101
|
Buas MF, Kadesch T. Regulation of skeletal myogenesis by Notch. Exp Cell Res 2010; 316:3028-33. [PMID: 20452344 DOI: 10.1016/j.yexcr.2010.05.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Accepted: 05/03/2010] [Indexed: 12/11/2022]
Abstract
Notch signaling has emerged as a key player in skeletal muscle development and regeneration. Simply stated, Notch signaling inhibits differentiation. Accordingly, fine-tuning the pathway is essential for proper muscle homeostasis. This review will address various aspects of Notch signaling, including our current views of the core pathway, its effects in muscle, its interactions with other signaling pathways, and its relationship with ageing.
Collapse
Affiliation(s)
- Matthew F Buas
- Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6145, USA
| | | |
Collapse
|
102
|
Yin L, Velazquez OC, Liu ZJ. Notch signaling: emerging molecular targets for cancer therapy. Biochem Pharmacol 2010; 80:690-701. [PMID: 20361945 DOI: 10.1016/j.bcp.2010.03.026] [Citation(s) in RCA: 126] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Revised: 03/23/2010] [Accepted: 03/24/2010] [Indexed: 02/07/2023]
Abstract
The Notch signaling pathway is a highly conserved developmental pathway, which plays a critical role in cell-fate decision, tissue patterning and morphogenesis. There is increasing evidence that this pathway is dysregulated in a variety of malignancies, and can behave as either an oncogene or a tumor suppressor depending upon cell context. This review highlights the current evidence for aberration of the Notch signaling pathway in a wide range of tumors from hematological cancers, such as leukemia and lymphoma through to skin, breast, lung, pancreas, colon and brain tumors. It proposes that the Notch signaling pathway may represent novel therapeutic targets and will be a welcome asset to the cancer therapeutic arena.
Collapse
Affiliation(s)
- Ling Yin
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | | | | |
Collapse
|
103
|
Delta-Notch signaling is involved in the segregation of the three germ layers in Xenopus laevis. Dev Biol 2010; 339:477-92. [PMID: 20079726 DOI: 10.1016/j.ydbio.2010.01.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Revised: 01/06/2010] [Accepted: 01/07/2010] [Indexed: 11/20/2022]
Abstract
In vertebrates, the induction of the three germ layers (ectoderm, mesoderm and endoderm) has been extensively studied, but less is known about how they segregate. Here, we investigated whether Delta-Notch signaling is involved in this process. Activating the pathway in the marginal zone with Notch(ICD) resulted in an expansion of endodermal and neural ectoderm precursors, leaving a thinner mesodermal ring around the blastopore at gastrula stage, when germ layers are segregated. On the other hand, when the pathway was blocked with Delta-1(STU) or with an antisense morpholino oligonucleotide against Notch, the pan-mesodermal brachyury (bra) domain was expanded and the neural border was moved animalwards. Strikingly, the suprablastoporal endoderm was either expanded when Delta-1 signaling was blocked, or reduced after the general knock-down of Notch. In addition, either activating or blocking the pathway delays the blastopore closure. We conclude that the process of delimiting the three germ layers requires Notch signaling, which may be finely regulated by ligands and/or involve non-canonical components of the pathway. Moreover, Notch activity must be modulated at appropriate levels during this process in order to keep normal morphogenetic movements during gastrulation.
Collapse
|
104
|
Notch signaling in pulmonary hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 661:279-98. [PMID: 20204737 DOI: 10.1007/978-1-60761-500-2_18] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Proteins of the Notch receptor family are cell surface receptors that transduce signals between neighboring cells. The Notch signaling pathway is highly evolutionarily conserved and critical for cell fate determination during embryogenesis and early postnatal life, including many aspects of vascular development. The interaction of Notch receptor with its membrane-bound ligands leads to cleavage of the receptor into an intracellular domain that translocates to the nucleus and activates the transcription factor, C-promoter binding factor 1 (CBF1; also known as Recombination signal-binding protein for immunoglobulin kappa J region, RBPJ). To date, four Notch receptors have been characterized in humans. Of these, Notch3 is expressed only in arterial smooth muscle cells in the human. The functional importance of Notch3 signaling in human vascular smooth muscle cells has been recognized. Notch3 receptor signaling has been shown in several model systems to control vascular smooth muscle cell proliferation and maintain smooth muscle cells in an undifferentiated state. This review focuses on recent findings of the role of Notch3 in regulating vascular smooth muscle cell behavior and phenotype and discusses the potential role of Notch3 signaling in the genesis of pulmonary arterial hypertension.
Collapse
|
105
|
|
106
|
Abstract
Stem cells are rare and unique precursor cells that participate in the building and rebuilding of tissues and organs during embryogenesis, postnatal growth, and injury repair. Stem cells are distinctively endowed with the ability to both self-renew and differentiate, such that they can replenish the stem cell pool while continuing to produce the differentiated daughter cells that are essential for tissue function. Stem cell self-renewal/differentiation decisions must be carefully controlled during organogenesis, tissue homeostasis, and regeneration, as failure in stem cell maintenance or activation can lead to progressive tissue wasting, while unchecked self-renewal is a hallmark of many cancers. Here, we review evidence implicating the Notch signaling pathway, an evolutionarily conserved cell fate determinant with widespread roles in a variety of tissues and organisms, as a crucial regulator of stem cell behavior. As discussed below, this pathway plays varied and critical roles at multiple stages of organismal development, in lineage-specific differentiation of pluripotent embryonic stem cells, and in controlling stem cell numbers and activity in the context of age-related tissue degeneration, injury-induced tissue repair, and malignancy.
Collapse
|
107
|
Akiho M, Nakashima H, Sakata M, Yamasa Y, Yamaguchi A, Sakuma K. Expression profile of Notch-1 in mechanically overloaded plantaris muscle of mice. Life Sci 2009; 86:59-65. [PMID: 19945468 DOI: 10.1016/j.lfs.2009.11.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Revised: 10/29/2009] [Accepted: 11/16/2009] [Indexed: 10/20/2022]
Abstract
AIM We investigated the expression pattern of Notch-1 in normal and hypertrophied plantaris muscle of mice. MAIN METHODS We performed immunofluorescence of both Notch-1 and the Notch-1-linking molecules. KEY FINDINGS Immunofluorescence labeling revealed Notch-1 protein in Pax7-positive satellite cells during days 2-6. We observed clear co-localization between Notch-1 and myogenin (4.9+/-1.3%) in the hypertrophied muscle at 4days. Several mononuclei (possibly satellite cells) possessed both Notch-1 and Foxo1 in the plantaris muscle subjected to mechanical overloading (4.1+/-1.2%). SIGNIFICANCE Notch-1 may play an important role in the maintenance of quiescent satellite cells.
Collapse
Affiliation(s)
- Mai Akiho
- Research Center for Physical Fitness, Sports and Health, Toyohashi, University of Technology, 1-1 Hibarigaoka, Tenpaku-cho, Toyohashi, 441-8580, Japan
| | | | | | | | | | | |
Collapse
|
108
|
Li X, Zhang X, Leathers R, Makino A, Huang C, Parsa P, Macias J, Yuan JXJ, Jamieson SW, Thistlethwaite PA. Notch3 signaling promotes the development of pulmonary arterial hypertension. Nat Med 2009; 15:1289-97. [PMID: 19855400 PMCID: PMC2780347 DOI: 10.1038/nm.2021] [Citation(s) in RCA: 278] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2009] [Accepted: 08/24/2009] [Indexed: 01/02/2023]
Abstract
Notch receptor signaling is implicated in controlling smooth muscle cell proliferation and in maintaining smooth muscle cells in an undifferentiated state. Pulmonary arterial hypertension is characterized by excessive vascular resistance, smooth muscle cell proliferation in small pulmonary arteries, leading to elevation of pulmonary vascular resistance, right ventricular failure and death. Here we show that human pulmonary hypertension is characterized by overexpression of NOTCH3 in small pulmonary artery smooth muscle cells and that the severity of disease in humans and rodents correlates with the amount of NOTCH3 protein in the lung. We further show that mice with homozygous deletion of Notch3 do not develop pulmonary hypertension in response to hypoxic stimulation and that pulmonary hypertension can be successfully treated in mice by administration of N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT), a gamma-secretase inhibitor that blocks activation of Notch3 in smooth muscle cells. We show a mechanistic link from NOTCH3 receptor signaling through the Hairy and enhancer of Split-5 (HES-5) protein to smooth muscle cell proliferation and a shift to an undifferentiated smooth muscle cell phenotype. These results suggest that the NOTCH3-HES-5 signaling pathway is crucial for the development of pulmonary arterial hypertension and provide a target pathway for therapeutic intervention.
Collapse
MESH Headings
- Animals
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Cell Proliferation/drug effects
- Dipeptides/pharmacology
- Dipeptides/therapeutic use
- Disease Models, Animal
- Enzyme Inhibitors/pharmacology
- Enzyme Inhibitors/therapeutic use
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/physiology
- Humans
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypoxia/drug therapy
- Hypoxia/physiopathology
- In Vitro Techniques
- Lung/metabolism
- Lung/pathology
- Mice
- Mice, Knockout
- Microscopy, Electron, Transmission/methods
- Myocytes, Smooth Muscle
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Pulmonary Artery/ultrastructure
- RNA, Messenger/metabolism
- Rats
- Receptor, Notch3
- Receptors, Notch/deficiency
- Receptors, Notch/metabolism
- Repressor Proteins/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Signal Transduction/physiology
- Time Factors
Collapse
Affiliation(s)
- Xiaodong Li
- Division of Cardiothoracic Surgery, University of California, San Diego, San Diego, CA 92103-8892
| | - Xiaoxue Zhang
- Division of Cardiothoracic Surgery, University of California, San Diego, San Diego, CA 92103-8892
| | - Robin Leathers
- Division of Cardiothoracic Surgery, University of California, San Diego, San Diego, CA 92103-8892
| | - Ayako Makino
- Division of Pulmonary and Critical Care Medicine, University of California, San Diego, San Diego, CA 92103-8892
| | - Chengqun Huang
- Division of Cardiothoracic Surgery, University of California, San Diego, San Diego, CA 92103-8892
| | - Pouria Parsa
- Division of Cardiothoracic Surgery, University of California, San Diego, San Diego, CA 92103-8892
| | - Jesus Macias
- Department of Pathology, University of California, San Diego, San Diego, CA 92103-8892
| | - Jason X.-J. Yuan
- Division of Pulmonary and Critical Care Medicine, University of California, San Diego, San Diego, CA 92103-8892
| | - Stuart W. Jamieson
- Division of Cardiothoracic Surgery, University of California, San Diego, San Diego, CA 92103-8892
| | | |
Collapse
|
109
|
Jeong HW, Jeon US, Koo BK, Kim WY, Im SK, Shin J, Cho Y, Kim J, Kong YY. Inactivation of Notch signaling in the renal collecting duct causes nephrogenic diabetes insipidus in mice. J Clin Invest 2009; 119:3290-300. [PMID: 19855135 DOI: 10.1172/jci38416] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Accepted: 08/26/2009] [Indexed: 01/06/2023] Open
Abstract
The heterogeneous cellular composition of the mammalian renal collecting duct enables regulation of fluid, electrolytes, and acid-base homeostasis, but the molecular mechanism of its development has yet to be elucidated. The Notch signaling pathway is involved in cell fate determination and has been implicated in proximal-distal patterning in the mammalian kidney. To investigate the role of Notch signaling in renal collecting duct development, we generated mice in which Mind bomb-1 (Mib1), an E3 ubiquitin ligase required for the initiation of Notch signaling, was specifically inactivated in the ureteric bud of the developing kidney. Mice lacking Mib1 in the renal collecting duct displayed increased urinary production, decreased urinary osmolality, progressive hydronephrosis, sodium wasting, and a severe urinary concentrating defect manifested as nephrogenic diabetes insipidus. Histological analysis revealed a diminished number of principal cells and corresponding increase in the number of intercalated cells. Transgenic overexpression of Notch intracellular domain reversed the altered cellular composition of mutant renal collecting duct, with principal cells occupying the entire region. Our data demonstrate that Notch signaling is required for the development of the mammalian renal collecting duct and principal cell differentiation and indicate that pathway dysregulation may contribute to distal renal tubular disorders.
Collapse
Affiliation(s)
- Hyun-Woo Jeong
- Department of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
110
|
Gang EJ, Darabi R, Bosnakovski D, Xu Z, Kamm KE, Kyba M, Perlingeiro RCR. Engraftment of mesenchymal stem cells into dystrophin-deficient mice is not accompanied by functional recovery. Exp Cell Res 2009; 315:2624-36. [PMID: 19460366 DOI: 10.1016/j.yexcr.2009.05.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Revised: 05/05/2009] [Accepted: 05/06/2009] [Indexed: 02/08/2023]
Abstract
Mesenchymal stem cell preparations have been proposed for muscle regeneration in musculoskeletal disorders. Although MSCs have great in vitro expansion potential and possess the ability to differentiate into several mesenchymal lineages, myogenesis has proven to be much more difficult to induce. We have recently demonstrated that Pax3, the master regulator of the embryonic myogenic program, enables the in vitro differentiation of a murine mesenchymal stem cell line (MSCB9-Pax3) into myogenic progenitors. Here we show that injection of these cells into cardiotoxin-injured muscles of immunodeficient mice leads to the development of muscle tumors, resembling rhabdomyosarcomas. We then extended these studies to primary human mesenchymal stem cells (hMSCs) isolated from bone marrow. Upon genetic modification with a lentiviral vector encoding PAX3, hMSCs activated the myogenic program as demonstrated by expression of myogenic regulatory factors. Upon transplantation, the PAX3-modified MSCs did not generate rhabdomyosarcomas but rather, resulted in donor-derived myofibers. These were found at higher frequency in PAX3-transduced hMSCs than in mock-transduced MSCs. Nonetheless, neither engraftment of PAX3-modified or unmodified MSCs resulted in improved contractility. Thus these findings suggest that limitations remain to be overcome before MSC preparations result in effective treatment for muscular dystrophies.
Collapse
Affiliation(s)
- Eun Ji Gang
- Department of Developmental Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9133, USA
| | | | | | | | | | | | | |
Collapse
|
111
|
Müller P, Crofts JD, Newman BS, Bridgewater LC, Lin CY, Gustafsson JA, Ström A. SOX9 mediates the retinoic acid-induced HES-1 gene expression in human breast cancer cells. Breast Cancer Res Treat 2009; 120:317-26. [PMID: 19322650 DOI: 10.1007/s10549-009-0381-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2008] [Accepted: 03/14/2009] [Indexed: 12/16/2022]
Abstract
We have previously shown that the anti-proliferative effect of retinoic acid in human breast cancer cell line MCF-7 is dependent on HES-1 expression. Here we show that retinoic acid induces HES-1 expression via upregulation of transcription factor SOX9. By expressing a dominant negative form of SOX9, disrupting endogenous SOX9 activity, the retinoic acid-induced HES-1 mRNA expression was inhibited. We found an enhancer regulating HES-1 expression: two SOX9 binding sites upstream of the HES-1 gene that were capable of binding SOX9 in vitro. By performing chromatin immunoprecipitation, we showed that SOX9 binding to the HES-1 enhancer was induced by retinoic acid in vivo. In reporter assays, transfection of a SOX9 expression plasmid increased the activity of the HES-1 enhancer. The enhancer responded to retinoic acid; furthermore, the expression of a dominant negative SOX9 abolished this response. Taken together, we present here a novel transcriptional mechanism in regulating hormone-dependent cancer cell proliferation.
Collapse
Affiliation(s)
- Patrick Müller
- Department of Biosciences and Nutrition, Karolinska Institutet, Novum, 141 57, Huddinge, Sweden.
| | | | | | | | | | | | | |
Collapse
|
112
|
Kuang S, Gillespie MA, Rudnicki MA. Niche regulation of muscle satellite cell self-renewal and differentiation. Cell Stem Cell 2009; 2:22-31. [PMID: 18371418 DOI: 10.1016/j.stem.2007.12.012] [Citation(s) in RCA: 349] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Muscle satellite cells have been shown to be a heterogeneous population of committed myogenic progenitors and noncommitted stem cells. This hierarchical composition of differentiating progenitors and self-renewable stem cells assures the extraordinary regenerative capacity of skeletal muscles. Recent studies have revealed a role for asymmetric division in satellite cell maintenance and offer novel insights into the regulation of satellite cell function by the niche. A thorough understanding of the molecular regulation and cell fate determination of satellite cells and other potential stem cells resident in muscle is essential for successful stem cell-based therapies to treat muscular diseases.
Collapse
Affiliation(s)
- Shihuan Kuang
- The Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Health Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
| | | | | |
Collapse
|
113
|
Abstract
Notch signaling is critical for skeletal muscle development and regeneration, permitting the expansion of progenitor cells by preventing premature differentiation. We have interrogated the pathways through which ligand-mediated signaling inhibits myogenesis by identifying Notch target genes and assessing their impact on differentiation in vitro. Notch activation led to the robust induction of the transcriptional repressors Hey1 and HeyL in myoblasts, but only constitutive expression of Hey1 blocked myogenesis. siRNA-mediated knockdown of Hey1 had no effect on Notch's ability to inhibit differentiation, suggesting the existence of additional, possibly redundant pathways. We identified 82 genes whose expression was activated when C2C12 myoblasts were cultured in the presence of the Notch ligand Dll4. One of these, MyoR, is a novel Notch-responsive gene, whose protein product is known to repress myogenesis in vitro. siRNA-mediated knockdown of MyoR alone, or in combination with Hey1, was also ineffective at rescuing differentiation in the presence of Dll4. Our data support a model in which Notch signaling inhibits myogenesis through multiple pathways, two of which are defined by the Notch target genes Hey1 and MyoR.
Collapse
Affiliation(s)
- Matthew F. Buas
- Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6145
| | | | - Tom Kadesch
- Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6145
| |
Collapse
|
114
|
Nakahara J, Kanekura K, Nawa M, Aiso S, Suzuki N. Abnormal expression of TIP30 and arrested nucleocytoplasmic transport within oligodendrocyte precursor cells in multiple sclerosis. J Clin Invest 2009; 119:169-81. [PMID: 19104151 PMCID: PMC2613458 DOI: 10.1172/jci35440] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Accepted: 10/15/2008] [Indexed: 12/18/2022] Open
Abstract
Oligodendrocyte precursor cells (OPCs) persist near the demyelinated axons arising in MS but inefficiently differentiate into oligodendrocytes and remyelinate these axons. The pathogenesis of differentiation failure remains elusive. We initially hypothesized that injured axons fail to present Contactin, a positive ligand for the oligodendroglial Notch1 receptor to induce myelination, and thus tracked axoglial Contactin/Notch1 signaling in situ, using immunohistochemistry in brain tissue from MS patients containing chronic demyelinated lesions. Instead, we found that Contactin was saturated on demyelinated axons, Notch1-positive OPCs accumulated in Contactin-positive lesions, and the receptor was engaged, as demonstrated by cleavage to Notch1-intracellular domain (NICD). However, nuclear translocalization of NICD, required for myelinogenesis, was virtually absent in these cells. NICD and related proteins carrying nuclear localization signals were associated with the nuclear transporter Importin but were trapped in the cytoplasm. Abnormal expression of TIP30, a direct inhibitor of Importin, was observed in these OPCs. Overexpression of TIP30 in a rat OPC cell line resulted in cytoplasmic entrapment of NICD and arrest of differentiation upon stimulation with Contactin-Fc. Our results suggest that extracellular inhibitory factors as well as an intrinsic nucleocytoplasmic transport blockade within OPCs may be involved in the pathogenesis of remyelination failure in MS.
Collapse
Affiliation(s)
- Jin Nakahara
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
115
|
KyoT3, an isoform of murine FHL1, associates with the transcription factor RBP-J and represses the RBP-J-mediated transactivation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2008; 1779:805-10. [DOI: 10.1016/j.bbagrm.2008.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2007] [Revised: 07/31/2008] [Accepted: 08/01/2008] [Indexed: 11/23/2022]
|
116
|
Lin KT, Sloniowski S, Ethell DW, Ethell IM. Ephrin-B2-induced cleavage of EphB2 receptor is mediated by matrix metalloproteinases to trigger cell repulsion. J Biol Chem 2008; 283:28969-79. [PMID: 18713744 PMCID: PMC2570862 DOI: 10.1074/jbc.m804401200] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2008] [Revised: 08/05/2008] [Indexed: 01/03/2023] Open
Abstract
EphB receptors provide crucial adhesive and repulsive signals during cell migration and axon guidance, but it is unclear how they switch between these opposing responses. Here we provide evidence of an important role for matrix metalloproteinases (MMPs) in repulsive EphB2 signaling. We found that EphB2 is cleaved by MMPs both in vitro and in vivo, and that this cleavage is induced by interaction with its ligand ephrin-B2. Our findings demonstrate that MMP-2/MMP-9-specific inhibition or cleavage-resistant mutations in the ectodomain of EphB2 can prevent EphB2-mediated cell-cell repulsion in HEK293 cells, and block ephrin-B1-induced growth cone withdrawal in cultured hippocampal neurons. Transient expression of wtEphB2, but not noncleavable EphB2-4/5 mutant, restored ephrin-B1-induced growth cone collapse and withdrawal in EphB-deficient neurons. The inhibition of EphB2 cleavage also had potent regulatory effects on EphB2 activity. This study provides the first evidence that MMP-mediated cleavage of EphB2 is induced by receptor-ligand interactions at the cell surface and that this event triggers cell-repulsive responses.
Collapse
Affiliation(s)
- Kai-Ti Lin
- Division of Biomedical Sciences, University of California, Riverside, California 92521-0121, USA
| | | | | | | |
Collapse
|
117
|
McElhinny AS, Li JL, Wu L. Mastermind-like transcriptional co-activators: emerging roles in regulating cross talk among multiple signaling pathways. Oncogene 2008; 27:5138-47. [PMID: 18758483 DOI: 10.1038/onc.2008.228] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A family of Mastermind-like (MAML) genes encodes critical transcriptional co-activators for Notch signaling, an evolutionarily conserved pathway with numerous roles in both development and human diseases. Notch receptors are cleaved upon ligand engagement and the intracellular domain of Notch shuttles to the nucleus. MAMLs form a functional DNA-binding complex with the cleaved Notch receptor and the transcription factor CSL, thereby regulating transcriptional events that are specific to the Notch pathway. Here, we review recent studies that have utilized molecular, cellular and physiological model system strategies to reveal the pivotal roles of the MAML proteins in Notch signaling. Unexpectedly, however, emerging evidence implicate MAML proteins as exciting key transcriptional co-activators in other signal transduction pathways including: muscle differentiation and myopathies (MEF2C), tumor suppressor pathway (p53) and colon carcinoma survival (beta-catenin). Thus, the MAML family appears to function in transcriptional co-activation in a multitude of cellular processes. It is hypothesized that MAML proteins mediate cross-talk among the various signaling pathways and the diverse activities of the MAML proteins converge to impact normal biological processes and human diseases, including cancers.
Collapse
|
118
|
Corbin JG, Gaiano N, Juliano SL, Poluch S, Stancik E, Haydar TF. Regulation of neural progenitor cell development in the nervous system. J Neurochem 2008; 106:2272-87. [PMID: 18819190 PMCID: PMC2640107 DOI: 10.1111/j.1471-4159.2008.05522.x] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The mammalian telencephalon, which comprises the cerebral cortex, olfactory bulb, hippocampus, basal ganglia, and amygdala, is the most complex and intricate region of the CNS. It is the seat of all higher brain functions including the storage and retrieval of memories, the integration and processing of sensory and motor information, and the regulation of emotion and drive states. In higher mammals such as humans, the telencephalon also governs our creative impulses, ability to make rational decisions, and plan for the future. Despite its massive complexity, exciting work from a number of groups has begun to unravel the developmental mechanisms for the generation of the diverse neural cell types that form the circuitry of the mature telencephalon. Here, we review our current understanding of four aspects of neural development. We first begin by providing a general overview of the broad developmental mechanisms underlying the generation of neuronal and glial cell diversity in the telencephalon during embryonic development. We then focus on development of the cerebral cortex, the most complex and evolved region of the brain. We review the current state of understanding of progenitor cell diversity within the cortical ventricular zone and then describe how lateral signaling via the Notch-Delta pathway generates specific aspects of neural cell diversity in cortical progenitor pools. Finally, we review the signaling mechanisms required for development, and response to injury, of a specialized group of cortical stem cells, the radial glia, which act both as precursors and as migratory scaffolds for newly generated neurons.
Collapse
Affiliation(s)
- Joshua G. Corbin
- Center for Neuroscience Research, Children’s National Medical Center, Washington, DC 20010, USA
| | - Nicholas Gaiano
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | - Sylvie Poluch
- Program in Neuroscience, USUHS, Bethesda, Maryland 20814
| | - Elizabeth Stancik
- Center for Neuroscience Research, Children’s National Medical Center, Washington, DC 20010, USA
| | - Tarik F. Haydar
- Center for Neuroscience Research, Children’s National Medical Center, Washington, DC 20010, USA
| |
Collapse
|
119
|
Simon MC, Keith B. The role of oxygen availability in embryonic development and stem cell function. Nat Rev Mol Cell Biol 2008; 9:285-96. [PMID: 18285802 PMCID: PMC2876333 DOI: 10.1038/nrm2354] [Citation(s) in RCA: 718] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Low levels of oxygen (O2) occur naturally in developing embryos. Cells respond to their hypoxic microenvironment by stimulating several hypoxia-inducible factors (and other molecules that mediate O2 homeostasis), which then coordinate the development of the blood, vasculature, placenta, nervous system and other organs. Furthermore, embryonic stem and progenitor cells frequently occupy hypoxic 'niches' and low O2 regulates their differentiation. Recent work has revealed an important link between factors that are involved in regulating stem and progenitor cell behaviour and hypoxia-inducible factors, which provides a molecular framework for the hypoxic control of differentiation and cell fate. These findings have important implications for the development of therapies for tissue regeneration and disease.
Collapse
Affiliation(s)
- M Celeste Simon
- Howard Hughes Medical Institute, Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, 421 Curie Boulevard, Philadelphia, Pennsylvania 19104, USA.
| | | |
Collapse
|
120
|
Bai S, Kopan R, Zou W, Hilton MJ, Ong CT, Long F, Ross FP, Teitelbaum SL. NOTCH1 regulates osteoclastogenesis directly in osteoclast precursors and indirectly via osteoblast lineage cells. J Biol Chem 2008; 283:6509-18. [PMID: 18156632 DOI: 10.1074/jbc.m707000200] [Citation(s) in RCA: 189] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
NOTCH signaling is a key regulator of cell fate decisions in prenatal skeletal development and is active during adult tissue renewal. In addition, its association with neoplasia suggests that it is a candidate therapeutic target. We find that attenuated NOTCH signaling enhances osteoclastogenesis and bone resorption in vitro and in vivo by a combination of molecular mechanisms. First, deletion of Notch1-3 in bone marrow macrophages directly promotes their commitment to the osteoclast phenotype. These osteoclast precursors proliferate more rapidly than the wild type in response to macrophage colony-stimulating factor and are sensitized to RANKL and macrophage colony-stimulating factor, undergoing enhanced differentiation in response to low doses of either cytokine. Conforming with a role for NOTCH in this process, presentation of the NOTCH ligand JAGGED1 blunts the capacity of wild-type bone marrow macrophages to become osteoclasts. Combined, these data establish that NOTCH suppresses osteoclastogenesis via ligand-mediated receptor activation. Although NOTCH1 and NOTCH3 collaborate in regulating osteoclast formation, NOTCH1 is the dominant paralog. In addition, NOTCH1 deficiency promotes osteoclastogenesis indirectly by enhancing the ability of osteoblast lineage cells to stimulate osteoclastogenesis. This is achieved by decreasing the osteoprotegerin/RANKL expression ratio. Thus, NOTCH1 acts as a net inhibitor of bone resorption, exerting its effect both directly in osteoclast precursors and indirectly via osteoblast lineage cells. These observations raise caution that therapeutic inhibition of NOTCH signaling may adversely accelerate bone loss in humans.
Collapse
Affiliation(s)
- Shuting Bai
- Departments of Pathology and Immunology, Molecular Biology and Pharmacology, and Internal Medicine, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
121
|
Zheng X, Linke S, Dias JM, Zheng X, Gradin K, Wallis TP, Hamilton BR, Gustafsson M, Ruas JL, Wilkins S, Bilton RL, Brismar K, Whitelaw ML, Pereira T, Gorman JJ, Ericson J, Peet DJ, Lendahl U, Poellinger L. Interaction with factor inhibiting HIF-1 defines an additional mode of cross-coupling between the Notch and hypoxia signaling pathways. Proc Natl Acad Sci U S A 2008; 105:3368-73. [PMID: 18299578 PMCID: PMC2265116 DOI: 10.1073/pnas.0711591105] [Citation(s) in RCA: 210] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Indexed: 11/18/2022] Open
Abstract
Cells adapt to hypoxia by a cellular response, where hypoxia-inducible factor 1alpha (HIF-1alpha) becomes stabilized and directly activates transcription of downstream genes. In addition to this "canonical" response, certain aspects of the pathway require integration with Notch signaling, i.e., HIF-1alpha can interact with the Notch intracellular domain (ICD) to augment the Notch downstream response. In this work, we demonstrate an additional level of complexity in this cross-talk: factor-inhibiting HIF-1 (FIH-1) regulates not only HIF activity, but also the Notch signaling output and, in addition, plays a role in how Notch signaling modulates the hypoxic response. We show that FIH-1 hydroxylates Notch ICD at two residues (N(1945) and N(2012)) that are critical for the function of Notch ICD as a transactivator within cells and during neurogenesis and myogenesis in vivo. FIH-1 negatively regulates Notch activity and accelerates myogenic differentiation. In its modulation of the hypoxic response, Notch ICD enhances recruitment of HIF-1alpha to its target promoters and derepresses HIF-1alpha function. Addition of FIH-1, which has a higher affinity for Notch ICD than for HIF-1alpha, abrogates the derepression, suggesting that Notch ICD sequesters FIH-1 away from HIF-1alpha. In conclusion, the data reveal posttranslational modification of the activated form of the Notch receptor and an intricate mode of cross-coupling between the Notch and hypoxia signaling pathways.
Collapse
Affiliation(s)
- Xiaofeng Zheng
- Departments of *Cell and Molecular Biology and
- Molecular Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Sarah Linke
- School of Molecular and Biomedical Science and Australian Research Council, Special Research Centre for Molecular Genetics of Development, University of Adelaide, Adelaide SA 5005, Australia; and
| | | | | | | | - Tristan P. Wallis
- Protein Discovery Centre, Queensland Institute of Medical Research, Brisbane, Queensland 4029, Australia
| | - Brett R. Hamilton
- Protein Discovery Centre, Queensland Institute of Medical Research, Brisbane, Queensland 4029, Australia
| | | | | | - Sarah Wilkins
- School of Molecular and Biomedical Science and Australian Research Council, Special Research Centre for Molecular Genetics of Development, University of Adelaide, Adelaide SA 5005, Australia; and
| | - Rebecca L. Bilton
- School of Molecular and Biomedical Science and Australian Research Council, Special Research Centre for Molecular Genetics of Development, University of Adelaide, Adelaide SA 5005, Australia; and
| | - Kerstin Brismar
- Molecular Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Murray L. Whitelaw
- School of Molecular and Biomedical Science and Australian Research Council, Special Research Centre for Molecular Genetics of Development, University of Adelaide, Adelaide SA 5005, Australia; and
| | | | - Jeffrey J. Gorman
- Protein Discovery Centre, Queensland Institute of Medical Research, Brisbane, Queensland 4029, Australia
| | | | - Daniel J. Peet
- School of Molecular and Biomedical Science and Australian Research Council, Special Research Centre for Molecular Genetics of Development, University of Adelaide, Adelaide SA 5005, Australia; and
| | | | | |
Collapse
|
122
|
Maroto M, Iimura T, Dale JK, Bessho Y. BHLH proteins and their role in somitogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 638:124-39. [PMID: 21038774 DOI: 10.1007/978-0-387-09606-3_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The most obvious manifestation of the existence of a segmented, or metameric, body plan in vertebrate embryos is seen during the formation of the somites. Somites are transient embryonic structures formed in a progressive manner from a nonsegmented mesoderm in a highly regulated process called somitogenesis. As development proceeds different compartments are formed within each somite and these progressively follow a variety of differentiation programs to form segmented organs, such as the different bones that make the axial skeleton, body skeletal muscles and part of the dermis. Transcription factors from the basic helix-loop-helix (bHLH) protein family have been described to be implicated in each of the processes involved in somite formation. bHLH proteins are a family of transcription factors characterized by the presence of a DNA binding domain and a dimerization motif that consists of a basic region adjacent to an amphipathic helix, a loop and a second amphipathic helix. In this chapter we will review a number of bHLH proteins known to play a role in somitogenesis.
Collapse
Affiliation(s)
- Miguel Maroto
- College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, Scotland, UK.
| | | | | | | |
Collapse
|
123
|
Michie AM, Chan AC, Ciofani M, Carleton M, Lefebvre JM, He Y, Allman DM, Wiest DL, Zúñiga-Pflücker JC, Izon DJ. Constitutive Notch signalling promotes CD4 CD8 thymocyte differentiation in the absence of the pre-TCR complex, by mimicking pre-TCR signals. Int Immunol 2007; 19:1421-30. [PMID: 17981791 DOI: 10.1093/intimm/dxm113] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Notch1 signalling is essential for the commitment of multipotent lymphocyte precursors towards the alphabeta T-cell lineage and plays an important role in regulating beta-selection in CD4(-)CD8(-) double-negative (DN) thymocytes. However, the role played by Notch in promoting the development of CD4(+)CD8(+) double-positive (DP) thymocytes is poorly characterized. Here, we demonstrate that the introduction of a constitutively active Notch1 (ICN1) construct into RAG(-/-) lymphocyte precursors resulted in the generation of DP thymocytes in in vitro T-cell culture systems. Notably, developmental rescue was dependent not only on the presence of an intact Notch1 RAM domain but also on Delta-like signals, as ICN1-induced DP development in RAG(-/-) thymocytes occurred within an intact thymus or in OP9-DL1 co-cultures, but not in OP9-control co-cultures. Interestingly, ICN1 expression in SLP-76(-/-) precursors resulted in only a minimal developmental rescue to the immature CD8(+) single-positive stage, suggesting that Notch is utilizing the same signalling pathway as the pre-TCR complex. In support of this, ICN1 introduction resulted in the activation of the ERK-MAPK-signalling cascade in RAG(-/-) thymocytes. Taken together, these studies demonstrate that constitutive Notch signalling can bypass beta-selection during early T-cell development by inducing pre-TCR-like signals within a T-cell-promoting environment.
Collapse
Affiliation(s)
- Alison M Michie
- Division of Cancer Sciences and Molecular Pathology, Section of Experimental Haematology, Royal Infirmary, University of Glasgow, Glasgow, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
124
|
Message in a bottle: long-range retrograde signaling in the nervous system. Trends Cell Biol 2007; 17:519-28. [PMID: 18029183 DOI: 10.1016/j.tcb.2007.09.003] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2007] [Revised: 09/07/2007] [Accepted: 09/07/2007] [Indexed: 01/08/2023]
Abstract
In many regions of the nervous system, signals produced by target cells and surrounding glia or in response to in jury are received at axon terminals and then retrogradely propagated to cell bodies where they regulate gene transcription and other cellular processes required for development and adult function. The cellular and molecular mechanisms of axonal retrograde signaling in neurons have traditionally been studied in the context of survival signals provided by target-derived neurotrophic factors, in which signaling endosomes containing endocytosed ligand-receptor complexes and downstream effectors are retrogradely tra nsported by dynein motors. In recent years, this notion has been refined and additional mechanisms for long-range retrograde signaling in axons have been described. This article discusses some outstanding issues in the signaling endosome hypothesis as well as recent findings suggesting the existence of a variety of mechanisms for the retrograde propagation of signals in the nervous system.
Collapse
|
125
|
Hodkinson PS, Elliott PA, Lad Y, McHugh BJ, MacKinnon AC, Haslett C, Sethi T. Mammalian NOTCH-1 Activates β1 Integrins via the Small GTPase R-Ras. J Biol Chem 2007; 282:28991-29001. [PMID: 17664272 DOI: 10.1074/jbc.m703601200] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Notch is a central regulator of important cell fate decisions. Notch activation produces diverse cellular effects suggesting the presence of context-dependent control mechanisms. Genetic studies have demonstrated that Notch and integrin mutations have related phenotypes in key developmental processes such as vascular development and somitogenesis. We show that the intracellular domain of mammalian Notch-1 activates integrins without affecting integrin expression. Integrin activation is dependent on gamma-secretase-mediated intramembranous cleavage of membrane-bound Notch releasing intracellular Notch that activates R-Ras, independent of CSL-transcription. Notch also reverses H-Ras and Raf-mediated integrin suppression without affecting ERK phosphorylation. Membrane-bound Notch mutants that are inefficiently cleaved or intracellular Notch mutants lacking the ankyrin repeat sequence do not activate R-Ras or integrins. Co-expression of Msx2-interacting nuclear target (MINT) protein with Notch or expression of intracellular Notch-1 truncation mutants lacking the C-terminal transactivation/PEST domain suppresses Notch transcriptional activity without affecting integrin activation. Notch ligand, Delta-like ligand-4, stimulates R-Ras-dependent alpha 5 beta 1 integrin-mediated adhesion, demonstrating the physiological relevance of this pathway. This new CSL-independent Notch/R-Ras pathway provides a molecular mechanism to explain Notch, integrin, and Ras cross-talk during the development of multicellular organisms.
Collapse
Affiliation(s)
- Philip S Hodkinson
- University of Edinburgh, MRC Centre for Inflammation Research, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4SA, Scotland, United Kingdom
| | - Paul A Elliott
- University of Edinburgh, MRC Centre for Inflammation Research, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4SA, Scotland, United Kingdom
| | - Yatish Lad
- University of Edinburgh, MRC Centre for Inflammation Research, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4SA, Scotland, United Kingdom
| | - Brian J McHugh
- University of Edinburgh, MRC Centre for Inflammation Research, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4SA, Scotland, United Kingdom
| | - Alison C MacKinnon
- University of Edinburgh, MRC Centre for Inflammation Research, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4SA, Scotland, United Kingdom
| | - Christopher Haslett
- University of Edinburgh, MRC Centre for Inflammation Research, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4SA, Scotland, United Kingdom
| | - Tariq Sethi
- University of Edinburgh, MRC Centre for Inflammation Research, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4SA, Scotland, United Kingdom.
| |
Collapse
|
126
|
White JD, Vuocolo T, McDonagh M, Grounds MD, Harper GS, Cockett NE, Tellam R. Analysis of the callipyge phenotype through skeletal muscle development; association of Dlk1 with muscle precursor cells. Differentiation 2007; 76:283-98. [PMID: 17697128 DOI: 10.1111/j.1432-0436.2007.00208.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The callipyge mutation in sheep in the form of the paternal heterozygote results in skeletal muscle hypertrophy, which is most pronounced in the hindquarters. Overexpression of one of the genes in the region of the causative single-nucleotide polymorphism, Dlk1, is postulated to be a primary cause of the muscle hypertrophy although the mechanism is not clear. This study examined the expression of Dlk1 mRNA and its encoded protein in skeletal muscles of callipyge and wild-type sheep. The muscles examined included those that demonstrate hypertrophy in callipyge sheep as well as an unaffected muscle. The expression pattern of Dlk1 protein in these muscles was also measured over a developmental time course ranging from 80 days of gestation to 12 weeks after birth. Quantitative reverse transcription-polymerase chain reaction demonstrated that Dlk1 mRNA was significantly increased in affected, but not unaffected, muscles from callipyge sheep at 120 days of gestation through to 12 weeks of age. Immuno-localization of Dlk1 was pronounced in the interstitial connective tissue of fetal muscle but was less intense at later ages. No clear difference in Dlk1 immuno-localization was noted between genotypes in the fetal samples. Strong myofiber-specific Dlk1 immuno-localization was observed in hypertrophied callipyge muscles at 12 weeks of age. This staining was exclusively associated with fast type II myofibers and these had a significantly larger mean cross-sectional area, compared with fast type II myofibers in control sheep that did not overexpress Dlk1. In addition, Dlk1 immuno-localization was associated with a sub-population of Pax7-positive mononucleated cells in all skeletal muscles examined during fetal development and at birth, but this was not apparent at 12 weeks. There were no genotype-dependent alterations in the mRNA expression patterns of a number of promyogenic transcription factors indicating that the callipyge mutation was not affecting muscle cell differentiation per se. We postulate that Dlk1 is implicated in the commitment and/or proliferation of fetal myoblasts as well as in the maintenance of hypertrophy in fully differentiated myofibers.
Collapse
Affiliation(s)
- Jason D White
- School of Veterinary Science, The University of Melbourne, Parkville, Vic. 3010, Australia.
| | | | | | | | | | | | | |
Collapse
|
127
|
Kim MY, Ann EJ, Kim JY, Mo JS, Park JH, Kim SY, Seo MS, Park HS. Tip60 histone acetyltransferase acts as a negative regulator of Notch1 signaling by means of acetylation. Mol Cell Biol 2007; 27:6506-19. [PMID: 17636029 PMCID: PMC2099611 DOI: 10.1128/mcb.01515-06] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The Notch signaling pathway appears to perform an important function in a wide variety of organisms and cell types. In our present study, we provide evidence that UV irradiation-induced Tip60 proteins reduced Notch1 activity to a marked degree. Accumulated UV irradiation-induced Tip60 suppresses Notch1 transcriptional activity via the dissociation of the Notch1-IC-CSL complex. The binding between endogenous Tip60 and Notch1-IC in UV radiation-exposed cells was verified in this study by coimmunoprecipitation. Interestingly, the physical interaction of Tip60 with Notch1-IC occurs to a more profound degree in the presence of CSL but does not exist in a trimeric complex. Using Notch1-IC and Tip60 deletion mutants, we also determined that the N terminus, which harbors the RAM domain and seven ankyrin repeats of Notch1-IC, interacts with the zinc finger and acetyl coenzyme A domains of Tip60. Furthermore, here we report that Notch1-IC is a direct target of the acetyltransferase activity of Tip60. Collectively, our data suggest that Tip60 is an inhibitor of the Notch1 signaling pathway and that Tip60-dependent acetylation of Notch1-IC may be relevant to the mechanism by which Tip60 suppresses Notch1 signaling.
Collapse
Affiliation(s)
- Mi-Yeon Kim
- Hormonre Research Center, School of Biological Sciences and Technology, Chonnam National University, Yongbong-dong, Buk-ku, Gwangju 500-757, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
128
|
Sun H, Li L, Vercherat C, Gulbagci NT, Acharjee S, Li J, Chung TK, Thin TH, Taneja R. Stra13 regulates satellite cell activation by antagonizing Notch signaling. ACTA ACUST UNITED AC 2007; 177:647-57. [PMID: 17502421 PMCID: PMC2064210 DOI: 10.1083/jcb.200609007] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Satellite cells play a critical role in skeletal muscle regeneration in response to injury. Notch signaling is vital for satellite cell activation and myogenic precursor cell expansion but inhibits myogenic differentiation. Thus, precise spatial and temporal regulation of Notch activity is necessary for efficient muscle regeneration. We report that the basic helix-loop-helix transcription factor Stra13 modulates Notch signaling in regenerating muscle. Upon injury, Stra13(-/-) mice exhibit increased cellular proliferation, elevated Notch signaling, a striking regeneration defect characterized by degenerated myotubes, increased mononuclear cells, and fibrosis. Stra13(-/-) primary myoblasts also exhibit enhanced Notch activity, increased proliferation, and defective differentiation. Inhibition of Notch signaling ex vivo and in vivo ameliorates the phenotype of Stra13(-/-) mutants. We demonstrate in vitro that Stra13 antagonizes Notch activity and reverses the Notch-imposed inhibition of myogenesis. Thus, Stra13 plays an important role in postnatal myogenesis by attenuating Notch signaling to reduce myoblast proliferation and promote myogenic differentiation.
Collapse
Affiliation(s)
- Hong Sun
- Department of Molecular, Cell, and Developmental Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Vasyutina E, Lenhard DC, Wende H, Erdmann B, Epstein JA, Birchmeier C. RBP-J (Rbpsuh) is essential to maintain muscle progenitor cells and to generate satellite cells. Proc Natl Acad Sci U S A 2007; 104:4443-8. [PMID: 17360543 PMCID: PMC1815471 DOI: 10.1073/pnas.0610647104] [Citation(s) in RCA: 191] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In the developing muscle, a pool of myogenic progenitor cells is formed and maintained. These resident progenitors provide a source of cells for muscle growth in development and generate satellite cells in the perinatal period. By the use of conditional mutagenesis in mice, we demonstrate here that the major mediator of Notch signaling, the transcription factor RBP-J, is essential to maintain this pool of progenitor cells in an undifferentiated state. In the absence of RBP-J, these cells undergo uncontrolled myogenic differentiation, leading to a depletion of the progenitor pool. This results in a lack of muscle growth in development and severe muscle hypotrophy. In addition, satellite cells are not formed late in fetal development in conditional RBP-J mutant mice. We conclude that RBP-J is required in the developing muscle to set aside proliferating progenitors and satellite cells.
Collapse
Affiliation(s)
- Elena Vasyutina
- *Max Delbrück Center for Molecular Medicine, Robert Rössle Strasse 10, 13125 Berlin, Germany; and
| | - Diana C. Lenhard
- *Max Delbrück Center for Molecular Medicine, Robert Rössle Strasse 10, 13125 Berlin, Germany; and
| | - Hagen Wende
- *Max Delbrück Center for Molecular Medicine, Robert Rössle Strasse 10, 13125 Berlin, Germany; and
| | - Bettina Erdmann
- *Max Delbrück Center for Molecular Medicine, Robert Rössle Strasse 10, 13125 Berlin, Germany; and
| | - Jonathan A. Epstein
- Department of Cell and Developmental Biology and the Cardiovascular Institute, University of Pennsylvania, 954 BRB II, 421 Curie Boulevard, Philadelphia, PA 19104
| | - Carmen Birchmeier
- *Max Delbrück Center for Molecular Medicine, Robert Rössle Strasse 10, 13125 Berlin, Germany; and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
130
|
Arnoys EJ, Wang JL. Dual localization: proteins in extracellular and intracellular compartments. Acta Histochem 2007; 109:89-110. [PMID: 17257660 DOI: 10.1016/j.acthis.2006.10.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2006] [Revised: 10/01/2006] [Accepted: 10/09/2006] [Indexed: 12/24/2022]
Abstract
The goal of this article is to provide a comprehensive catalog of those proteins documented to exhibit dual localization, being found in both the extracellular compartment (cell surface and extracellular medium) as well as the intracellular compartment (cytosol and nucleus). A large subset of these proteins that show dual localization is found both in the nucleus and outside of cells. Proteins destined to be secreted out of the cell or to be expressed at the cell surface usually enter the endomembrane pathway on the basis of a signal sequence that targets them into the endoplasmic reticulum. Proteins destined for import into the nucleus, on the other hand, usually carry a nuclear localization signal. We have organized our catalog in terms of the presence and absence of these trafficking signals: (a) proteins that contain a signal sequence but no nuclear localization signal; (b) proteins that contain both a signal sequence as well as a nuclear localization signal; (c) proteins that contain a nuclear localization signal but lack a signal sequence; and (d) proteins containing neither a signal sequence nor a nuclear localization signal. Novel insights regarding the activities of several classes of proteins exhibiting dual localization can be derived when one targeting signal is experimentally abrogated. For example, the mitogenic activity of both fibroblasts growth factor-1 and schwannoma-derived growth factor clearly requires nuclear localization, independent of the activation of the receptor tyrosine kinase signaling pathway. In addition, there is a growing list of integral membrane receptors that undergo translocation to the nucleus, with bona fide nuclear localization signals and transcription activation activity. The information provided in this descriptive catalog will, hopefully, stimulate investigations into the pathways and mechanisms of transport between these compartments and the physiological significance of dual localization.
Collapse
Affiliation(s)
- Eric J Arnoys
- Department of Chemistry and Biochemistry, Calvin College, Grand Rapids, MI 49546, USA
| | | |
Collapse
|
131
|
Taqvi S, Dixit L, Roy K. Biomaterial-based notch signaling for the differentiation of hematopoietic stem cells into T cells. J Biomed Mater Res A 2007; 79:689-97. [PMID: 16845670 DOI: 10.1002/jbm.a.30916] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Thymocyte development takes place in a complex milieu of supportive cells and ECM that are responsible for the proliferation, adhesion, migration, and selection processes these cells undergo before reaching maturity. In recent years, the role of notch signaling in lymphocyte development, specifically T-cell development, has been extensively characterized. Although notch ligand mediated signals have been shown to be a necessary component of T-cell generation from stem cells, high-throughput, synthetic biomaterial-based systems for notch-directed stem-cell differentiation into lymphocytes are yet to be reported. Here, we present a microbead-based, artificial notch signaling system to study stem-cell differentiation into the T-cell lineage. Magnetic microbeads were functionalized with the notch ligand DLL4 using streptavidin-biotin binding and antibody-antigen coupling. Immunohistochemistry and flow cytometry analysis indicated approximately 65% conjugation efficiency. Efficient notch signaling through these functionalized microbeads was demonstrated through a myotube inhibition assay in C2C12 myoblasts. Thy1.2(+) early T cells were successfully generated from mouse bone marrow hematopoietic stem cells (BMHSCs) using DLL4 functionalized beads using both insert-based and mixed stromal cell (OP9) coculture conditions, indicating that stem cell-stromal cell physical contact is not necessary for DLL4 directed T-cell differentiation. Coculture studies with bead-to-cell ratios of 1:1 generated higher T-cell differentiation efficiencies, compared to bead-to-cell ratios of 5:1. These data demonstrate the promising potential of this biomaterial-based notch signaling system to generate T cells from stem cells and to elucidate the molecular interactions in T-cell development.
Collapse
Affiliation(s)
- Sabia Taqvi
- Department of Biomedical Engineering, University of Texas, Austin, Texas 78712, USA
| | | | | |
Collapse
|
132
|
Kang MA, Jeoung NH, Kim JY, Lee JE, Jung UJ, Choi MS, Lee WH, Kwon OS, Lee H, Park YB. Up-regulation of skeletal muscle LIM protein 1 gene by 25-hydroxycholesterol may mediate morphological changes of rat aortic smooth muscle cells. Life Sci 2007; 80:460-7. [PMID: 17161435 DOI: 10.1016/j.lfs.2006.09.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Revised: 07/26/2006] [Accepted: 09/27/2006] [Indexed: 11/22/2022]
Abstract
Changes in the expression level of the skeletal muscle LIM protein 1 (SLIM1) in cultured A10 cells were monitored in response to 25-hydroxycholesterol (25-HC), an oxidized form of cholesterol present in the oxidized low-density lipoproteins. The level of SLIM1 mRNA was elevated in a time- and concentration-dependent manner by treatment of 25-HC. Expressions of smooth muscle (SM) alpha-actin and calponin-1 (CNN-1), early markers for SMC differentiation, were also increased by the 25-HC treatments. Expressions of all three genes (SLIM1, SM alpha-actin and CNN-1) were simultaneously elevated in the cells treated with 9-cis retinoic acid (RA). On the other hand, the SLIM1 expression induced by the 25-HC or 9-cis RA (as well as SM alpha-actin and CNN-1) was decreased by the treatment of 15d-PGJ2. Since the 25-HC, 9-cis RA and 15d-PGJ2 were ligands for the LXR, RXRalpha and PPARgamma respectively, there might be a functional positive cross-talk between LXR and RXRalpha pathways and a negative cross-talk between PPARgamma and LXR and/or RXRalpha pathways in the regulation of SLIM1 expression. The cells stably transfected with the expressional vector for SLIM1 also showed an elevation in the levels of SM alpha-actin and CNN-1. In addition, an over-production of SLIM1 in the cells resulted in a change in the cell-shape into a spindle-like form, which is identical to that observed after a prolonged treatment of the cells with cholesterol.
Collapse
MESH Headings
- Actins/genetics
- Alitretinoin
- Animals
- Aorta/cytology
- Aorta/drug effects
- Aorta/metabolism
- Calcium-Binding Proteins/genetics
- Cell Differentiation/drug effects
- Cell Line
- Cell Shape/drug effects
- Cell Shape/genetics
- Dose-Response Relationship, Drug
- Gene Expression Regulation/drug effects
- Hydroxycholesterols/pharmacology
- LIM Domain Proteins
- Microfilament Proteins/genetics
- Muscle Proteins/genetics
- Muscle, Skeletal/cytology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Prostaglandin D2/analogs & derivatives
- Prostaglandin D2/pharmacology
- Rats
- Time Factors
- Tretinoin/pharmacology
- Up-Regulation
- Calponins
Collapse
Affiliation(s)
- Mi Ae Kang
- Department of Genetic Engineering, Kyungpook National University, 702-701, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
133
|
Dyczynska E, Sun D, Yi H, Sehara-Fujisawa A, Blobel CP, Zolkiewska A. Proteolytic processing of delta-like 1 by ADAM proteases. J Biol Chem 2007; 282:436-44. [PMID: 17107962 PMCID: PMC2692894 DOI: 10.1074/jbc.m605451200] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Delta-like 1 (Dll1) is a mammalian ligand for Notch receptors. Interactions between Dll1 and Notch in trans activate the Notch pathway, whereas Dll1 binding to Notch in cis inhibits Notch signaling. Dll1 undergoes proteolytic processing in its extracellular domain by ADAM10. In this work we demonstrate that Dll1 represents a substrate for several other members of the ADAM family. In co-transfected cells, Dll1 is constitutively cleaved by ADAM12, and the N-terminal fragment of Dll1 is released to medium. ADAM12-mediated cleavage of Dll1 is cell density-dependent, takes place in cis orientation, and does not require the presence of the cytoplasmic domain of ADAM12. Full-length Dll1, but not its N- or C-terminal proteolytic fragment, co-immunoprecipitates with ADAM12. By using a Notch reporter construct, we show that Dll1 processing by ADAM12 increases Notch signaling in a cell-autonomous manner. Furthermore, ADAM9 and ADAM17 have the ability to process Dll1. In contrast, ADAM15 does not cleave Dll1, although the two proteins still co-immunoprecipitate with each other. Asn-353 present in the catalytic motif of ADAM12 and other Dll1-processing ADAMs, but absent in ADAM15, is necessary for Dll1 cleavage. Dll1 cleavage is reduced in ADAM9/12/15(-/-) mouse embryonic fibroblasts (MEFs), suggesting that the endogenous ADAM9 and/or ADAM12 present in wild type MEFs contribute to Dll1 processing. Finally, the endogenous Dll1 present in primary mouse myoblasts undergoes cleavage in confluent, differentiating myoblast cultures, and this cleavage is decreased by ADAM12 small interfering RNAs. Our findings expand the role of ADAM proteins in the regulation of Notch signaling.
Collapse
Affiliation(s)
- Emilia Dyczynska
- Department of Biochemistry, Kansas State University, Manhattan, KS, USA
| | - Danqiong Sun
- Department of Biochemistry, Kansas State University, Manhattan, KS, USA
| | - Haiqing Yi
- Department of Biochemistry, Kansas State University, Manhattan, KS, USA
| | - Atsuko Sehara-Fujisawa
- Department of Growth Regulation, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Carl P. Blobel
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, Weill Medical College of Cornell University, New York, NY, USA
| | - Anna Zolkiewska
- Department of Biochemistry, Kansas State University, Manhattan, KS, USA
| |
Collapse
|
134
|
Kondoh K, Sunadome K, Nishida E. Notch signaling suppresses p38 MAPK activity via induction of MKP-1 in myogenesis. J Biol Chem 2006; 282:3058-65. [PMID: 17158101 DOI: 10.1074/jbc.m607630200] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Cross-talks among intracellular signaling pathways are important for the regulation of cell fate decisions and cellular responses to extracellular signals. Both the Notch pathway and the MAPK pathways play important roles in many biological processes, and the Notch pathway has been shown to interact with the ERK-type MAPK pathway. However, its interaction with the other MAPK pathways is unknown. Here we show that Notch signaling activation in C2C12 cells suppresses the activity of p38 MAPK to inhibit myogenesis. Our results show that Notch specifically induces expression of MKP-1, a member of the dual-specificity MAPK phosphatase, which directly inactivates p38 to negatively regulate C2C12 myogenesis. The Notch-induced expression of MKP-1 is shown to depend on RBP-J. Moreover, inhibition of MKP-1 expression by short interfering RNA suppresses p38 inactivation and partially rescues the negative regulation of myogenesis. These results reveal a novel cross-talk between the Notch pathway and the p38 MAPK pathway that is mediated by Notch induction of MKP-1.
Collapse
Affiliation(s)
- Kunio Kondoh
- Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | | | | |
Collapse
|
135
|
Lupien M, Diévart A, Morales CR, Hermo L, Calvo E, Kay DG, Hu C, Jolicoeur P. Expression of constitutively active Notch1 in male genital tracts results in ectopic growth and blockage of efferent ducts, epididymal hyperplasia and sterility. Dev Biol 2006; 300:497-511. [PMID: 17046738 DOI: 10.1016/j.ydbio.2006.09.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2004] [Revised: 09/01/2006] [Accepted: 09/07/2006] [Indexed: 11/27/2022]
Abstract
The Notch signaling pathway is involved in a variety of developmental processes. Here, we characterize the phenotypes developing in the reproductive organs of male transgenic (Tg) mice constitutively expressing the activated mouse Notch1 intracellular domain (Notch1(intra)) under the regulatory control of the mouse mammary tumor virus (MMTV) long terminal repeat (LTR). Tg expression was detected in testis, vas deferens and epididymis by Northern blot analysis. In situ hybridization with a Notch1-specific probe lacked sensitivity to detect expression in normal-appearing cells, but demonstrated expression in hyperplastic epithelial cells of the vas deferens, epididymis and efferent ducts. Tg males from three independent founder lines were sterile. Histological analysis of reproductive organs of young Tg males (postnatal ages 8 and 21) showed no difference compared to those of non-Tg males. In contrast, in adult Tg mice from day 38 onwards, the efferent ducts, the vas deferens and most epididymal segments revealed bilateral epithelial cell hyperplasia with absence of fully differentiated epithelial cells. Electron microscopy confirmed the uniformly undifferentiated state of these cells. Immunohistochemistry with anti-PCNA antibody also revealed enhanced proliferation of Tg epididymis. In adult Tg testis, the different generations of germ cells of seminiferous tubules appeared normal, although some tubules were highly dilated and revealed an absence of early and/or late spermatids. The epithelial cells of the Tg tubuli recti and rete testis were not abnormal, but the rete testis was highly dilated and contained numerous spermatozoa, suggesting a downstream blockage. Consistent with a blockage of efferent ducts often seen at the rete testis/efferent duct interface, spermatozoa were absent in epididymis of all adult Tg mice and in all highly hyperplastic efferent duct tubules of these Tg mice. Such a blockage was visualized by injection of Evans blue dye into the rete testis lumen. Finally, the presence of ectopic hyperplastic efferent duct tubules was observed within the testicular parenchyma itself, outside their normal territory, suggesting that Notch1 signaling is involved in the establishment of these borders. This phenotype seems to represent a novel developmental defect in mammals. Together, these results show that constitutive Notch1 signaling significantly affects the development of male reproductive organs.
Collapse
Affiliation(s)
- Mathieu Lupien
- Laboratory of Molecular Biology, Clinical Research Institute of Montreal, Canada
| | | | | | | | | | | | | | | |
Collapse
|
136
|
Lubman OY, Ilagan MXG, Kopan R, Barrick D. Quantitative dissection of the Notch:CSL interaction: insights into the Notch-mediated transcriptional switch. J Mol Biol 2006; 365:577-89. [PMID: 17070841 PMCID: PMC1851696 DOI: 10.1016/j.jmb.2006.09.071] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2006] [Revised: 09/25/2006] [Accepted: 09/25/2006] [Indexed: 12/11/2022]
Abstract
Complex formation between the intracellular domain of the Notch receptor (NICD) and the transcription factor CSL is indispensable for transcriptional activation. To understand how NICD displaces CSL-associated co-repressors, we have quantified the binding of different Notch1 ICD regions to a key interaction domain (the beta trefoil domain, or BTD) of human CSL. Electrophoresis, scattering, and titration calorimetry indicate that NICD and BTD combine to form a 1:1 heterodimer. Neither the Notch1 ankyrin domain (ANK) nor C-terminal region contributes binding energy towards BTD. In contrast, binding energy is attributed largely to a short segment including the conserved WFP sequence motif within the RAM region (the approximately 140 residue polypeptide segment N-terminal to the ANK domain); substitution of this motif substantially reduces affinity. Short (< or =25 residues) WFP-containing peptides encoded by the four mammalian Notch genes have similar affinities to BTD; thus, activity differences between paralogues either result from other regions of NICD and CSL or from differences in interaction with downstream components. The importance of RAM was demonstrated by the ability of a short RAM peptides to dissociate NICD:CSL interaction in cellular lysates. These results support an emerging molecular mechanism for the displacement of co-repressors from DNA-bound CSL by NICD.
Collapse
Affiliation(s)
- Olga Y. Lubman
- T.C Jenkins Department of Biophysics, Johns Hopkins University, Baltimore, MD, USA
- Department of Molecular Biology and Pharmacology and Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ma. Xenia G. Ilagan
- Department of Molecular Biology and Pharmacology and Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Raphael Kopan
- Department of Molecular Biology and Pharmacology and Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
- *Authors to whom correspondence should be addressed: Doug Barrick, Phone: (410) 516-0409; Fax: (410) 516-4118: E-mail: , Raphael Kopan, Phone: (314) 747-5520; Fax: (314) 362-7058: E-mail:
| | - Doug Barrick
- T.C Jenkins Department of Biophysics, Johns Hopkins University, Baltimore, MD, USA
- *Authors to whom correspondence should be addressed: Doug Barrick, Phone: (410) 516-0409; Fax: (410) 516-4118: E-mail: , Raphael Kopan, Phone: (314) 747-5520; Fax: (314) 362-7058: E-mail:
| |
Collapse
|
137
|
Chau MDL, Tuft R, Fogarty K, Bao ZZ. Notch signaling plays a key role in cardiac cell differentiation. Mech Dev 2006; 123:626-40. [PMID: 16843648 PMCID: PMC1567976 DOI: 10.1016/j.mod.2006.06.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2006] [Revised: 05/22/2006] [Accepted: 06/07/2006] [Indexed: 11/17/2022]
Abstract
Results from lineage tracing studies indicate that precursor cells in the ventricles give rise to both cardiac muscle and conduction cells. Cardiac conduction cells are specialized cells responsible for orchestrating the rhythmic contractions of the heart. Here, we show that Notch signaling plays an important role in the differentiation of cardiac muscle and conduction cell lineages in the ventricles. Notch1 expression coincides with a conduction marker, HNK-1, at early stages. Misexpression of constitutively active Notch1 (NIC) in early heart tubes in chick exhibited multiple effects on cardiac cell differentiation. Cells expressing NIC had a significant decrease in expression of cardiac muscle markers, but an increase in expression of conduction cell markers, HNK-1, and SNAP-25. However, the expression of the conduction marker connexin 40 was inhibited. Loss-of-function study, using a dominant-negative form of Suppressor-of-Hairless, further supports that Notch1 signaling is important for the differentiation of these cardiac cell types. Functional studies show that the expression of constitutively active Notch1 resulted in abnormalities in ventricular conduction pathway patterns.
Collapse
Affiliation(s)
- Mary D L Chau
- Department of Medicine and Cell Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | |
Collapse
|
138
|
Luo D, Renault VM, Rando TA. The regulation of Notch signaling in muscle stem cell activation and postnatal myogenesis. Semin Cell Dev Biol 2006; 16:612-22. [PMID: 16087370 DOI: 10.1016/j.semcdb.2005.07.002] [Citation(s) in RCA: 144] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The Notch signaling pathway is an evolutionarily conserved pathway that is critical for tissue morphogenesis during development, but is also involved in tissue maintenance and repair in the adult. In skeletal muscle, regulation of Notch signaling is involved in somitogenesis, muscle development, and the proliferation and cell fate determination of muscle stems cells during regeneration. During each of these processes, the spatial and temporal control of Notch signaling is essential for proper tissue formation. That control is mediated by a series of regulatory proteins and protein complexes that enhance or inhibit Notch signaling by regulating protein processing, localization, activity, and stability. In this review, we focus on the regulation of Notch signaling during postnatal muscle regeneration when muscle stem cells ("satellite cells") must activate, proliferate, progress along a myogenic lineage pathway, and ultimately differentiate to form new muscle. We review the regulators of Notch signaling, such as Numb and Deltex, that have documented roles in myogenesis as well as other regulators that may play a role in modulating Notch signaling during satellite cell activation and postnatal myogenesis.
Collapse
Affiliation(s)
- Dan Luo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305-5235, USA
| | | | | |
Collapse
|
139
|
Abstract
Two papers in this issue of Cell, (Nam et al., 2006; Wilson and Kovall, 2006) report the crystal structure of the Notch transcriptional activation complex. These structures show that the ANK domain of Notch is an integral part of the transcription complex and supports MAM binding, whereas the RAM domain may trigger allosteric changes in the structure needed for the derepression of transcription.
Collapse
Affiliation(s)
- Doug Barrick
- T.C. Jenkins Department of Biophysics, The Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | | |
Collapse
|
140
|
Lowell S, Benchoua A, Heavey B, Smith AG. Notch promotes neural lineage entry by pluripotent embryonic stem cells. PLoS Biol 2006; 4:e121. [PMID: 16594731 PMCID: PMC1431581 DOI: 10.1371/journal.pbio.0040121] [Citation(s) in RCA: 205] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2005] [Accepted: 02/15/2006] [Indexed: 12/31/2022] Open
Abstract
A central challenge in embryonic stem (ES) cell biology is to understand how to impose direction on primary lineage commitment. In basal culture conditions, the majority of ES cells convert asynchronously into neural cells. However, many cells resist differentiation and others adopt nonneural fates. Mosaic activation of the neural reporter Sox-green fluorescent protein suggests regulation by cell-cell interactions. We detected expression of Notch receptors and ligands in mouse ES cells and investigated the role of this pathway. Genetic manipulation to activate Notch constitutively does not alter the stem cell phenotype. However, upon withdrawal of self-renewal stimuli, differentiation is directed rapidly and exclusively into the neural lineage. Conversely, pharmacological or genetic interference with Notch signalling suppresses the neural fate choice. Notch promotion of neural commitment requires parallel signalling through the fibroblast growth factor receptor. Stromal cells expressing Notch ligand stimulate neural specification of human ES cells, indicating that this is a conserved pathway in pluripotent stem cells. These findings define an unexpected and decisive role for Notch in ES cell fate determination. Limiting activation of endogenous Notch results in heterogeneous lineage commitment. Manipulation of Notch signalling is therefore likely to be a key factor in taking command of ES cell lineage choice.
Collapse
Affiliation(s)
- Sally Lowell
- 1Centre Development in Stem Cell Biology, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Alexandra Benchoua
- 1Centre Development in Stem Cell Biology, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- 2INSERM U 421/I-STEM, Faculté de Médecine, Evry-Cedex, France
| | - Barry Heavey
- 1Centre Development in Stem Cell Biology, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Austin G Smith
- 1Centre Development in Stem Cell Biology, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- 3Institute for Stem Cell Biology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
141
|
Hu C, Diévart A, Lupien M, Calvo E, Tremblay G, Jolicoeur P. Overexpression of activated murine Notch1 and Notch3 in transgenic mice blocks mammary gland development and induces mammary tumors. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 168:973-90. [PMID: 16507912 PMCID: PMC1606519 DOI: 10.2353/ajpath.2006.050416] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The mouse mammary tumor virus (MMTV) provirus was found to target the Notch1 gene, producing insertional mutations in mammary tumors of MMTV/neu transgenic (Tg) mice. In these mammary tumors, the Notch1 gene is truncated upstream of the transmembrane domain, and the resulting Notch1 intracellular domain (Notch1(intra)), deleted of most extracellular sequences, is overexpressed. Although Notch1(intra) transforms mammary epithelial cells in vitro, its role in mammary gland tumor formation in vivo was not studied. Therefore, we generated MMTV/Notch1(intra) Tg mice that overexpress murine Notch1(intra) in the mammary glands. We observed that MMTV/Notch1(intra) Tg females were unable to feed their pups because of impaired ductal and lobulo-alveolar mammary gland development. This was associated with decreased proliferation of ductal and alveolar epithelial cells during rapid expansion at puberty and in early pregnancy, as well as decreased production of beta-casein. Notch1(intra) repressed expression of the beta-casein gene promoter, as assessed in vitro with a beta-casein/luciferase reporter construct. The MMTV/Notch1(intra) Tg females developed mammary gland tumors, confirming the oncogenic potential of Notch1(intra) in vivo. Furthermore, MMTV/Notch3(intra) Tg mice exhibited a very similar phenotype. Thus, these Tg mice represent novel models for studying the role of Notch1 or Notch3 in the development and transformation of the mammary gland.
Collapse
MESH Headings
- Aging
- Animals
- Apoptosis
- Caseins/genetics
- Cell Proliferation
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Evolution, Molecular
- Female
- Gene Expression Regulation, Developmental
- Gene Expression Regulation, Neoplastic
- Mammary Glands, Animal/abnormalities
- Mammary Glands, Animal/growth & development
- Mammary Glands, Animal/pathology
- Mammary Neoplasms, Animal/etiology
- Mammary Neoplasms, Animal/genetics
- Mammary Neoplasms, Animal/pathology
- Mammary Tumor Virus, Mouse/genetics
- Mice
- Mice, Transgenic
- Oncogenes/genetics
- Pregnancy
- Promoter Regions, Genetic/genetics
- Receptor, Notch1/genetics
- Receptor, Notch1/metabolism
- Receptor, Notch3
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- Transcription, Genetic
- Transcriptional Activation
Collapse
Affiliation(s)
- Chunyan Hu
- Laboratory of Molecular Biology, Clinical Research Institute of Montréal, QC, Canada
| | | | | | | | | | | |
Collapse
|
142
|
Langdon T, Hayward P, Brennan K, Wirtz-Peitz F, Sanders P, Zecchini V, Friday A, Balayo T, Martinez Arias A. Notch receptor encodes two structurally separable functions inDrosophila: A genetic analysis. Dev Dyn 2006; 235:998-1013. [PMID: 16534797 DOI: 10.1002/dvdy.20735] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The Notch gene of Drosophila encodes a single transmembrane receptor that plays a central role in the process of lateral inhibition. This process results in the selection of individual mesodermal and neural precursors during the development of the muscular and nervous systems. The activation of Notch during lateral inhibition is mediated by the transmembrane ligand Delta (Dl) and effected by the transcription factor Suppressor of Hairless (Su(H)). The same functional cassette plays a role in other processes, in particular, the development and patterning of the wing. Genetic analysis has suggested that, in addition to the Su(H)-dependent pathway, Notch can signal in an Su(H)-independent manner. This process seems to be tightly associated with signalling by Wingless, a member of the Wnt family of signalling molecules. Here, we have analyzed further the possibility that the Notch protein encodes two different functions. To do so, we have studied the activities and genetic properties of different Notch receptors bearing deletions of specific regions of the intracellular and the extracellular domains in different developmental processes, and have sought to correlate the activity of these mutant proteins with those of existing mutants in Notch. Our results support the existence of at least two different activities of Notch each of which can be associated with specific structural domains.
Collapse
Affiliation(s)
- Tim Langdon
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, U.K
| | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Böhm C, Seibel NM, Henkel B, Steiner H, Haass C, Hampe W. SorLA signaling by regulated intramembrane proteolysis. J Biol Chem 2006; 281:14547-53. [PMID: 16531402 DOI: 10.1074/jbc.m601660200] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The single-transmembrane receptor SorLA/LR11 contains binding domains typical for lipoprotein receptors and a VPS10 domain, which binds the neuropeptide head-activator. This undecapeptide enhances proliferation of neuronal precursor cells in a SorLA-dependent manner. Using specific inhibitors we found previously that head activator activates shedding of SorLA by the metalloprotease TACE close to the transmembrane domain releasing the large extra-cellular part of the receptor. Here we show that the remaining COOH-terminal membrane fragment of SorLA is processed by gamma-secretase. Inhibition of gamma-secretase by specific inhibitors or overexpression of dominant negative presenilin mutants and knock out of the presenilin genes led to accumulation of the SorLA membrane fragment and also of full-length SorLA in the membrane. In an in vitro assay we observed the gamma-secretase-dependent release of the two soluble cleavage products, the SorLA cytoplasmic domain and the SorLA beta-peptide. These processing steps are reminiscent of a novel signaling pathway that has been described for the notch receptor. Here, the notch cytoplasmic domain is released into the cytoplasm by the gamma-secretase and migrates to the nucleus where it acts as a transcriptional regulator. In parallel we found that a fusion protein of the released cytoplasmic tail of SorLA with EGFP located to the nucleus only if the nuclear localization signal of SorLA was intact. In a reporter gene assay the cytoplasmic domain of SorLA acted as a transcriptional activator indicating that SorLA might directly regulate transcription after activation by gamma-secretase.
Collapse
Affiliation(s)
- Christopher Böhm
- University Medical Center Hamburg-Eppendorf, Center of Experimental Medicine, Department of Biochemistry and Molecular Biology II: Molecular Cell Biology, Martinistrasse 52, D-20246 Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
144
|
Holowacz T, Zeng L, Lassar AB. Asymmetric localization of numb in the chick somite and the influence of myogenic signals. Dev Dyn 2006; 235:633-45. [PMID: 16425215 PMCID: PMC2561193 DOI: 10.1002/dvdy.20672] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Whereas Notch signaling is known to play an essential role in the formation of somites, its role during later stages of somite maturation is less well understood. Here, we examine the signals and transcription factors that control the expression of the Notch antagonist, Numb, during somite maturation in the chick embryo. Numb mRNA is present in the epithelial somite and is increased in expression in the forming myotome. Numb protein displays a very specific subcellular localization and dynamic expression during somite maturation. Numb protein is asymmetrically localized in a cortical crescent on the basal side of dividing cells in the dorsomedial lip of the dermomyotome and is subsequently uniformly distributed throughout differentiated myotomal cells. Treatment of somites with either the combination of Wnt-3a and Shh, or ectodermal signals plus noggin, both of which induce somitic myogenesis, did not significantly affect Numb transcript levels but did lead to a dramatic increase in the levels of Numb protein, which was uniformly distributed throughout the cytoplasm of the resultant myotubes. Forced expression of MyoD in somites similarly induced high levels of Numb protein throughout the cytoplasm, without affecting Numb mRNA levels. We also found that signals that promote somitic myogenesis or forced MyoD expression induced expression of the Notch ligand, Serrate-2. Our findings suggest that Notch signals are specifically repressed in the myotome and that asymmetric expression of Numb in dividing cells of the dorsomedial lip of the dermomyotome may modulate whether these cells continue to divide or differentiate into myotomal cells.
Collapse
Affiliation(s)
- Tamara Holowacz
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
145
|
Shen H, McElhinny AS, Cao Y, Gao P, Liu J, Bronson R, Griffin JD, Wu L. The Notch coactivator, MAML1, functions as a novel coactivator for MEF2C-mediated transcription and is required for normal myogenesis. Genes Dev 2006; 20:675-88. [PMID: 16510869 PMCID: PMC1413284 DOI: 10.1101/gad.1383706] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The MAML (mastermind-like) proteins are a family of three co-transcriptional regulators that are essential for Notch signaling, a pathway critical for cell fate determination. Though the functions of MAML proteins in normal development remain unresolved, their distinct tissue distributions and differential activities in cooperating with various Notch receptors suggest that they have unique roles. Here we show that mice with a targeted disruption of the Maml1 gene have severe muscular dystrophy. In vitro, Maml1-null embryonic fibroblasts failed to undergo MyoD-induced myogenic differentiation, further suggesting that Maml1 is required for muscle development. Interestingly, overexpression of MAML1 in C2C12 cells dramatically enhanced myotube formation and increased the expression of muscle-specific genes, while RNA interference (RNAi)-mediated MAML1 knockdown abrogated differentiation. Moreover, we determined that MAML1 interacts with MEF2C (myocyte enhancer factor 2C), functioning as its potent co-transcriptional regulator. Surprisingly, however, MAML1's promyogenic effects were completely blocked upon activation of Notch signaling, which was associated with recruitment of MAML1 away from MEF2C to the Notch transcriptional complex. Our study thus reveals novel and nonredundant functions for MAML1: It acts as a coactivator for MEF2C transcription and is essential for proper muscle development. Mechanistically, MAML1 appears to mediate cross-talk between Notch and MEF2 to influence myogenic differentiation.
Collapse
Affiliation(s)
- Huangxuan Shen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
146
|
Ehebauer M, Chirgadze D, Hayward P, Martinez Arias A, Blundell T. High-resolution crystal structure of the human Notch 1 ankyrin domain. Biochem J 2006; 392:13-20. [PMID: 16011479 PMCID: PMC1317659 DOI: 10.1042/bj20050515] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The Notch receptor is part of a highly conserved signalling system of central importance to animal development. Its ANK (ankyrin) domain is required for Notch-mediated signal transduction. The crystal structure of the human Notch 1 ANK domain was solved by molecular replacement at 1.9 A (1 A=0.1 nm) resolution, and it shows that the features identified in the Drosophila homologue are conserved. The domain has six of the seven ANK repeats predicted from sequence. The putative first repeat, which has only part of the consensus and a long insertion, is disordered in both molecules in the asymmetric unit, possibly due to the absence of the RAM (RBPJkappa-associated molecule) region N-terminal to it. The exposed hydrophobic core is involved in intermolecular interactions in the crystal. Evolutionary trace analysis identified several residues that map to the hairpins of the structure and may be of functional importance. Based on the Notch 1 ANK structure and analysis of homologous Notch ANK sequences, we predict two possible binding sites on the domain: one on the concave surface of repeat 2 and the other below the hairpins of repeats 6-7.
Collapse
Affiliation(s)
- Matthias T. Ehebauer
- *Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, U.K
| | - Dimitri Y. Chirgadze
- *Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, U.K
| | - Penny Hayward
- †Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, U.K
| | - Alfonso Martinez Arias
- †Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, U.K
| | - Tom L. Blundell
- *Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, U.K
- To whom correspondence should be addressed (email )
| |
Collapse
|
147
|
Gustafsson MV, Zheng X, Pereira T, Gradin K, Jin S, Lundkvist J, Ruas JL, Poellinger L, Lendahl U, Bondesson M. Hypoxia requires notch signaling to maintain the undifferentiated cell state. Dev Cell 2006; 9:617-28. [PMID: 16256737 DOI: 10.1016/j.devcel.2005.09.010] [Citation(s) in RCA: 814] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2005] [Revised: 07/08/2005] [Accepted: 09/12/2005] [Indexed: 02/06/2023]
Abstract
In addition to controlling a switch to glycolytic metabolism and induction of erythropoiesis and angiogenesis, hypoxia promotes the undifferentiated cell state in various stem and precursor cell populations. Here, we show that the latter process requires Notch signaling. Hypoxia blocks neuronal and myogenic differentiation in a Notch-dependent manner. Hypoxia activates Notch-responsive promoters and increases expression of Notch direct downstream genes. The Notch intracellular domain interacts with HIF-1alpha, a global regulator of oxygen homeostasis, and HIF-1alpha is recruited to Notch-responsive promoters upon Notch activation under hypoxic conditions. Taken together, these data provide molecular insights into how reduced oxygen levels control the cellular differentiation status and demonstrate a role for Notch in this process.
Collapse
Affiliation(s)
- Maria V Gustafsson
- Department of Cell and Molecular Biology, Medical Nobel Institute, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Abstract
Unraveling the complex tissue interactions necessary to generate the structural and functional diversity present among craniofacial muscles is challenging. These muscles initiate their development within a mesenchymal population bounded by the brain, pharyngeal endoderm, surface ectoderm, and neural crest cells. This set of spatial relations, and in particular the segmental properties of these adjacent tissues, are unique to the head. Additionally, the lack of early epithelialization in head mesoderm necessitates strategies for generating discrete myogenic foci that may differ from those operating in the trunk. Molecular data indeed indicate dissimilar methods of regulation, yet transplantation studies suggest that some head and trunk myogenic populations are interchangeable. The first goal of this review is to present key features of these diversities, identifying and comparing tissue and molecular interactions regulating myogenesis in the head and trunk. Our second focus is on the diverse morphogenetic movements exhibited by craniofacial muscles. Precursors of tongue muscles partly mimic migrations of appendicular myoblasts, whereas myoblasts destined to form extraocular muscles condense within paraxial mesoderm, then as large cohorts they cross the mesoderm:neural crest interface en route to periocular regions. Branchial muscle precursors exhibit yet another strategy, establishing contacts with neural crest populations before branchial arch formation and maintaining these relations through subsequent stages of morphogenesis. With many of the prerequisite stepping-stones in our knowledge of craniofacial myogenesis now in place, discovering the cellular and molecular interactions necessary to initiate and sustain the differentiation and morphogenesis of these neglected craniofacial muscles is now an attainable goal.
Collapse
Affiliation(s)
- Drew M Noden
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA.
| | | |
Collapse
|
149
|
Abstract
Notch signaling regulates cell fate determination and many developmental processes. Here we report that lateral inhibition, a major mechanism for Notch activity, is modulated by Hairy, a bHLH-WRPW protein. In Xenopus, Notch can have from inhibitory, permissive to enhancing roles in muscle or neural differentiation. These cell context-dependent effects correlate with Hairy expression levels from high to low, respectively, in the cells. Moreover, Notch effects can be altered upon manipulation of Hairy expression. We propose that Hairy provides a cell context in which a cell can interpret Notch and other extrinsic signals by controlling responsiveness of its target genes; this mode of Hairy-Notch interaction may apply in other systems.
Collapse
Affiliation(s)
- Yanzhen Cui
- Department of Environmental and Biomolecular Systems, Oregon Graduate Institute School of Science and Engineering, Oregon Health and Science University, 20000 NW Walker Road, Beaverton, OR 97006, USA.
| |
Collapse
|
150
|
Francis JC, Radtke F, Logan MPO. Notch1 signals through Jagged2 to regulate apoptosis in the apical ectodermal ridge of the developing limb bud. Dev Dyn 2005; 234:1006-15. [PMID: 16245338 DOI: 10.1002/dvdy.20590] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The Notch family of receptors is involved in a wide variety of developmental processes, including cell fate specification, cell proliferation, and cell survival decisions during cell differentiation and tissue morphogenesis. Notch1 and Notch ligands are expressed in the developing limbs, and Notch signalling has been implicated in the formation of a variety of tissues that comprise the limb, such as the skeleton, musculature, and vasculature. Notch signalling has also been implicated in regulating overall limb size. We have used a conditional allele of Notch1 in combination with two different Cre transgenic lines to delete Notch1 function either in the limb mesenchyme or in the apical ectodermal ridge (AER) and limb ectoderm. We demonstrate that Notch signalling, involving Notch1 and Jagged2, is required to regulate the number of Fgf8-expressing cells that comprise the AER and that regulation of the levels of fibroblast growth factor signalling is important for the freeing of the digits during normal limb formation. Regulation of the extent of the AER is achieved by Notch signalling positively regulating apoptosis in the AER. We also demonstrate that Notch1 is not required for proper formation of all the derivatives of the limb mesenchyme.
Collapse
Affiliation(s)
- Jeffrey C Francis
- Division of Developmental Biology, National Institute for Medical Research, Mill Hill, London, UK
| | | | | |
Collapse
|