42 results on '"Van Loocke W"'
Search Results
2. Targeting BET proteins improves the therapeutic efficacy of BCL-2 inhibition in T-cell acute lymphoblastic leukemia
- Author
-
Peirs, S, Frismantas, V, Matthijssens, F, Van Loocke, W, Pieters, T, Vandamme, N, Lintermans, B, Dobay, M P, Berx, G, Poppe, B, Goossens, S, Bornhauser, B C, Bourquin, J-P, and Van Vlierberghe, P
- Published
- 2017
- Full Text
- View/download PDF
3. Long noncoding RNA signatures define oncogenic subtypes in T-cell acute lymphoblastic leukemia
- Author
-
Wallaert, A, Durinck, K, Van Loocke, W, Van de Walle, I, Matthijssens, F, Volders, P J, Avila Cobos, F, Rombaut, D, Rondou, P, Mestdagh, P, Vandesompele, J, Poppe, B, Taghon, T, Soulier, J, Van Vlierberghe, P, and Speleman, F
- Published
- 2016
- Full Text
- View/download PDF
4. Characterization of the genome-wide TLX1 binding profile in T-cell acute lymphoblastic leukemia
- Author
-
Durinck, K, Van Loocke, W, Van der Meulen, J, Van de Walle, I, Ongenaert, M, Rondou, P, Wallaert, A, de Bock, C E, Van Roy, N, Poppe, B, Cools, J, Soulier, J, Taghon, T, Speleman, F, and Van Vlierberghe, P
- Published
- 2015
- Full Text
- View/download PDF
5. RUNX2 regulates leukemic cell metabolism and chemotaxis in high-risk T cell acute lymphoblastic leukemia
- Author
-
Matthijssens, F, Sharma, ND, Nysus, M, Nickl, CK, Kang, H, Perez, DR, Lintermans, B, Van Loocke, W, Roels, J, Peirs, S, Demoen, L, Pieters, T, Reunes, L, Lammens, T, De Moerloose, B, Van Nieuwerburgh, F, Deforce, DL, Cheung, LC, Kotecha, RS, Risseeuw, MDP, Van Calenbergh, S, Takarada, T, Yoneda, Y, Van Delft, FW, Lock, RB ; https://orcid.org/0000-0002-3436-9071, Merkley, SD, Chigaev, A, Sklar, LA, Mullighan, CG, Loh, ML, Winter, SS, Hunger, SP, Goossens, S, Castillo, EF, Ornatowski, W, Van Vlierberghe, P, Matlawska-Wasowska, K, Matthijssens, F, Sharma, ND, Nysus, M, Nickl, CK, Kang, H, Perez, DR, Lintermans, B, Van Loocke, W, Roels, J, Peirs, S, Demoen, L, Pieters, T, Reunes, L, Lammens, T, De Moerloose, B, Van Nieuwerburgh, F, Deforce, DL, Cheung, LC, Kotecha, RS, Risseeuw, MDP, Van Calenbergh, S, Takarada, T, Yoneda, Y, Van Delft, FW, Lock, RB ; https://orcid.org/0000-0002-3436-9071, Merkley, SD, Chigaev, A, Sklar, LA, Mullighan, CG, Loh, ML, Winter, SS, Hunger, SP, Goossens, S, Castillo, EF, Ornatowski, W, Van Vlierberghe, P, and Matlawska-Wasowska, K
- Published
- 2021
6. MEIS2 Is an Adrenergic Core Regulatory Transcription Factor Involved in Early Initiation of TH-MYCN-Driven Neuroblastoma Formation.
- Author
-
De Wyn, J, Zimmerman, MW, Weichert-Leahey, N, Nunes, C, Cheung, BB, Abraham, BJ, Beckers, A, Volders, P-J, Decaesteker, B, Carter, DR, Look, AT, De Preter, K, Van Loocke, W, Marshall, GM, Durbin, AD, Speleman, F, Durinck, K, De Wyn, J, Zimmerman, MW, Weichert-Leahey, N, Nunes, C, Cheung, BB, Abraham, BJ, Beckers, A, Volders, P-J, Decaesteker, B, Carter, DR, Look, AT, De Preter, K, Van Loocke, W, Marshall, GM, Durbin, AD, Speleman, F, and Durinck, K
- Abstract
Roughly half of all high-risk neuroblastoma patients present with MYCN amplification. The molecular consequences of MYCN overexpression in this aggressive pediatric tumor have been studied for decades, but thus far, our understanding of the early initiating steps of MYCN-driven tumor formation is still enigmatic. We performed a detailed transcriptome landscaping during murine TH-MYCN-driven neuroblastoma tumor formation at different time points. The neuroblastoma dependency factor MEIS2, together with ASCL1, was identified as a candidate tumor-initiating factor and shown to be a novel core regulatory circuit member in adrenergic neuroblastomas. Of further interest, we found a KEOPS complex member (gm6890), implicated in homologous double-strand break repair and telomere maintenance, to be strongly upregulated during tumor formation, as well as the checkpoint adaptor Claspin (CLSPN) and three chromosome 17q loci CBX2, GJC1 and LIMD2. Finally, cross-species master regulator analysis identified FOXM1, together with additional hubs controlling transcriptome profiles of MYCN-driven neuroblastoma. In conclusion, time-resolved transcriptome analysis of early hyperplastic lesions and full-blown MYCN-driven neuroblastomas yielded novel components implicated in both tumor initiation and maintenance, providing putative novel drug targets for MYCN-driven neuroblastoma.
- Published
- 2021
7. PRL3 enhances T-cell acute lymphoblastic leukemia growth through suppressing T-cell signaling pathways and apoptosis
- Author
-
Garcia, E. G., primary, Veloso, A., additional, Oliveira, M. L., additional, Allen, J. R., additional, Loontiens, S., additional, Brunson, D., additional, Do, D., additional, Yan, C., additional, Morris, R., additional, Iyer, S., additional, Garcia, S. P., additional, Iftimia, N., additional, Van Loocke, W., additional, Matthijssens, F., additional, McCarthy, K., additional, Barata, J. T., additional, Speleman, F., additional, Taghon, T., additional, Gutierrez, A., additional, Van Vlierberghe, P., additional, Haas, W., additional, Blackburn, J. S., additional, and Langenau, D. M., additional
- Published
- 2020
- Full Text
- View/download PDF
8. S860 TARGETING ABERRANT DNA METHYLATION AS A NOVEL AND UNIFORM THERAPEUTIC STRATEGY FOR THE TREATMENT OF T-CELL ACUTE LYMPHOBLASTIC LEUKEMIA AND LYMPHOMA
- Author
-
Thenoz, M., primary, Roels, J., additional, Reunes, L., additional, Pieters, T., additional, Peirs, S., additional, Milani, G., additional, Van Loocke, W., additional, Lammens, T., additional, Deforce, D., additional, Van Nieuwerburgh, F., additional, Mansour, M., additional, Taghon, T., additional, Gossens, S., additional, and Van Vlierberghe, P., additional
- Published
- 2019
- Full Text
- View/download PDF
9. A comprehensive inventory of TLX1 controlled long non-coding RNAs in T-cell acute lymphoblastic leukemia through polyA+ and total RNA sequencing
- Author
-
Verboom, K, Van Loocke, W, Volders, PJ, Decaesteker, B, Avila Cobos, F, Bornschein, S, De Bock, CE ; https://orcid.org/0000-0001-5182-8535, Kalender Atak, Z, Clappier, E, Aerts, S, Cools, J, Soulier, J, Taghon, T, Van Vlierberghe, P, Vandesompele, J, Speleman, F, Durinck, K, Verboom, K, Van Loocke, W, Volders, PJ, Decaesteker, B, Avila Cobos, F, Bornschein, S, De Bock, CE ; https://orcid.org/0000-0001-5182-8535, Kalender Atak, Z, Clappier, E, Aerts, S, Cools, J, Soulier, J, Taghon, T, Van Vlierberghe, P, Vandesompele, J, Speleman, F, and Durinck, K
- Published
- 2018
10. The Notch driven long non-coding RNA repertoire in T-cell acute lymphoblastic leukemia
- Author
-
Durinck, K., primary, Wallaert, A., additional, Van de Walle, I., additional, Van Loocke, W., additional, Volders, P.-J., additional, Vanhauwaert, S., additional, Geerdens, E., additional, Benoit, Y., additional, Van Roy, N., additional, Poppe, B., additional, Soulier, J., additional, Cools, J., additional, Mestdagh, P., additional, Vandesompele, J., additional, Rondou, P., additional, Van Vlierberghe, P., additional, Taghon, T., additional, and Speleman, F., additional
- Published
- 2014
- Full Text
- View/download PDF
11. Myb overexpression synergizes with the loss of Pten and is a dependency factor and therapeutic target in T-cell lymphoblastic leukemia.
- Author
-
Almeida A, T'Sas S, Pagliaro L, Fijalkowski I, Sleeckx W, Van Steenberge H, Zamponi R, Lintermans B, Van Loocke W, Palhais B, Reekmans A, Bardelli V, Demoen L, Reunes L, Deforce D, Van Nieuwerburgh F, Kentsis A, Ntziachristos P, Van Roy N, De Moerloose B, Mecucci C, La Starza R, Roti G, Goossens S, Van Vlierberghe P, and Pieters T
- Abstract
T-lineage acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy that accounts for 10%-15% of pediatric and 25% of adult ALL cases. Although the prognosis of T-ALL has improved over time, the outcome of T-ALL patients with primary resistant or relapsed leukemia remains poor. Therefore, further progress in the treatment of T-ALL requires a better understanding of its biology and the development of more effective precision oncologic therapies. The proto-oncogene MYB is highly expressed in diverse hematologic malignancies, including T-ALLs with genomic aberrations that further potentiate its expression and activity. Previous studies have associated MYB with a malignant role in the pathogenesis of several cancers. However, its role in the induction and maintenance of T-ALL remains relatively poorly understood. In this study, we found that an increased copy number of MYB is associated with higher MYB expression levels, and might be associated with inferior event-free survival of pediatric T-ALL patients. Using our previously described conditional Myb overexpression mice, we generated two distinct MYB-driven T-ALL mouse models. We demonstrated that the overexpression of Myb synergizes with Pten deletion but not with the overexpression of Lmo2 to accelerate the development of T-cell lymphoblastic leukemias. We also showed that MYB is a dependency factor in T-ALL since RNA interference of Myb blocked cell cycle progression and induced apoptosis in both human and murine T-ALL cell lines. Finally, we provide preclinical evidence that targeting the transcriptional activity of MYB can be a useful therapeutic strategy for the treatment of T-ALL., Competing Interests: The authors declare no conflict of interest., (© 2024 The Authors. HemaSphere published by John Wiley & Sons Ltd on behalf of European Hematology Association.)
- Published
- 2024
- Full Text
- View/download PDF
12. Human iPSC modeling recapitulates in vivo sympathoadrenal development and reveals an aberrant developmental subpopulation in familial neuroblastoma.
- Author
-
Van Haver S, Fan Y, Bekaert SL, Everaert C, Van Loocke W, Zanzani V, Deschildre J, Maestre IF, Amaro A, Vermeirssen V, De Preter K, Zhou T, Kentsis A, Studer L, Speleman F, and Roberts SS
- Abstract
Studies defining normal and disrupted human neural crest cell development have been challenging given its early timing and intricacy of development. Consequently, insight into the early disruptive events causing a neural crest related disease such as pediatric cancer neuroblastoma is limited. To overcome this problem, we developed an in vitro differentiation model to recapitulate the normal in vivo developmental process of the sympathoadrenal lineage which gives rise to neuroblastoma. We used human in vitro pluripotent stem cells and single-cell RNA sequencing to recapitulate the molecular events during sympathoadrenal development. We provide a detailed map of dynamically regulated transcriptomes during sympathoblast formation and illustrate the power of this model to study early events of the development of human neuroblastoma, identifying a distinct subpopulation of cell marked by SOX2 expression in developing sympathoblast obtained from patient derived iPSC cells harboring a germline activating mutation in the anaplastic lymphoma kinase (ALK) gene., Competing Interests: A.K. is a consultant to Novartis, Rgenta, and Blueprint Medicines. L.S. is a scientific co-founder and consultant of Bluerock Therapeutics Inc. All other authors declare no conflict of interest., (© 2023 The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
13. SOX11 regulates SWI/SNF complex components as member of the adrenergic neuroblastoma core regulatory circuitry.
- Author
-
Decaesteker B, Louwagie A, Loontiens S, De Vloed F, Bekaert SL, Roels J, Vanhauwaert S, De Brouwer S, Sanders E, Berezovskaya A, Denecker G, D'haene E, Van Haver S, Van Loocke W, Van Dorpe J, Creytens D, Van Roy N, Pieters T, Van Neste C, Fischer M, Van Vlierberghe P, Roberts SS, Schulte J, Ek S, Versteeg R, Koster J, van Nes J, Zimmerman M, De Preter K, and Speleman F
- Subjects
- Humans, Child, Transcription Factors genetics, Chromatin, Cell Nucleus, Chromosome Aberrations, Adrenergic Agents, DNA Helicases, Nuclear Proteins genetics, SOXC Transcription Factors genetics, Histone Demethylases, Neuroblastoma genetics
- Abstract
The pediatric extra-cranial tumor neuroblastoma displays a low mutational burden while recurrent copy number alterations are present in most high-risk cases. Here, we identify SOX11 as a dependency transcription factor in adrenergic neuroblastoma based on recurrent chromosome 2p focal gains and amplifications, specific expression in the normal sympatho-adrenal lineage and adrenergic neuroblastoma, regulation by multiple adrenergic specific (super-)enhancers and strong dependency on high SOX11 expression in adrenergic neuroblastomas. SOX11 regulated direct targets include genes implicated in epigenetic control, cytoskeleton and neurodevelopment. Most notably, SOX11 controls chromatin regulatory complexes, including 10 SWI/SNF core components among which SMARCC1, SMARCA4/BRG1 and ARID1A. Additionally, the histone deacetylase HDAC2, PRC1 complex component CBX2, chromatin-modifying enzyme KDM1A/LSD1 and pioneer factor c-MYB are regulated by SOX11. Finally, SOX11 is identified as a core transcription factor of the core regulatory circuitry (CRC) in adrenergic high-risk neuroblastoma with a potential role as epigenetic master regulator upstream of the CRC., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
14. Pre-Clinical Evaluation of the Hypomethylating Agent Decitabine for the Treatment of T-Cell Lymphoblastic Lymphoma.
- Author
-
Provez L, Putteman T, Landfors M, Roels J, Reunes L, T'Sas S, Van Loocke W, Lintermans B, De Coninck S, Thenoz M, Sleeckx W, Maćkowska-Maślak N, Taghon T, Mansour MR, Farah N, Norga K, Vandenberghe P, Kotecha RS, Goossens S, Degerman S, De Smedt R, and Van Vlierberghe P
- Abstract
T-cell lymphoblastic lymphoma (T-LBL) is a rare and aggressive lymphatic cancer, often diagnosed at a young age. Patients are treated with intensive chemotherapy, potentially followed by a hematopoietic stem cell transplantation. Although prognosis of T-LBL has improved with intensified treatment protocols, they are associated with side effects and 10-20% of patients still die from relapsed or refractory disease. Given this, the search toward less toxic anti-lymphoma therapies is ongoing. Here, we targeted the recently described DNA hypermethylated profile in T-LBL with the DNA hypomethylating agent decitabine. We evaluated the anti-lymphoma properties and downstream effects of decitabine, using patient derived xenograft (PDX) models. Decitabine treatment resulted in prolonged lymphoma-free survival in all T-LBL PDX models, which was associated with downregulation of the oncogenic MYC pathway. However, some PDX models showed more benefit of decitabine treatment compared to others. In more sensitive models, differentially methylated CpG regions resulted in more differentially expressed genes in open chromatin regions. This resulted in stronger downregulation of cell cycle genes and upregulation of immune response activating transcripts. Finally, we suggest a gene signature for high decitabine sensitivity in T-LBL. Altogether, we here delivered pre-clinical proof of the potential use of decitabine as a new therapeutic agent in T-LBL.
- Published
- 2023
- Full Text
- View/download PDF
15. IRF2 is required for development and functional maturation of human NK cells.
- Author
-
Persyn E, Wahlen S, Kiekens L, Van Loocke W, Siwe H, Van Ammel E, De Vos Z, Van Nieuwerburgh F, Matthys P, Taghon T, Vandekerckhove B, Van Vlierberghe P, and Leclercq G
- Subjects
- Humans, Gene Expression Regulation, Cell Differentiation genetics, Cytokines metabolism, Interferon Regulatory Factor-2 genetics, Interferon Regulatory Factor-2 metabolism, Killer Cells, Natural metabolism, Transcription Factors metabolism
- Abstract
Natural killer (NK) cells are cytotoxic and cytokine-producing lymphocytes that play an important role in the first line of defense against malignant or virus-infected cells. A better understanding of the transcriptional regulation of human NK cell differentiation is crucial to improve the efficacy of NK cell-mediated immunotherapy for cancer treatment. Here, we studied the role of the transcription factor interferon regulatory factor (IRF) 2 in human NK cell differentiation by stable knockdown or overexpression in cord blood hematopoietic stem cells and investigated its effect on development and function of the NK cell progeny. IRF2 overexpression had limited effects in these processes, indicating that endogenous IRF2 expression levels are sufficient. However, IRF2 knockdown greatly reduced the cell numbers of all early differentiation stages, resulting in decimated NK cell numbers. This was not caused by increased apoptosis, but by decreased proliferation. Expression of IRF2 is also required for functional maturation of NK cells, as the remaining NK cells after silencing of IRF2 had a less mature phenotype and showed decreased cytotoxic potential, as well as a greatly reduced cytokine secretion. Thus, IRF2 plays an important role during development and functional maturation of human NK cells., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Persyn, Wahlen, Kiekens, Van Loocke, Siwe, Van Ammel, De Vos, Van Nieuwerburgh, Matthys, Taghon, Vandekerckhove, Van Vlierberghe and Leclercq.)
- Published
- 2022
- Full Text
- View/download PDF
16. TXNIP Promotes Human NK Cell Development but Is Dispensable for NK Cell Functionality.
- Author
-
Persyn E, Wahlen S, Kiekens L, Taveirne S, Van Loocke W, Van Ammel E, Van Nieuwerburgh F, Taghon T, Vandekerckhove B, Van Vlierberghe P, and Leclercq G
- Subjects
- Animals, Carrier Proteins genetics, Cell Differentiation genetics, Cytokines metabolism, Gene Expression, Humans, Mice, Killer Cells, Natural metabolism, Thioredoxins genetics, Thioredoxins metabolism
- Abstract
The ability of natural killer (NK) cells to kill tumor cells without prior sensitization makes them a rising player in immunotherapy. Increased understanding of the development and functioning of NK cells will improve their clinical utilization. As opposed to murine NK cell development, human NK cell development is still less understood. Here, we studied the role of thioredoxin-interacting protein (TXNIP) in human NK cell differentiation by stable TXNIP knockdown or overexpression in cord blood hematopoietic stem cells, followed by in vitro NK cell differentiation. TXNIP overexpression only had marginal effects, indicating that endogenous TXNIP levels are sufficient in this process. TXNIP knockdown, however, reduced proliferation of early differentiation stages and greatly decreased NK cell numbers. Transcriptome analysis and experimental confirmation showed that reduced protein synthesis upon TXNIP knockdown likely caused this low proliferation. Contrary to its profound effects on the early differentiation stages, TXNIP knockdown led to limited alterations in NK cell phenotype, and it had no effect on NK cell cytotoxicity or cytokine production. Thus, TXNIP promotes human NK cell differentiation by affecting protein synthesis and proliferation of early NK cell differentiation stages, but it is redundant for functional NK cell maturation.
- Published
- 2022
- Full Text
- View/download PDF
17. RRM2 enhances MYCN-driven neuroblastoma formation and acts as a synergistic target with CHK1 inhibition.
- Author
-
Nunes C, Depestel L, Mus L, Keller KM, Delhaye L, Louwagie A, Rishfi M, Whale A, Kara N, Andrews SR, Dela Cruz F, You D, Siddiquee A, Cologna CT, De Craemer S, Dolman E, Bartenhagen C, De Vloed F, Sanders E, Eggermont A, Bekaert SL, Van Loocke W, Bek JW, Dewyn G, Loontiens S, Van Isterdael G, Decaesteker B, Tilleman L, Van Nieuwerburgh F, Vermeirssen V, Van Neste C, Ghesquiere B, Goossens S, Eyckerman S, De Preter K, Fischer M, Houseley J, Molenaar J, De Wilde B, Roberts SS, Durinck K, and Speleman F
- Abstract
High-risk neuroblastoma, a pediatric tumor originating from the sympathetic nervous system, has a low mutation load but highly recurrent somatic DNA copy number variants. Previously, segmental gains and/or amplifications allowed identification of drivers for neuroblastoma development. Using this approach, combined with gene dosage impact on expression and survival, we identified ribonucleotide reductase subunit M2 (RRM2) as a candidate dependency factor further supported by growth inhibition upon in vitro knockdown and accelerated tumor formation in a neuroblastoma zebrafish model coexpressing human RRM2 with MYCN. Forced RRM2 induction alleviates excessive replicative stress induced by CHK1 inhibition, while high RRM2 expression in human neuroblastomas correlates with high CHK1 activity. MYCN-driven zebrafish tumors with RRM2 co-overexpression exhibit differentially expressed DNA repair genes in keeping with enhanced ATR-CHK1 signaling activity. In vitro, RRM2 inhibition enhances intrinsic replication stress checkpoint addiction. Last, combinatorial RRM2-CHK1 inhibition acts synergistic in high-risk neuroblastoma cell lines and patient-derived xenograft models, illustrating the therapeutic potential.
- Published
- 2022
- Full Text
- View/download PDF
18. The transcription factor RUNX2 drives the generation of human NK cells and promotes tissue residency.
- Author
-
Wahlen S, Matthijssens F, Van Loocke W, Taveirne S, Kiekens L, Persyn E, Van Ammel E, De Vos Z, De Munter S, Matthys P, Van Nieuwerburgh F, Taghon T, Vandekerckhove B, Van Vlierberghe P, and Leclercq G
- Subjects
- Humans, Core Binding Factor Alpha 1 Subunit metabolism, Gene Expression Regulation, Killer Cells, Natural metabolism, Transcription Factors metabolism
- Abstract
Natural killer (NK) cells are innate lymphocytes that eliminate virus-infected and cancer cells by cytotoxicity and cytokine secretion. In addition to circulating NK cells, distinct tissue-resident NK subsets have been identified in various organs. Although transcription factors regulating NK cell development and function have been extensively studied in mice, the role of RUNX2 in these processes has not been investigated, neither in mice nor in human. Here, by manipulating RUNX2 expression with either knockdown or overexpression in human haematopoietic stem cell-based NK cell differentiation cultures, combined with transcriptomic and ChIP-sequencing analyses, we established that RUNX2 drives the generation of NK cells, possibly through induction of IL-2Rβ expression in NK progenitor cells. Importantly, RUNX2 promotes tissue residency in human NK cells. Our findings have the potential to improve existing NK cell-based cancer therapies and can impact research fields beyond NK cell biology, since tissue-resident subsets have also been described in other lymphocyte subpopulations., Competing Interests: SW, FM, WV, ST, LK, EP, EV, ZD, SD, PM, FV, TT, BV, PV, GL No competing interests declared, (© 2022, Wahlen et al.)
- Published
- 2022
- Full Text
- View/download PDF
19. ETV6-NCOA2 fusion induces T/myeloid mixed-phenotype leukemia through transformation of nonthymic hematopoietic progenitor cells.
- Author
-
Fishman H, Madiwale S, Geron I, Bari V, Van Loocke W, Kirschenbaum Y, Ganmore I, Kugler E, Rein-Gil A, Friedlander G, Schiby G, Birger Y, Strehl S, Soulier J, Knoechel B, Ferrando A, Noy-Lotan S, Nagler A, Mulloy JC, Van Vlierberghe P, and Izraeli S
- Subjects
- Animals, Cell Transformation, Neoplastic, Cells, Cultured, Female, Hematopoietic Stem Cells pathology, Humans, Leukemia, Myeloid pathology, Mice, Mice, Inbred C57BL, Mice, SCID, ETS Translocation Variant 6 Protein, Gene Expression Regulation, Leukemic, Hematopoietic Stem Cells metabolism, Leukemia, Myeloid genetics, Nuclear Receptor Coactivator 2 genetics, Oncogene Proteins, Fusion genetics, Proto-Oncogene Proteins c-ets genetics, Repressor Proteins genetics
- Abstract
Mixed-phenotype acute leukemia is a rare subtype of leukemia in which both myeloid and lymphoid markers are co-expressed on the same malignant cells. The pathogenesis is largely unknown, and the treatment is challenging. We previously reported the specific association of the recurrent t(8;12)(q13;p13) chromosomal translocation that creates the ETV6-NCOA2 fusion with T/myeloid leukemias. Here we report that ETV6-NCOA2 initiates T/myeloid leukemia in preclinical models; ectopic expression of ETV6-NCOA2 in mouse bone marrow hematopoietic progenitors induced T/myeloid lymphoma accompanied by spontaneous Notch1-activating mutations. Similarly, cotransduction of human cord blood CD34+ progenitors with ETV6-NCOA2 and a nontransforming NOTCH1 mutant induced T/myeloid leukemia in immunodeficient mice; the immunophenotype and gene expression pattern were similar to those of patient-derived ETV6-NCOA2 leukemias. Mechanistically, we show that ETV6-NCOA2 forms a transcriptional complex with ETV6 and the histone acetyltransferase p300, leading to derepression of ETV6 target genes. The expression of ETV6-NCOA2 in human and mouse nonthymic hematopoietic progenitor cells induces transcriptional dysregulation, which activates a lymphoid program while failing to repress the expression of myeloid genes such as CSF1 and MEF2C. The ETV6-NCOA2 induced arrest at an early immature T-cell developmental stage. The additional acquisition of activating NOTCH1 mutations transforms the early immature ETV6-NCOA2 cells into T/myeloid leukemias. Here, we describe the first preclinical model to depict the initiation of T/myeloid leukemia by a specific somatic genetic aberration., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
20. Cyclin D2 overexpression drives B1a-derived MCL-like lymphoma in mice.
- Author
-
Pieters T, T'Sas S, Vanhee S, Almeida A, Driege Y, Roels J, Van Loocke W, Daneels W, Baens M, Marchand A, Van Trimpont M, Matthijssens F, Morscio J, Lemeire K, Lintermans B, Reunes L, Chaltin P, Offner F, Van Dorpe J, Hochepied T, Berx G, Beyaert R, Staal J, Van Vlierberghe P, and Goossens S
- Subjects
- Allografts, Animals, B-Lymphocytes metabolism, B-Lymphocytes pathology, Cyclin D2 metabolism, Gene Expression Regulation, Neoplastic, Lymphoma, Mantle-Cell drug therapy, Mice, Inbred C57BL, Mice, Transgenic, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein metabolism, Neoplasms, Experimental drug therapy, Neoplasms, Experimental genetics, Neoplasms, Experimental pathology, Neoplastic Cells, Circulating, Tumor Suppressor Protein p53 genetics, Xenograft Model Antitumor Assays, Mice, Cyclin D2 genetics, Lymphoma, Mantle-Cell genetics, Lymphoma, Mantle-Cell pathology, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein antagonists & inhibitors
- Abstract
Mantle cell lymphoma (MCL) is an aggressive B cell lymphoma with poor long-term overall survival. Currently, MCL research and development of potential cures is hampered by the lack of good in vivo models. MCL is characterized by recurrent translocations of CCND1 or CCND2, resulting in overexpression of the cell cycle regulators cyclin D1 or D2, respectively. Here, we show, for the first time, that hematopoiesis-specific activation of cyclin D2 is sufficient to drive murine MCL-like lymphoma development. Furthermore, we demonstrate that cyclin D2 overexpression can synergize with loss of p53 to form aggressive and transplantable MCL-like lymphomas. Strikingly, cyclin D2-driven lymphomas display transcriptional, immunophenotypic, and functional similarities with B1a B cells. These MCL-like lymphomas have B1a-specific B cell receptors (BCRs), show elevated BCR and NF-κB pathway activation, and display increased MALT1 protease activity. Finally, we provide preclinical evidence that inhibition of MALT1 protease activity, which is essential for the development of early life-derived B1a cells, can be an effective therapeutic strategy to treat MCL., Competing Interests: Disclosures: R. Beyaert reported grants from Galapagos nv outside the submitted work; in addition, R. Beyaert had a patent to WO09065897 issued. No other disclosures were reported., (© 2021 Pieters et al.)
- Published
- 2021
- Full Text
- View/download PDF
21. MEIS2 Is an Adrenergic Core Regulatory Transcription Factor Involved in Early Initiation of TH-MYCN-Driven Neuroblastoma Formation.
- Author
-
De Wyn J, Zimmerman MW, Weichert-Leahey N, Nunes C, Cheung BB, Abraham BJ, Beckers A, Volders PJ, Decaesteker B, Carter DR, Look AT, De Preter K, Van Loocke W, Marshall GM, Durbin AD, Speleman F, and Durinck K
- Abstract
Roughly half of all high-risk neuroblastoma patients present with MYCN amplification. The molecular consequences of MYCN overexpression in this aggressive pediatric tumor have been studied for decades, but thus far, our understanding of the early initiating steps of MYCN-driven tumor formation is still enigmatic. We performed a detailed transcriptome landscaping during murine TH-MYCN-driven neuroblastoma tumor formation at different time points. The neuroblastoma dependency factor MEIS2, together with ASCL1, was identified as a candidate tumor-initiating factor and shown to be a novel core regulatory circuit member in adrenergic neuroblastomas. Of further interest, we found a KEOPS complex member ( gm6890 ), implicated in homologous double-strand break repair and telomere maintenance, to be strongly upregulated during tumor formation, as well as the checkpoint adaptor Claspin ( CLSPN ) and three chromosome 17q loci CBX2 , GJC1 and LIMD2 . Finally, cross-species master regulator analysis identified FOXM1, together with additional hubs controlling transcriptome profiles of MYCN-driven neuroblastoma. In conclusion, time-resolved transcriptome analysis of early hyperplastic lesions and full-blown MYCN-driven neuroblastomas yielded novel components implicated in both tumor initiation and maintenance, providing putative novel drug targets for MYCN-driven neuroblastoma.
- Published
- 2021
- Full Text
- View/download PDF
22. T-BET and EOMES Accelerate and Enhance Functional Differentiation of Human Natural Killer Cells.
- Author
-
Kiekens L, Van Loocke W, Taveirne S, Wahlen S, Persyn E, Van Ammel E, De Vos Z, Matthys P, Van Nieuwerburgh F, Taghon T, Van Vlierberghe P, Vandekerckhove B, and Leclercq G
- Subjects
- Animals, Antibody-Dependent Cell Cytotoxicity, Cell Lineage, Chromatin Assembly and Disassembly, Coculture Techniques, Epigenesis, Genetic, Fetal Blood cytology, GPI-Linked Proteins genetics, GPI-Linked Proteins metabolism, Hematopoietic Stem Cells immunology, Humans, Interferon-gamma metabolism, K562 Cells, Killer Cells, Natural immunology, Mice, Phenotype, Receptors, IgG genetics, Receptors, IgG metabolism, Receptors, KIR genetics, Receptors, KIR metabolism, T-Box Domain Proteins genetics, Transcriptome, T-bet Transcription Factor, Cell Differentiation, Hematopoietic Stem Cells metabolism, Killer Cells, Natural metabolism, T-Box Domain Proteins metabolism
- Abstract
T-bet and Eomes are transcription factors that are known to be important in maturation and function of murine natural killer (NK) cells. Reduced T-BET and EOMES expression results in dysfunctional NK cells and failure to control tumor growth. In contrast to mice, the current knowledge on the role of T-BET and EOMES in human NK cells is rudimentary. Here, we ectopically expressed either T-BET or EOMES in human hematopoietic progenitor cells. Combined transcriptome, chromatin accessibility and protein expression analyses revealed that T-BET or EOMES epigenetically represses hematopoietic stem cell quiescence and non-NK lineage differentiation genes, while activating an NK cell-specific transcriptome and thereby drastically accelerating NK cell differentiation. In this model, the effects of T-BET and EOMES are largely overlapping, yet EOMES shows a superior role in early NK cell maturation and induces faster NK receptor and enhanced CD16 expression. T-BET particularly controls transcription of terminal maturation markers and epigenetically controls strong induction of KIR expression. Finally, NK cells generated upon T-BET or EOMES overexpression display improved functionality, including increased IFN-γ production and killing, and especially EOMES overexpression NK cells have enhanced antibody-dependent cellular cytotoxicity. Our findings reveal novel insights on the regulatory role of T-BET and EOMES in human NK cell maturation and function, which is essential to further understand human NK cell biology and to optimize adoptive NK cell therapies., Competing Interests: An international patent application WO2020070070 was filed by Ghent University (Ghent, Belgium) on 30/09/2019, with title ‘Accelerated human hematopoietic stem cell differentiation towards mature natural killer cells with enhanced antibody-dependent cytotoxic activity’, with LK and GL as inventors. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Kiekens, Van Loocke, Taveirne, Wahlen, Persyn, Van Ammel, De Vos, Matthys, Van Nieuwerburgh, Taghon, Van Vlierberghe, Vandekerckhove and Leclercq.)
- Published
- 2021
- Full Text
- View/download PDF
23. RUNX2 regulates leukemic cell metabolism and chemotaxis in high-risk T cell acute lymphoblastic leukemia.
- Author
-
Matthijssens F, Sharma ND, Nysus M, Nickl CK, Kang H, Perez DR, Lintermans B, Van Loocke W, Roels J, Peirs S, Demoen L, Pieters T, Reunes L, Lammens T, De Moerloose B, Van Nieuwerburgh F, Deforce DL, Cheung LC, Kotecha RS, Risseeuw MD, Van Calenbergh S, Takarada T, Yoneda Y, van Delft FW, Lock RB, Merkley SD, Chigaev A, Sklar LA, Mullighan CG, Loh ML, Winter SS, Hunger SP, Goossens S, Castillo EF, Ornatowski W, Van Vlierberghe P, and Matlawska-Wasowska K
- Subjects
- Animals, Cell Line, Tumor, Chemotaxis, Leukocyte, Child, Core Binding Factor Alpha 1 Subunit genetics, Core Binding Factor beta Subunit metabolism, Disease Progression, Gene Expression Regulation, Developmental, Gene Expression Regulation, Neoplastic, Gene Rearrangement, Hematopoiesis, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, Humans, In Vitro Techniques, Mice, Myeloid-Lymphoid Leukemia Protein genetics, Myeloid-Lymphoid Leukemia Protein metabolism, Organelle Biogenesis, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma pathology, RNA, Messenger genetics, RNA, Messenger metabolism, Receptors, CXCR4 metabolism, Signal Transduction, Core Binding Factor Alpha 1 Subunit metabolism, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma metabolism
- Abstract
T cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic malignancy with inferior outcome compared with that of B cell ALL. Here, we show that Runt-related transcription factor 2 (RUNX2) was upregulated in high-risk T-ALL with KMT2A rearrangements (KMT2A-R) or an immature immunophenotype. In KMT2A-R cells, we identified RUNX2 as a direct target of the KMT2A chimeras, where it reciprocally bound the KMT2A promoter, establishing a regulatory feed-forward mechanism. Notably, RUNX2 was required for survival of immature and KMT2A-R T-ALL cells in vitro and in vivo. We report direct transcriptional regulation of CXCR4 signaling by RUNX2, thereby promoting chemotaxis, adhesion, and homing to medullary and extramedullary sites. RUNX2 enabled these energy-demanding processes by increasing metabolic activity in T-ALL cells through positive regulation of both glycolysis and oxidative phosphorylation. Concurrently, RUNX2 upregulation increased mitochondrial dynamics and biogenesis in T-ALL cells. Finally, as a proof of concept, we demonstrate that immature and KMT2A-R T-ALL cells were vulnerable to pharmacological targeting of the interaction between RUNX2 and its cofactor CBFβ. In conclusion, we show that RUNX2 acts as a dependency factor in high-risk subtypes of human T-ALL through concomitant regulation of tumor metabolism and leukemic cell migration.
- Published
- 2021
- Full Text
- View/download PDF
24. Long non-coding RNAs as novel therapeutic targets in juvenile myelomonocytic leukemia.
- Author
-
Hofmans M, Lammens T, Depreter B, Wu Y, Erlacher M, Caye A, Cavé H, Flotho C, de Haas V, Niemeyer CM, Stary J, Van Nieuwerburgh F, Deforce D, Van Loocke W, Van Vlierberghe P, Philippé J, and De Moerloose B
- Subjects
- Adolescent, Antineoplastic Agents therapeutic use, Bone Marrow pathology, Case-Control Studies, Child, Child, Preschool, Female, Gene Knockdown Techniques, Healthy Volunteers, Humans, Infant, Leukemia, Myelomonocytic, Juvenile blood, Leukemia, Myelomonocytic, Juvenile drug therapy, Leukemia, Myelomonocytic, Juvenile pathology, Leukocytes, Mononuclear, Male, Primary Cell Culture, RNA, Long Noncoding antagonists & inhibitors, RNA, Long Noncoding genetics, RNA-Seq, Tumor Cells, Cultured, Antineoplastic Agents pharmacology, Gene Expression Regulation, Leukemic drug effects, Leukemia, Myelomonocytic, Juvenile genetics, RNA, Long Noncoding metabolism
- Abstract
Juvenile myelomonocytic leukemia (JMML) treatment primarily relies on hematopoietic stem cell transplantation and results in long-term overall survival of 50-60%, demonstrating a need to develop novel treatments. Dysregulation of the non-coding RNA transcriptome has been demonstrated before in this rare and unique disorder of early childhood. In this study, we investigated the therapeutic potential of targeting overexpressed long non-coding RNAs (lncRNAs) in JMML. Total RNA sequencing of bone marrow and peripheral blood mononuclear cell preparations from 19 untreated JMML patients and three healthy children revealed 185 differentially expressed lncRNA genes (131 up- and 54 downregulated). LNA GapmeRs were designed for 10 overexpressed and validated lncRNAs. Molecular knockdown (≥ 70% compared to mock control) after 24 h of incubation was observed with two or more independent GapmeRs in 6 of them. For three lncRNAs (lnc-THADA-4, lnc-ACOT9-1 and NRIR) knockdown resulted in a significant decrease of cell viability after 72 h of incubation in primary cultures of JMML mononuclear cells, respectively. Importantly, the extent of cellular damage correlated with the expression level of the lncRNA of interest. In conclusion, we demonstrated in primary JMML cell cultures that knockdown of overexpressed lncRNAs such as lnc-THADA-4, lnc-ACOT9-1 and NRIR may be a feasible therapeutic strategy.
- Published
- 2021
- Full Text
- View/download PDF
25. A novel TLX1-driven T-ALL zebrafish model: comparative genomic analysis with other leukemia models.
- Author
-
Loontiens S, Vanhauwaert S, Depestel L, Dewyn G, Van Loocke W, Moore FE, Garcia EG, Batchelor L, Borga C, Squiban B, Malone-Perez M, Volders PJ, Olexiouk V, Van Vlierberghe P, Langenau DM, Frazer JK, Durinck K, and Speleman F
- Subjects
- Animals, Comparative Genomic Hybridization methods, Gene Transfer Techniques, Genomics methods, Zebrafish, Homeodomain Proteins genetics, Leukemia-Lymphoma, Adult T-Cell genetics, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics, Proto-Oncogene Proteins genetics
- Published
- 2020
- Full Text
- View/download PDF
26. Aging of preleukemic thymocytes drives CpG island hypermethylation in T-cell acute lymphoblastic leukemia.
- Author
-
Roels J, Thénoz M, Szarzyńska B, Landfors M, De Coninck S, Demoen L, Provez L, Kuchmiy A, Strubbe S, Reunes L, Pieters T, Matthijssens F, Van Loocke W, Erarslan-Uysal B, Richter-Pechańska P, Declerck K, Lammens T, De Moerloose B, Deforce D, Van Nieuwerburgh F, Cheung LC, Kotecha RS, Mansour MR, Ghesquière B, Van Camp G, Berghe WV, Kowalczyk JR, Szczepański T, Davé UP, Kulozik AE, Goossens S, Curtis DJ, Taghon T, Dawidowska M, Degerman S, and Van Vlierberghe P
- Subjects
- CpG Islands genetics, DNA Methylation genetics, Humans, Aging, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics, Thymocytes
- Abstract
Cancer cells display DNA hypermethylation at specific CpG islands in comparison to their normal healthy counterparts, but the mechanism that drives this so-called CpG island methylator phenotype (CIMP) remains poorly understood. Here, we show that CpG island methylation in human T-cell acute lymphoblastic leukemia (T-ALL) mainly occurs at promoters of Polycomb Repressor Complex 2 (PRC2) target genes that are not expressed in normal or malignant T-cells and which display a reciprocal association with H3K27me3 binding. In addition, we revealed that this aberrant methylation profile reflects the epigenetic history of T-ALL and is established already in pre-leukemic, self-renewing thymocytes that precede T-ALL development. Finally, we unexpectedly uncover that this age-related CpG island hypermethylation signature in T-ALL is completely resistant to the FDA-approved hypomethylating agent Decitabine. Altogether, we here provide conceptual evidence for the involvement of a pre-leukemic phase characterized by self-renewing thymocytes in the pathogenesis of human T-ALL., Competing Interests: Conflicts of Interest The authors declare no potential conflicts of interest.
- Published
- 2020
- Full Text
- View/download PDF
27. The transcription factor ETS1 is an important regulator of human NK cell development and terminal differentiation.
- Author
-
Taveirne S, Wahlen S, Van Loocke W, Kiekens L, Persyn E, Van Ammel E, De Mulder K, Roels J, Tilleman L, Aumercier M, Matthys P, Van Nieuwerburgh F, Kerre TCC, Taghon T, Van Vlierberghe P, Vandekerckhove B, and Leclercq G
- Subjects
- Apoptosis genetics, Apoptosis immunology, Cell Differentiation genetics, Cell Line, Gene Expression Profiling, Genome-Wide Association Study, Human Embryonic Stem Cells cytology, Humans, Killer Cells, Natural cytology, Protein Isoforms genetics, Protein Isoforms immunology, Proto-Oncogene Protein c-ets-1 genetics, Cell Differentiation immunology, Gene Expression Regulation immunology, Human Embryonic Stem Cells immunology, Killer Cells, Natural immunology, Lymphocyte Activation, Proto-Oncogene Protein c-ets-1 immunology
- Abstract
Natural killer (NK) cells are important in the immune defense against tumor cells and pathogens, and they regulate other immune cells by cytokine secretion. Although murine NK cell biology has been extensively studied, knowledge about transcriptional circuitries controlling human NK cell development and maturation is limited. By generating ETS1-deficient human embryonic stem cells and by expressing the dominant-negative ETS1 p27 isoform in cord blood hematopoietic progenitor cells, we show that the transcription factor ETS1 is critically required for human NK cell differentiation. Genome-wide transcriptome analysis determined by RNA-sequencing combined with chromatin immunoprecipitation-sequencing analysis reveals that human ETS1 directly induces expression of key transcription factors that control NK cell differentiation (ie, E4BP4, TXNIP, TBET, GATA3, HOBIT, BLIMP1). In addition, ETS1 regulates expression of genes involved in apoptosis and NK cell activation. Our study provides important molecular insights into the role of ETS1 as an important regulator of human NK cell development and terminal differentiation., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
28. The EMT Transcription Factor ZEB2 Promotes Proliferation of Primary and Metastatic Melanoma While Suppressing an Invasive, Mesenchymal-Like Phenotype.
- Author
-
Vandamme N, Denecker G, Bruneel K, Blancke G, Akay Ö, Taminau J, De Coninck J, De Smedt E, Skrypek N, Van Loocke W, Wouters J, Nittner D, Köhler C, Darling DS, Cheng PF, Raaijmakers MIG, Levesque MP, Mallya UG, Rafferty M, Balint B, Gallagher WM, Brochez L, Huylebroeck D, Haigh JJ, Andries V, Rambow F, Van Vlierberghe P, Goossens S, van den Oord JJ, Marine JC, and Berx G
- Subjects
- Animals, Humans, Lung Neoplasms genetics, Lung Neoplasms metabolism, Melanoma genetics, Melanoma metabolism, Mice, Neoplasm Invasiveness, Transcription Factors genetics, Tumor Cells, Cultured, Zinc Finger E-box Binding Homeobox 2 genetics, Cell Proliferation, Epithelial-Mesenchymal Transition, Gene Expression Regulation, Neoplastic, Lung Neoplasms secondary, Melanoma pathology, Transcription Factors metabolism, Zinc Finger E-box Binding Homeobox 2 metabolism
- Abstract
Epithelial-to-mesenchymal transition (EMT)-inducing transcription factors (TF) are well known for their ability to induce mesenchymal states associated with increased migratory and invasive properties. Unexpectedly, nuclear expression of the EMT-TF ZEB2 in human primary melanoma has been shown to correlate with reduced invasion. We report here that ZEB2 is required for outgrowth for primary melanomas and metastases at secondary sites. Ablation of Zeb2 hampered outgrowth of primary melanomas in vivo , whereas ectopic expression enhanced proliferation and growth at both primary and secondary sites. Gain of Zeb2 expression in pulmonary-residing melanoma cells promoted the development of macroscopic lesions. In vivo fate mapping made clear that melanoma cells undergo a conversion in state where ZEB2 expression is replaced by ZEB1 expression associated with gain of an invasive phenotype. These findings suggest that reversible switching of the ZEB2/ZEB1 ratio enhances melanoma metastatic dissemination. SIGNIFICANCE: ZEB2 function exerts opposing behaviors in melanoma by promoting proliferation and expansion and conversely inhibiting invasiveness, which could be of future clinical relevance. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/14/2983/F1.large.jpg., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
29. Targeting cytokine- and therapy-induced PIM1 activation in preclinical models of T-cell acute lymphoblastic leukemia and lymphoma.
- Author
-
De Smedt R, Morscio J, Reunes L, Roels J, Bardelli V, Lintermans B, Van Loocke W, Almeida A, Cheung LC, Kotecha RS, Mansour MR, Uyttebroeck A, Vandenberghe P, La Starza R, Mecucci C, Lammens T, Van Roy N, De Moerloose B, Barata JT, Taghon T, Goossens S, and Van Vlierberghe P
- Subjects
- Animals, Apoptosis, Cell Proliferation, Drug Therapy, Combination, Humans, Mice, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma immunology, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma pathology, T-Lymphocytes drug effects, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antineoplastic Combined Chemotherapy Protocols pharmacology, Cytokines pharmacology, Gene Expression Regulation, Neoplastic drug effects, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Proto-Oncogene Proteins c-pim-1 metabolism, T-Lymphocytes immunology
- Abstract
T-cell acute lymphoblastic leukemia (T-ALL) and T-cell acute lymphoblastic lymphoma (T-LBL) are aggressive hematological malignancies that are currently treated with high-dose chemotherapy. Over the last several years, the search toward novel and less-toxic therapeutic strategies for T-ALL/T-LBL patients has largely focused on the identification of cell-intrinsic properties of the tumor cell. However, non-cell-autonomous activation of specific oncogenic pathways might also offer opportunities that could be exploited at the therapeutic level. In line with this, we here show that endogenous interleukin 7 (IL7) can increase the expression of the oncogenic kinase proviral integration site for Moloney-murine leukemia 1 (PIM1) in CD127+ T-ALL/T-LBL, thereby rendering these tumor cells sensitive to in vivo PIM inhibition. In addition, using different CD127+ T-ALL/T-LBL xenograft models, we also reveal that residual tumor cells, which remain present after short-term in vivo chemotherapy, display consistent upregulation of PIM1 as compared with bulk nontreated tumor cells. Notably, this effect was transient as increased PIM1 levels were not observed in reestablished disease after abrogation of the initial chemotherapy. Furthermore, we uncover that this phenomenon is, at least in part, mediated by the ability of glucocorticoids to cause transcriptional upregulation of IL7RA in T-ALL/T-LBL patient-derived xenograft (PDX) cells, ultimately resulting in non-cell-autonomous PIM1 upregulation by endogenous IL7. Finally, we confirm in vivo that chemotherapy in combination with a pan-PIM inhibitor can improve leukemia survival in a PDX model of CD127+ T-ALL. Altogether, our work reveals that IL7 and glucocorticoids coordinately drive aberrant activation of PIM1 and suggests that IL7-responsive CD127+ T-ALL and T-LBL patients could benefit from PIM inhibition during induction chemotherapy., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
30. The ETS transcription factor ETV5 is a target of activated ALK in neuroblastoma contributing to increased tumour aggressiveness.
- Author
-
Mus LM, Lambertz I, Claeys S, Kumps C, Van Loocke W, Van Neste C, Umapathy G, Vaapil M, Bartenhagen C, Laureys G, De Wever O, Bexell D, Fischer M, Hallberg B, Schulte J, De Wilde B, Durinck K, Denecker G, De Preter K, and Speleman F
- Subjects
- Anaplastic Lymphoma Kinase genetics, Animals, Apoptosis, Biomarkers, Tumor genetics, DNA-Binding Proteins genetics, Female, Humans, Mice, Mice, Nude, Neuroblastoma genetics, Neuroblastoma metabolism, Transcription Factors genetics, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Anaplastic Lymphoma Kinase metabolism, Biomarkers, Tumor metabolism, Cell Proliferation, DNA-Binding Proteins metabolism, Gene Expression Regulation, Neoplastic, Neuroblastoma pathology, Transcription Factors metabolism
- Abstract
Neuroblastoma is an aggressive childhood cancer arising from sympatho-adrenergic neuronal progenitors. The low survival rates for high-risk disease point to an urgent need for novel targeted therapeutic approaches. Detailed molecular characterization of the neuroblastoma genomic landscape indicates that ALK-activating mutations are present in 10% of primary tumours. Together with other mutations causing RAS/MAPK pathway activation, ALK mutations are also enriched in relapsed cases and ALK activation was shown to accelerate MYCN-driven tumour formation through hitherto unknown ALK-driven target genes. To gain further insight into how ALK contributes to neuroblastoma aggressiveness, we searched for known oncogenes in our previously reported ALK-driven gene signature. We identified ETV5, a bona fide oncogene in prostate cancer, as robustly upregulated in neuroblastoma cells harbouring ALK mutations, and show high ETV5 levels downstream of the RAS/MAPK axis. Increased ETV5 expression significantly impacted migration, invasion and colony formation in vitro, and ETV5 knockdown reduced proliferation in a murine xenograft model. We also established a gene signature associated with ETV5 knockdown that correlates with poor patient survival. Taken together, our data highlight ETV5 as an intrinsic component of oncogenic ALK-driven signalling through the MAPK axis and propose that ETV5 upregulation in neuroblastoma may contribute to tumour aggressiveness.
- Published
- 2020
- Full Text
- View/download PDF
31. Genetic characterization and therapeutic targeting of MYC-rearranged T cell acute lymphoblastic leukaemia.
- Author
-
Milani G, Matthijssens F, Van Loocke W, Durinck K, Roels J, Peirs S, Thénoz M, Pieters T, Reunes L, Lintermans B, Vandamme N, Lammens T, Van Roy N, Van Nieuwerburgh F, Deforce D, Schwab C, Raimondi S, Dalla Pozza L, Carroll AJ 3rd, De Moerloose B, Benoit Y, Goossens S, Berx G, Harrison CJ, Basso G, Cavé H, Sutton R, Asnafi V, Meijerink J, Mullighan C, Loh M, and Van Vlierberghe P
- Subjects
- Animals, Biomarkers, Tumor, DNA Copy Number Variations, Disease Models, Animal, Genetic Association Studies, Humans, Mice, Molecular Targeted Therapy, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma pathology, Translocation, Genetic, Xenograft Model Antitumor Assays, Gene Rearrangement, Genes, myc, Genetic Predisposition to Disease, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics
- Published
- 2019
- Full Text
- View/download PDF
32. Purification of high-quality RNA from a small number of fluorescence activated cell sorted zebrafish cells for RNA sequencing purposes.
- Author
-
Loontiens S, Depestel L, Vanhauwaert S, Dewyn G, Gistelinck C, Verboom K, Van Loocke W, Matthijssens F, Willaert A, Vandesompele J, Speleman F, and Durinck K
- Subjects
- Animals, Cell Count, Poly A genetics, Quality Control, Flow Cytometry, RNA genetics, RNA isolation & purification, Sequence Analysis, RNA, Zebrafish genetics
- Abstract
Background: Transgenic zebrafish lines with the expression of a fluorescent reporter under the control of a cell-type specific promoter, enable transcriptome analysis of FACS sorted cell populations. RNA quality and yield are key determinant factors for accurate expression profiling. Limited cell number and FACS induced cellular stress make RNA isolation of sorted zebrafish cells a delicate process. We aimed to optimize a workflow to extract sufficient amounts of high-quality RNA from a limited number of FACS sorted cells from Tg(fli1a:GFP) zebrafish embryos, which can be used for accurate gene expression analysis., Results: We evaluated two suitable RNA isolation kits (the RNAqueous micro and the RNeasy plus micro kit) and determined that sorting cells directly into lysis buffer is a critical step for success. For low cell numbers, this ensures direct cell lysis, protects RNA from degradation and results in a higher RNA quality and yield. We showed that this works well up to 0.5× dilution of the lysis buffer with sorted cells. In our sort settings, this corresponded to 30,000 and 75,000 cells for the RNAqueous micro kit and RNeasy plus micro kit respectively. Sorting more cells dilutes the lysis buffer too much and requires the use of a collection buffer. We also demonstrated that an additional genomic DNA removal step after RNA isolation is required to completely clear the RNA from any contaminating genomic DNA. For cDNA synthesis and library preparation, we combined SmartSeq v4 full length cDNA library amplification, Nextera XT tagmentation and sample barcoding. Using this workflow, we were able to generate highly reproducible RNA sequencing results., Conclusions: The presented optimized workflow enables to generate high quality RNA and allows accurate transcriptome profiling of small populations of sorted zebrafish cells.
- Published
- 2019
- Full Text
- View/download PDF
33. A comprehensive inventory of TLX1 controlled long non-coding RNAs in T-cell acute lymphoblastic leukemia through polyA+ and total RNA sequencing.
- Author
-
Verboom K, Van Loocke W, Volders PJ, Decaesteker B, Cobos FA, Bornschein S, de Bock CE, Atak ZK, Clappier E, Aerts S, Cools J, Soulier J, Taghon T, Van Vlierberghe P, Vandesompele J, Speleman F, and Durinck K
- Subjects
- Azepines pharmacology, Cell Line, Tumor, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Humans, RNA Interference, T-Lymphocytes drug effects, T-Lymphocytes metabolism, Triazoles pharmacology, Homeodomain Proteins genetics, Poly A genetics, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics, Proto-Oncogene Proteins genetics, RNA, Long Noncoding genetics, Sequence Analysis, RNA methods
- Published
- 2018
- Full Text
- View/download PDF
34. Deletion 6q Drives T-cell Leukemia Progression by Ribosome Modulation.
- Author
-
Gachet S, El-Chaar T, Avran D, Genesca E, Catez F, Quentin S, Delord M, Thérizols G, Briot D, Meunier G, Hernandez L, Pla M, Smits WK, Buijs-Gladdines JG, Van Loocke W, Menschaert G, André-Schmutz I, Taghon T, Van Vlierberghe P, Meijerink JP, Baruchel A, Dombret H, Clappier E, Diaz JJ, Gazin C, de Thé H, Sigaux F, and Soulier J
- Subjects
- Animals, Cell Line, Tumor, Chromosomes, Human, Pair 6, Disease Progression, Gene Expression Profiling, Haploinsufficiency, Heterogeneous-Nuclear Ribonucleoproteins metabolism, Humans, Leukemia, T-Cell metabolism, Leukemia, T-Cell pathology, Mice, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma pathology, RNA Interference, RNA, Long Noncoding metabolism, Transplantation, Heterologous, Chromosome Deletion, Heterogeneous-Nuclear Ribonucleoproteins genetics, Leukemia, T-Cell genetics, RNA, Long Noncoding genetics, Ribosomes metabolism
- Abstract
Deletion of chromosome 6q is a well-recognized abnormality found in poor-prognosis T-cell acute lymphoblastic leukemia (T-ALL). Using integrated genomic approaches, we identified two candidate haploinsufficient genes contiguous at 6q14, SYNCRIP (encoding hnRNP-Q) and SNHG5 (that hosts snoRNAs), both involved in regulating RNA maturation and translation. Combined silencing of both genes, but not of either gene alone, accelerated leukemogeneis in a Tal1/Lmo1/Notch1 -driven mouse model, demonstrating the tumor-suppressive nature of the two-gene region. Proteomic and translational profiling of cells in which we engineered a short 6q deletion by CRISPR/Cas9 genome editing indicated decreased ribosome and mitochondrial activities, suggesting that the resulting metabolic changes may regulate tumor progression. Indeed, xenograft experiments showed an increased leukemia-initiating cell activity of primary human leukemic cells upon coextinction of SYNCRIP and SNHG5. Our findings not only elucidate the nature of 6q deletion but also highlight the role of ribosomes and mitochondria in T-ALL tumor progression. SIGNIFICANCE: The oncogenic role of 6q deletion in T-ALL has remained elusive since this chromosomal abnormality was first identified more than 40 years ago. We combined genomic analysis and functional models to show that the codeletion of two contiguous genes at 6q14 enhances malignancy through deregulation of a ribosome-mitochondria axis, suggesting the potential for therapeutic intervention. This article is highlighted in the In This Issue feature, p. 1494 ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
35. TBX2 is a neuroblastoma core regulatory circuitry component enhancing MYCN/FOXM1 reactivation of DREAM targets.
- Author
-
Decaesteker B, Denecker G, Van Neste C, Dolman EM, Van Loocke W, Gartlgruber M, Nunes C, De Vloed F, Depuydt P, Verboom K, Rombaut D, Loontiens S, De Wyn J, Kholosy WM, Koopmans B, Essing AHW, Herrmann C, Dreidax D, Durinck K, Deforce D, Van Nieuwerburgh F, Henssen A, Versteeg R, Boeva V, Schleiermacher G, van Nes J, Mestdagh P, Vanhauwaert S, Schulte JH, Westermann F, Molenaar JJ, De Preter K, and Speleman F
- Subjects
- Antineoplastic Agents pharmacology, Azepines pharmacology, Brain Neoplasms drug therapy, Brain Neoplasms metabolism, Brain Neoplasms pathology, Cell Line, Tumor, Cell Survival drug effects, Cyclin-Dependent Kinases genetics, Cyclin-Dependent Kinases metabolism, DNA Copy Number Variations, Epigenesis, Genetic, Forkhead Box Protein M1 metabolism, HEK293 Cells, Histones genetics, Histones metabolism, Humans, Kv Channel-Interacting Proteins metabolism, N-Myc Proto-Oncogene Protein metabolism, Neuroblastoma drug therapy, Neuroblastoma metabolism, Neuroblastoma pathology, Organoids drug effects, Organoids metabolism, Organoids pathology, Panobinostat pharmacology, Phenylenediamines pharmacology, Pyrimidines pharmacology, Repressor Proteins metabolism, Signal Transduction, T-Box Domain Proteins metabolism, Triazoles pharmacology, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Cyclin-Dependent Kinase-Activating Kinase, Brain Neoplasms genetics, Forkhead Box Protein M1 genetics, Gene Expression Regulation, Neoplastic, Kv Channel-Interacting Proteins genetics, N-Myc Proto-Oncogene Protein genetics, Neuroblastoma genetics, Repressor Proteins genetics, T-Box Domain Proteins genetics
- Abstract
Chromosome 17q gains are almost invariably present in high-risk neuroblastoma cases. Here, we perform an integrative epigenomics search for dosage-sensitive transcription factors on 17q marked by H3K27ac defined super-enhancers and identify TBX2 as top candidate gene. We show that TBX2 is a constituent of the recently established core regulatory circuitry in neuroblastoma with features of a cell identity transcription factor, driving proliferation through activation of p21-DREAM repressed FOXM1 target genes. Combined MYCN/TBX2 knockdown enforces cell growth arrest suggesting that TBX2 enhances MYCN sustained activation of FOXM1 targets. Targeting transcriptional addiction by combined CDK7 and BET bromodomain inhibition shows synergistic effects on cell viability with strong repressive effects on CRC gene expression and p53 pathway response as well as several genes implicated in transcriptional regulation. In conclusion, we provide insight into the role of the TBX2 CRC gene in transcriptional dependency of neuroblastoma cells warranting clinical trials using BET and CDK7 inhibitors.
- Published
- 2018
- Full Text
- View/download PDF
36. A Novel t(8;14)(q24;q11) Rearranged Human Cell Line as a Model for Mechanistic and Drug Discovery Studies of NOTCH1-Independent Human T-Cell Leukemia.
- Author
-
Tosello V, Milani G, Martines A, Macri N, Van Loocke W, Matthijssens F, Buldini B, Minuzzo S, Bongiovanni D, Schumacher RF, Amadori A, Van Vlierberghe P, and Piovan E
- Abstract
MYC -translocated T-lineage acute lymphoblastic leukemia (T-ALL) is a rare subgroup of T-ALL associated with CDKN2A/B deletions, PTEN inactivation, and absence of NOTCH1 or FBXW7 mutations. This subtype of T-ALL has been associated with induction failure and aggressive disease. Identification of drug targets and mechanistic insights for this disease are still limited. Here, we established a human NOTCH1-independent MYC -translocated T-ALL cell line that maintains the genetic and phenotypic characteristics of the parental leukemic clone at diagnosis. The University of Padua T-cell acute lymphoblastic leukemia 13 (UP-ALL13) cell line has all the main features of the above described MYC -translocated T-ALL. Interestingly, UP-ALL13 was found to harbor a heterozygous R882H DNMT3A mutation typically found in myeloid leukemia. Chromatin immunoprecipitation coupled with high-throughput sequencing for histone H3 lysine 27 (H3K27) acetylation revealed numerous putative super-enhancers near key transcription factors, including MYC, MYB, and LEF1. Marked cytotoxicity was found following bromodomain-containing protein 4 (BRD4) inhibition with AZD5153, suggesting a strict dependency of this particular subtype of T-ALL on the activity of super-enhancers. Altogether, this cell line may be a useful model system for dissecting the signaling pathways implicated in NOTCH1-independent T-ALL and for the screening of targeted anti-leukemia agents specific for this T-ALL subgroup.
- Published
- 2018
- Full Text
- View/download PDF
37. Evaluation of relative quantification of alternatively spliced transcripts using droplet digital PCR.
- Author
-
Van Heetvelde M, Van Loocke W, Trypsteen W, Baert A, Vanderheyden K, Crombez B, Vandesompele J, De Leeneer K, and Claes KBM
- Abstract
Introduction: For the relative quantification of isoform expression, RT-qPCR has been the gold standard for over a decade. More recently, digital PCR is becoming widely implemented, as it is promised to be more accurate, sensitive and less affected by inhibitors, without the need for standard curves. In this study we evaluated RT-qPCR versus RT-droplet digital PCR (ddPCR) for the relative quantification of isoforms in controls and carriers of the splice site mutation BRCA1 c.212+3A>G, associated with increased expression of several isoforms., Materials and Methods: RNA was extracted from EBV cell lines of controls and heterozygous BRCA1 c.212+3A>G carriers. Transcript-specific plasmids were available to determine the efficiency, precision, reproducibility and accuracy of each method., Results: Both ddPCR and RT-qPCR were able to accurately quantify all targets and showed the same LOB, LOD and LOQ; also precision and reproducibility were similar. Both techniques have the same dynamic range and linearity at biologically relevant template concentrations. However, a significantly higher cost and workload was required for ddPCR experiments., Conclusions: Our study recognizes the potential and validity of digital PCR but shows the value of a highly optimized qPCR for the relative quantification of isoforms. Cost efficiency and simplicity turned out to be better for RT-qPCR.
- Published
- 2017
- Full Text
- View/download PDF
38. Comprehensive miRNA expression profiling in human T-cell acute lymphoblastic leukemia by small RNA-sequencing.
- Author
-
Wallaert A, Van Loocke W, Hernandez L, Taghon T, Speleman F, and Van Vlierberghe P
- Subjects
- Humans, MicroRNAs genetics, Sequence Analysis, RNA, Gene Expression Profiling, MicroRNAs analysis, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, T-Lymphocytes pathology
- Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a genetically heterogeneous disease that can be classified into different molecular genetic subtypes according to their mRNA gene expression profile. In this study, we applied RNA sequencing to investigate the full spectrum of miRNA expression in primary T-ALL patient samples, T-ALL leukemia cell lines and healthy donor thymocytes. Notably, this analysis revealed that genetic subtypes of human T-ALL also display unique miRNA expression signatures, which are largely conserved in human T-ALL cell lines with corresponding genetic background. Furthermore, small RNA-sequencing also unraveled the variety of isoforms that are expressed for each miRNA in T-ALL and showed that a significant number of miRNAs are actually represented by an alternative isomiR. Finally, comparison of CD34
+ and CD4+ CD8+ healthy donor thymocytes and T-ALL miRNA profiles allowed identifying several novel miRNAs with putative oncogenic or tumor suppressor functions in T-ALL. Altogether, this study provides a comprehensive overview of miRNA expression in normal and malignant T-cells and sets the stage for functional evaluation of novel miRNAs in T-ALL disease biology.- Published
- 2017
- Full Text
- View/download PDF
39. Oncogenic ZEB2 activation drives sensitivity toward KDM1A inhibition in T-cell acute lymphoblastic leukemia.
- Author
-
Goossens S, Peirs S, Van Loocke W, Wang J, Takawy M, Matthijssens F, Sonderegger SE, Haigh K, Nguyen T, Vandamme N, Costa M, Carmichael C, Van Nieuwerburgh F, Deforce D, Kleifeld O, Curtis DJ, Berx G, Van Vlierberghe P, and Haigh JJ
- Subjects
- Animals, Benzoates therapeutic use, Cell Line, Tumor, Cyclopropanes therapeutic use, Gene Expression Regulation, Leukemic, Homeodomain Proteins genetics, Humans, Mice, Mice, Inbred NOD, Mice, SCID, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics, Protein Interaction Maps drug effects, Repressor Proteins genetics, Up-Regulation, Zinc Finger E-box Binding Homeobox 2, Benzoates pharmacology, Cyclopropanes pharmacology, Histone Demethylases antagonists & inhibitors, Histone Demethylases metabolism, Homeodomain Proteins metabolism, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Repressor Proteins metabolism
- Abstract
Elevated expression of the Zinc finger E-box binding homeobox transcription factor-2 (ZEB2) is correlated with poor prognosis and patient outcome in a variety of human cancer subtypes. Using a conditional gain-of-function mouse model, we recently demonstrated that ZEB2 is an oncogenic driver of immature T-cell acute lymphoblastic leukemia (T-ALL), a heterogenic subgroup of human leukemia characterized by a high incidence of remission failure or hematological relapse after conventional chemotherapy. Here, we identified the lysine-specific demethylase KDM1A as a novel interaction partner of ZEB2 and demonstrated that mouse and human T-ALLs with increased ZEB2 levels critically depend on KDM1A activity for survival. Therefore, targeting the ZEB2 protein complex through direct disruption of the ZEB2-KDM1A interaction or pharmacological inhibition of the KDM1A demethylase activity itself could serve as a novel therapeutic strategy for this aggressive subtype of human leukemia and possibly other ZEB2-driven malignancies., (© 2017 by The American Society of Hematology.)
- Published
- 2017
- Full Text
- View/download PDF
40. Unique long non-coding RNA expression signature in ETV6/RUNX1-driven B-cell precursor acute lymphoblastic leukemia.
- Author
-
Ghazavi F, De Moerloose B, Van Loocke W, Wallaert A, Helsmoortel HH, Ferster A, Bakkus M, Plat G, Delabesse E, Uyttebroeck A, Van Nieuwerburgh F, Deforce D, Van Roy N, Speleman F, Benoit Y, Lammens T, and Van Vlierberghe P
- Subjects
- Cell Line, Tumor, Child, Computational Biology methods, Gene Expression, Gene Expression Regulation, Leukemic, Humans, Molecular Sequence Annotation, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology, RNA Interference, Sequence Analysis, RNA, Core Binding Factor Alpha 2 Subunit genetics, Oncogene Proteins, Fusion genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics, RNA, Long Noncoding genetics, Transcriptome
- Abstract
Overwhelming evidence indicates that long non-coding RNAs have essential roles in tumorigenesis. Nevertheless, their role in the molecular pathogenesis of pediatric B-cell precursor acute lymphoblastic leukemia has not been extensively explored. Here, we conducted a comprehensive analysis of the long non-coding RNA transcriptome in ETV6/RUNX1-positive BCP-ALL, one of the most frequent subtypes of pediatric leukemia. First, we used primary leukemia patient samples to identify an ETV6/RUNX1 specific expression signature consisting of 596 lncRNA transcripts. Next, integration of this lncRNA signature with RNA sequencing of BCP-ALL cell lines and lncRNA profiling of an in vitro model system of ETV6/RUNX1 knockdown, revealed that lnc-NKX2-3-1, lnc-TIMM21-5, lnc-ASTN1-1 and lnc-RTN4R-1 are truly regulated by the oncogenic fusion protein. Moreover, sustained inactivation of lnc-RTN4R-1 and lnc-NKX2-3-1 in ETV6/RUNX1 positive cells caused profound changes in gene expression. All together, our study defined a unique lncRNA expression signature associated with ETV6/RUNX1-positive BCP-ALL and identified lnc-RTN4R-1 and lnc-NKX2-3-1 as lncRNAs that might be functionally implicated in the biology of this prevalent subtype of human leukemia.
- Published
- 2016
- Full Text
- View/download PDF
41. GATA3 induces human T-cell commitment by restraining Notch activity and repressing NK-cell fate.
- Author
-
Van de Walle I, Dolens AC, Durinck K, De Mulder K, Van Loocke W, Damle S, Waegemans E, De Medts J, Velghe I, De Smedt M, Vandekerckhove B, Kerre T, Plum J, Leclercq G, Rothenberg EV, Van Vlierberghe P, Speleman F, and Taghon T
- Subjects
- Cell Differentiation, Cell Lineage immunology, Cellular Reprogramming, Child, GATA3 Transcription Factor immunology, Gene Expression Regulation, Developmental, Hematopoietic Stem Cells cytology, Hepatocyte Nuclear Factor 1-alpha genetics, Hepatocyte Nuclear Factor 1-alpha immunology, Humans, Killer Cells, Natural cytology, Killer Cells, Natural immunology, Primary Cell Culture, Receptor, Notch1 genetics, Receptor, Notch1 immunology, Signal Transduction, Thymocytes cytology, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases immunology, Cell Lineage genetics, Feedback, Physiological, GATA3 Transcription Factor genetics, Hematopoietic Stem Cells immunology, Thymocytes immunology
- Abstract
The gradual reprogramming of haematopoietic precursors into the T-cell fate is characterized by at least two sequential developmental stages. Following Notch1-dependent T-cell lineage specification during which the first T-cell lineage genes are expressed and myeloid and dendritic cell potential is lost, T-cell specific transcription factors subsequently induce T-cell commitment by repressing residual natural killer (NK)-cell potential. How these processes are regulated in human is poorly understood, especially since efficient T-cell lineage commitment requires a reduction in Notch signalling activity following T-cell specification. Here, we show that GATA3, in contrast to TCF1, controls human T-cell lineage commitment through direct regulation of three distinct processes: repression of NK-cell fate, upregulation of T-cell lineage genes to promote further differentiation and restraint of Notch activity. Repression of the Notch1 target gene DTX1 hereby is essential to prevent NK-cell differentiation. Thus, GATA3-mediated positive and negative feedback mechanisms control human T-cell lineage commitment.
- Published
- 2016
- Full Text
- View/download PDF
42. Novel biological insights in T-cell acute lymphoblastic leukemia.
- Author
-
Durinck K, Goossens S, Peirs S, Wallaert A, Van Loocke W, Matthijssens F, Pieters T, Milani G, Lammens T, Rondou P, Van Roy N, De Moerloose B, Benoit Y, Haigh J, Speleman F, Poppe B, and Van Vlierberghe P
- Subjects
- Adolescent, Adult, Animals, Child, Child, Preschool, Humans, Infant, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cellular Reprogramming, Epigenesis, Genetic, Hematopoietic Stem Cell Transplantation, Precursor Cells, T-Lymphoid metabolism, Precursor Cells, T-Lymphoid pathology, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma pathology, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma therapy
- Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive type of blood cancer that accounts for about 15% of pediatric and 25% of adult acute lymphoblastic leukemia (ALL) cases. It is considered as a paradigm for the multistep nature of cancer initiation and progression. Genetic and epigenetic reprogramming events, which transform T-cell precursors into malignant T-ALL lymphoblasts, have been extensively characterized over the past decade. Despite our comprehensive understanding of the genomic landscape of human T-ALL, leukemia patients are still treated by high-dose multiagent chemotherapy, potentially followed by hematopoietic stem cell transplantation. Even with such aggressive treatment regimens, which are often associated with considerable acute and long-term side effects, about 15% of pediatric and 40% of adult T-ALL patients still relapse, owing to acquired therapy resistance, and present with very dismal survival perspectives. Unfortunately, the molecular mechanisms by which residual T-ALL tumor cells survive chemotherapy and act as a reservoir for leukemic progression and hematologic relapse remain poorly understood. Nevertheless, it is expected that enhanced molecular understanding of T-ALL disease biology will ultimately facilitate a targeted therapy driven approach that can reduce chemotherapy-associated toxicities and improve survival of refractory T-ALL patients through personalized salvage therapy. In this review, we summarize recent biological insights into the molecular pathogenesis of T-ALL and speculate how the genetic landscape of T-ALL could trigger the development of novel therapeutic strategies for the treatment of human T-ALL., (Copyright © 2015 ISEH - International Society for Experimental Hematology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.