Notch1 Receptor

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 14133 Experts worldwide ranked by ideXlab platform

Tien-shun Yeh - One of the best experts on this subject based on the ideXlab platform.

  • the autonomous notch signal pathway is activated by baicalin and baicalein but is suppressed by niclosamide in k562 cells
    Journal of Cellular Biochemistry, 2009
    Co-Authors: Anming Wang, Yuchih Liang, Yenchou Chen, Yuhming Hwu, Tien-shun Yeh
    Abstract:

    The Notch signaling pathway plays important roles in a variety of cellular processes. Aberrant transduction of Notch signaling contributes to many diseases and cancers in humans. The Notch Receptor intracellular domain, the activated form of Notch Receptor, is extremely difficult to detect in normal cells. However, it can activate signaling at very low protein concentration to elicit its biological effects. In the present study, a cell based luciferase reporter gene assay was established in K562 cells to screen drugs which could modulate the endogenous CBF1-dependent Notch signal pathway. Using this system, we found that the luciferase activity of CBF1-dependent reporter gene was activated by baicalin and baicalein but suppressed by niclosamide in both dose- and time-dependent manners. Treatment with these drugs modulated endogenous Notch signaling and affected mRNA expression levels of Notch1 Receptor and Notch target genes in K562 cells. Additionally, erythroid differentiation of K562 cells was suppressed by baicalin and baicalein yet was promoted by niclosamide. Colony-forming ability in soft agar was decreased after treatment with baicalin and baicalein, but was not affected in the presence of niclosamide. Thus, modulation of Notch signaling after treatment with any of these three drugs may affect tumorigenesis of K562 cells suggesting that these drugs may have therapeutic potential for those tumors associated with Notch signaling. Taken together, this system could be beneficial for screening of drugs with potential to treat Notch signal pathway-associated diseases.

  • the activated Notch1 Receptor cooperates with α enolase and mbp 1 in modulating c myc activity
    Molecular and Cellular Biology, 2008
    Co-Authors: Kai Wen Hsu, Rong-hong Hsieh, Min-jen Tseng, Yan-hwa Wu Lee, Chihong Chao, Tien-shun Yeh
    Abstract:

    The Notch signal pathway plays multifaceted roles to promote or suppress tumorigenesis. The Notch1 Receptor intracellular domain (N1IC), the activated form of the Notch1 Receptor, activates the c-myc proto-oncogene. The complex of N1IC and transcription factor YY1 binds to the human c-myc promoter to enhance c-myc expression in a CBF1-independent manner. Here we demonstrated that N1IC interacted with the c-Myc-regulating proteins alpha-enolase and c-myc promoter binding protein 1 (MBP-1). Both alpha-enolase and MBP-1 suppressed the N1IC-enhanced activity of the c-myc promoter in a CBF1-independent manner. The YY1 response element in front of the P2 c-myc promoter was essential and sufficient for the modulation of c-myc by N1IC and alpha-enolase or MBP-1. Furthermore, N1IC, YY1, and alpha-enolase or MBP-1 but not CBF1 bound to the c-myc promoter through associating with the YY1 response element. Hemin-induced erythroid differentiation was suppressed by N1IC in K562 cells. This suppression was relieved by the expression of alpha-enolase and MBP-1. In addition, both alpha-enolase and MBP-1 suppressed the N1IC-enhanced colony-forming ability through c-myc. These results indicate that the activated Notch1 Receptor and alpha-enolase or MBP-1 cooperate in controlling c-myc expression through binding the YY1 response element of the c-myc promoter to regulate tumorigenesis.

  • The CBF1-independent Notch1 signal pathway activates human c-myc expression partially via transcription factor YY1.
    Carcinogenesis, 2007
    Co-Authors: Wan Ru Liao, Rong-hong Hsieh, Min-jen Tseng, Ru-tsun Mai, Yan-hwa Wu Lee, Kai Wen Hsu, Tien-shun Yeh
    Abstract:

    Transcription factor Ying Yang 1 (YY1) indirectly regulates the C promoter-binding factor 1 (CBF1)-dependent Notch1 signaling via direct interaction with the Notch1 Receptor intracellular domain (N1IC) on CBF1-response elements. To evaluate the possibility that the N1IC might modulate the gene expression of YY1 target genes through associating with YY1 on the YY1-response elements, we herein investigated the effect of Notch1 signaling on the expression of YY1 target genes. We found that the N1IC bound to the double-stranded oligonucleotides of YY1-response element to activate luciferase activity of the reporter gene with YY1-response elements through a CBF1-independent manner. Furthermore, the N1IC also bound to the promoter of human c-myc oncogene, a YY1 target gene, to elevate c-myc expression via a CBF1-independent pathway. The activation of reporter genes with YY1-response elements or human c-myc promoter by N1IC depended on the formation of N1IC-YY1-associated complex. To delineate the role of the Notch signal pathway in tumorigenesis, K562 cell lines expressing the N1IC were established. Compared with control cells, the proliferation and the tumor growth of N1IC-expressing K562 cells were suppressed. Taken together, these results suggest that the N1IC enhances the human c-myc promoter activity that is partially modulated by YY1 through a CBF1-independent pathway. However, the enhancement of c-myc expression by N1IC is insufficient to promote the tumor growth of K562 cells.

  • Association of transcription factor YY1 with the high molecular weight Notch complex suppresses the transactivation activity of Notch.
    The Journal of biological chemistry, 2003
    Co-Authors: Tien-shun Yeh, Yu-min Lin, Rong-hong Hsieh, Min-jen Tseng
    Abstract:

    Notch Receptors are evolutionarily conserved from Drosophila to human and play important roles in cell fate decisions. After ligand binding, Notch Receptors are cleaved to release their intracellular domains. The intracellular domains, the activated form of Notch Receptors, are then translocated into the nucleus where they interact with other transcriptional machinery to regulate the expression of cellular genes. To dissect the molecular mechanisms of Notch signaling, the cellular targets that interact with Notch1 Receptor intracellular domain (N1IC) were screened. In this study, we found that endogenous transcription factor Ying Yang 1 (YY1) was associated with exogenous N1IC in human K562 erythroleukemic cells. The ankyrin (ANK) domain of N1IC and zinc finger domains of YY1 were essential for the association of N1IC and YY1 according to the pull-down assay of glutathione S-transferase fusion proteins. Furthermore, both YY1 and N1IC were present in a large complex of the nucleus to suppress the luciferase reporter activity transactivated by Notch signaling. The transcription factor YY1 indirectly regulated the transcriptional activity of the wild-type CBF1-response elements via the direct interaction of N1IC and CBF1. We also demonstrated the association between endogenous N1IC and intrinsic YY1 in human acute T-cell lymphoblastic leukemia cell lines. Taken together, these results indicate that transcription factor YY1 may modulate Notch signaling via association with the high molecular weight Notch complex.

Rebeca Pineirosabaris - One of the best experts on this subject based on the ideXlab platform.

  • coronary arterial development is regulated by a dll4 jag1 ephrinb2 signaling cascade
    eLife, 2019
    Co-Authors: Stanislao Igor Travisano, Rebeca Pineirosabaris, Vera Lúcia De Oliveira, Joaquim Gregobessa, Belen Prados
    Abstract:

    Coronaries are essential for myocardial growth and heart function. Notch is crucial for mouse embryonic angiogenesis, but its role in coronary development remains uncertain. We show Jag1, Dll4 and activated Notch1 Receptor expression in sinus venosus (SV) endocardium. Endocardial Jag1 removal blocks SV capillary sprouting, while Dll4 inactivation stimulates excessive capillary growth, suggesting that ligand antagonism regulates coronary primary plexus formation. Later endothelial ligand removal, or forced expression of Dll4 or the glycosyltransferase Mfng, blocks coronary plexus remodeling, arterial differentiation, and perivascular cell maturation. Endocardial deletion of Efnb2 phenocopies the coronary arterial defects of Notch mutants. Angiogenic rescue experiments in ventricular explants, or in primary human endothelial cells, indicate that EphrinB2 is a critical effector of antagonistic Dll4 and Jag1 functions in arterial morphogenesis. Thus, coronary arterial precursors are specified in the SV prior to primary coronary plexus formation and subsequent arterial differentiation depends on a Dll4-Jag1-EphrinB2 signaling cascade.

Larry H Matherly - One of the best experts on this subject based on the ideXlab platform.

  • Notch1 Receptor regulates akt protein activation loop thr308 dephosphorylation through modulation of the pp2a phosphatase in phosphatase and tensin homolog pten null t cell acute lymphoblastic leukemia cells
    Journal of Biological Chemistry, 2013
    Co-Authors: Eric C Hales, Amanda Larson Gedman, Jeffrey W Taub, Larry H Matherly
    Abstract:

    Abstract Notch1 activating mutations occur in more than 50% of T-cell acute lymphoblastic leukemia (T-ALL) cases and increase expression of Notch1 target genes, some of which activate AKT. HES1 transcriptionally silences phosphatase and tensin homolog (PTEN), resulting in AKT activation, which is reversed by Notch1 inhibition with γ-secretase inhibitors (GSIs). Mutational loss of PTEN is frequent in T-ALL and promotes resistance to GSIs due to AKT activation. GSI treatments increased AKT-Thr308 phosphorylation and signaling in PTEN-deficient, GSI-resistant T-ALL cell lines (Jurkat, CCRF-CEM, and MOLT3), suggesting that Notch1 represses AKT independent of its PTEN transcriptional effects. AKT-Thr308 phosphorylation and downstream signaling were also increased by knocking down Notch1 in Jurkat (N1KD) cells. This was blocked by treatment with the AKT inhibitor perifosine. The PI3K inhibitor wortmannin and the protein phosphatase type 2A (PP2A) inhibitor okadaic acid both impacted AKT-Thr308 phosphorylation to a greater extent in nontargeted control than N1KD cells, suggesting decreased dephosphorylation of AKT-Thr308 by PP2A in the latter. Phosphorylations of AMP-activated protein kinaseα (AMPKα)-Thr172 and p70S6K-Thr389, both PP2A substrates, were also increased in both N1KD and GSI-treated cells and responded to okadaic acid treatment. A transcriptional regulatory mechanism was implied because ectopic expression of dominant-negative mastermind-like protein 1 increased and wild-type HES1 decreased phosphorylation of these PP2A targets. This was independent of changes in PP2A subunit levels or in vitro PP2A activity, but was accompanied by decreased association of PP2A with AKT in N1KD cells. These results suggest that Notch1 can regulate PP2A dephosphorylation of critical cellular regulators including AKT, AMPKα, and p70S6K.

  • Notch1 Receptor regulates akt protein activation loop thr308 dephosphorylation through modulation of the pp2a phosphatase in phosphatase and tensin homolog pten null t cell acute lymphoblastic leukemia cells
    Journal of Biological Chemistry, 2013
    Co-Authors: Eric C Hales, Amanda Larson Gedman, Jeffrey W Taub, Steven Orr, Larry H Matherly
    Abstract:

    Notch1 activating mutations occur in more than 50% of T-cell acute lymphoblastic leukemia (T-ALL) cases and increase expression of Notch1 target genes, some of which activate AKT. HES1 transcriptionally silences phosphatase and tensin homolog (PTEN), resulting in AKT activation, which is reversed by Notch1 inhibition with γ-secretase inhibitors (GSIs). Mutational loss of PTEN is frequent in T-ALL and promotes resistance to GSIs due to AKT activation. GSI treatments increased AKT-Thr308 phosphorylation and signaling in PTEN-deficient, GSI-resistant T-ALL cell lines (Jurkat, CCRF-CEM, and MOLT3), suggesting that Notch1 represses AKT independent of its PTEN transcriptional effects. AKT-Thr308 phosphorylation and downstream signaling were also increased by knocking down Notch1 in Jurkat (N1KD) cells. This was blocked by treatment with the AKT inhibitor perifosine. The PI3K inhibitor wortmannin and the protein phosphatase type 2A (PP2A) inhibitor okadaic acid both impacted AKT-Thr308 phosphorylation to a greater extent in nontargeted control than N1KD cells, suggesting decreased dephosphorylation of AKT-Thr308 by PP2A in the latter. Phosphorylations of AMP-activated protein kinaseα (AMPKα)-Thr172 and p70S6K-Thr389, both PP2A substrates, were also increased in both N1KD and GSI-treated cells and responded to okadaic acid treatment. A transcriptional regulatory mechanism was implied because ectopic expression of dominant-negative mastermind-like protein 1 increased and wild-type HES1 decreased phosphorylation of these PP2A targets. This was independent of changes in PP2A subunit levels or in vitro PP2A activity, but was accompanied by decreased association of PP2A with AKT in N1KD cells. These results suggest that Notch1 can regulate PP2A dephosphorylation of critical cellular regulators including AKT, AMPKα, and p70S6K. Background: PTEN loss promotes resistance to γ-secretase inhibitors by increasing AKT signaling in T-cell acute lymphoblastic leukemia (T-ALL) with mutant activated Notch1. Results: Notch1 inhibition increases AKT phosphorylation and involves the PP2A phosphatase. Conclusion: Notch1 regulates PP2A dephosphorylation of AKT-Thr308 by impacting association of PP2A with AKT. Significance: Better understanding of regulation of AKT signaling by Notch1 may lead to new therapies for T-ALL.

Jeffrey W Taub - One of the best experts on this subject based on the ideXlab platform.

  • Notch1 Receptor regulates akt protein activation loop thr308 dephosphorylation through modulation of the pp2a phosphatase in phosphatase and tensin homolog pten null t cell acute lymphoblastic leukemia cells
    Journal of Biological Chemistry, 2013
    Co-Authors: Eric C Hales, Amanda Larson Gedman, Jeffrey W Taub, Larry H Matherly
    Abstract:

    Abstract Notch1 activating mutations occur in more than 50% of T-cell acute lymphoblastic leukemia (T-ALL) cases and increase expression of Notch1 target genes, some of which activate AKT. HES1 transcriptionally silences phosphatase and tensin homolog (PTEN), resulting in AKT activation, which is reversed by Notch1 inhibition with γ-secretase inhibitors (GSIs). Mutational loss of PTEN is frequent in T-ALL and promotes resistance to GSIs due to AKT activation. GSI treatments increased AKT-Thr308 phosphorylation and signaling in PTEN-deficient, GSI-resistant T-ALL cell lines (Jurkat, CCRF-CEM, and MOLT3), suggesting that Notch1 represses AKT independent of its PTEN transcriptional effects. AKT-Thr308 phosphorylation and downstream signaling were also increased by knocking down Notch1 in Jurkat (N1KD) cells. This was blocked by treatment with the AKT inhibitor perifosine. The PI3K inhibitor wortmannin and the protein phosphatase type 2A (PP2A) inhibitor okadaic acid both impacted AKT-Thr308 phosphorylation to a greater extent in nontargeted control than N1KD cells, suggesting decreased dephosphorylation of AKT-Thr308 by PP2A in the latter. Phosphorylations of AMP-activated protein kinaseα (AMPKα)-Thr172 and p70S6K-Thr389, both PP2A substrates, were also increased in both N1KD and GSI-treated cells and responded to okadaic acid treatment. A transcriptional regulatory mechanism was implied because ectopic expression of dominant-negative mastermind-like protein 1 increased and wild-type HES1 decreased phosphorylation of these PP2A targets. This was independent of changes in PP2A subunit levels or in vitro PP2A activity, but was accompanied by decreased association of PP2A with AKT in N1KD cells. These results suggest that Notch1 can regulate PP2A dephosphorylation of critical cellular regulators including AKT, AMPKα, and p70S6K.

  • Notch1 Receptor regulates akt protein activation loop thr308 dephosphorylation through modulation of the pp2a phosphatase in phosphatase and tensin homolog pten null t cell acute lymphoblastic leukemia cells
    Journal of Biological Chemistry, 2013
    Co-Authors: Eric C Hales, Amanda Larson Gedman, Jeffrey W Taub, Steven Orr, Larry H Matherly
    Abstract:

    Notch1 activating mutations occur in more than 50% of T-cell acute lymphoblastic leukemia (T-ALL) cases and increase expression of Notch1 target genes, some of which activate AKT. HES1 transcriptionally silences phosphatase and tensin homolog (PTEN), resulting in AKT activation, which is reversed by Notch1 inhibition with γ-secretase inhibitors (GSIs). Mutational loss of PTEN is frequent in T-ALL and promotes resistance to GSIs due to AKT activation. GSI treatments increased AKT-Thr308 phosphorylation and signaling in PTEN-deficient, GSI-resistant T-ALL cell lines (Jurkat, CCRF-CEM, and MOLT3), suggesting that Notch1 represses AKT independent of its PTEN transcriptional effects. AKT-Thr308 phosphorylation and downstream signaling were also increased by knocking down Notch1 in Jurkat (N1KD) cells. This was blocked by treatment with the AKT inhibitor perifosine. The PI3K inhibitor wortmannin and the protein phosphatase type 2A (PP2A) inhibitor okadaic acid both impacted AKT-Thr308 phosphorylation to a greater extent in nontargeted control than N1KD cells, suggesting decreased dephosphorylation of AKT-Thr308 by PP2A in the latter. Phosphorylations of AMP-activated protein kinaseα (AMPKα)-Thr172 and p70S6K-Thr389, both PP2A substrates, were also increased in both N1KD and GSI-treated cells and responded to okadaic acid treatment. A transcriptional regulatory mechanism was implied because ectopic expression of dominant-negative mastermind-like protein 1 increased and wild-type HES1 decreased phosphorylation of these PP2A targets. This was independent of changes in PP2A subunit levels or in vitro PP2A activity, but was accompanied by decreased association of PP2A with AKT in N1KD cells. These results suggest that Notch1 can regulate PP2A dephosphorylation of critical cellular regulators including AKT, AMPKα, and p70S6K. Background: PTEN loss promotes resistance to γ-secretase inhibitors by increasing AKT signaling in T-cell acute lymphoblastic leukemia (T-ALL) with mutant activated Notch1. Results: Notch1 inhibition increases AKT phosphorylation and involves the PP2A phosphatase. Conclusion: Notch1 regulates PP2A dephosphorylation of AKT-Thr308 by impacting association of PP2A with AKT. Significance: Better understanding of regulation of AKT signaling by Notch1 may lead to new therapies for T-ALL.

Jon C Aster - One of the best experts on this subject based on the ideXlab platform.

  • leukemia associated mutations within the Notch1 heterodimerization domain fall into at least two distinct mechanistic classes
    Molecular and Cellular Biology, 2006
    Co-Authors: Michael J Malecki, Jon C Aster, Cheryll Sanchezirizarry, Jennifer L Mitchell, Gavin Histen, Stephen C Blacklow
    Abstract:

    The Notch1 Receptor is cleaved within its extracellular domain by furin during its maturation, yielding two subunits that are held together noncovalently by a juxtamembrane heterodimerization (HD) domain. Normal Notch1 signaling is initiated by the binding of ligand to the extracellular subunit, which renders the transmembrane subunit susceptible to two successive cleavages within and C terminal to the heterodimerization domain, catalyzed by metalloproteases and gamma-secretase, respectively. Because mutations in the heterodimerization domain of Notch1 occur frequently in human T-cell acute lymphoblastic leukemia (T-ALL), we assessed the effect of 16 putative tumor-associated mutations on Notch1 signaling and HD domain stability. We show here that 15 of the 16 mutations activate canonical Notch1 signaling. Increases in signaling occur in a ligand-independent fashion, require gamma-secretase activity, and correlate with an increased susceptibility to cleavage by metalloproteases. The activating mutations cause soluble Notch1 heterodimers to dissociate more readily, either under native conditions (n = 3) or in the presence of urea (n = 11). One mutation, an insertion of 14 residues immediately N terminal to the metalloprotease cleavage site, increases metalloprotease sensitivity more than all others, despite a negligible effect on heterodimer stability by comparison, suggesting that the insertion may expose the S2 site by repositioning it relative to protective Notch1 ectodomain residues. Together, these studies show that leukemia-associated HD domain mutations render Notch1 sensitive to ligand-independent proteolytic activation through two distinct mechanisms.

  • t cell acute lymphoblastic leukemia lymphoma a human cancer commonly associated with aberrant Notch1 signaling
    Current Opinion in Hematology, 2004
    Co-Authors: Warren S Pear, Jon C Aster
    Abstract:

    Purpose of review Although constitutively activated forms of the Notch1 Receptor are potent inducers of T cell acute lymphoblastic leukemia/lymphoma when expressed in the bone marrow stem cells of mice, the known involvement of Notch1 in human T cell acute lymphoblastic leukemia/lymphoma has been restricted to very rare tumors associated with a (7;9) chromosomal translocation involving the Notch1 gene. This picture has changed dramatically in the past year with the discovery of frequent mutations involving Notch1 in human T cell acute lymphoblastic leukemia/lymphoma. Recent findings Notch1 point mutations, insertions, and deletions producing aberrant increases in Notch1 signaling are frequently present in both childhood and adult T cell acute lymphoblastic leukemia/lymphoma and are detected in tumors from all major molecular subtypes. These observations are particularly important in the light of experiments using human and murine T cell acute lymphoblastic leukemia/lymphoma cell lines indicating that Notch1 signals are required for sustained growth and, in a subset of lines, survival. This inference is based in part on experiments conducted with small molecule inhibitors of gamma-secretase, a protease required for normal NOTCH signal transduction and the activity of the mutated forms of Notch1 found commonly in human T cell acute lymphoblastic leukemia/lymphoma. Summary These findings support a central role for aberrant NOTCH signaling in the pathogenesis of human T cell acute lymphoblastic leukemia/lymphoma, and they provide a rationale for trials of NOTCH inhibitors, such as gamma-secretase antagonists, in this aggressive human malignancy.

  • isolation and functional analysis of a cdna for human jagged2 a gene encoding a ligand for the Notch1 Receptor
    Molecular and Cellular Biology, 1997
    Co-Authors: B Luo, Jon C Aster, Robert P Hasserjian, Frank C Kuo, Jeffrey Sklar
    Abstract:

    Signaling through Notch Receptors has been implicated in the control of cellular differentiation in animals ranging from nematodes to humans. Starting from a human expressed sequence tag-containing sequence resembling that of Serrate, the gene for a ligand of Drosophila melanogaster Notch, we assembled a full-length cDNA, now called human Jagged2, from overlapping cDNA clones. The full-length cDNA encodes a polypeptide having extensive sequence homology to Serrate (40.6% identity and 58.7% similarity) and even greater homology to several putative mammalian Notch ligands that have subsequently been described. When in situ hybridization was performed, expression of the murine Jagged2 homolog was found to be highest in fetal thymus, epidermis, foregut, dorsal root ganglia, and inner ear. In Northern blot analysis of RNA from tissues of 2-week-old mice, the 5.0-kb Jagged2 transcript was most abundant in heart, lung, thymus, skeletal muscle, brain, and testis. Immunohistochemistry revealed coexpression of Jagged2 and Notch1 within thymus and other fetal murine tissues, consistent with interaction of the two proteins in vivo. Coculture of fibroblasts expressing human Jagged2 with murine C2C12 myoblasts inhibited myogenic differentiation, accompanied by increased Notch1 and the appearance of a novel 115-kDa Notch1 fragment. Exposure of C2C12 cells to Jagged2 led to increased amounts of Notch mRNA as well as mRNAs for a second Notch Receptor, Notch3, and a second Notch ligand, Jagged1. Constitutively active forms of Notchl in C2C12 cells also induced increased levels of the same set of mRNAs, suggesting positive feedback control of these genes initiated by binding of Jagged2 to Notch1. This feedback control may function in vivo to coordinate differentiation across certain groups of progenitor cells adopting identical cell fates.