Academic literature on the topic 'Notch signaling, Leukemia, Chemosensitivity'

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 signaling, Leukemia, Chemosensitivity.'

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 signaling, Leukemia, Chemosensitivity"

1

Láinez-González, Daniel, Juana Serrano-López, and Juan Manuel Alonso-Dominguez. "Understanding the Notch Signaling Pathway in Acute Myeloid Leukemia Stem Cells: From Hematopoiesis to Neoplasia." Cancers 14, no. 6 (March 12, 2022): 1459. http://dx.doi.org/10.3390/cancers14061459.

Full text
Abstract:
The Notch signaling pathway is fundamental to early fetal development, but its role in acute myeloid leukemia is still unclear. It is important to elucidate the function that contains Notch, not only in acute myeloid leukemia, but in leukemic stem cells (LSCs). LSCs seem to be the principal cause of patient relapse. This population is in a quiescent state. Signaling pathways that govern this process must be understood to increase the chemosensitivity of this compartment. In this review, we focus on the conserved Notch signaling pathway, and its repercussions in hematopoiesis and hematological neoplasia. We found in the literature both visions regarding Notch activity in acute myeloid leukemia. On one hand, the activation of Notch leads to cell proliferation, on the other hand, the activation of Notch leads to cell cycle arrest. This dilemma requires further experiments to be answered, in order to understand the role of Notch not only in acute myeloid leukemia, but especially in LSCs.
APA, Harvard, Vancouver, ISO, and other styles
2

Takam Kamga, Paul, Giada Dal Collo, Martina Midolo, Annalisa Adamo, Pietro Delfino, Angela Mercuri, Simone Cesaro, et al. "Inhibition of Notch Signaling Enhances Chemosensitivity in B-cell Precursor Acute Lymphoblastic Leukemia." Cancer Research 79, no. 3 (December 18, 2018): 639–49. http://dx.doi.org/10.1158/0008-5472.can-18-1617.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Takam Kamga, Paul, Bassi Giulio, Adriana Cassaro, Roberta Stradoni, Martina Midolo, Omar Perbellini, and Mauro Krampera. "Role of Stromal Cell-Mediated Notch Signaling in AML Resistance to Chemotherapy." Blood 124, no. 21 (December 6, 2014): 1044. http://dx.doi.org/10.1182/blood.v124.21.1044.1044.

Full text
Abstract:
Abstract Introduction: Our group has recently shown that bone marrow-mesenchymal stromal cell (BM-MSCs)-mediated Notch signaling may control survival and chemoresistance of B-acute lymphoblastic leukemia (B-ALL) and chronic lymphocytic leukemia (CLL) cells. Conversely, the role of Notch signaling in acute myeloid leukemia (AML) remains controversial, as its contribution to the crosstalk between BM-MSCs and AML cells is still unknown. Thus, we evaluated the role of the Notch pathway in the proliferation, survival and chemoresistance of AML primary blast cells in co-culture with BM-MSCs. Methods: AML blast cells were obtained after informed consent from bone marrow samples (30) and peripheral blood (20) of AML patients, according to the Institutional guidelines. BM-MSCs were expanded from bone marrow of 12 healthy donors (BM-MSCs) and of 12 AML-patients (BM-MSCs*). PCR, FACS analysis and western immunoblotting were used to study the expression of Notch receptors and ligands, as well as Notch activation status, in AML cells and BM-MSCs. AML cells were co-cultured with BM-MSCs or BM-MSCs* at 10:1 (AML:BM-MSCs) ratio for 2 to 3 days in presence of Cytarabine, Etoposide, Idarubicin, as well as in presence or absence of anti-Notch-1, -2, -3, -4, anti-Jagged1, -2 and anti-DLL3 blocking antibodies or gamma secretase inhibitor-XII (GSI-XII). Cell viability was evaluated by Annexin-V/Propidium Iodide (PI) and MTT; proliferation and cell cycle were assessed through CFSE dilution and PI methods, respectively. Results: AML cells expressed Notch receptors and ligands, showing Notch-1, -2, Jagged-1, -2 and DLL-3 signature, while BM-MSCs/ BM-MSCs* showed expression of Notch-1, -2, -3, -4 and Jagged-1, -2 and DLL-1. We then analyzed Notch activation in different cell types by evaluating the expression of NICD-1, -2, -3 as well as Hes-1. We found that at least 50% of AML samples showed basal Notch activation while BM-MSCs/ BM-MSCs* showed slight Notch activation. The expression and activation pattern were modulated after 3 days of co-culture with either BM-MSCs or BM-MSCs*. The pan blockade of Notch signaling by GSI-XII were capable to inhibit AML cell proliferation as well as induce AML cell apoptosis in culture or in co-culture with BM-MSCs/ BM-MSCs*. The addition of chemotherapeutic agents decreased AML cell viability in culture, while a significant rescue from apoptosis was observed when cocultured with BM-MSCs or BM-MSCs*. Pan Notch signaling blockade by either GSI-XII or combination of Notch receptor-blocking antibodies in presence of chemotherapeutic agents significantly lowered the supportive role of BM-MSCs towards AML cell lines. The specific blockade of Notch-1, -2, -3 or Jagged-1, -2 rescued partially the chemosensibility, while blockade of Notch-4 or DLL-3 rescued totally the chemosensitivity of primary AML cells in co-culture with BM-MSCs. Conclusions: These results suggest that Notch signaling may represent a potential therapeutic strategy to overcome bone marrow stromal-mediated survival and chemoresistance of AML. Moreover Notch blocking antibodies were able to impair the survival benefit imparted by bone marrow stromal cells. Therefore blocking Notch antibodies could be a useful strategy to improve the efficiency of AML chemotherapy. Disclosures No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
4

Nishimura, Akira, Kazuaki Yokoyama, Chika Yamagishi, Takuya Naruto, Tomohiro Morio, Akinori Kanai, Hirotaka Matsui, et al. "Clinical Feature and Genetic Alterations in Myeloid/Natural Killer (NK) Cell Precursor Acute Leukemia and Myeloid/NK Cell Acute Leukemia." Blood 132, Supplement 1 (November 29, 2018): 2824. http://dx.doi.org/10.1182/blood-2018-99-118627.

Full text
Abstract:
Abstract Introduction: Myeloid/Natural killer cell precursor acute leukemia (MNKPL) and myeloid/NK cell acute leukemia (MNKL) is a rare hematologic malignancy prevalent in East Asia. MNKPL is characterized by marked extramedullary involvement, immature lymphoblastoid morphology without myeloperoxidase (MPO) reactivity, a CD7+/CD33+/CD34+/CD16−/CD15−/+/HLA-DR+ phenotype, myeloid chemosensitivity, and a poor prognosis. By contrast, MNKL shows no extramedullary involvement, a HLA‐DR−/CD33+/CD16−/CD34−/+ phenotype, myeloid chemosensitivity, and a good prognosis. However, analysis of outcome and genetic alterations in these leukemias are limited. Here, we report outcome and genetic alterations in the patients with MNKPL and MNKL. Methods: The Leukemia and Lymphoma Committee of the Japanese Society of Pediatric Hematology and Oncology (JSPHO) sent out two questionnaires to 110 JSPHO affiliated hospitals. The first questionnaire requested details of the number of pediatric patients with MNKPL or MNKL had been diagnosed during the period 2000-2013. The second questionnaire requested more detailed information about clinical curses. Overall survival (OS) and event free survival (EFS) defined as relapse or death was analyzed. The protocol of this retrospective study was approved by the review boards of JSPHO and Ehime Prefectural Central Hospital. We also performed whole exome sequence (WES) using 7 children's samples (5 MNKPL, 2 MNKL) and target sequence using 2 adult's samples (2 MNKPL) from this and another independent cohort. The research protocol was approved by the review board of TMDU. Results: Thirteen children with MNKPL and 6 children with MNKL were identified. Median age of MNKPL was 8 year-old (range; 0.5-17) and median age of MNKL was 10 year-old (range; 2-13). There are 8 males and 5 females in MNKPL and 4 males and 2 females in MNKL. In MNKPL, central nervous system, mediastinum and lymph node involvement was observed in 1 case respectively. Nasal sinus involvement was observed in 1 case in MNKL. Eleven patients with MNKPL and 3 patients with MNKL were treated with acute myeloid leukemia style chemotherapy and 1 MNKPL patients and 3 MNKL patients were treated with acute lymphoblastic leukemia/non-Hodgkin lymphoma style chemotherapy. Complete remission after induction therapy was achieved in 8/13 MNKPL children and 4/6 MNKL children. Twelve out of 13 MNKPL children and all 6 MNKL children underwent hematopoietic cell transplantation (HCT) with myeloablative conditioning regimen. Median follow up period was 5.3 years in MNKPL and 3.8 years in MNKL patients. 5-year OS of MNKPL and MNKL was 67.3 % and 41.7 %, 5-year EFS of MNKPL and MNKL was 52.7 % and 41.7 % respectively. In genetic analysis, average 148 somatic mutations in MNKPL and 88 somatic mutations in MNKL were identified by WES. In combined analysis using adult cases, the recurrent mutations were observed in NOTCH1, NRAS (n=3, respectively), MAML2, MAP3K1, SIRPA (n=2, respectively) as activating signal genes, and CLTCL1 (n=2) as cell adhesion molecules, and RECQL4 (n=2) as cell cycle/DNA repair molecules, and PRDM2, CREBBP, SETBP1 (n=2, respectively) as epigenetic modifiers, and WT1, ZNF384, BCLAF1 (n=2, respectively) as transcription factors. Conclusions: Previously, it has been reported that outcome of MNKL is relatively good than MNKPL. MNKPL and MNKL children had a poor prognosis in our cohort even though most patients received HCT. We identified alteration of molecules involved in NOTCH signaling and RAS-MAPK pathways. In addition, mutations of several transcription factors such as WT1 were identified. The drugs targeting RAS pathway and epigenetic factors may have the potential to improve outcome. An international collaboration for clinical and cytogenetic research of MNKPL and MNKL is needed as they are complex and rare diseases. Disclosures No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
5

Guerrero, Carmina Louise Hugo, Yoshiko Yamashita, Megumi Kuba-Miyara, Naoki Imaizumi, Shugo Sakihama, Masaki Hayashi, Takashi Miyagi, et al. "Proteomic Profiling of HTLV-1 Carriers and ATL Patients Reveal TNFR2 As a Novel Diagnostic and Chemosensitivity Biomarker for ATL." Blood 134, Supplement_1 (November 13, 2019): 660. http://dx.doi.org/10.1182/blood-2019-129817.

Full text
Abstract:
Adult T-cell leukemia/lymphoma (ATL) is a peripheral T-cell malignancy associated with the human T-cell leukemia virus type I (HTLV-1). Classification of ATL into clinical subtypes acute, lymphoma, chronic and smoldering types was proposed based on prognostic factors, clinical features and natural history of the disease. Although HTLV-1 infection alone is not sufficient to cause ATL and only about 5% of HTLV-1 carriers progress to ATL, the prognosis is generally poor especially for patients with aggressive ATL (i.e., acute, lymphoma or unfavorable chronic types), with a median survival time at around 1 year, even after chemotherapy. Currently, biomarkers to predict ATL onset and progression are limited, making early diagnosis and treatment for ATL challenging. To develop early diagnostic biomarkers for ATL, we performed, for the first time, an extensive proteomic profiling of HTLV-1 carriers and ATL patients as a foundation for establishing a blood-based biomarker panel for ATL. Expression levels of 1305 plasma proteins in HTLV-1 carriers (n=40), untreated ATL patients (n=40, 28 acute; 4 lymphoma; 5 chronic; 3 smoldering), and remission status (n=5) were measured by SOMAscan assay (SomaLogic Inc, Boulder, CO). ATL diagnosis was based on criteria proposed by the Japan Clinical Oncology Group (JCOG) and identification of monoclonal integration of HTLV-1 proviral DNA using Southern blot hybridization method. Deregulated proteins in HTLV-1 versus ATL versus remission states were ranked by significance (Welch's t-test) and discrimination capacity (area under the curve [AUC]). In addition, machine learning algorithms were used to set discrimination boundaries for HTLV-1, ATL, and remission states using some of the top deregulated proteins. Statistical analyses were performed using Python 3.6.2 software. To elucidate on ATL pathogenesis, we further analyzed our proteomic data using Gene Set Enrichment Analysis (GSEA 3.0 hallmarks, curated gene sets) and Gene Ontology (GO Panther Pathways) and determined pathway deregulation among disease states as well as among ATL subtypes. Overrepresented pathways in ATL versus HTLV-1 included inflammation mediated by cytokine and chemokine signaling, angiogenesis, notch signaling, and IL6/JAK/STAT3, among others. Among a total of 176 proteins which were categorized as extremely significant (p<0.00001) with AUC scores ranging from 0.90-0.99, we further confirmed plasma protein concentrations of CD223 (LAG3) (n=40, 2 healthy individuals; 8 HTLV-1 carriers; 10 acute; 8 lymphoma; 6 chronic; 6 smoldering), CD30 (TNFRSF8), TIM-3 (HAVCR2) (n=40, 2 healthy individuals; 8 HTLV-1 carriers; 12 acute; 6 lymphoma, 6 chronic; 6 smoldering), and TNFR2 (TNFRSF1B) (n=79, 5 healthy individuals; 16 HTLV-1 carriers; 26 acute; 9 lymphoma; 11 chronic; 9 smoldering), through ELISA. We discovered significantly higher CD30 and TIM-3 levels in acute ATL versus HTLV-1 carriers (p<0.05) and remarkably high TNFR2 levels among aggressive ATL patients, acute (p<0.001) and lymphoma types (p<0.01) versus HTLV-1 carriers. In addition, a significant decrease in TNFR2 levels among ATL patients who have achieved remission was also seen (p<0.001). These results indicate the potential value of TNFR2 not only as a diagnostic biomarker for ATL, but also for predicting response or failure to therapy. Furthermore, this study represents a novel proteomic approach in developing candidate biomarkers for ATL, by combining data from high-throughput SOMAmer technology, machine learning, proteomic pathway analysis, in addition to previously well-established techniques such as ELISA. Further investigation of TNFR2 and other potential biomarkers in this study need to be done in the near future. Disclosures Fukushima: Daiichi-Sankyo: Research Funding.
APA, Harvard, Vancouver, ISO, and other styles
6

Aster, Jon C., Warren S. Pear, and Stephen C. Blacklow. "Notch Signaling in Leukemia." Annual Review of Pathology: Mechanisms of Disease 3, no. 1 (February 2008): 587–613. http://dx.doi.org/10.1146/annurev.pathmechdis.3.121806.154300.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Aster, Jon C., and Warren S. Pear. "Notch signaling in leukemia." Current Opinion in Hematology 8, no. 4 (July 2001): 237–44. http://dx.doi.org/10.1097/00062752-200107000-00010.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Grieselhuber, N. R., J. M. Klco, A. M. Verdoni, T. Lamprecht, S. M. Sarkaria, L. D. Wartman, and T. J. Ley. "Notch signaling in acute promyelocytic leukemia." Leukemia 27, no. 7 (March 4, 2013): 1548–57. http://dx.doi.org/10.1038/leu.2013.68.

Full text
APA, Harvard, Vancouver, ISO, and other styles
9

Lobry, Camille, Panagiotis Ntziachristos, Delphine Ndiaye-Lobry, Philmo Oh, Luisa Cimmino, Nan Zhu, Elisa Araldi, et al. "Notch pathway activation targets AML-initiating cell homeostasis and differentiation." Journal of Experimental Medicine 210, no. 2 (January 28, 2013): 301–19. http://dx.doi.org/10.1084/jem.20121484.

Full text
Abstract:
Notch signaling pathway activation is known to contribute to the pathogenesis of a spectrum of human malignancies, including T cell leukemia. However, recent studies have implicated the Notch pathway as a tumor suppressor in myeloproliferative neoplasms and several solid tumors. Here we report a novel tumor suppressor role for Notch signaling in acute myeloid leukemia (AML) and demonstrate that Notch pathway activation could represent a therapeutic strategy in this disease. We show that Notch signaling is silenced in human AML samples, as well as in AML-initiating cells in an animal model of the disease. In vivo activation of Notch signaling using genetic Notch gain of function models or in vitro using synthetic Notch ligand induces rapid cell cycle arrest, differentiation, and apoptosis of AML-initiating cells. Moreover, we demonstrate that Notch inactivation cooperates in vivo with loss of the myeloid tumor suppressor Tet2 to induce AML-like disease. These data demonstrate a novel tumor suppressor role for Notch signaling in AML and elucidate the potential therapeutic use of Notch receptor agonists in the treatment of this devastating leukemia.
APA, Harvard, Vancouver, ISO, and other styles
10

Kamdje, Armel Hervé Nwabo, and Mauro Krampera. "Notch signaling in acute lymphoblastic leukemia: any role for stromal microenvironment?" Blood 118, no. 25 (December 15, 2011): 6506–14. http://dx.doi.org/10.1182/blood-2011-08-376061.

Full text
Abstract:
Abstract Notch signaling pathway regulates many different events of embryonic and adult development; among them, Notch plays an essential role in the onset of hematopoietic stem cells and influences multiple maturation steps of developing lymphoid and myeloid cells. Deregulation of Notch signaling determines several human disorders, including cancer. In the last decade it became evident that Notch signaling plays pivotal roles in the onset and development of T- and B-cell acute lymphoblastic leukemia by regulating the intracellular molecular pathways involved in leukemia cell survival and proliferation. On the other hand, bone marrow stromal cells are equally necessary for leukemia cell survival by preventing blast cell apoptosis and favoring their reciprocal interactions and cross-talk with bone marrow microenvironment. Quite surprisingly, the link between Notch signaling pathway and bone marrow stromal cells in acute lymphoblastic leukemia has been pointed out only recently. In fact, bone marrow stromal cells express Notch receptors and ligands, through which they can interact with and influence normal and leukemia T- and B-cell survival. Here, the data concerning the development of T- and B-cell acute lymphoblastic leukemia has been critically reviewed in light of the most recent findings on Notch signaling in stromal microenvironment.
APA, Harvard, Vancouver, ISO, and other styles

Dissertations / Theses on the topic "Notch signaling, Leukemia, Chemosensitivity"

1

Wang, Shu-Fang. "Development of Notch-dependent T-cell leukemia by deregulated Rap1 signaling." 京都大学 (Kyoto University), 2008. http://hdl.handle.net/2433/137035.

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

Chan, Steven Man Cheong. "Protein microarray technology for profiling signaling patwhays [sic] : insights into pro-oncogenic notch signaling in T cell acute lymphoblastic leukemia /." May be available electronically:, 2006. http://proquest.umi.com/login?COPT=REJTPTU1MTUmSU5UPTAmVkVSPTI=&clientId=12498.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Zimmerman, Grant Robert. "T-ALL LEUKEMIA DYSREGULATES STROMAL BONE MARROW ENVIRONMENT AND DISRUPTS NICHE-STEM CELL SIGNALING AXIS." Case Western Reserve University School of Graduate Studies / OhioLINK, 2015. http://rave.ohiolink.edu/etdc/view?acc_num=case1436293859.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Wen-Hwei, Chang, and 張文輝. "The members of Notch signaling system in Acute Promyelocyte Leukemia (APL) cell lines." Thesis, 2001. http://ndltd.ncl.edu.tw/handle/87458487310842413407.

Full text
Abstract:
碩士
台北醫學院
細胞及分子生物研究所
89
The promyelocytic cells in acute promyelocytic leukemia (APL) devoid the ability for terminal differentiation. The PKC activator, 12-O-tetradecanoylphorbol-13-acetate (TPA) can induce the differentiation of APL cell lines into macrophage. The Vitamin A derivatives, all-trans retinoic acid (ATRA), and the polar-planer compound, dimethyl sulfoxide (DMSO), have the ability to induce granulocyte maturation from promyelocytic cells. In the past decade, clinical therapy for APL patients used ATRA to induce cells maturation. Nevertheless, despite an initial good response, most patients that received continuous treatment with ATRA relapse and develop RA-resistant disease. The real mechanism of those inducer drugs influence cells differentiation still unsolved. The classical view holds that Notch signaling keeps cells in an undifferentiated state and as an important switch controlling cell fate decisions in a wide variety of tissues and cell types. Notch signaling pathway involves Notch receptors, Notch ligands, intracellular effectors and Notch modulators. In mammals the Notch gene family comprise four related genes, Notch1-4, as the Notch ligands that encode transmembrane proteins. Ligand-receptor interaction mediated Notch activation ultimately leads to the conversion of effectors-CSL (CBF1, Su(H), Lag-1) proteins, from repressors to transcriptional activators, and subsequent up-regulation of downstream targets. We are interesting in exploring the role of Notch signaling on the ATRA or DMSO- and TPA-induced differentiation of APL cell lines. In this study, HL-60 and KG-1 cells were induced to differentiate into macrophage-like cells after exposure to 2x 10-8 M TPA for 2 days. Five days of treatment with 10-6 M ATRA or 1.5 % DMSO can stimulate HL-60 to differentiate into granulocyte cells without affecting KG-1 cells. The differentiation of APL cell lines was confirmed by Liu’s stain on cytospin preparation. The total RNA from those cells was isolated and the transcriptional levels of Notch molecules, Notch ligands, as well as Notch modulates were elucidated by the RT-PCR methodology. We found that both APL cell lines expressed Notch receptors: Notch-1, -2, -4; Notch ligand Jagged-1 and Notch modulator manic fringe. After differentiation of APL cells to either lineage, the transcriptional levels of Notch receptors: Notch-1, -2, -4; Notch ligand Jagged-1 and Notch modulator manic fringe were not affected in this study. Our data may not indicate the regulation in the transcriptional levels of Notch system correlate to the differentiation stages of APL cells. The post-translational mechanisms, especially proteins modification, were implicated to be the major candidates for regulation of Notch signaling activity in recent studies. We assume that the post-translational regulation of Notch signaling system and interaction of the signaling molecules activated by differentiation inducers with molecules in Notch pathway are the players involve in inducing differentiation of APL cells.
APA, Harvard, Vancouver, ISO, and other styles
5

Steinbuck, Martin. "Novel T-cell receptor mediated mechanisms of Notch activation and signaling." Thesis, 2016. https://hdl.handle.net/2144/19064.

Full text
Abstract:
The Notch receptor is an evolutionarily highly conserved transmembrane protein essential to a wide spectrum of cellular systems. Notch is especially important to T-cell development, and its deregulation leads to leukemia. Although not well characterized, Notch signaling continues to play an integral role in peripheral T-cells, in which a unique mode of Notch activation can occur. In contrast to canonical Notch activation initiated by adjacent ligand-expressing cells, T-cell receptor (TCR)-stimulation is sufficient to induce robust Notch signaling. However, the interactions between these two pathways have not been defined. In this dissertation, we show that Notch activation occurs in peripheral T-cells within a few hours post TCR-stimulation and is required for optimal T-cell activation. Utilizing a panel of inhibitors against components of the TCR signaling cascade, we demonstrate that Notch activation is facilitated through initiation of protein kinase C-induced ADAM-metalloprotease activity. Moreover, internalization of Notch via endocytosis is indispensible for this process. Whereas ligand-mediated Notch stimulation relies on mechanical pulling forces that disrupt the autoinhibitory domain of Notch, we hypothesized that in T-cells in the absence of ligands, these conformational changes are induced through chemical adjustments in the endosome, causing alleviation of autoinhibition and receptor activation. Our data show that endocytosis is not only a prerequisite for TCR-induced Notch processing during normal T-cell function, but is essential even in Notch-mutated T-leukemia cells exhibiting constitutively active Notch signaling. Our work has also focused on signaling mechanisms of Notch following receptor activation. The Notch signal is transduced via cleavage of the intracellular portion of the receptor that subsequently translocates to the nucleus where it regulates gene transcription via interactions with its DNA-binding partner, RBPJκ. Utilizing RBPJκ-deficient T-cells, we show that, although Notch signaling is required, RBPJκ-dependent signaling is dispensable for peripheral T-cell proliferation and activation. Using retroviral constructs that encode modified, active forms of Notch restricted to the nucleus or cytoplasm, we provide evidence that Notch signaling may utilize RBPJκ-independent pathways for signal transduction. In conclusion, T-cells have evolved a unique method of Notch receptor activation, described for the first time in this dissertation, as well as novel mechanisms that facilitate downstream signaling.
APA, Harvard, Vancouver, ISO, and other styles
6

NOCE, CLAUDIA. "NOTCH SIGNALING DEREGULATION IN T CELL ACUTE LYMPHOBLASTIC LEUKEMIA PROMOTES THE GENERATION OF MYELOID DERIVED SUPPRESSOR CELLS." Doctoral thesis, 2016. http://hdl.handle.net/11573/924450.

Full text
Abstract:
Notch receptors play an important role in both T cell differentiation and T-cell leukemia development. Notch3 constitutive activation inside T cell compartment of transgenic (N3tg) mice induces an aggressive form of ‘T-cell acute lymphoblastic leukemia’ (T-ALL), characterized by the ‘aberrant’ presence of extrathymic immature CD4+CD8+ T cells. ‘Myeloid Derived Suppressor Cells’ (MDSCs) include a heterogeneous group of immature/progenitor myeloid cells, that in mice are broadly identified as CD11b+Gr-1+ cells. MDSCs are expanded and activated in tumor microenvironment, where they suppress host immune responses, including that of T cells, thus facilitating disease progression. MDSCs have been extensively described in solid tumors and, more recently in hematological malignancies. However, nothing is known about MDSCs in acute T-cell leukemia, so far. We observed alterations inside myeloid compartment of N3-tg mice, with a systemic expansion of immature CD11b+Gr1+ cells, that display features of MDSCs, such as the overexpression of molecules involved in their functios (i.e. Arginase-1, ROS), and the ability to suppress in vitro proliferating T cells. To the best of our knowledge, this represents the first demonstration of the presence of MDSCs in associaton with T-cell leukemia. Moreover, we suggest that in our model of Notch3-dependent T-ALL, the massive presence of aberrant CD4+CD8+ (DP) T cells in BM ‘tumor’ microenvironment induces alterations in differentiation of myeloid compartment, through a non-cell autonomous mechanism. Indeed, BM CD4+CD8+ T cells from N3-tg mice promote the generation of MDSCs, at least in vitro, through a mechanism that is partially dependent on the IL-6 produced. Collectively, our data shed new light on the cross-talk between T cell and myeloid cell compartment and report the appearence of MDSCs in the context of Notch-dependent T-ALL, that may influence the disease outcome and thus would have an important impact on the development of innovative therapies.
APA, Harvard, Vancouver, ISO, and other styles
7

Pinto, Inês Garcia Brandão e. Silva. "The role of NRARP in the pathogenesis of T-cell acute lymphoblastic leukemia." Master's thesis, 2017. http://hdl.handle.net/10451/32852.

Full text
Abstract:
Tese de mestrado, Oncobiologia, Universidade de Lisboa, Faculdade de Medicina, 2017
T-cell acute lymphoblastic leukemia (T-ALL) is a severe hematologic malignancy, commonly associated with high frequency of relapse. NOTCH signaling is a hallmark of T-ALL pathogenesis and more than 50% of T-ALL cases harbor NOTCH activating mutations. Despite considerable clinical advances throughout the years, NOTCH signaling inhibition cannot be efficiently achieved, due to weak therapeutic effects and severe toxicity. NRARP is a negative regulator of the NOTCH pathway and a transcriptional target of NOTCH. It is known to regulate both T-cell lineage commitment and early differentiation. Importantly it has been shown that in NOTCH-induced T-ALL, loss of the microRNA181ab1 inhibits T-ALL development, partly by de-repressing NRARP. NRARP further modulates WNT signaling, by stabilizing LEF1, a transcription factor acting downstream of WNT pathway, shown to induce the malignant transformation of murine thymocytes. Altogether, these findings suggest that a deregulation of NRARP function may play a role in T-ALL. Thus, the main goal of this project is to understand the role of NRARP in T-ALL pathogenesis and evaluate its therapeutic potential. We started by determining NRARP expression in human T-ALL cells. We observed that NRARP protein levels are significantly increased in T-ALL comparing with human thymocytes. This result, although consistent with the fact that NRARP is a transcriptional target of NOTCH, suggests that NRARP is not sufficient to block NOTCH oncogenic signals. To test this hypothesis, we overexpressed NRARP in human T-ALL cell lines. Intriguingly, even though we observed a block in NOTCH signaling in all cell lines analyzed, functional studies revealed that while some T-ALL cells proliferate less upon NRARP overexpression, some proliferate more. Importantly, this suppression or promotion of cell expansion correlated with the changes observed in cell cycle progression (delayed or accelerated, respectively) and cMYC expression (downregulated or upregulated, respectively) in NRARP overexpressing cells and, curiously, with the mutational status of NOTCH (mutated or wild-type, respectively). Altogether, our results suggest that NRARP is capable of blocking NOTCH signaling independently of NOTCH mutational status. Nonetheless, in T-ALL cells that depend on NOTCH oncogenic signals (NOTCH mutated cells), NRARP overexpression has a “tumor-suppressor”-like function, whereas in leukemia cells that do not depend on NOTCH oncogenic signals (NOTCH wild-type) it plays an oncogene-like role. Further studies are necessary to understand if WNT signaling contributes to this dual role of NRARP in T-ALL.
A leucemia linfoblástica aguda de células T (LLA-T) é uma neoplasia hematológica severa, resultante da expansão clonal de progenitores linfóides de células T, representando cerca de 15% dos casos de adultos e 25% dos casos pediátricos de leucemia linfoblástica aguda (LLA). Apesar do prognóstico de doentes com LLA-T ter melhorado significativamente ao longo dos anos, a taxa de recidiva é ainda elevada e o prognóstico dos doentes com doença resistente à terapia ou recidivante é muito reservado. A via de sinalização NOTCH é fundamental para a regulação de processos de homeostasia e regeneração de tecidos e desempenha funções muito importantes na hematopoiese. O compromisso de progenitores linfóides para com a linhagem T e os passos iniciais do desenvolvimento de células T no interior do timo são processos regulados pela sinalização da via NOTCH. Desta forma, não é de estranhar que a sua desregulação seja uma característica da patogénese de LLA-T, e que mutações em NOTCH, levando a uma activação constitutiva da via, se verifiquem em mais de 50% dos casos. A identificação destas mutações levou ao desenvolvimento de terapias dirigidas para esta via. A inibição da via NOTCH foi alcançada essencialmente pelo uso de inibidores da γ-secretase. Estes inibem a via NOTCH ao bloquearem a clivagem final do receptor NOTCH, impedindo desta forma que a sua forma activa seja translocada para o núcleo e, consequentemente, que actue como factor de transcrição. No entanto, verificou-se posteriormente que a sua utilização terapêutica tem efeitos anti-leucémicos reduzidos, associados a efeitos secundários graves, a nível intestinal. Outras abordagens terapêuticas, tais como a utilização de anticorpos que têm como alvo receptores e ligandos da via NOTCH, foram desenvolvidas, mas também estas apresentam efeitos secundários severos, tais como a indução de tumores vasculares e toxicidade hepática. Desta forma, uma melhor compreensão dos mecanismos envolvidos na sinalização via NOTCH em LLA-T poderá originar terapias mais eficazes e seguras. Recentemente, foi demonstrado que a perda de expressão do microRNA miR-181ab1 inibe o desenvolvimento de LLA-T induzido pela activação de NOTCH1, em parte devido ao aumento da expressão de NRARP. Este último é um alvo transcricional da via de sinalização NOTCH e um regulador negativo da mesma. É também importante realçar que a sobre-expressão deste gene em células estaminais hematopoiéticas de ratinho compromete a especificação de células na linhagem T e a sua posterior diferenciação em timócitos. Igualmente importante é o facto de ter sido demonstrado que NRARP está associado à via de sinalização WNT através da estabilização do factor de transcrição LEF1, capaz de induzir transformação maligna de timócitos de ratinho. Desta forma, a desregulação da função de NRARP pode desempenhar um papel na patogénese de LLA-T. Assim, o principal objectivo da presente dissertação é estudar o papel de NRARP no desenvolvimento desta doença. Começámos por caracterizar a expressão de NRARP em células de LLA-T, quantificando para tal os níveis de expressão de mRNA e proteína de NRARP em linhas celulares e amostras primárias. Além disso, para investigar a sua função nestas células, NRARP foi sobre-expresso em algumas linhas celulares de LLA-T, por transdução lentiviral. Estas células foram usadas para avaliar os efeitos de NRARP na sinalização de NOTCH, através da quantificação da expressão da forma intracelular de NOTCH, bem como de alvos a jusante desta via. Foram ainda executados ensaios funcionais nas mesmas células, nomeadamente ensaios de proliferação e viabilidade, bem como de progressão do ciclo celular. Verificámos que os níveis de proteína de NRARP estão significativamente mais elevados em células de LLA-T, comparando com timócitos saudáveis. Este resultado é consistente com o facto de NRARP ser um alvo transcricional da via NOTCH, comumente hiper-activado num contexto de LLA-T. Por outro lado, sendo NRARP um regulador negativo da via NOTCH, estes resultados sugerem que os níveis de NRARP não são suficientes para bloquear os sinais oncogénicos produzidos por esta via. Para testar essa hipótese, procedemos à sobre-expressão de NRARP em linhas celulares humanas de LLA-T e analisámos os efeitos na via NOTCH. No geral, observámos que a sobre-expressão de NRARP suprime a via NOTCH, resultando não só numa diminuição dos níveis da forma intracelular e activa de NOTCH, bem como no decréscimo da maioria dos alvos transcricionais analisados. Contudo, é importante realçar que os níveis do oncogene cMYC, um importante alvo desta via, estavam diminuídos num grupo de linhas celulares, mas aumentados num outro grupo. Curiosamente, a análise funcional da sobre-expressão de NRARP revelou que, apesar da via de sinalização NOTCH estar bloqueada, em algumas linhas celulares NRARP inibe a proliferação, mas noutras promove a sua expansão. A análise da viabilidade e do ciclo celular, demonstrou que a sobre-expressão de NRARP não tem um impacto significativo a nível da viabilidade na maioria das linhas celulares analisadas, mas afecta a progressão do ciclo celular. Estes resultados são consistentes com as observações a nível proliferativo e com as diferenças observadas na expressão de cMYC. Ou seja, nas linhas celulares em que a sobre-expressão de NRARP induz uma diminuição na proliferação, observámos atrasos na progressão do ciclo celular e uma diminuição da expressão de cMYC. Nas linhas celulares em que a sobre-expressão de NRARP resulta num aumento da proliferação, verificámos uma progressão mais rápida no ciclo celular e um aumento na expressão de cMYC. Uma observação interessante é o facto de as linhas celulares cujo crescimento é diminuído por NRARP apresentarem mutações em NOTCH, enquanto as linhas cujo crescimento é favorecido por NRARP, não apresentarem mutações neste gene. Com base nestes resultados, formulámos a hipótese de que a sobre-expressão de NRARP, para além de bloquear a via de sinalização NOTCH, pode afectar directa ou indirectamente outra(s) via(s). Uma vez que NRARP regula LEF1, um factor de transcrição da via WNT, investigámos os efeitos da sobre-expressão de NRARP na via de sinalização WNT. Ainda que bastante preliminares, os nossos resultados sugerem uma diminuição na activação da via WNT nas linhas celulares com um crescimento mais lento. Curiosamente, observámos ainda nestas linhas uma ligação entre NRARP e uma isoforma de LEF1, descrita como tendo um papel negativo na via WNT. Por outro lado, nas linhas celulares com crescimento mais rápido. observámos a interacção de NRARP com outra isoforma de LEF1, possivelmente a isoforma full-lenght. Contudo, é importante ressalvar que estes resultados são preliminares e necessitam de ser validados. Em suma, os resultados obtidos sugerem que NRARP pode desempenhar funções opostas em LLA-T. Apesar de NRARP ser capaz de bloquear a via NOTCH em todas as linhas celulares analisadas, em células que não dependam de sinais oncogénicos produzidos por NOTCH (NOTCH wild-type), NRARP poderá afectar outra via de sinalização, tendo um papel oncogénico. Em células dependentes dos sinais oncogénicos de NOTCH (NOTCH mutado), NRARP terá um papel de supressor tumoral. A importância da via WNT e o envolvimento de LEF1 nesta dualidade na função de NRARP requer estudos mais extensivos.
APA, Harvard, Vancouver, ISO, and other styles
8

Gonçalves, Joana Margarida Luís. "The role of NRARP in the regulation of Wnt signaling pathway in T-cell acute lymphoblastic leukemia." Master's thesis, 2018. http://hdl.handle.net/10451/35556.

Full text
Abstract:
Tese de mestrado, Biologia Molecular e Genética, Universidade de Lisboa, Faculdade de Ciências, 2018
A leucemia linfoblástica aguda de células T (LLA-T) é uma neoplasia hematológica agressiva que se caracteriza por uma proliferação anormal e descontrolada de progenitores de linfócitos T. LLA-T representa cerca de 15% dos casos de leucemia linfoblástica aguda (LLA) em doentes pediátricos e 25% dos casos de LLA em adultos. Apesar dos regimes intensivos de quimioterapia utilizados atualmente no tratamento de LLAT, o prognóstico dos doentes que apresentam resistência primária ou adquirida ao tratamento é reservado, o que requer a procura de novos alvos e/ou estratégias terapêuticas. A via de sinalização Notch, que tem um papel fundamental na hematopoiese e em particular no processo de diferenciação e maturação dos linfócitos T, está associada ao desenvolvimento de LLA-T. Mutações que levam à ativação constitutiva desta via de sinalização verificam-se em aproximadamente 60% dos doentes com LLA-T. Por este motivo, a via Notch tem sido um importante alvo terapêutico, contra a qual têm sido desenvolvidos agentes terapêuticos capazes de bloquear a sua ativação, como os inibidores das enzimas γ- secretase. No entanto, estes fármacos são altamente tóxicos para o trato gastrointestinal e em contexto clínico a sua eficácia terapêutica tem demonstrado ser limitada. Nem só a via Notch tem sido associada a LLA-T. A via de sinalização Wnt, também com um papel crucial no desenvolvimento de linfócitos T, está alterada em amostras de doentes com LLAT, nomeadamente no que diz respeito aos níveis de transcrição e expressão de elementos necessários à ativação da via (β-catenina e LEF1). A ocorrência de mutações nestes mesmos elementos também foi verificada. Durante o estudo do papel do microRNA miR-181ab1 em LLA-T induzida por mutações em NOTCH1 verificou-se que este microRNA promove o desenvolvimento de LLA-T, ao inibir a expressão de NRARP, um regulador negativo da via Notch. A proteína NRARP está envolvida no processo de desenvolvimento dos linfócitos T, tendo sido verificado que quando é sobreexpressa em células estaminais hematopoiéticas, a diferenciação destas em linfócitos T maturos fica comprometida. Além disto, NRARP tem sido associada à via de sinalização Wnt, mas como um regulador positivo desta, através da promoção da estabilidade do fator de transcrição LEF1. Para investigar o papel da proteína NRARP em LLA-T, investigadores do laboratório onde foi desenvolvida esta tese analisaram amostras primárias e linhas celulares de LLA-T tendo sido verificado que os níveis de NRARP estavam significativamente aumentados em comparação com timócitos normais. Neste contexto, estabeleceram-se linhas celulares de LLA-T com sobreexpressão de NRARP onde se concluiu que em células de LLA-T com mutações em NOTCH1, a sobre-expressão desta proteína leva à diminuição da proliferação celular. Curiosamente, por outro lado, em células de LLA-T sem mutações em NOTCH1, quando NRARP é sobre-expressa ocorre um aumento da proliferação celular. Em termos de sinalização celular, verifica-se uma diminuição da expressão da proteína cMYC (promotor de proliferação celular) em células de LLA-T com mutações em NOTCH1. No entanto, verifica-se um aumento da expressão desta proteína em células de LLA-T sem mutações em NOTCH1. Considerando que cMYC é um alvo transcricional da via Notch, mas também de outras vias de sinalização, incluindo a via Wnt, foi proposto o envolvimento desta via no aumento da proliferação de células LLA-T sem mutações em NOTCH1. Resultados preliminares, mostram que a sobre-expressão de NRARP em células de LLA-T sem mutações em NOTCH1 promove um aumento dos níveis proteicos de β-catenina, o que indica um aumento da ativação da via Wnt. Assim, o objetivo desta tese é perceber o papel da proteína NRARP em termos funcionais e mecanísticos, na regulação da via de sinalização Wnt, em LLA-T. Em primeiro lugar, analisámos o impacto funcional da inibição da via Wnt em células de LLAT com e sem sobre-expressão de NRARP, de modo a conseguirmos correlacionar a importância desta via na proliferação e viabilidade das células, com a presença de mutações em NOTCH1. Verificámos que as células sem mutações em NOTCH1 e com sobre-expressão de NRARP são mais sensíveis à inibição da via Wnt do que as células sem sobre-expressão de NRARP, demonstrando não só que NRARP tem um papel crucial na ativação desta via, mas também que a ativação da via Wnt é necessária para a proliferação e viabilidade destas células. Relativamente às células com mutações em NOTCH1, observou-se que estas, apesar de inicialmente sensíveis ao inibidor têm a capacidade de recuperar dos efeitos deste, atingindo níveis de proliferação e viabilidade superiores aos das células controlo (células não tratadas). Este fenómeno sugere que a inibição da via Wnt pode ter um efeito protetor nestas células. Posteriormente, com o objetivo de dissecar o mecanismo pelo qual NRARP regula a via Wnt, investigamos se, tal como descrito noutro contexto, NRARP estabiliza a proteína LEF1. Para tal, utilizaram-se células com e sem mutações em NOTCH1 com sobre-expressão de NRARP onde se procedeu à diminuição da expressão de LEF1, utilizando um RNA de interferência (shRNA). Verificou-se que nas células sem mutações em NOTCH1, após diminuição dos níveis de LEF1, ocorre uma diminuição da proliferação celular, sendo este efeito mais significativo nas mesmas células quando NRARP é sobre-expressa. Estes resultados apoiam hipótese de que células de LLA-T sem mutações em NOTCH1 necessitam da ativação da via Wnt para proliferarem e que NRARP regula a ativação desta via através da proteína LEF1. Nas células com mutações em NOTCH1, a diminuição dos níveis de LEF1 não promove alterações na proliferação celular quando NRARP é sobre-expressa. No entanto, nas células parentais, a diminuição da expressão de LEF1 induz um aumento da proliferação celular, sugerindo que a via de sinalização Wnt tem um efeito inibitório na proliferação de células com mutações em NOTCH1. Após confirmar que é através de LEF1 que NRARP regula a via de sinalização Wnt, fomos avaliar se NRARP regula diretamente LEF1 e se interage com alguma isoforma especifica deste. Em linha com os resultados dos ensaios funcionais, colocámos a hipótese de que em células LLAT sem mutações em NOTCH1, NRARP deveria interagir com a isoforma longa de LEF1 que promove ativação da via Wnt. Por outro lado, em células com mutações, NRARP interagiria com a isoforma curta que não promove a via Wnt. Assim, realizámos ensaios de co-imunoprecipitação onde concluímos que NRARP se liga a LEF1 em células de LLA-T com e sem mutações em NOTCH1, quando NRARP é sobre-expressa ou não. No que diz respeito à isoforma envolvida na interação verificámos que NRARP interage com a isoforma de comprimento longo em células de LLA-T com e sem mutações em NOTCH1, o que contraria em parte a hipótese colocada. Para confirmar estes resultados, procedemos a ensaios de PLA (Proximity ligation assay) que permitem avaliar se duas proteínas interagem e ainda quantificar o número de interações. Desta forma, conseguimos confirmar que LEF1 e NRARP interagem e aferir que a percentagem de células com interações aumenta significativamente quando NRARP é sobre-expressa, em células de LLA-T com ou sem mutações em NOTCH1. Para determinar se o aumento de interações entre LEF1 e NRARP é traduzido na ativação da via de sinalização Wnt, realizaram-se ensaios de co-imunoprecipitação onde se procedeu à precipitação de LEF1 e de β-catenina, seguida da deteção de β-catenina e LEF1, respetivamente. Verificou-se que estas duas proteínas interagem em células de LLA-T com e sem mutações em NOTCH1. Procedemos ainda à realização de ensaios de PLA para quantificar as interações entre LEF1 e β-catenina, e deste modo avaliar o grau de ativação da via Wnt nas diferentes células de LLA-T e de que forma NRARP modula essa ativação. Numa perspetiva geral observámos que o número de interações LEF1-β-catenina é significativamente maior em células sem mutações em NOTCH1 comparativamente a células com mutações. Verificámos ainda que quando a proteína NRARP é sobre-expressa, o número de interações aumenta em células sem mutações em NOTCH1. No entanto o mesmo não se verifica em células com mutações. Isto permitiu-nos concluir que nas células sem mutações em NOTCH1, a proteína NRARP promove o aumento da ativação da via da Wnt. Nas células com mutações em NOTCH1, apesar do número de interações LEF1-NRARP aumentar após sobre-expressão de NRARP, isso não se traduz num aumento do número de interações LEF1-β-catenina e consequentemente na ativação da via Wnt. Assim sendo, são necessárias mais experiências para elucidar o mecanismo responsável pela não ativação da via Wnt nestas células, sabendo que NRARP está a interagir com LEF1. Em suma, estes resultados demonstram que nas células sem mutações em NOTCH1, NRARP promove a ativação da via Wnt, através da interação com LEF1. Além disso, a ativação desta via é necessária para que estas células proliferem e se mantenham vivas. Em células com mutações em NOTCH1, NRARP não promove a ativação da via Wnt, apesar da sobre-expressão desta proteína resultar num aumento da sua interação com LEF1.Verificou-se ainda que a via Wnt não é fundamental para a manutenção funcional destas células e ainda que a sua inibição parece acarretar benefícios ao nível da proliferação e viabilidade celular.
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy characterized by the abnormal and uncontrolled proliferation of immature T-cell progenitors, accounting for 15% of pediatric and 25% of adult cases of ALL. Despite the recent advances in therapeutic approaches, patients’ prognosis remains poor due to primary or acquired resistance to treatment. Deregulations in Notch and Wnt signaling pathways have been shown to contribute to T-ALL development. Of note, NOTCH1-activating mutations are present in approximately 60% of TALL cases and upregulation of Wnt signaling main drivers, LEF1 and β-catenin, have also been reported in both pediatric and adult T-ALL patients. The loss of the mir-181ab1 inhibits NOTCH1-induced T-ALL development, by de-repressing NRARP which is a negative regulator of Notch signaling. Importantly, NRARP also function as a positive regulator of Wnt signaling by promoting LEF1 stability, a transcription factor absolutely required for the activation of this pathway. NRARP has a dual role in the proliferation of T-ALL cells. In cells harboring NOTCH1 mutations, NRARP overexpression blocks cell proliferation. In contrary, in T-ALL cells without NOTCH1 mutations, it promotes cell proliferation. These results suggest the involvement of different signaling pathway(s) in the regulation of these cells proliferation. Because NRARP is a positive regulator of Wnt signaling, it was hypothesized that this pathway may be involved in the proliferation of T-ALL cells without NOTCH1 mutations. Here, we aimed at exploring if and how NRARP modulates Wnt signaling in T-ALL. First, by evaluating the functional importance of this pathway in T-ALL cells, and then by dissecting the mechanism underlying Wnt signaling regulation by NRARP. Our results show that NRARP is involved in Wnt signaling activation in T-ALL cells without NOTCH1 mutations. Furthermore, Wnt pathway is required for proliferation and survival of these cells. Regarding NOTCH1 mutant cells, NRARP does not seems to have a role in the regulation of Wnt signaling. Interestingly, blocking this pathway has a protective effect on these cells, promoting an increase in cell growth and viability. We also found that NRARP regulates Wnt signaling through LEF1 protein. In addition, NRARP overexpression leads to an increase in the number of LEF1-NRARP interactions in T-ALL cells. However, this increase is only translated into Wnt signaling activation in T-ALL cells without NOTCH1 mutations. Overall, we conclude that proliferation of T-ALL cells without NOTCH1 mutations is promoted by NRARP that positively modulates Wnt signaling, through interaction with LEF1.
APA, Harvard, Vancouver, ISO, and other styles

Book chapters on the topic "Notch signaling, Leukemia, Chemosensitivity"

1

Hoofd, Catherine, Vincenzo Giambra, and Andrew P. Weng. "Notch Signaling in T-Cell Acute Lymphoblastic Leukemia and Other Hematologic Malignancies." In Targeting Notch in Cancer, 199–225. New York, NY: Springer New York, 2018. http://dx.doi.org/10.1007/978-1-4939-8859-4_8.

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

Conference papers on the topic "Notch signaling, Leukemia, Chemosensitivity"

1

Wei, Ping, Ming Qiu, Qinghai Peng, John Lippincott, Joseph Zachwieja, Eugenia Kraynov, Aidong Wu, et al. "Abstract 1765: Inhibition of Notch signaling by a Notch1 monoclonal antibody induces robust anti-tumor efficacy in T-cell acute lymphoblastic leukemia and breast cancer." In Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL. American Association for Cancer Research, 2011. http://dx.doi.org/10.1158/1538-7445.am2011-1765.

Full text
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