation was measured by 3-(4, 5-Dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide Methylthiazolyl tetrazolium (MTT) assay, and cell cycle distribution and apoptosis were analyzed by flow cytometry analysis (FACS). CDK8 and β-catenin protein levels were also examined by real-time PCR and immunohistochemistry (IHC) in colon cancer tissues and adjacent normal tissues. RESULTS After CDK8 specific siRNA transfection, mRNA and protein expression levels of CDK8 and β-catenin in HCT116 cells were noticeably decreased (P < 0.05). CDK8 specific siRNA transfection inhibited HCT116 cells' proliferation and facilitated their apoptosis significantly (P < 0.05). In addition, the proportion of HCT116 cells in the G0/G1 phase was remarkably increased after CDK8-siRNA transfection (P < 0.05). The expression levels of CDK8 and β-catenin in adjacent normal tissues were lower than in tumor tissues (P < 0.05). Moreover, the expression of CDK8 was correlated with the expression of β-catenin in both tumor and adjacent normal tissues (P < 0.05). CONCLUSIONS CDK8 and β-catenin were expressed in colon cancer at a high frequency. CDK8 specific siRNA transfection down-regulated the expression of CDK8 in colon cancer cells, which was also associated with a decrease in the expression of β-catenin Moreover, CDK8 specific siRNA inhibited the proliferation of colon cancer cells, promoted their apoptosis and arrested these cells in the G0/G1 phase. Interference of CDK8 might be an effective strategy through β-catenin regulation of colon cancer.
Collapse
Affiliation(s)
- Song-Bing He
- Department of General Surgery, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | | | | | | | | | | |
Collapse
|
318
|
Popko-Scibor AE, Lindberg MJ, Hansson ML, Holmlund T, Wallberg AE. Ubiquitination of Notch1 is regulated by MAML1-mediated p300 acetylation of Notch1. Biochem Biophys Res Commun 2011; 416:300-6. [PMID: 22100894 DOI: 10.1016/j.bbrc.2011.11.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 11/04/2011] [Indexed: 02/02/2023]
Abstract
Earlier studies demonstrated the involvement of the p300 histone acetyltransferase in Notch signaling but the precise mechanisms by which p300 might modulate Notch function remains to be investigated. In this study, we show that p300 acetylates Notch1 ICD in cell culture assay and in vitro, and conserved lysines located within the Notch C-terminal nuclear localization signal are essential for Notch acetylation. MAML1 and CSL, which are components of the Notch transcription complex, enhance Notch acetylation and we suggest that MAML1 increases Notch acetylation by potentiating p300 autoacetylation. Furthermore, MAML1-dependent acetylation of Notch1 ICD by p300 decreases the ubiquitination of Notch1 ICD in cellular assays. CDK8 has been shown to target Notch1 for ubiquitination and proteosomal degradation. We show that CDK8 inhibits Notch acetylation and Notch transcription enhanced by p300. Therefore, we speculate that acetylation of Notch1 might be a mechanism to regulate Notch activity by interfering with ubiquitin-dependent pathways.
Collapse
Affiliation(s)
- Anita E Popko-Scibor
- Division of Molecular Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | | | | | | |
Collapse
|
319
|
Zhang Y, Wheatley R, Fulkerson E, Tapp A, Estes PA. Mastermind mutations generate a unique constellation of midline cells within the Drosophila CNS. PLoS One 2011; 6:e26197. [PMID: 22046261 PMCID: PMC3203113 DOI: 10.1371/journal.pone.0026197] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 09/22/2011] [Indexed: 02/05/2023] Open
Abstract
Background The Notch pathway functions repeatedly during the development of the central nervous system in metazoan organisms to control cell fate and regulate cell proliferation and asymmetric cell divisions. Within the Drosophila midline cell lineage, which bisects the two symmetrical halves of the central nervous system, Notch is required for initial cell specification and subsequent differentiation of many midline lineages. Methodology/Principal Findings Here, we provide the first description of the role of the Notch co-factor, mastermind, in the central nervous system midline of Drosophila. Overall, zygotic mastermind mutations cause an increase in midline cell number and decrease in midline cell diversity. Compared to mutations in other components of the Notch signaling pathway, such as Notch itself and Delta, zygotic mutations in mastermind cause the production of a unique constellation of midline cell types. The major difference is that midline glia form normally in zygotic mastermind mutants, but not in Notch and Delta mutants. Moreover, during late embryogenesis, extra anterior midline glia survive in zygotic mastermind mutants compared to wild type embryos. Conclusions/Significance This is an example of a mutation in a signaling pathway cofactor producing a distinct central nervous system phenotype compared to mutations in major components of the pathway.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Genetics, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Randi Wheatley
- Department of Genetics, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Eric Fulkerson
- Department of Genetics, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Amanda Tapp
- Department of Genetics, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Patricia A. Estes
- Department of Genetics, North Carolina State University, Raleigh, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
320
|
Xu W, Ji JY. Dysregulation of CDK8 and Cyclin C in tumorigenesis. J Genet Genomics 2011; 38:439-52. [PMID: 22035865 PMCID: PMC9792140 DOI: 10.1016/j.jgg.2011.09.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Revised: 09/05/2011] [Accepted: 09/06/2011] [Indexed: 01/23/2023]
Abstract
Appropriately controlled gene expression is fundamental for normal growth and survival of all living organisms. In eukaryotes, the transcription of protein-coding mRNAs is dependent on RNA polymerase II (Pol II). The multi-subunit transcription cofactor Mediator complex is proposed to regulate most, if not all, of the Pol II-dependent transcription. Here we focus our discussion on two subunits of the Mediator complex, cyclin-dependent kinase 8 (CDK8) and its regulatory partner Cyclin C (CycC), because they are either mutated or amplified in a variety of human cancers. CDK8 functions as an oncoprotein in melanoma and colorectal cancers, thus there are considerable interests in developing drugs specifically targeting the CDK8 kinase activity. However, to evaluate the feasibility of targeting CDK8 for cancer therapy and to understand how their dysregulation contributes to tumorigenesis, it is essential to elucidate the in vivo function and regulation of CDK8-CycC, which are still poorly understood in multi-cellular organisms. We summarize the evidence linking their dysregulation to various cancers and present our bioinformatics and computational analyses on the structure and evolution of CDK8. We also discuss the implications of these observations in tumorigenesis. Because most of the Mediator subunits, including CDK8 and CycC, are highly conserved during eukaryotic evolution, we expect that investigations using model organisms such as Drosophila will provide important insights into the function and regulation of CDK8 and CycC in different cellular and developmental contexts.
Collapse
Affiliation(s)
- Wu Xu
- Department of Chemistry, University of Louisiana at Lafayette, P.O. Box 44370, Lafayette, LA 70504, USA
| | - Jun-Yuan Ji
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M Health Science Center, College Station, TX 77843, USA
- Corresponding author: Tel: +1 979 845 6389, fax: +1 979 847 9481. (J.-Y. Ji)
| |
Collapse
|
321
|
Andersson ER, Sandberg R, Lendahl U. Notch signaling: simplicity in design, versatility in function. Development 2011; 138:3593-612. [PMID: 21828089 DOI: 10.1242/dev.063610] [Citation(s) in RCA: 712] [Impact Index Per Article: 50.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Notch signaling is evolutionarily conserved and operates in many cell types and at various stages during development. Notch signaling must therefore be able to generate appropriate signaling outputs in a variety of cellular contexts. This need for versatility in Notch signaling is in apparent contrast to the simple molecular design of the core pathway. Here, we review recent studies in nematodes, Drosophila and vertebrate systems that begin to shed light on how versatility in Notch signaling output is generated, how signal strength is modulated, and how cross-talk between the Notch pathway and other intracellular signaling systems, such as the Wnt, hypoxia and BMP pathways, contributes to signaling diversity.
Collapse
Affiliation(s)
- Emma R Andersson
- Department of Cell and Molecular Biology, Karolinska Institute, SE-171 77 Stockholm, Sweden
| | | | | |
Collapse
|
322
|
Cummins TD, Mendenhall MD, Lowry MN, Korte EA, Barati MT, Khundmiri SJ, Salyer SA, Klein JB, Powell DW. Elongin C is a mediator of Notch4 activity in human renal tubule cells. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2011; 1814:1748-57. [PMID: 22001063 DOI: 10.1016/j.bbapap.2011.09.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 09/12/2011] [Accepted: 09/23/2011] [Indexed: 10/17/2022]
Abstract
Notch proteins (Notch 1-4) are a family of trans-membrane cell surface receptors that are converted into transcriptional regulators when activated by interactions with cell surface ligands on adjacent cells. Ligand-binding stimulates proteolytic cleavage of the trans-membrane domain, releasing an active intracellular domain (ICD) that translocates to the nucleus and impacts transcription. In transit, the ICD may interact with regulatory proteins that modulate the expression and transcriptional activity. We have found that Notch4(ICD) expression is enhanced in the tubule cells of fibrotic kidneys from diabetic mice and humans and identified Notch4(ICD) interacting proteins that could be pertinent to normal and pathological functions. Using proteomic techniques, several components of the Elongin C complex were identified as candidate Notch4(ICD) interactors. Elongin C complexes can function as ubiquitin ligases capable of regulating proteasomal degradation of specific protein substrates. Our studies indicate that ectopic Elongin C expression stimulates Notch4(ICD) degradation and inhibits its transcriptional activity in human kidney tubule HK11 cells. Blocking Elongin C mediated degradation by MG132 indicates the potential for ubiquitin-mediated Elongin C regulation of Notch4(ICD). Functional interaction of Notch4(ICD) and Elongin C provides novel insight into regulation of Notch signaling in epithelial cell biology and disease.
Collapse
Affiliation(s)
- Timothy D Cummins
- Departments of Biochemistry and Molecular Biology, University of Kentucky, KY, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
323
|
Wang MM. Notch signaling and Notch signaling modifiers. Int J Biochem Cell Biol 2011; 43:1550-62. [PMID: 21854867 DOI: 10.1016/j.biocel.2011.08.005] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 07/28/2011] [Accepted: 08/05/2011] [Indexed: 02/07/2023]
Abstract
Originally discovered nearly a century ago, the Notch signaling pathway is critical for virtually all developmental programs and modulates an astounding variety of pathogenic processes. The DSL (Delta, Serrate, LAG-2 family) proteins have long been considered canonical activators of the core Notch pathway. More recently, a wide and expanding network of non-canonical extracellular factors has also been shown to modulate Notch signaling, conferring newly appreciated complexity to this evolutionarily conserved signal transduction system. Here, I review current concepts in Notch signaling, with a focus on work from the last decade elucidating novel extracellular proteins that up- or down-regulate signal potency.
Collapse
Affiliation(s)
- Michael M Wang
- Neurology Service, Veterans Administration Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA.
| |
Collapse
|
324
|
Chen W, Roeder RG. Mediator-dependent nuclear receptor function. Semin Cell Dev Biol 2011; 22:749-58. [PMID: 21854863 DOI: 10.1016/j.semcdb.2011.07.026] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 07/20/2011] [Indexed: 12/24/2022]
Abstract
As gene-specific transcription factors, nuclear receptors are broadly involved in many important biological processes. Their function on target genes requires the stepwise assembly of different coactivator complexes that facilitate chromatin remodeling and subsequent preinitiation complex (PIC) formation and function. Mediator has proved to be a crucial, and general, nuclear receptor-interacting coactivator, with demonstrated functions in transcription steps ranging from chromatin remodeling to subsequent PIC formation and function. Here we discuss our current understanding of (i) pathways involved in Mediator recruitment and function through nuclear receptor target gene enhancers and promoters, (ii) conditional requirements for the strong nuclear receptor-Mediator interactions mediated by NR AF2 domains and the MED1 LXXLL motifs, (iii) Mediator functions, through different nuclear receptor-interacting subunits, in different metabolic pathways, (iv) emerging functions of Mediator as a corepressor in addition to its major role as a coactivator and (v) mechanisms by which Mediator acts to transmit signals from enhancer-bound nuclear receptors to the general transcription machinery at core promoters to effect PIC formation and function. As a nuclear receptor coregulator with increasingly diverse functions, Mediator may thus modulate nuclear receptor signaling through several different mechanisms.
Collapse
Affiliation(s)
- Wei Chen
- Laboratory of Biochemistry and Molecular Biology, The Rockefeller University, 1230 York Ave., New York, NY 10065, USA.
| | | |
Collapse
|
325
|
Critical roles of NOTCH1 in acute T-cell lymphoblastic leukemia. Int J Hematol 2011; 94:118-125. [PMID: 21814881 DOI: 10.1007/s12185-011-0899-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 07/06/2011] [Indexed: 10/18/2022]
Abstract
NOTCH1 plays a central role in T-cell development and, when aberrantly activated, in acute T-cell lymphoblastic leukemia (T-ALL). As a transmembrane receptor that is ultimately converted into a transcription factor, NOTCH1 directly activates a spectrum of target genes, which function to mediate NOTCH1 signaling in normal or transformed T cells. During physiologic T-cell development, NOTCH1 has important functions in cell fate determination, proliferation, survival and metabolism. Activating NOTCH1 mutations occur in more than half of human patients with T-ALL, suggesting an important role for aberrant NOTCH1 signaling in the pathogenesis of this disease. Inhibiting NOTCH1 signaling in patient-derived cell lines and murine T-ALLs leads to growth arrest and/or apoptosis suggesting that NOTCH1 inhibitors can improve T-ALL treatment. However, there are challenges to translate NOTCH1 inhibitors to the clinic because of toxicity and resistance. This review focuses on molecular mechanisms of oncogenic NOTCH1 signaling, molecular and functional analysis of NOTCH1 transcriptional targets in T-ALL, and recent advances in therapeutic targeting of NOTCH1.
Collapse
|
326
|
Interactions between subunits of the Mediator complex with gene-specific transcription factors. Semin Cell Dev Biol 2011; 22:759-68. [PMID: 21839847 DOI: 10.1016/j.semcdb.2011.07.022] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 07/26/2011] [Accepted: 07/28/2011] [Indexed: 11/24/2022]
Abstract
The Mediator complex forms the bridge between gene-specific transcription factors and the RNA polymerase II (RNAP II) machinery. Mediator is a large polypetide complex consisting of about thirty polypeptides that are mostly conserved from yeast to human. Mediator coordinates RNAP II recruitment, phosphorylation of the C-terminal domain of RNAP II, enhancer-loop formation and post-initiation events. The focus of the review is to summarize the current knowledge of transcription factor/Mediator interactions in higher eukaryotes and illuminate the physiological and gene-selective roles of Mediator.
Collapse
|
327
|
Kato Y. The multiple roles of Notch signaling during left-right patterning. Cell Mol Life Sci 2011; 68:2555-67. [PMID: 21544546 PMCID: PMC11114802 DOI: 10.1007/s00018-011-0695-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Revised: 03/28/2011] [Accepted: 04/19/2011] [Indexed: 10/18/2022]
Abstract
The establishment of left-right (LR) asymmetry is regulated by intricate signaling mechanisms during embryogenesis and this asymmetry is critical for morphogenesis as well as the positioning of internal organs within the organism. Recent progress including elucidation of ion transporters, leftward nodal flow, and regulation of asymmetric gene expression contributes to our understanding of how the breaking of the symmetry is initiated and how this laterality information is subsequently transmitted to the organ primordium. A number of developmental signaling pathways have been implicated in this complex process. In this review, we will focus on the roles of the Notch signaling pathway during development of LR asymmetry. The Notch signaling pathway is a short-range communication system between neighboring cells. While Notch signaling plays essential roles in regulating the morphogenesis of the node and left-specific expression of Nodal in the lateral plate mesoderm, a hallmark gene in LR patterning, Notch signaling also suppresses the expression of Pitx2 that is a direct downstream target of Nodal during later stages of development. This negative activity of Notch signaling towards left-specific activity was recently shown to be inhibited by the B cell lymphoma 6 (BCL6)/BCL6 co-repressor (BcoR) transcriptional repressor complex in a target-specific manner. The complex regulation of Notch-dependent gene expression for LR asymmetry will be highlighted in this review.
Collapse
Affiliation(s)
- Yoichi Kato
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA.
| |
Collapse
|
328
|
Janody F, Treisman JE. Requirements for mediator complex subunits distinguish three classes of notch target genes at the Drosophila wing margin. Dev Dyn 2011; 240:2051-9. [PMID: 21793099 DOI: 10.1002/dvdy.22705] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2011] [Indexed: 11/11/2022] Open
Abstract
Spatial and temporal gene regulation relies on a combinatorial code of sequence-specific transcription factors that must be integrated by the general transcriptional machinery. A key link between the two is the mediator complex, which consists of a core complex that reversibly associates with the accessory kinase module. We show here that genes activated by Notch signaling at the dorsal-ventral boundary of the Drosophila wing disc fall into three classes that are affected differently by the loss of kinase module subunits. One class requires all four kinase module subunits for activation, while the others require only Med12 and Med13, either for activation or for repression. These distinctions do not result from different requirements for the Notch coactivator Mastermind or the corepressors Hairless and Groucho. We propose that interactions with the kinase module through distinct cofactors allow the DNA-binding protein Suppressor of Hairless to carry out both its activator and repressor functions.
Collapse
Affiliation(s)
- Florence Janody
- Kimmel Center for Biology and Medicine of the Skirball Institute, NYU School of Medicine, Department of Cell Biology, New York, New York, USA
| | | |
Collapse
|
329
|
Leem YE, Choi HK, Jung SY, Kim BJ, Lee KY, Yoon K, Qin J, Kang JS, Kim ST. Esco2 promotes neuronal differentiation by repressing Notch signaling. Cell Signal 2011; 23:1876-84. [PMID: 21777673 DOI: 10.1016/j.cellsig.2011.07.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 07/05/2011] [Indexed: 10/18/2022]
Abstract
Esco2 is an acetyltransferase that is required for the establishment of sister chromatid cohesion. Roberts-SC phocomelia (RBS) syndrome caused by the mutations of Esco2 gene, is an autosomal recessive development disorder characterized by growth retardation, limb reduction and craniofacial abnormalities including cleft lip and palate. Here, we show that Esco2 protein co-immunoprecipitates with Notch but not with CBF1. Esco2 represses the transactivational activity of Notch protein in an acetyltransferase-independent manner. Chromatin immunoprecipitation experiments suggest that Esco2 might regulate the activity of NICD-CBF1 via attenuating NICD binding to CBF1 on the promoter of Hes1, the downstream target gene of Notch. Furthermore, we demonstrate that the overexpression of Esco2 promotes the neuronal differentiation of P19 embryonic carcinoma cells and C17.2 neural progenitor cells and the knockdown of Esco2 by siRNA blocks the differentiation. The inhibitory effects of Notch protein on neuronal differentiation of P19 cells was suppressed by Esco2 overexpression. Taken together, our study suggests that Esco2 may play an important role in neurogenesis by attenuating Notch signaling to promote neuronal differentiation.
Collapse
Affiliation(s)
- Young-Eun Leem
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon, 440-746, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
330
|
Ilagan MXG, Lim S, Fulbright M, Piwnica-Worms D, Kopan R. Real-time imaging of notch activation with a luciferase complementation-based reporter. Sci Signal 2011; 4:rs7. [PMID: 21775282 DOI: 10.1126/scisignal.2001656] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Notch signaling regulates many cellular processes during development and adult tissue renewal. Upon ligand binding, Notch receptors undergo ectodomain shedding followed by γ-secretase-mediated release of the Notch intracellular domain (NICD), which translocates to the nucleus and associates with the DNA binding protein CSL [CBF1/RBPjκ/Su(H)/Lag1] to activate gene expression. Mammalian cells contain four Notch receptors that can have both redundant and specific activities. To monitor activation of specific Notch paralogs in live cells and in real time, we developed luciferase complementation imaging (LCI) reporters for NICD-CSL association and validated them as a specific, robust, and sensitive assay system that enables structure-function and pharmacodynamic analyses. Detailed kinetic analyses of various mechanistic aspects of Notch signaling, including nuclear translocation and inhibition of the activities of γ-secretase and ADAM metalloproteases, as well as agonist- and ligand-dependent activation, were conducted in live cells. These experiments showed that Notch-LCI is an effective approach for characterizing modulators that target Notch signaling and for studying pathway dynamics in normal and disease contexts.
Collapse
Affiliation(s)
- Ma Xenia G Ilagan
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | | | | | | | | |
Collapse
|
331
|
Wesley CS, Guo H, Chaudhry KA, Thali MJ, Yin JC, Clason T, Wesley UV. Loss of PTB or negative regulation of Notch mRNA reveals distinct zones of Notch and actin protein accumulation in Drosophila embryo. PLoS One 2011; 6:e21876. [PMID: 21750738 PMCID: PMC3130057 DOI: 10.1371/journal.pone.0021876] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Accepted: 06/12/2011] [Indexed: 11/19/2022] Open
Abstract
Polypyrimidine Tract Binding (PTB) protein is a regulator of mRNA processing and translation. Genetic screens and studies of wing and bristle development during the post-embryonic stages of Drosophila suggest that it is a negative regulator of the Notch pathway. How PTB regulates the Notch pathway is unknown. Our studies of Drosophila embryogenesis indicate that (1) the Notch mRNA is a potential target of PTB, (2) PTB and Notch functions in the dorso-lateral regions of the Drosophila embryo are linked to actin regulation but not their functions in the ventral region, and (3) the actin-related Notch activity in the dorso-lateral regions might require a Notch activity at or near the cell surface that is different from the nuclear Notch activity involved in cell fate specification in the ventral region. These data raise the possibility that the Drosophila embryo is divided into zones of different PTB and Notch activities based on whether or not they are linked to actin regulation. They also provide clues to the almost forgotten role of Notch in cell adhesion and reveal a role for the Notch pathway in cell fusions.
Collapse
Affiliation(s)
- Cedric S Wesley
- Department of Genetics, University of Wisconsin, Madison, Wisconsin, United States of America.
| | | | | | | | | | | | | |
Collapse
|
332
|
Ranganathan P, Vasquez-Del Carpio R, Kaplan FM, Wang H, Gupta A, VanWye JD, Capobianco AJ. Hierarchical phosphorylation within the ankyrin repeat domain defines a phosphoregulatory loop that regulates Notch transcriptional activity. J Biol Chem 2011; 286:28844-28857. [PMID: 21685388 DOI: 10.1074/jbc.m111.243600] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Notch signal transduction pathway mediates important cellular functions through direct cell-to-cell contact. Deregulation of Notch activity can lead to an altered cell proliferation and has been linked to many human cancers. Casein kinase 2 (CK2), a ubiquitous kinase, regulates several cellular processes by phosphorylating proteins involved in signal transduction, gene expression, and protein synthesis. In this report we identify Notch(ICD) as a novel target of phosphorylation by CK2. Using mapping and mutational studies, we identified serine 1901, located in the ankyrin domain of Notch, as the target amino acid. Interestingly, phosphorylation of serine 1901 by CK2 appears to generate a second phosphorylation site at threonine 1898. Furthermore, threonine 1898 phosphorylation only occurs when Notch forms a complex with Mastermind and CSL. Phosphorylation of both threonine 1898 and serine 1901 resulted in decreased binding of the Notch-Mastermind-CSL ternary complex to DNA and consequently lower transcriptional activity. These data indicate that the phosphorylation of serine 1901 and threonine 1898 negatively regulates Notch function by dissociating the complex from DNA. This study identifies a new component involved in regulation of Notch(ICD) transcriptional activity, reinforcing the notion that a precise and tight regulation is required for this essential signaling pathway.
Collapse
Affiliation(s)
- Prathibha Ranganathan
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family of Surgery and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami Florida 33136
| | - Rodrigo Vasquez-Del Carpio
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family of Surgery and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami Florida 33136
| | - Fred M Kaplan
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family of Surgery and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami Florida 33136
| | - Hong Wang
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family of Surgery and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami Florida 33136
| | - Ashu Gupta
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family of Surgery and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami Florida 33136
| | - Jeffrey D VanWye
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family of Surgery and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami Florida 33136
| | - Anthony J Capobianco
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family of Surgery and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami Florida 33136.
| |
Collapse
|
333
|
Sprinzak D, Lakhanpal A, LeBon L, Garcia-Ojalvo J, Elowitz MB. Mutual inactivation of Notch receptors and ligands facilitates developmental patterning. PLoS Comput Biol 2011; 7:e1002069. [PMID: 21695234 PMCID: PMC3111533 DOI: 10.1371/journal.pcbi.1002069] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Accepted: 04/12/2011] [Indexed: 11/18/2022] Open
Abstract
Developmental patterning requires juxtacrine signaling in order to tightly coordinate the fates of neighboring cells. Recent work has shown that Notch and Delta, the canonical metazoan juxtacrine signaling receptor and ligand, mutually inactivate each other in the same cell. This cis-interaction generates mutually exclusive sending and receiving states in individual cells. It generally remains unclear, however, how this mutual inactivation and the resulting switching behavior can impact developmental patterning circuits. Here we address this question using mathematical modeling in the context of two canonical pattern formation processes: boundary formation and lateral inhibition. For boundary formation, in a model motivated by Drosophila wing vein patterning, we find that mutual inactivation allows sharp boundary formation across a broader range of parameters than models lacking mutual inactivation. This model with mutual inactivation also exhibits robustness to correlated gene expression perturbations. For lateral inhibition, we find that mutual inactivation speeds up patterning dynamics, relieves the need for cooperative regulatory interactions, and expands the range of parameter values that permit pattern formation, compared to canonical models. Furthermore, mutual inactivation enables a simple lateral inhibition circuit architecture which requires only a single downstream regulatory step. Both model systems show how mutual inactivation can facilitate robust fine-grained patterning processes that would be difficult to implement without it, by encoding a difference-promoting feedback within the signaling system itself. Together, these results provide a framework for analysis of more complex Notch-dependent developmental systems. Multicellular development requires tightly regulated spatial pattern formation, frequently including the generation of sharp differences over short length scales. Classic examples include boundary formation in the Drosophila wing veins and lateral inhibition patterning in the differentiation of sensory cells. These processes and a diverse variety of others are mediated by the Notch signaling system which allows neighboring cells to exchange information, via interaction between the Notch receptor on one cell and its ligands such as Delta, on another. Interestingly, recent evidence has shown that Notch and Delta within the same cell (in cis) also interact, mutually inactivating each other. However, the significance of this interaction for pattern formation has remained unclear. Here we show, by analytical and computational modeling, how this cis interaction intrinsically generates a difference-promoting logic that optimizes the system for use in fine-grained pattern formation. Specifically, boundary formation and lateral inhibition patterning both operate more effectively and with simpler circuit architectures than they could without this interaction. Our results provide a foundation for understanding these and other Notch-dependent pattern formation processes.
Collapse
Affiliation(s)
- David Sprinzak
- Howard Hughes Medical Institute, California Institute of Technology, Pasadena, California, United States of America
- Division of Biology and Department of Applied Physics, California Institute of Technology, Pasadena, California, United States of America
| | - Amit Lakhanpal
- Howard Hughes Medical Institute, California Institute of Technology, Pasadena, California, United States of America
- Division of Biology and Department of Applied Physics, California Institute of Technology, Pasadena, California, United States of America
| | - Lauren LeBon
- Howard Hughes Medical Institute, California Institute of Technology, Pasadena, California, United States of America
- Division of Biology and Department of Applied Physics, California Institute of Technology, Pasadena, California, United States of America
| | - Jordi Garcia-Ojalvo
- Departament de Física i Enginyeria Nuclear, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Michael B. Elowitz
- Howard Hughes Medical Institute, California Institute of Technology, Pasadena, California, United States of America
- Division of Biology and Department of Applied Physics, California Institute of Technology, Pasadena, California, United States of America
- * E-mail:
| |
Collapse
|
334
|
Abstract
Notch is a crucial cell signaling pathway in metazoan development. By means of cell-cell interactions, Notch signaling regulates cellular identity, proliferation, differentiation and apoptosis. Within the last decade, numerous studies have shown an important role for this pathway in the development and homeostasis of mammalian stem cell populations. Hematopoietic stem cells (HSCs) constitute a well-defined population that shows self-renewal and multi-lineage differentiation potential, with the clinically relevant capacity to repopulate the hematopoietic system of an adult organism. Here, we review the emergence, development and maintenance of HSCs during mammalian embryogenesis and adulthood, with respect to the role of Notch signaling in hematopoietic biology.
Collapse
|
335
|
Young RA. Control of the embryonic stem cell state. Cell 2011; 144:940-54. [PMID: 21414485 DOI: 10.1016/j.cell.2011.01.032] [Citation(s) in RCA: 900] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 11/23/2010] [Accepted: 01/03/2011] [Indexed: 12/25/2022]
Abstract
Embryonic stem cells and induced pluripotent stem cells hold great promise for regenerative medicine. These cells can be propagated in culture in an undifferentiated state but can be induced to differentiate into specialized cell types. Moreover, these cells provide a powerful model system for studies of cellular identity and early mammalian development. Recent studies have provided insights into the transcriptional control of embryonic stem cell state, including the regulatory circuitry underlying pluripotency. These studies have, as a consequence, uncovered fundamental mechanisms that control mammalian gene expression, connect gene expression to chromosome structure, and contribute to human disease.
Collapse
Affiliation(s)
- Richard A Young
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.
| |
Collapse
|
336
|
Glycogen synthase kinase 3 beta positively regulates Notch signaling in vascular smooth muscle cells: role in cell proliferation and survival. Basic Res Cardiol 2011; 106:773-85. [PMID: 21557011 DOI: 10.1007/s00395-011-0189-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 04/13/2011] [Accepted: 04/29/2011] [Indexed: 01/11/2023]
Abstract
The role of glycogen synthase kinase 3 beta (GSK-3β) in modulating Notch control of vascular smooth muscle cell (vSMC) growth (proliferation and apoptosis) was examined in vitro under varying conditions of cyclic strain and validated in vivo following changes in medial tension and stress. Modulation of GSK-3β in vSMC following ectopic expression of constitutively active GSK-3β, siRNA knockdown and pharmacological inhibition with SB-216763 demonstrated that GSK-3β positively regulates Notch intracellular domain expression, CBF-1/RBP-Jκ transactivation and downstream target gene mRNA levels, while concomitantly promoting vSMC proliferation and inhibiting apoptosis. In contrast, inhibition of GSK-3β attenuated Notch signaling and decreased vSMC proliferation and survival. Exposure of vSMC to cyclic strain environments in vitro using both a Flexercell™ Tension system and a novel Sylgard™ phantom vessel following bare metal stent implantation revealed that cyclic strain inhibits GSK-3β activity independent of p42/p44 MAPK and p38 activation concomitant with reduced Notch signaling and decreased vSMC proliferation and survival. Exposure of vSMC to changes in medial strain microenvironments in vivo following carotid artery ligation revealed that enhanced GSK-3β activity was predominantly localized to medial and neointimal vSMC concomitant with increased Notch signaling, proliferating nuclear antigen and decreased Bax expression, respectively, as vascular remodeling progressed. GSK-3β is an important modulator of Notch signaling leading to altered vSMC cell growth where low strain/tension microenvironments prevail.
Collapse
|
337
|
Calderwood MA, Lee S, Holthaus AM, Blacklow SC, Kieff E, Johannsen E. Epstein-Barr virus nuclear protein 3C binds to the N-terminal (NTD) and beta trefoil domains (BTD) of RBP/CSL; only the NTD interaction is essential for lymphoblastoid cell growth. Virology 2011; 414:19-25. [PMID: 21440926 DOI: 10.1016/j.virol.2011.02.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 02/04/2011] [Accepted: 02/21/2011] [Indexed: 01/31/2023]
Abstract
Association of EBV nuclear proteins EBNA2, EBNA3A and EBNA3C with RBP/CSL, is essential for lymphoblastoid cell line (LCL) proliferation. Conserved residues in the EBNA3 homology domain, required for RBP/CSL interaction, lack the WΦP motif that mediates EBNA2 and Notch binding to the RBP/CSL beta-trefoil domain (BTD). We map RBP/CSL interacting residues within EBNA3A(aa128-204) and EBNA3C(aa211-233). The EBNA3A results are consistent with an earlier report (aa125-222), but the EBNA3C domain is unexpectedly small and includes a "WTP" sequence. This EBNA3C WTP motif confers RBP/CSL binding in vitro, in yeast, and in mammalian cells. Further, an EBNA3C WTP→STP(W227S) mutation impaired BTD binding whereas EBNA3 homology domain mutations disrupted RBP/CSL N-terminal domain (NTD) binding. WTP was not essential for EBNA3C repression of EBNA2 in reporter assays or for maintenance of LCL growth. Our results indicate that EBNA3 proteins interact with multiple RBP/CSL domains, but only NTD interactions are required for LCL growth.
Collapse
Affiliation(s)
- Michael A Calderwood
- Department of Medicine, Channing Laboratory, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Avenue, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
338
|
Waters KA, Reinke V. Extrinsic and intrinsic control of germ cell proliferation in Caenorhabditis elegans. Mol Reprod Dev 2011; 78:151-60. [PMID: 21337453 DOI: 10.1002/mrd.21289] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Accepted: 01/06/2011] [Indexed: 12/23/2022]
Abstract
The germ cells of Caenorhabditis elegans serve as a useful model to study the balance between proliferation and differentiation within the context of development and changing environmental signals experienced by the animal. Germ cells adjacent to a stem cell niche in the distal region of the gonad retain the capacity to divide during adulthood, making them unique from other cells in the organism. We will highlight recent advances in our understanding of mechanisms that control proliferation, as well as the signaling pathways involved in promoting mitosis at the expense of differentiation.
Collapse
|
339
|
Rakowski LA, Lehotzky EA, Chiang MY. Transient responses to NOTCH and TLX1/HOX11 inhibition in T-cell acute lymphoblastic leukemia/lymphoma. PLoS One 2011; 6:e16761. [PMID: 21326611 PMCID: PMC3033898 DOI: 10.1371/journal.pone.0016761] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Accepted: 12/29/2010] [Indexed: 11/26/2022] Open
Abstract
To improve the treatment strategies of T-cell acute lymphoblastic leukemia/lymphoma (T-ALL), further efforts are needed to identify therapeutic targets. Dysregulated expression of HOX-type transcription factors occurs in 30–40% of cases of T-ALL. TLX1/HOX11 is the prototypical HOX-type transcription factor. TLX1 may be an attractive therapeutic target because mice that are deficient in TLX1 are healthy. To test this possibility, we developed a conditional doxycycline-regulated mouse model of TLX1-initiated T-ALL. TLX1 induced T-ALL after ∼5–7 months with penetrance of 15–60%. Similar to human TLX1-type T-ALLs, the TLX1-induced tumors were arrested at the cortical stage of T-cell development and acquired activating NOTCH1 mutations. Inhibition of NOTCH signaling abrogated growth of cell lines derived from the TLX1-induced tumors. NOTCH inhibition also transiently delayed leukemia progression in vivo. Suppression of TLX1 expression slowed the growth of TLX1 tumor cell lines. Suppression of TLX1 in vivo also transiently delayed leukemia progression. We have shown that TLX1 functions as a T-cell oncogene that is active during both the induction and the maintenance phases of leukemia. However, the effect of suppressing NOTCH or TLX1 was transient. The tumors eventually “escaped” from inhibition. These data imply that the biological pathways and gene sets impacted by TLX1 and NOTCH have largely lost their importance in the fully established tumor. They have been supplanted by stronger oncogenic pathways. Although TLX1 or NOTCH inhibitors may not be effective as single agents, they may still contribute to combination therapy for TLX1-driven acute leukemia.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Bone Marrow Transplantation/methods
- Cells, Cultured
- Disease Progression
- Gene Expression Regulation, Leukemic/drug effects
- Homeodomain Proteins/antagonists & inhibitors
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Homeodomain Proteins/physiology
- Humans
- Immunotherapy, Adoptive/methods
- Leukemia-Lymphoma, Adult T-Cell/drug therapy
- Leukemia-Lymphoma, Adult T-Cell/pathology
- Leukemia-Lymphoma, Adult T-Cell/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Molecular Targeted Therapy
- Proto-Oncogene Proteins/antagonists & inhibitors
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins/physiology
- Receptor, Notch1/antagonists & inhibitors
- Receptor, Notch1/metabolism
- Receptor, Notch1/physiology
- Time Factors
Collapse
Affiliation(s)
- Lesley A. Rakowski
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, United States of America
| | - Erica A. Lehotzky
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, United States of America
| | - Mark Y. Chiang
- Division of Hematology-Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
340
|
Li X, von Boehmer H. Notch Signaling in T-Cell Development and T-ALL. ISRN HEMATOLOGY 2011; 2011:921706. [PMID: 22111016 PMCID: PMC3200084 DOI: 10.5402/2011/921706] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 12/15/2010] [Indexed: 11/23/2022]
Abstract
The Notch signaling pathway is an evolutionarily conserved cell signaling system present in most multicellular organisms, as it controls cell fate specification by regulating cell proliferation, differentiation, apoptosis, and survival. Regulation of the Notch signaling pathway can be achieved at multiple levels. Notch proteins are involved in lineage fate decisions in a variety of tissues in various species. Notch is essential for T lineage cell differentiation including T versus B and αβ versus γδ lineage specification. In this paper, we discuss Notch signaling in normal T-cell maturation and differentiation as well as in T-cell acute lymphoblastic lymphoma/leukemia.
Collapse
Affiliation(s)
- Xiaoyu Li
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA
| | - Harald von Boehmer
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA
| |
Collapse
|
341
|
Greene DM, Bloomfield G, Skelton J, Ivens A, Pears CJ. Targets downstream of Cdk8 in Dictyostelium development. BMC DEVELOPMENTAL BIOLOGY 2011; 11:2. [PMID: 21255384 PMCID: PMC3037916 DOI: 10.1186/1471-213x-11-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Accepted: 01/21/2011] [Indexed: 11/11/2022]
Abstract
Background Cdk8 is a component of the mediator complex which facilitates transcription by RNA polymerase II and has been shown to play an important role in development of Dictyostelium discoideum. This eukaryote feeds as single cells but starvation triggers the formation of a multicellular organism in response to extracellular pulses of cAMP and the eventual generation of spores. Strains in which the gene encoding Cdk8 have been disrupted fail to form multicellular aggregates unless supplied with exogenous pulses of cAMP and later in development, cdk8- cells show a defect in spore production. Results Microarray analysis revealed that the cdk8- strain previously described (cdk8-HL) contained genome duplications. Regeneration of the strain in a background lacking detectable gene duplication generated strains (cdk8-2) with identical defects in growth and early development, but a milder defect in spore generation, suggesting that the severity of this defect depends on the genetic background. The failure of cdk8- cells to aggregate unless rescued by exogenous pulses of cAMP is consistent with a failure to express the catalytic subunit of protein kinase A. However, overexpression of the gene encoding this protein was not sufficient to rescue the defect, suggesting that this is not the only important target for Cdk8 at this stage of development. Proteomic analysis revealed two potential targets for Cdk8 regulation, one regulated post-transcriptionally (4-hydroxyphenylpyruvate dioxygenase (HPD)) and one transcriptionally (short chain dehydrogenase/reductase (SDR1)). Conclusions This analysis has confirmed the importance of Cdk8 at multiple stages of Dictyostelium development, although the severity of the defect in spore production depends on the genetic background. Potential targets of Cdk8-mediated gene regulation have been identified in Dictyostelium which will allow the mechanism of Cdk8 action and its role in development to be determined.
Collapse
Affiliation(s)
- David M Greene
- Biochemistry Department, Oxford University, South Parks Road, Oxford OX1 3QU UK.
| | | | | | | | | |
Collapse
|
342
|
Assembly of a Notch transcriptional activation complex requires multimerization. Mol Cell Biol 2011; 31:1396-408. [PMID: 21245387 DOI: 10.1128/mcb.00360-10] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Notch transmembrane receptors direct essential cellular processes, such as proliferation and differentiation, through direct cell-to-cell interactions. Inappropriate release of the intracellular domain of Notch (N(ICD)) from the plasma membrane results in the accumulation of deregulated nuclear N(ICD) that has been linked to human cancers, notably T-cell acute lymphoblastic leukemia (T-ALL). Nuclear N(ICD) forms a transcriptional activation complex by interacting with the coactivator protein Mastermind-like 1 and the DNA binding protein CSL (for CBF-1/Suppressor of Hairless/Lag-1) to regulate target gene expression. Although it is well understood that N(ICD) forms a transcriptional activation complex, little is known about how the complex is assembled. In this study, we demonstrate that N(ICD) multimerizes and that these multimers function as precursors for the stepwise assembly of the Notch activation complex. Importantly, we demonstrate that the assembly is mediated by N(ICD) multimers interacting with Skip and Mastermind. These interactions form a preactivation complex that is then resolved by CSL to form the Notch transcriptional activation complex on DNA.
Collapse
|
343
|
Abstract
Early genetics in flies revealed that Notch is a complex pleiotropic locus. We now know that Notch is a receptor that plays prominent roles during development and functions locally in many tissues to instruct cell fate decisions. Drosophila has been an excellent model to identify genetically the elements that contribute to the canonical Notch signaling transduction machinery defined as DSL-Notch-CSL-MAML axis. This core pathway is required in many biological events in all animals. Though the canonical Notch pathway is relatively simple, and as the steps of the events are now more deeply understood, an increasing number of reports in the last decade show that many other molecules can influence Notch signaling, some by competing with a given element of the cascade. This may occur at any step bringing more diversity and plasticity to the Notch pathway. Most of these regulatory molecules act in a context-specific manner and/or are themselves key regulators in other pathways, providing increasing examples of how connections among distinct pathway modulate each other ("cross talk"). The noncanonical signals discussed in this chapter are broadly defined and correspond to the following: DSL-independent activations, interactions with non-DSL ligands, CSL-independent signaling, signal transduction without cleavage, differential posttranslational modifications, competition/protection for a cofactor, and cross talk with other signaling pathways [Wnt, bone morphogenic protein (BMP), NF-kappaB, etc.]. Though some deemed controversial, these events may impact human diseases. Understanding the molecular nature of these events will allow avoidance of adverse effects during possible clinical treatments. In this review, we will focus on some noncanonical Notch activities and their in vivo significance during developmental and pathological processes.
Collapse
Affiliation(s)
- Pascal Heitzler
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| |
Collapse
|
344
|
Nicholson SC, Nicolay BN, Frolov MV, Moberg KH. Notch-dependent expression of the archipelago ubiquitin ligase subunit in the Drosophila eye. Development 2010; 138:251-60. [PMID: 21148181 DOI: 10.1242/dev.054429] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
archipelago (ago)/Fbw7 encodes a conserved protein that functions as the substrate-receptor component of a polyubiquitin ligase that suppresses tissue growth in flies and tumorigenesis in vertebrates. Ago/Fbw7 targets multiple proteins for degradation, including the G1-S regulator Cyclin E and the oncoprotein dMyc/c-Myc. Despite prominent roles in growth control, little is known about the signals that regulate Ago/Fbw7 abundance in developing tissues. Here we use the Drosophila eye as a model to identify developmental signals that regulate ago expression. We find that expression of ago mRNA and protein is induced by passage of the morphogenetic furrow (MF) and identify the hedgehog (hh) and Notch (N) pathways as elements of this inductive mechanism. Cells mutant for N pathway components, or hh-defective cells that express reduced levels of the Notch ligand Delta, fail to upregulate ago transcription in the region of the MF; reciprocally, ectopic N activation in eye discs induces expression of ago mRNA. A fragment of the ago promoter that contains consensus binding sites for the N pathway transcription factor Su(H) is bound by Su(H) and confers N-inducibility in cultured cells. The failure to upregulate ago in N pathway mutant cells correlates with accumulation of the SCF-Ago target Cyclin E in the area of the MF, and this is rescued by re-expression of ago. These data suggest a model in which N acts through ago to restrict levels of the pro-mitotic factor Cyclin E. This N-Ago-Cyclin E link represents a significant new cell cycle regulatory mechanism in the developing eye.
Collapse
Affiliation(s)
- Sarah C Nicholson
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
345
|
Mo JS, Ann EJ, Yoon JH, Jung J, Choi YH, Kim HY, Ahn JS, Kim SM, Kim MY, Hong JA, Seo MS, Lang F, Choi EJ, Park HS. Serum- and glucocorticoid-inducible kinase 1 (SGK1) controls Notch1 signaling by downregulation of protein stability through Fbw7 ubiquitin ligase. J Cell Sci 2010; 124:100-12. [PMID: 21147854 DOI: 10.1242/jcs.073924] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Notch is a transmembrane protein that acts as a transcriptional factor in the Notch signaling pathway for cell survival, cell death and cell differentiation. Notch1 and Fbw7 mutations both lead the activation of the Notch1 pathway and are found in the majority of patients with the leukemia T-ALL. However, little is known about the mechanisms and regulators that are responsible for attenuating the Notch signaling pathway through Fbw7. Here, we report that the serum- and glucocorticoid-inducible protein kinase SGK1 remarkably reduced the protein stability of the active form of Notch1 through Fbw7. The protein level and transcriptional activity of the Notch1 intracellular domain (Notch1-IC) were higher in SGK1-deficient cells than in SGK1 wild-type cells. Notch1-IC was able to form a trimeric complex with Fbw7 and SGK1, thereby SGK1 enhanced the protein degradation of Notch1-IC via a Fbw7-dependent proteasomal pathway. Furthermore, activated SGK1 phosphorylated Fbw7 at serine 227, an effect inducing Notch1-IC protein degradation and ubiquitylation. Moreover, accumulated dexamethasone-induced SGK1 facilitated the degradation of Notch1-IC through phosphorylation of Fbw7. Together our results suggest that SGK1 inhibits the Notch1 signaling pathway via phosphorylation of Fbw7.
Collapse
Affiliation(s)
- Jung-Soon Mo
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
346
|
The Notch signaling pathway: molecular basis of cell context dependency. Eur J Cell Biol 2010; 90:572-81. [PMID: 21126799 DOI: 10.1016/j.ejcb.2010.10.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Revised: 10/05/2010] [Accepted: 10/05/2010] [Indexed: 11/21/2022] Open
Abstract
Notch receptor signaling controls cell-fate specification, self-renewal, differentiation, proliferation and apoptosis throughout development and regeneration in all animal species studied to date. Its dysfunction causes several developmental defects and diseases in the adult. A key feature of Notch signaling is its remarkable cell-context dependency. In this review, we summarize the influences of the cellular context that regulate Notch activity and propose a model how the interplay between the cell-intrinsically established chromatin state and the cell-extrinsic signals that modify chromatin may select for Notch target accessibility and activation in different cellular contexts.
Collapse
|
347
|
RITA, a novel modulator of Notch signalling, acts via nuclear export of RBP-J. EMBO J 2010; 30:43-56. [PMID: 21102556 DOI: 10.1038/emboj.2010.289] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Accepted: 10/26/2010] [Indexed: 12/14/2022] Open
Abstract
The evolutionarily conserved Notch signal transduction pathway regulates fundamental cellular processes during embryonic development and in the adult. Ligand binding induces presenilin-dependent cleavage of the receptor and a subsequent nuclear translocation of the Notch intracellular domain (NICD). In the nucleus, NICD binds to the recombination signal sequence-binding protein J (RBP-J)/CBF-1 transcription factor to induce expression of Notch target genes. Here, we report the identification and functional characterization of RBP-J interacting and tubulin associated (RITA) (C12ORF52) as a novel RBP-J/CBF-1-interacting protein. RITA is a highly conserved 36 kDa protein that, most interestingly, binds to tubulin in the cytoplasm and shuttles rapidly between cytoplasm and nucleus. This shuttling RITA exports RBP-J/CBF-1 from the nucleus. Functionally, we show that RITA can reverse a Notch-induced loss of primary neurogenesis in Xenopus laevis. Furthermore, RITA is able to downregulate Notch-mediated transcription. Thus, we propose that RITA acts as a negative modulator of the Notch signalling pathway, controlling the level of nuclear RBP-J/CBF-1, where its amounts are limiting.
Collapse
|
348
|
Zhou ZD, Kumari U, Xiao ZC, Tan EK. Notch as a molecular switch in neural stem cells. IUBMB Life 2010; 62:618-23. [PMID: 20681026 DOI: 10.1002/iub.362] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Notch signaling pathway, originally discovered in Drosophila, impinges on a wide array of cellular processes including cell proliferation, cell differentiation, and cell apoptosis. Recent accumulating evidences implicated the important roles of Notch signal pathway in different aspects of stem cell biology of neural stem cell (NSC). In vivo and in vitro studies illustrated that Notch signal pathway could promote gliogenesis, inhibit premature neurogenesis, and be involved in self-renew of NSC. This short review summarizes the roles of the Notch signaling pathway on gliogenesis, neurogenesis, and self-renew of NSC and their underlying molecular mechanisms.
Collapse
Affiliation(s)
- Zhi-Dong Zhou
- National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore
| | | | | | | |
Collapse
|
349
|
Aster JC, Blacklow SC, Pear WS. Notch signalling in T-cell lymphoblastic leukaemia/lymphoma and other haematological malignancies. J Pathol 2010; 223:262-73. [PMID: 20967796 DOI: 10.1002/path.2789] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Revised: 09/10/2010] [Accepted: 09/16/2010] [Indexed: 12/21/2022]
Abstract
Notch receptors participate in a highly conserved signalling pathway that regulates normal development and tissue homeostasis in a context- and dose-dependent manner. Deregulated Notch signalling has been implicated in many diseases, but the clearest example of a pathogenic role is found in T-cell lymphoblastic leukaemia/lymphoma (T-LL), in which the majority of human and murine tumours have acquired mutations that lead to aberrant increases in Notch1 signalling. Remarkably, it appears that the selective pressure for Notch mutations is virtually unique among cancers to T-LL, presumably reflecting a special context-dependent role for Notch in normal T-cell progenitors. Nevertheless, there are some recent reports suggesting that Notch signalling has subtle, yet important roles in other forms of haematological malignancy as well. Here, we review the role of Notch signalling in various blood cancers, focusing on T-LL with an eye towards targeted therapeutics.
Collapse
Affiliation(s)
- Jon C Aster
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA.
| | | | | |
Collapse
|
350
|
Abstract
The Putzig (Pzg) protein is associated with the NURF nucleosome remodeling complex, thereby promoting Notch target gene expression. Our findings suggest a novel Pzg-NURF complex that is responsible for the epigenetic regulation of Notch target genes. Drosophila putzig was identified as a member of the TRF2–DREF complex that is involved in core promoter selection. Additionally, putzig regulates Notch signaling, however independently of DREF. Here, we show that Putzig associates with the NURF complex. Loss of any NURF component including the NURF-specific subunit Nurf 301 impedes binding of Putzig to Notch target genes, suggesting that NURF recruits Putzig to these sites. Accordingly, Putzig can be copurified with any NURF member. Moreover, Nurf 301 mutants show reduced Notch target gene activity and enhance Notch mutant phenotypes. These data suggest a novel Putzig–NURF chromatin complex required for epigenetic activation of Notch targets.
Collapse
Affiliation(s)
- Sabrina J Kugler
- Institute of Genetics, University of Hohenheim, 70599 Stuttgart, Germany
| | | |
Collapse
|