Academic literature on the topic 'NOTCH 1 Intracellular Domain Indicate'

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the lists of relevant articles, books, theses, conference reports, and other scholarly sources on the topic 'NOTCH 1 Intracellular Domain Indicate.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Journal articles on the topic "NOTCH 1 Intracellular Domain Indicate"

1

Kitagawa, Motoo, Toshinao Oyama, Taichi Kawashima, Barry Yedvobnick, Anumeha Kumar, Kenji Matsuno, and Kenichi Harigaya. "A Human Protein with Sequence Similarity to DrosophilaMastermind Coordinates the Nuclear Form of Notch and a CSL Protein To Build a Transcriptional Activator Complex on Target Promoters." Molecular and Cellular Biology 21, no. 13 (July 1, 2001): 4337–46. http://dx.doi.org/10.1128/mcb.21.13.4337-4346.2001.

Full text
Abstract:
ABSTRACT Mastermind (Mam) has been implicated as an important positive regulator of the Notch signaling pathway by genetic studies usingDrosophila melanogaster. Here we describe a biochemical mechanism of action of Mam within the Notch signaling pathway. Expression of a human sequence related to Drosophila Mam (hMam-1) in mammalian cells augments induction of Hairy Enhancer of split (HES) promoters by Notch signaling. hMam-1 stabilizes and participates in the DNA binding complex of the intracellular domain of human Notch1 and a CSL protein. Truncated versions of hMam-1 that can maintain an association with the complex behave in a dominant negative fashion and depress transactivation. Furthermore,Drosophila Mam forms a similar complex with the intracellular domain of Drosophila Notch andDrosophila CSL protein during activation of Enhancer of split, the Drosophila counterpart ofHES. These results indicate that Mam is an essential component of the transcriptional apparatus of Notch signaling.
APA, Harvard, Vancouver, ISO, and other styles
2

Blokzijl, Andries, Camilla Dahlqvist, Eva Reissmann, Anna Falk, Annalena Moliner, Urban Lendahl, and Carlos F. Ibáñez. "Cross-talk between the Notch and TGF-β signaling pathways mediated by interaction of the Notch intracellular domain with Smad3." Journal of Cell Biology 163, no. 4 (November 24, 2003): 723–28. http://dx.doi.org/10.1083/jcb.200305112.

Full text
Abstract:
The Notch and transforming growth factor-β (TGF-β) signaling pathways play critical roles in the control of cell fate during metazoan development. However, mechanisms of cross-talk and signal integration between the two systems are unknown. Here, we demonstrate a functional synergism between Notch and TGF-β signaling in the regulation of Hes-1, a direct target of the Notch pathway. Activation of TGF-β signaling up-regulated Hes-1 expression in vitro and in vivo. This effect was abrogated in myogenic cells by a dominant-negative form of CSL, an essential DNA-binding component of the Notch pathway. TGF-β regulated transcription from the Hes-1 promoter in a Notch-dependent manner, and the intracellular domain of Notch1 (NICD) cooperated synergistically with Smad3, an intracellular transducer of TGF-β signals, to induce the activation of synthetic promoters containing multimerized CSL- or Smad3-binding sites. NICD and Smad3 were shown to interact directly, both in vitro and in cells, in a ligand-dependent manner, and Smad3 could be recruited to CSL-binding sites on DNA in the presence of CSL and NICD. These findings indicate that Notch and TGF-β signals are integrated by direct protein–protein interactions between the signal-transducing intracellular elements from both pathways.
APA, Harvard, Vancouver, ISO, and other styles
3

Chandiran, Karthik, and Lisa M. Minter. "Notch-1 regulation of microRNAs alter the expression of proinflammatory cytokines and Th1 response." Journal of Immunology 196, no. 1_Supplement (May 1, 2016): 127.1. http://dx.doi.org/10.4049/jimmunol.196.supp.127.1.

Full text
Abstract:
Abstract Notch signaling is required for activation and differentiation of T cells. Upon its S3 cleavage by gamma secretase, Notch intracellular domain triggers the expression of various molecules depending on its binding partners. The data published from our lab states that Notch1 can induce Th1 immune response and regulates the expression of IFNγ by modulating the expression of the transcription factor T-bet. However the detailed mechanism behind Notch1 mediated Th1 differentiation is not clear. Here we report that Notch1 can regulate Th1 differentiation by targeting microRNAs. miR-29 is a family of micro RNAs (miR29a,b,c) that targets ifng and both its transcription factors tbx21 and eomes directly by binding to their respective 3′ UTRs. By using novel gamma secretase inhibitors (GSIs) we show that Notch can modulate the expression of miR-29. Overexpression of Notch1 in T cell hybridoma cell lines further validates Notch1- mediated mirR-29 regulation. These results indicate a novel regulatory mechanism of T cell differentiation through Notch1 mediated microRNA expression.
APA, Harvard, Vancouver, ISO, and other styles
4

Koshizaka, Masaya, Minoru Takemoto, Seiya Sato, Hirotake Tokuyama, Masaki Fujimoto, Emiko Okabe, Ryoichi Ishibashi, et al. "An Angiotensin II Type 1 Receptor Blocker Prevents Renal Injury via Inhibition of the Notch Pathway in Ins2 Akita Diabetic Mice." Experimental Diabetes Research 2012 (2012): 1–10. http://dx.doi.org/10.1155/2012/159874.

Full text
Abstract:
Recently, it has been reported that the Notch pathway is involved in the pathogenesis of diabetic nephropathy. In this study, we investigated the activation of the Notch pathway in Ins2 Akita diabetic mouse (Akita mouse) and the effects of telmisartan, an angiotensin II type1 receptor blocker, on the Notch pathway. The intracellular domain of Notch1 (ICN1) is proteolytically cleaved from the cell plasma membrane in the course of Notch activation. The expression of ICN1 and its ligand, Jagged1, were increased in the glomeruli of Akita mice, especially in the podocytes. Administration of telmisartan significantly ameliorated the expression of ICN1 and Jagged1. Telmisartan inhibited the angiotensin II-induced increased expression of transforming growth factorβand vascular endothelial growth factor A which could directly activate the Notch signaling pathway in cultured podocytes. Our results indicate that the telmisartan prevents diabetic nephropathy through the inhibition of the Notch pathway.
APA, Harvard, Vancouver, ISO, and other styles
5

Nobta, Masuhiro, Tomoo Tsukazaki, Yasuaki Shibata, Chang Xin, Takeshi Moriishi, Seiji Sakano, Hiroyuki Shindo, and Akira Yamaguchi. "Critical Regulation of Bone Morphogenetic Protein-induced Osteoblastic Differentiation by Delta1/Jagged1-activated Notch1 Signaling." Journal of Biological Chemistry 280, no. 16 (February 4, 2005): 15842–48. http://dx.doi.org/10.1074/jbc.m412891200.

Full text
Abstract:
Functional involvement of the Notch pathway in osteoblastic differentiation has been previously investigated using the truncated intracellular domain, which mimics Notch signaling by interacting with the DNA-binding protein CBF-1. However, it is unclear whether Notch ligands Delta1 and Jagged1 also induce an identical cellular response in osteoblastic differentiation. We have shown that both Delta1 and Jagged1 were expressed concomitantly with Notch1 in maturating osteoblastic cells during bone regeneration and that overexpressed and immobilized recombinant Delta1 and Jagged1 alone did not alter the differentiated state of MC3T3-E1 and C2C12 cells. However, they augmented bone morphogenetic protein-2 (BMP2)-induced alkaline phosphatase activity and the expression of several differentiation markers, except for osteocalcin, and ultimately enhanced calcified nodule andin vivoectopic bone formation of MC3T3-E1. In addition, both ligands transmitted signal through the CBF-1-dependent pathway and stimulated the expression of HES-1, a direct target of Notch pathway. To test the necessity of Notch signaling in BMP2-induced differentiation, Notch signaling was inhibited by the dominant negative extracellular domain of Notch1, specific inhibitor, or small interference RNA. These treatments decreased alkaline phosphatase activity as well as the expression of other differentiation markers and inhibited the promoter activity of Id-1, a target gene of the BMP pathway. These results indicate the functional redundancy between Delta1 and Jagged1 in osteoblastic differentiation whereby Delta1/Jagged1-activated Notch1 enhances BMP2-induced differentiation through the identical signaling pathway. Furthermore, our data also suggest that functional Notch signaling is essential not only for BMP2-induced osteoblast differentiation but also for BMP signaling itself.
APA, Harvard, Vancouver, ISO, and other styles
6

Kusano, Shuichi, and Nancy Raab-Traub. "An Epstein-Barr Virus Protein Interacts with Notch." Journal of Virology 75, no. 1 (January 1, 2001): 384–95. http://dx.doi.org/10.1128/jvi.75.1.384-395.2001.

Full text
Abstract:
ABSTRACT The Epstein-Barr virus (EBV) BamHI A mRNAs were originally identified in cDNA libraries from nasopharyngeal carcinoma, where they are expressed at high levels. The RNAs are differentially spliced to form several open reading frames and also contain the BARF0 open reading frame at the 3′ end. One cDNA, RK-BARF0, included a potential endoplasmic reticulum-targeting signal peptide sequence. The RK-BARF0 protein is shown here to interact with the Notch4 ligand binding domain, using yeast two-hybrid screening, coimmunoprecipitation, and confocal microscopy. This interaction induces translocation of a portion of the full-length unprocessed Notch4 to the nucleus by using the Notch nuclear localization signal. These effects of RK-BARF0 on Notch intracellular location indicate that EBV possibly modulates Notch signaling. Unprocessed Notch4 was also detected in immunoprecipitated complexes from EBV-infected cells by using a rabbit antiserum raised against a BARF0-specific peptide. This finding provides additional evidence for expression of RK-BARF0 and its interaction with Notch during EBV infection. In EBV-infected, EBNA2-negative cells, RK-BARF0 induced the expression of EBV latent membrane protein 1 (LMP1), and this induction was dependent on the RK-BARF0/Notch interaction domain. The activation of LMP1 expression by RK-BARF0 may be responsible for expression of LMP1 in EBV latent infections in the absence of EBNA2.
APA, Harvard, Vancouver, ISO, and other styles
7

Kim, Su Hwan, Jin Hyun Kang, Tae Ki Uhm, Do Jin Kim, and Il Yup Chung. "Notch signaling promotes MUC5AC expression through epidermal growth factor receptor-extracellular regulated kinase pathway (163.1)." Journal of Immunology 186, no. 1_Supplement (April 1, 2011): 163.1. http://dx.doi.org/10.4049/jimmunol.186.supp.163.1.

Full text
Abstract:
Abstract Airway mucus hypersecretion and goblet cell hyperplasia plays an important role in the pathogenesis of several major airway inflammatory diseases. MUC5AC is a major component of the mucus produced by airway epithelial cells and considered to be a marker of mucus cell hyperplasia. In the study we investigate the involvement of Notch signaling in EGF-mediated expression of MUC5AC in human epithelial NCI-H292 cells. EGF stimulated the generation of Notch intracellular domain (NICD) in a RBP-Jκ-dependent manner. Treatment with γ-secretase inhibitor (GSI) or introduction of small interfering RNA directed against Notch1 reduced EGF-induced expression of MUC5AC mRNA and protein. The inhibitory effect of both GSI and Notch1 silencing on MUC5AC expression was accompanied by reduced generation of NICD and reduced expression of a Notch downstream target, Hes-1. Blockage of the Notch signaling with the two agents resulted in a decrease in ERK phosphorylation induced by EGF stimulation. ERK inhibitor inhibited MUC5AC production. These results indicate that ERK activation is necessary for Notch signaling to regulate the EGFR-mediated MUC5AC expression. Collectively, these results suggest that Notch signaling regulates the EGF-induced expression of MUC5AC through modulation of ERK activity.
APA, Harvard, Vancouver, ISO, and other styles
8

Hoebeke, Inge, Magda De Smedt, Inge Van de Walle, Katia Reynvoet, Greet De Smet, Jean Plum, and Georges Leclercq. "Overexpression of HES-1 is not sufficient to impose T-cell differentiation on human hematopoietic stem cells." Blood 107, no. 7 (April 1, 2006): 2879–81. http://dx.doi.org/10.1182/blood-2005-05-1815.

Full text
Abstract:
Abstract By retroviral overexpression of the Notch-1 intracellular domain (ICN) in human CD34+ hematopoietic stem cells (HSCs), we have shown previously that Notch-1 signaling promotes the T-cell fate and inhibits the monocyte and B-cell fate in several in vitro and in vivo differentiation assays. Here, we investigated whether the effects of constitutively active Notch-1 can be mimicked by overexpression of its downstream target gene HES1. Upon HES-1 retroviral transduction, human CD34+ stem cells had a different outcome in the differentiation assays as compared to ICN-transduced cells. Although HES-1 induced a partial block in B-cell development, it did not inhibit monocyte development and did not promote T/NK-cell-lineage differentiation. On the contrary, a higher percentage of HES-1-transduced stem cells remained CD34+. These experiments indicate that HES-1 alone is not able to substitute for Notch-1 signaling to induce T-cell differentiation of human CD34+ hematopoietic stem cells.
APA, Harvard, Vancouver, ISO, and other styles
9

Kindler, Thomas, Melanie G. Cornejo, Claudia Scholl, Jianing Liu, Dena S. Leeman, J. Erika Haydu, Stefan Fröhling, Benjamin H. Lee, and D. Gary Gilliland. "K-RasG12D–induced T-cell lymphoblastic lymphoma/leukemias harbor Notch1 mutations and are sensitive to γ-secretase inhibitors." Blood 112, no. 8 (October 15, 2008): 3373–82. http://dx.doi.org/10.1182/blood-2008-03-147587.

Full text
Abstract:
Abstract To study the impact of oncogenic K-Ras on T-cell leukemia/lymphoma development and progression, we made use of a conditional K-RasG12D murine knockin model, in which oncogenic K-Ras is expressed from its endogenous promoter. Transplantation of whole bone marrow cells that express oncogenic K-Ras into wild-type recipient mice resulted in a highly penetrant, aggressive T-cell leukemia/lymphoma. The lymphoblasts were composed of a CD4/CD8 double-positive population that aberrantly expressed CD44. Thymi of primary donor mice showed reduced cellularity, and immunophenotypic analysis demonstrated a block in differentiation at the double-negative 1 stage. With progression of disease, approximately 50% of mice acquired Notch1 mutations within the PEST domain. Of note, primary lymphoblasts were hypersensitive to γ-secretase inhibitor treatment, which is known to impair Notch signaling. This inhibition was Notch-specific as assessed by down-regulation of Notch1 target genes and intracellular cleaved Notch. We also observed that the oncogenic K-Ras-induced T-cell disease was responsive to rapamycin and inhibitors of the RAS/MAPK pathway. These data indicate that patients with T-cell leukemia with K-Ras mutations may benefit from therapies that target the NOTCH pathway alone or in combination with inhibition of the PI3K/AKT/MTOR and RAS/MAPK pathways.
APA, Harvard, Vancouver, ISO, and other styles
10

Wallberg, Annika E., Kia Pedersen, Urban Lendahl, and Robert G. Roeder. "p300 and PCAF Act Cooperatively To Mediate Transcriptional Activation from Chromatin Templates by Notch Intracellular Domains In Vitro." Molecular and Cellular Biology 22, no. 22 (November 15, 2002): 7812–19. http://dx.doi.org/10.1128/mcb.22.22.7812-7819.2002.

Full text
Abstract:
ABSTRACT Ligand activation of Notch receptors leads to release of the intracellular receptor domain (Notch IC), which translocates to the nucleus and interacts with the DNA-binding protein RBP-Jκ to control expression of specific target genes. A number of proteins have been shown to interact with Notch ICs and to modulate target gene activation, but the precise function of and interplay between these factors is not known. This report investigates the Notch IC-interacting proteins, p300, PCAF, and Mastermind-like 1 (MAML1), in an in vitro transcription system with purified factors and naked DNA or chromatin templates. MAML1, RBP-Jκ, and Notch IC are all required for optimal transcription from DNA, whereas transcription from chromatin requires, in addition, p300, which interacts with MAML1. The transcriptional activity of p300 requires acetyl coenzyme A, indicating that it functions as a histone acetyltransferase when mediating Notch IC function. PCAF is unable to promote transcription on its own but enhances Notch IC-mediated transcription from chromatin in conjunction with p300. These data define a critical role for p300 in the potentiation of Notch IC function by MAML1 and PCAF, provide the first evidence for cooperativity between PCAF and p300 in Notch IC function, and also indicate direct effects of RBP-Jκ, Notch IC, and MAML1 on the general transcription machinery.
APA, Harvard, Vancouver, ISO, and other styles

Dissertations / Theses on the topic "NOTCH 1 Intracellular Domain Indicate"

1

Petit-Paitel, Agnès. "Étude du rôle des présénilines dans la maturation de la protéine précurseur du peptide amyloi͏̈de dans la maladie d'Alzheimer et dans la maturation du récepteur Notch-1 : une fonction controversée." Paris 7, 2002. http://www.theses.fr/2002PA077149.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

KUAN, YU-HSIN, and 官予馨. "To investigate the proliferation mechanism of hMSC by transducing Notch 1 intracellular domain-coupled magnetic nanoparticle." Thesis, 2018. http://ndltd.ncl.edu.tw/handle/k66q4j.

Full text
Abstract:
碩士
國立中正大學
化學工程研究所
106
Notch family proteins play an important role in cell proliferation, differentiation, and apoptosis. The intracellular domain of activated Notch 1 receptor (NICD) can form complexes with transcription factors of the CSL family, resulting in subsequent activation of downstream target genes: Myc, p21, and the HES family. In this study, glutathione (GSH)-bound superoxide paramagnetic iron oxide nanoparticles (SPION@SiO2-MA-GSH) were prepared, and then separately coupled with the various lengths of Notch 1 intracellular domain fragments: N1IC, RA, and ANK via the interaction of GSH and GST-tagged proteins. Impulsed magnetic field was employed to deliver these protein-magnetic nanoparticle conjugates to human mesenchymal stem cells (hMSC) derived from bone marrow. After a culture for two days, the cell viability was measured by MTT assay. Then the regulated effect of NICD fragment to downstream gene and protein were checked by RT-PCR and Western-Blot respectively. The results show that the cell viability of hMSC was most significantly enhanced by transduction with N1IC-coupled SPION@SiO2-MA-GSH. The cell viability was improved from 100% to 139.63±19.69 %, which was than those transduced with conjugates of RA and ANK, 123.64±12.95 and 109.57±10.93 respectively, suggesting that the activation of downstream gene of N1IC caused hMSC to proliferate. From the results of RT-PCR and Western-Blot, we found that the expression level of HES 1 gene in transduced hMSC was much higher than that non-transduced hMSC. The higher HES 1 expression level resulted in the enhanced expression of phosphorylated-Akt protein, and then consequently promoted the proliferation of hMSC.
APA, Harvard, Vancouver, ISO, and other styles
3

Yu, Ching-ping, and 于青平. "Promote the directed differentiation of mesenchymal stem cells by the transduction of Notch-1 intracellular domain using magnetic nanoparticles." Thesis, 2007. http://ndltd.ncl.edu.tw/handle/75495558410076850980.

Full text
Abstract:
碩士
國立中正大學
化學工程所
95
Notch signaling has been shown to play an important role in cell proliferation and differentiation, as well as in cell-fate decision. Previous studies revealed that the transfection of Notch-1 intracellular domain (N1IC) could impair osteoblast differentiation and enhance adipogenesis in culture of stromal cells from murine bone marrow, and that bone marrow stromal cells from both rat and human sources can be induced to differentiate into cells with neuronal characteristics using gene transfection with N1IC and subsequent with certain trophic factors. Our study consists of two major parts. We first investigated Notch express in mesenchymal stem cell (MSC) and liver cells. We found that the Notch expression decreased during the differentiation from MSCs into liver cells, while Notch was expressed in both normal and liver tumor tissues. According to these results, we used a high efficient method for the transduction of MSCs with protein of N1IC and investigated the role of N1IC in the in vitro differentiation. The results suggest that N1IC has different roles in different differentiated cells. The transduction of N1IC could enhance the proliferation of MSCs and maintain MSCs in the state of progenitor. The transduced N1IC could also promote bone differentiation of MSCs.
APA, Harvard, Vancouver, ISO, and other styles
4

Mungamuri, Sathish Kumar. "Notch1-Induced Survival Signaling And Its Implications In Cancer Therapeutics." Thesis, 2006. http://hdl.handle.net/2005/455.

Full text
Abstract:
Notch receptors and ligands are type I transmembrane proteins that regulate development and differentiation during cell-cell contact. There are four Notch receptor homologues and five notch ligands, identified in humans till date. Upon ligand activation, Notch1 intracellular domain (NIC-1) is released into the cytoplasm, which binds to several proteins as well as translocates into the nucleus to effect the Notch signaling. In the absence of the activated Notch signaling, the Notch target genes are kept repressed by the transcriptional repressor C protein binding factor 1 (CBF1) also known as RBPjk or CSL for CBF1/Su(H)/Lag1. RBPjk binds to the sequence “CGTGGGAA” and acts as a constitutive repressor. Upon ligand dependent activation, NIC-1 enters into the nucler and converts RBPjk from transcriptional repressor to an activator. Notch binding to CSL replaces the SMRT corepressor complex with a coactivator complex including SKIP, Mastermind like 1 (MAML1) (Mastermind in Drosophila), and histone acetyl transferases PCAF, GCN5 and p300 activating the transcription of target genes. Mastermind-like (MAML), a family of transcriptional activator proteins comprising of 3 members 1 to 3, has been shown to be required for Notch signaling. MAML forms a ternary complex with RBPjk-NIC by directly interacting with NIC. In turn, MAML recruits the histone acetyl transferase p300/CBP, which acetylates the histones, thereby altering the structure of chromatin amenable for transcription. Activation of Notch pathway induces oncogenesis, which can be divided into two categories including 1) Inhibition of Apoptosis and 2) Induction of proliferation. In T cells, activation of Notch1 protects cells from T cell receptor, dexamethasone and etoposide-mediated apoptosis, Fas receptor-mediated signaling by up regulating IAP (Inhibitor of Apoptosis) and Bcl-2 families, as well as FLIP (FLICE-like inhibitor protein). Notch signaling also promotes the survival of T cells through maintenance of cell size as well as through the promotion of glucose uptake and metabolism. Notch-1 has been shown to protect against anoikis (apoptosis induced by matrix withdrawal) or p53-mediated apoptosis in immortalized epithelial cells, T cell receptor-induced apoptosis in mature cells and dexamethasone-mediated apoptosis in thymocytes. This study was carried out to functionally characterize NIC-1 (human Notch1-intracellular domain) as an inhibitor of apoptosis and to evaluate the therapeutic potential of reversal of this apoptosis inhibition. The main objectives of this study are 1. Construction of recombinant adenovirus expressing human Notch1-intracellular domain (Ad-NIC-1) and characterization of NIC-1 as an inhibitor of chemotherapy and p53-induced cytotoxicity and apoptosis. 2. Role of PI3 kinase -Akt/PKB -mTOR pathway in NIC-1-mediated inhibition of p53-induced apoptosis. 3. Essential role of association between mTOR and NIC-1 and the dependent NIC-1 phosphorylation in Notch1-mediated transcription and survival signaling. 4. Identification of NIC-1 as an inhibitor of E1A-induced apoptosis and the role of mTOR in NIC-1-mediated inhibition of E1A-induced apoptosis. Activated Notch1 was first linked to tumorigenesis through identification of a recurrent t(7;9)(q34;q34.3) chromosomal translocation involving the human Notch1 gene that is found in a subset of human pre-T-cell acute lymphoblastic leukemia’s (T-ALL). Deregulated Notch signaling is oncogenic, inhibits apoptosis and promotes survival. In order to understand survival signaling induced by Notch1 and its possible role in chemoresistance, we have generated a replication deficient recombinant adenovirus expressing human Notch1-intracellular domain (Ad-NIC-1) and shown that it produces functional NIC-1 protein. Using this overexpression system, we characterized that activated Notch1-inhibits chemotherapy and in particular p53 induced apoptosis. Notch1-mediated inhibition of p53-induced apoptosis does not include coactivator squelching. p53 was inefficient in binding to its DNA in NIC-1 overexpressing cells. The levels of phosphorylation at Ser15, Ser20, and Ser392 of p53 expressed from Ad-p53 significantly reduced in NIC-1 preinfected cells. These results suggest that NIC-1-mediated inhibition of p53-mediated apoptosis involves reduced DNA binding, reduced nuclear localization and reduced post translational modifications and thus reduced transactivation of its target genes. Notch1-mediated inhibition of p53 was found to occur mainly through mammalian target of rapamycin (mTOR) using PI3 kinase-Akt/PKB pathway, as the mTOR inhibitor; rapamycin treatment was able to reverse Notch-1 mediated inhibition of p53 and chemoresistance. Consistent with this, rapamycin failed to reverse NIC-1 induced chemoresistance in cells expressing rapamycin resistant mTOR. Our results also suggest that the N-terminal HEAT repeat and the kinase function of mTOR are essential for Notch mediated inhibition of p53. Further, ectopic expression of eIF4E, a translational regulator that acts downstream of mTOR, inhibited p53-induced apoptosis and conferred protection against p53-mediated cytotoxicity to similar extent as that of NIC-1 overexpression, but was not reversed by rapamycin, which indicates that eIF4E is the major target of mTOR in Notch1-mediated survival signaling. Notch1-intracellular domain (NIC-1), following proteolytic cleavage, binds to RBPjk and regulates transcription. Active NIC-1 located in the nucleus is phosphorylated, which makes it more stable and bind better to RBPjk. NIC-1 was also shown to bind to Deltex1 in the cytoplasm. Next, we studied the requirement of components of Notch1 signaling pathway for this function. By using variety of approaches, we found that both RBPjk and Maml1 and hence transcription activation is required for NIC-1-mediated survival signaling and inhibition of p53 functions. Interestingly, while we found the other Notch1 effector, Deltex1 is also required for above functions, Notch1 failed to activate PI3 kinase -Akt/PKB -mTOR pathway in Deltex1, but not in RBPjk silenced cells. Our results suggest that Notch-Deltex1 pathway activates PI3 kinase. Previous studies show that NIC-1 interacts with Deltex1 and Grb2 interacts with PI3 kinase. Our data shows that Deltex1 interacts with SH3 domain of Grb2. Since Notch1-Deltex1 and PI3 kinase-Grb2 interactions are known, we conclude that Notch1 activation of PI3 kinase involves Deltex1 and Grb2. We found activated mTOR was able to binds to NIC-1 and regulates its phosphorylation. Inhibition of mTOR either by PI3 kinase inhibitors or mTOR inhibitor treatment or silencing of Akt/PKB or mTOR reduced the phosphorylation of NIC-1 with the concomitant reduction in NIC-1-mediated transcription. Further, endogenous Notch1 receptor activated by the DSL ligand failed to activate transcription efficiently in rapamycin treated cells, implying a positive role for mTOR in mammalian Notch signaling. These studies reveal that Notch1 activates PI3 kinase -Akt/PKB -mTOR signaling through Deltex1 and subsequently activated mTOR modulates Notch1 signaling by direct binding and possibly thorough phosphorylation of the intracellular domain of Notch. Adenoviral E1A, in the absence of cooperating oncogene, suppresses primary tumor growth and reverses the transformed phenotype of human tumor cells by inducing apoptosis. E1A requires p53 for efficient induction of apoptosis and was shown to induce apoptosis by down regulating Akt and the activation of pro apoptotic factor p38 MAP kinase. Since our results suggest Notch1 inhibits chemotherapy and p53-induced apoptosis, we analyzed the ability of Notch1 to protect cells from E1A-induced apoptosis. Here we show that NIC-1 suppresses the ability of E1A to induce apoptosis. NIC-1 requires mTOR-dependent signal to inhibit E1A-mediated apoptosis, as the rapamycin, an mTOR inhibitor was able to completely reverse the ability of Notch1 to protect cells against E1A-induced apoptosis. The role of mTOR in NIC-1-mediated survival signaling was further confirmed by using the cells stably expressing rapamycin resistant mTOR. Rapamycin was able to reverse Notch1-mediated protection in cells expressing wild type mTOR but not in rapamycin resistant mTOR expressing cells. We also found that E1A was able to induce apoptosis in cells silenced for the pro apoptotic factor p38 and NIC-1 continued to inhibit E1A-induced apoptosis in these cells. These results confirm that Notch1 requires the activation of mTOR signaling but not p38 MAP kinase for inhibition of E1A-induced apoptosis. These results also suggest that the combination therapy utilizing E1A-mediated gene delivery in combination with inhibition of mTOR pathway may prove successful in treating Notch overexpressing cancers. Chemotherapy remains a major treatment modality for human cancers. Chemoresistance is a clinical problem that severely limits treatment success. It can be divided into two forms: intrinsic and acquired. Intrinsic resistance is the essence of oncogenic transformation, resulting from activation of oncogenes and the loss of tumor suppressors, and manifests itself as alterations in cell cycle checkpoints and apoptotic pathways. It is now widely accepted that the apoptotic capacity of the cancer cell is crucial in determining the response to chemotherapeutic agents. Indeed, several gene products that regulate apoptosis, i.e., p53, Akt and PI3K are frequently altered in cancer cells. In this study, we identified that cells with aberrant Notch1 signaling are chemoresistant. Activated Notch1 overexpression makes cells resistant to chemotherapy in a wild type p53 dependent manner. Notch protected p53 wild type cells but not p53 mutated or p53 deleted cells against chemotherapy induced cytotoxicity. Further, inactivation of p53 by specific silencing abrogated the ability of NIC-1 to protect H460 cells against adriamycin induced cytotoxicity. Most importantly, NIC-1 mediated chemoresistance can be reversed by blocking PI3 kinase -Akt/PKB -mTOR pathway. Collectively, these results suggest that cancers with activated Notch1 signaling are chemoresistant and provide basis for the reversal of chemoresistance.
APA, Harvard, Vancouver, ISO, and other styles

Book chapters on the topic "NOTCH 1 Intracellular Domain Indicate"

1

Hasan, Sana S., and Andreas Fischer. "Notch Signaling in Familial Cerebral Cavernous Malformations and Immunohistochemical Detection of Cleaved Notch1 Intracellular Domain." In Methods in Molecular Biology, 427–35. New York, NY: Springer US, 2020. http://dx.doi.org/10.1007/978-1-0716-0640-7_31.

Full text
APA, Harvard, Vancouver, ISO, and other styles

Conference papers on the topic "NOTCH 1 Intracellular Domain Indicate"

1

Jorgensen, M. J., MJ Rabiet, A. B. Cantor, B. Furie, C. L. Brown, C. B. Shoemaker, and B. C. Furie. "VITAMIN K-DEPENDENT γ-CARBOXYLATION OF FACTOR IX REQUIRES A RECOGNITION SITE CONTAINED WITHIN THE PROPEPTIDE." In XIth International Congress on Thrombosis and Haemostasis. Schattauer GmbH, 1987. http://dx.doi.org/10.1055/s-0038-1643564.

Full text
Abstract:
The vitamin K-dependent proteins, including Factor IX (FIX), are calcium-binding proteins that undergo vitamin K-dependent post-translational modification to convert amino terminal glutamic aoid residues to Gla residues. Sequence homology among the propeptides of these proteins suggests a role for this region in designating the adjacent glutamic acid-rich domain for γ-carboxylation during intraoellular processing. Mutations vere made in the propeptide (residues -1 to -18) of FIX, and the effects on γ-carboxylation were assessed. The human FIX cDNA coding sequenoe was modified using oligonucleotide-directed site-specific mutagenesis and was expressed in Chinese hamster ovary cells. The extent of γ-carboxylation of secreted FIX was determined by (1) ability to interact with conformation-specific antibodies directed against the Gla-dependent, metal-stabilized, native structure of FIX, and (2) direct Gla analysis of the alkaline hydrolysate. Using the unmodified coding sequence, 64 ± 17 % of recombinant Factor IX bound to the conformation-specific antibodies, and 9.4 ± 0.7 Gla residues were found (compared with 12 Gla in plasma FIX). When the 18-residue propeptide was deleted, secreted FIX contained no detectable native FIX antigen and no detectable Gla. Similarly, point mutations leading to substitution of Ala for Phe at residue -16 or Glu for Ala at residue -10 led to secretion of FIX containing 2% and 6% native antigen, respectively, and approximately 1-2 Gla residues. The molecular weight of each of the reoombinant FIX species, as estimated by SDS-PAGE, was identical to that of plasma FIX. NH2-terminal sequence analysis of the mutant FIX speoies yielded the NH2-terminal sequence of plasma FIX. These data indicate that the mutations made in the propeptide did not interfere with intracellular proteolytic prooessing of FIX. We conolude that the FIX propeptide participates in defining a recognition site that designates an adjacent glutamic acid-rich domain for γ-carboxylation. The association of the propeptide with the γ-carboxylation recognition site provides the first demonstration of a specific function served by a propeptide in post-translational protein processing.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography