scholarly journals Constitutive Notch pathway activation in murine ZMYM2-FGFR1–induced T-cell lymphomas associated with atypical myeloproliferative disease

Blood ◽  
2011 ◽  
Vol 117 (25) ◽  
pp. 6837-6847 ◽  
Author(s):  
MingQiang Ren ◽  
John K. Cowell

Abstract The ZMYM2-FGFR1 (formerly known as ZNF198-FGFR1) fusion kinase induces stem cell leukemia–lymphoma syndrome (SCLL), a hematologic malignancy characterized by rapid transformation to acute myeloid leukemia and T-lymphoblastic lymphoma. In the present study, we demonstrate frequent, constitutive activation of Notch1 and its downstream target genes in T-cell lymphomas that arose in a murine model of ZMYM2-FGFR1 SCLL. Notch up-regulation was also demonstrated in human SCLL- and FGFR1OP2-FGFR1-expressing KG-1 cells. To study the role of Notch in T-cell lymphomagenesis, we developed a highly tumorigenic cell line from ZMYM2-FGFR1–expressing cells. Pharmacologic inhibition of Notch signaling in these cells using γ-secretase inhibitors significantly delayed leukemogenesis in vivo. shRNA targeting of Notch1, as well as c-promoter–binding factor 1 (CBF1) and mastermind-like 1 (MAML1), 2 essential cofactors involved in transcriptional activation of Notch target genes, also significantly delayed or inhibited tumorigenesis in vivo. Mutation analysis demonstrated that 5′ promoter deletions and alternative promoter usage were responsible for constitutive activation of Notch1 in all T-cell lymphomas. These data demonstrate the importance of Notch signaling in the etiology of SCLL, and suggest that targeting this pathway could provide a novel strategy for molecular therapies to treat SCLL patients.

2011 ◽  
Vol 22 (17) ◽  
pp. 3242-3252 ◽  
Author(s):  
Dieter Maier ◽  
Patricia Kurth ◽  
Adriana Schulz ◽  
Andrew Russell ◽  
Zhenyu Yuan ◽  
...  

In metazoans, the highly conserved Notch pathway drives cellular specification. On receptor activation, the intracellular domain of Notch assembles a transcriptional activator complex that includes the DNA-binding protein CSL, a composite of human C-promoter binding factor 1, Suppressor of Hairless of Drosophila melanogaster [Su(H)], and lin-12 and Glp-1 phenotype of Caenorhabditis elegans. In the absence of ligand, CSL represses Notch target genes. However, despite the structural similarity of CSL orthologues, repression appears largely diverse between organisms. Here we analyze the Notch repressor complex in Drosophila, consisting of the fly CSL protein, Su(H), and the corepressor Hairless, which recruits general repressor proteins. We show that the C-terminal domain of Su(H) is necessary and sufficient for forming a high-affinity complex with Hairless. Mutations in Su(H) that affect interactions with Notch and Mastermind have no effect on Hairless binding. Nonetheless, we demonstrate that Notch and Hairless compete for CSL in vitro and in cell culture. In addition, we identify a site in Hairless that is crucial for binding Su(H) and subsequently show that this Hairless mutant is strongly impaired, failing to properly assemble the repressor complex in vivo. Finally, we demonstrate Hairless-mediated inhibition of Notch signaling in a cell culture assay, which hints at a potentially similar repression mechanism in mammals that might be exploited for therapeutic purposes.


Blood ◽  
2010 ◽  
Vol 116 (25) ◽  
pp. 5443-5454 ◽  
Author(s):  
Robin Jeannet ◽  
Jérôme Mastio ◽  
Alejandra Macias-Garcia ◽  
Attila Oravecz ◽  
Todd Ashworth ◽  
...  

Abstract The Notch pathway is frequently activated in T-cell acute lymphoblastic leukemias (T-ALLs). Of the Notch receptors, Notch1 is a recurrent target of gain-of-function mutations and Notch3 is expressed in all T-ALLs, but it is currently unclear how these receptors contribute to T-cell transformation in vivo. We investigated the role of Notch1 and Notch3 in T-ALL progression by a genetic approach, in mice bearing a knockdown mutation in the Ikaros gene that spontaneously develop Notch-dependent T-ALL. While deletion of Notch3 has little effect, T cell–specific deletion of floxed Notch1 promoter/exon 1 sequences significantly accelerates leukemogenesis. Notch1-deleted tumors lack surface Notch1 but express γ-secretase–cleaved intracellular Notch1 proteins. In addition, these tumors accumulate high levels of truncated Notch1 transcripts that are caused by aberrant transcription from cryptic initiation sites in the 3′ part of the gene. Deletion of the floxed sequences directly reprograms the Notch1 locus to begin transcription from these 3′ promoters and is accompanied by an epigenetic reorganization of the Notch1 locus that is consistent with transcriptional activation. Further, spontaneous deletion of 5′ Notch1 sequences occurs in approximately 75% of Ikaros-deficient T-ALLs. These results reveal a novel mechanism for the oncogenic activation of the Notch1 gene after deletion of its main promoter.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2700-2700
Author(s):  
Jun Xia ◽  
Matthew RM Jotte ◽  
Stephanie Sun ◽  
Geoffrey L. Uy ◽  
Abi Vainstein ◽  
...  

Abstract T cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic malignancy that accounts for 10-15% of pediatric and 25% of adult ALL cases. Approximately 20-25% of pediatric and 50% of adult patients with T-ALL relapse following induction therapy, and the prognosis after relapse is dismal, with 3-year event-free survival of only 10-15%. Thus, there is a clear unmet clinical need for better therapies for T-ALL. CXCL12 (stromal-derived factor-1, SDF1) is a CXC chemokine that is constitutively expressed at high levels in the bone marrow. CXCR4 is the major receptor for CXCL12 and is by far the most highly expressed chemokine receptor on T-ALL cells. Two groups recently showed that genetic loss of CXCR4 signaling in murine or human T-ALL cells markedly suppressed their growth in vivo. Here we explore the hypothesis that inhibition of the CXCR4 signaling with a potent new CXCR4 antagonist, BL-8040 alone, or in combination with ABT263 (navitoclax), a BCL2/BCLXL antagonist, will have therapeutic activity against T-ALL. To test this hypothesis, we xenotransplanted a human T-ALL cell line (P12/Ichikawa cells) or 5 different patient-derived T-ALL xenografts into non-irradiated NSG mice. In each case, the T-ALL cells were allowed to engraft and then mice were randomly assigned to one of four treatment groups: 1) vehicle control; 2) BL-8040 alone; 3) ABT263 alone; 4) combined BL-8040 and ABT263. Treatment was given for 2 weeks, and leukemia burden monitored weekly by bioluminescent imaging or by flow cytometry to quantify human CD45+ T-ALL cells in the blood. A minimum of 10 mice in each cohort from two independent experiments were analyzed (Figure 1). A significant reduction in T-ALL burden was observed in all T ALL samples after treatment with BL8040 alone, with a mean fold-decrease compared with control mice after 2 weeks of treatment of 966 ± 651 (range 16-3,486). Consistent with a prior report, the response to ABT263 alone was more variable, with responses seen in 4 of 6 T-ALL samples. All T-ALL samples had a marked response to the combination of BL-8040 and ABT263, with a mean fold decrease of 4,389 ± 2,602 (range, 139-15,199). A previous study suggested that inhibition of CXCR4 signaling may suppress Myc expression (Pitt et al, Cancer Cell, 2015), which is relevant, since overexpression of Myc secondary to mutations in the Notch pathway are common in T-ALL. However, we observed no difference in Myc protein expression or expression of Myc target genes following BL-8040 treatment. Consistent with this observation, treatment with BL8040 had no impact on the cell cycle status of T-ALL cells in vivo. Of note, we observed a marked decrease in Akt and Erk phosphorylation following BL8040, suggesting that CXCR4 signaling may regulate T ALL survival in vivo by suppressing these pathways. Conclusion. Collectively, these data suggest that BL-8040, either alone or in combination with ABT263, is highly active in T-ALL and support our ongoing clinical trial of BL-8040 in combination with nelarabine for patients with relapsed T-ALL (NCT02763384). Figure 1. Figure 1. Disclosures Uy: GlycoMimetics: Consultancy; Curis: Consultancy. Vainstein:Biolinerx: Employment. Sorani:BioLineRx Ltd.,: Employment. Bohana-Kashtan:BioLineRx Ltd.,: Employment, Equity Ownership; Cell Cure Neurosciences: Equity Ownership. Shaw:BioLineRx Ltd.: Employment, Equity Ownership.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. sci-30-sci-30
Author(s):  
Adolfo A. Ferrando

The NOTCH pathway is an evolutionarily conserved mechanism responsible for the direct transduction of developmental signals at the cell surface into changes in gene expression in the nucleus. In the hematopoietic system, the NOTCH1 signaling pathway is required for the commitment of multipotent hematopoietic progenitors to the T cell lineage and, later on, to support cell growth, proliferation, and survival at multiple stages of thymocyte development. These physiological functions are disrupted by activating mutations in the NOTCH1 gene in over 50% of T-cell acute lymphoblastic leukemias (T-ALL). NOTCH1 mutations result in ligand-independent activation or increased protein stability, leading to deregulated cell proliferation and survival in T cell progenitors and T cell transformation. Importantly, small molecule inhibitors of the γ-secretase complex (GSIs) effectively abrogate NOTCH1 signaling, making NOTCH1 a promising therapeutic target for the treatment of T-ALL. However, the clinical development of GSIs has been hampered by our incomplete understanding of the effector pathways controlled by NOTCH1, the lack of clinical responses to GSI therapy, and the development of gastrointestinal toxicity secondary to inhibition of NOTCH signaling in the gut. Recent progress in the identification of the transcriptional regulatory networks that control T cell transformation downstream of NOTCH1 has shown a close relationship between oncogenic NOTCH1 signaling and the transcriptional control of cell growth and metabolism. Thus, NOTCH1 directly controls multiple genes involved in anabolic pathways and further promotes cell growth via direct transcriptional upregulation of MYC and indirect upregulation of the PI3K-AKT signaling pathway via HES1-mediated downregulation of PTEN. Moreover, oncogenic NOTCH1 signaling promotes activation of the NFKB pathway and interacts with the function of p53. Although the specific mechanisms mediating some of these interactions have not been elucidated yet, these results suggest that combinations of GSIs with chemotherapy or drugs targeting the PI3K-AKT and NFKB pathways may have a synergistic effect in the treatment of T-ALL. Moreover, inhibitory antibodies capable of selectively blocking the activation of NOTCH1 and inhibitors of the nuclear transcriptional complex mediating the activation of NOTCH1 target genes offer alternative approaches for the inhibition of oncogenic NOTCH1 signaling. Each of these strategies offers the promise to induce potent antileukemic effects and/or to ameliorate the gastrointestinal toxicity associated with systemic inhibition of NOTCH signaling and may ultimately lead to the development of rationally-designed and highly effective anti-NOTCH1 therapies for T-ALL.


2006 ◽  
Vol 203 (10) ◽  
pp. 2239-2245 ◽  
Author(s):  
Ivan Maillard ◽  
LiLi Tu ◽  
Arivazhagan Sambandam ◽  
Yumi Yashiro-Ohtani ◽  
John Millholland ◽  
...  

Genetic inactivation of Notch signaling in CD4−CD8− double-negative (DN) thymocytes was previously shown to impair T cell receptor (TCR) gene rearrangement and to cause a partial block in CD4+CD8+ double-positive (DP) thymocyte development in mice. In contrast, in vitro cultures suggested that Notch was absolutely required for the generation of DP thymocytes independent of pre-TCR expression and activity. To resolve the respective role of Notch and the pre-TCR, we inhibited Notch-mediated transcriptional activation in vivo with a green fluorescent protein–tagged dominant-negative Mastermind-like 1 (DNMAML) that allowed us to track single cells incapable of Notch signaling. DNMAML expression in DN cells led to decreased production of DP thymocytes but only to a modest decrease in intracellular TCRβ expression. DNMAML attenuated the pre-TCR–associated increase in cell size and CD27 expression. TCRβ or TCRαβ transgenes failed to rescue DNMAML-related defects. Intrathymic injections of DNMAML− or DNMAML+ DN thymocytes revealed a complete DN/DP transition block, with production of DNMAML+ DP thymocytes only from cells undergoing late Notch inactivation. These findings indicate that the Notch requirement during the β-selection checkpoint in vivo is absolute and independent of the pre-TCR, and it depends on transcriptional activation by Notch via the CSL/RBP-J–MAML complex.


1998 ◽  
Vol 18 (12) ◽  
pp. 6939-6950 ◽  
Author(s):  
Yuichi Ono ◽  
Norio Fukuhara ◽  
Osamu Yoshie

ABSTRACT Previously, we have shown that TAL1 and the LIM-only protein gene (LMO) are regularly coactivated in T-cell acute lymphoblastic leukemia (T-ALL). This observation is likely to relate to the findings that TAL1 and LMO are highly synergistic in T-cell tumorigenesis in double-transgenic mice. To understand the molecular mechanisms of functional synergy between TAL1 and LMO in tumorigenesis and transcriptional regulation, we tried to identify downstream target genes regulated by TAL1 and LMO by a subtractive PCR method. One of the isolated genes, that for retinaldehyde dehydrogenase 2 (RALDH2), was regularly expressed in most of the T-ALL cell lines that coexpressed TAL1 and LMO. Exogenously transfected TAL1 and LMO, but not either alone, inducedRALDH2 expression in a T-ALL cell line, HPB-ALL, not expressing endogeneous TAL1 or LMO. The RALDH2 transcripts in T-ALL were, however, mostly initiated within the second intron. Promoter analysis revealed that a GATA site in a cryptic promoter in the second intron was essential and sufficient for the TAL1- and LMO-dependent transcriptional activation, and GATA3 binds to this site. In addition, forced expression of GATA3 potentiated the induction ofRALDH2 by TAL1 and LMO, and these three factors formed a complex in vivo. Furthermore, a TAL1 mutant not binding to DNA also activated the transcription of RALDH2 in the presence of LMO and GATA3. Collectively, we have identified the RALDH2 gene as a first example of direct transcriptional target genes regulated by TAL1 and LMO in T-ALL. In this case, TAL1 and LMO act as cofactors for GATA3 to activate the transcription ofRALDH2.


Blood ◽  
2011 ◽  
Vol 118 (4) ◽  
pp. 1154-1162 ◽  
Author(s):  
Wei Zheng ◽  
Tuomas Tammela ◽  
Masahiro Yamamoto ◽  
Andrey Anisimov ◽  
Tanja Holopainen ◽  
...  

Abstract Notch signaling plays a central role in cell-fate determination, and its role in lateral inhibition in angiogenic sprouting is well established. However, the role of Notch signaling in lymphangiogenesis, the growth of lymphatic vessels, is poorly understood. Here we demonstrate Notch pathway activity in lymphatic endothelial cells (LECs), as well as induction of delta-like ligand 4 (Dll4) and Notch target genes on stimulation with VEGF or VEGF-C. Suppression of Notch signaling by a soluble form of Dll4 (Dll4-Fc) synergized with VEGF in inducing LEC sprouting in 3-dimensional (3D) fibrin gel assays. Expression of Dll4-Fc in adult mouse ears promoted lymphangiogenesis, which was augmented by coexpressing VEGF. Lymphangiogenesis triggered by Notch inhibition was suppressed by a monoclonal VEGFR-2 Ab as well as soluble VEGF and VEGF-C/VEGF-D ligand traps. LECs transduced with Dll4 preferentially adopted the tip cell position over nontransduced cells in 3D sprouting assays, suggesting an analogous role for Dll4/Notch in lymphatic and blood vessel sprouting. These results indicate that the Notch pathway controls lymphatic endothelial quiescence, and explain why LECs are poorly responsive to VEGF compared with VEGF-C. Understanding the role of the Notch pathway in lymphangiogenesis provides further insight for the therapeutic manipulation of the lymphatic vessels.


Blood ◽  
2011 ◽  
Vol 118 (3) ◽  
pp. 795-803 ◽  
Author(s):  
Katia Urso ◽  
Arantzazu Alfranca ◽  
Sara Martínez-Martínez ◽  
Amelia Escolano ◽  
Inmaculada Ortega ◽  
...  

Abstract The nuclear factor of activated T cells (NFAT) family of transcription factors plays important roles in many biologic processes, including the development and function of the immune and vascular systems. Cells usually express more than one NFAT member, raising the question of whether NFATs play overlapping roles or if each member has selective functions. Using mRNA knock-down, we show that NFATc3 is specifically required for IL2 and cyclooxygenase-2 (COX2) gene expression in transformed and primary T cells and for T-cell proliferation. We also show that NFATc3 regulates COX2 in endothelial cells, where it is required for COX2, dependent migration and angiogenesis in vivo. These results indicate that individual NFAT members mediate specific functions through the differential regulation of the transcription of target genes. These effects, observed on short-term suppression by mRNA knock-down, are likely to have been masked by compensatory effects in gene-knockout studies.


2018 ◽  
Vol 19 (10) ◽  
pp. 3153 ◽  
Author(s):  
J. Muñoz-Bello ◽  
Leslie Olmedo-Nieva ◽  
Leonardo Castro-Muñoz ◽  
Joaquín Manzo-Merino ◽  
Adriana Contreras-Paredes ◽  
...  

The Wnt/β-catenin signaling pathway regulates cell proliferation and differentiation and its aberrant activation in cervical cancer has been described. Persistent infection with high risk human papillomavirus (HR-HPV) is the most important factor for the development of this neoplasia, since E6 and E7 viral oncoproteins alter cellular processes, promoting cervical cancer development. A role of HPV-16 E6 in Wnt/β-catenin signaling has been proposed, although the participation of HPV-18 E6 has not been previously studied. The aim of this work was to investigate the participation of HPV-18 E6 and E6*I, in the regulation of the Wnt/β-catenin signaling pathway. Here, we show that E6 proteins up-regulate TCF-4 transcriptional activity and promote overexpression of Wnt target genes. In addition, it was demonstrated that E6 and E6*I bind to the TCF-4 (T cell factor 4) and β-catenin, impacting TCF-4 stabilization. We found that both E6 and E6*I proteins interact with the promoter of Sp5, in vitro and in vivo. Moreover, although differences in TCF-4 transcriptional activation were found among E6 intratype variants, no changes were observed in the levels of regulated genes. Furthermore, our data support that E6 proteins cooperate with β-catenin to promote cell proliferation.


1999 ◽  
Vol 31 (3) ◽  
pp. 1618-1619
Author(s):  
B Otová ◽  
M Sladká ◽  
J Damoiseaux ◽  
A Panczak ◽  
V Mandys ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document