111 results on '"João T. Barata"'
Search Results
2. Anatomy of a crime: how IL7R and NRAS join forces to drive T-cell acute lymphoblastic leukemia
- Author
-
João T. Barata
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2024
- Full Text
- View/download PDF
3. CASZ1 upregulates PI3K-AKT-mTOR signaling and promotes T-cell acute lymphoblastic leukemia
- Author
-
Bruno A. Cardoso, Mafalda Duque, Ana Gírio, Rita Fragoso, Mariana L. Oliveira, James R. Allen, Leila R. Martins, Nádia C. Correia, André Bortolini Silveira, Alexandra Veloso, Shunsuke Kimura, Lisa Demoen, Filip Matthijssens, Sima Jeha, Cheng Cheng, Ching-Hon Pui, Ana R. Grosso, João L. Neto, Sérgio F. de Almeida, Pieter Van Vlieberghe, Charles G. Mullighan, J. Andres Yunes, David M. Langenau, Françoise Pflumio, and João T. Barata
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
CASZ1 is a conserved transcription factor involved in neural development, blood vessel assembly and heart morphogenesis. CASZ1 has been implicated in cancer, either suppressing or promoting tumor development depending on the tissue. However, the impact of CASZ1 on hematological tumors remains unknown. Here, we show that the T-cell oncogenic transcription factor TAL1 is a direct positive regulator of CASZ1, that T-cell acute lymphoblastic leukemia (T-ALL) samples at diagnosis overexpress CASZ1b isoform, and that CASZ1b expression in patient samples correlates with PI3K-AKT-mTOR signaling pathway activation. In agreement, overexpression of CASZ1b in both Ba/F3 and T-ALL cells leads to the activation of PI3K signaling pathway, which is required for CASZ1b-mediated transformation of Ba/F3 cells in vitro and malignant expansion in vivo. We further demonstrate that CASZ1b cooperates with activated NOTCH1 to promote T-ALL development in zebrafish, and that CASZ1b protects human T-ALL cells from serum deprivation and treatment with chemotherapeutic drugs. Taken together, our studies indicate that CASZ1b is a TAL1-regulated gene that promotes T-ALL development and resistance to chemotherapy.
- Published
- 2023
- Full Text
- View/download PDF
4. P339: DEVELOPMENT OF ANTI-IL-7RΑ ANTIBODIES AS A TARGETED THERAPY FOR T-CELL ACUTE LYMPHOBLASTIC LEUKEMIA
- Author
-
Mafalda Duque, João Santos, Sílvia Andrade, Daniela Teixeira, Maria Pajuelo, Rita Fragoso, and João T. Barata
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
5. P1379: DEVELOPMENT OF A GENE THERAPY CELL DEATH-INDUCING SYSTEM REGULATED BY MICRORNAS FOR T-CELL ACUTE LYMPHOBLASTIC LEUKEMIA.
- Author
-
Mafalda Duque, Joana Ferreira Da Silva, João T. Barata, and Rita Fragoso
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
6. Primary cilia contribute to the aggressiveness of atypical teratoid/rhabdoid tumors
- Author
-
Lena Blümel, Nan Qin, Johannes Berlandi, Eunice Paisana, Rita Cascão, Carlos Custódia, David Pauck, Daniel Picard, Maike Langini, Kai Stühler, Frauke-Dorothee Meyer, Sarah Göbbels, Bastian Malzkorn, Max C. Liebau, João T. Barata, Astrid Jeibmann, Kornelius Kerl, Serap Erkek, Marcel Kool, Stefan M. Pfister, Pascal D. Johann, Michael C. Frühwald, Arndt Borkhardt, Guido Reifenberger, Claudia C. Faria, Ute Fischer, Martin Hasselblatt, Jasmin Bartl, and Marc Remke
- Subjects
Cytology ,QH573-671 - Abstract
Abstract Atypical teratoid/rhabdoid tumor (AT/RT) is a highly malignant brain tumor in infants that is characterized by loss of nuclear expression of SMARCB1 or SMARCA4 proteins. Recent studies show that AT/RTs comprise three molecular subgroups, namely AT/RT-TYR, AT/RT-MYC and AT/RT-SHH. The subgroups show distinct expression patterns of genes involved in ciliogenesis, however, little is known about the functional roles of primary cilia in the biology of AT/RT. Here, we show that primary cilia are present across all AT/RT subgroups with specific enrichment in AT/RT-TYR patient samples. Furthermore, we demonstrate that primary ciliogenesis contributes to AT/RT biology in vitro and in vivo. Specifically, we observed a significant decrease in proliferation and clonogenicity following disruption of primary ciliogenesis in AT/RT cell line models. Additionally, apoptosis was significantly increased via the induction of STAT1 and DR5 signaling, as detected by proteogenomic profiling. In a Drosophila model of SMARCB1 deficiency, concomitant knockdown of several cilia-associated genes resulted in a substantial shift of the lethal phenotype with more than 20% of flies reaching adulthood. We also found significantly extended survival in an orthotopic xenograft mouse model of AT/RT upon disruption of primary ciliogenesis. Taken together, our findings indicate that primary ciliogenesis or its downstream signaling contributes to the aggressiveness of AT/RT and, therefore, may constitute a novel therapeutic target.
- Published
- 2022
- Full Text
- View/download PDF
7. An instructive role for Interleukin-7 receptor α in the development of human B-cell precursor leukemia
- Author
-
Ifat Geron, Angela Maria Savino, Hila Fishman, Noa Tal, John Brown, Virginia A. Turati, Chela James, Jolanda Sarno, Michal Hameiri-Grossman, Yu Nee Lee, Avigail Rein, Hillary Maniriho, Yehudit Birger, Anna Zemlyansky, Inna Muler, Kara L. Davis, Victoria Marcu-Malina, Nicole Mattson, Oren Parnas, Rabea Wagener, Ute Fischer, João T. Barata, Catriona H. M. Jamieson, Markus Müschen, Chun-Wei Chen, Arndt Borkhardt, Ilan Richard Kirsch, Arnon Nagler, Tariq Enver, and Shai Izraeli
- Subjects
Science - Abstract
Activating mutations in Interleukin-7 receptor alpha (IL7Ra) have been reported in B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) but its role in leukaemogenesis is not clear. Here, the authors show that activation of IL7Ra in primary human hematopoietic progenitors initiates preleukaemia and cooperates with CDKN2A silencing to develop BCP-ALL.
- Published
- 2022
- Full Text
- View/download PDF
8. Interleukin-7 receptor α mutational activation can initiate precursor B-cell acute lymphoblastic leukemia
- Author
-
Afonso R. M. Almeida, João L. Neto, Ana Cachucho, Mayara Euzébio, Xiangyu Meng, Rathana Kim, Marta B. Fernandes, Beatriz Raposo, Mariana L. Oliveira, Daniel Ribeiro, Rita Fragoso, Priscila P. Zenatti, Tiago Soares, Mafalda R. de Matos, Juliana Ronchi Corrêa, Mafalda Duque, Kathryn G. Roberts, Zhaohui Gu, Chunxu Qu, Clara Pereira, Susan Pyne, Nigel J. Pyne, Vasco M. Barreto, Isabelle Bernard-Pierrot, Emannuelle Clappier, Charles G. Mullighan, Ana R. Grosso, J. Andrés Yunes, and João T. Barata
- Subjects
Science - Abstract
Interleukin-7 receptor alpha (IL7Ra) is important for lymphoid cell development but its role in leukaemogenesis is not clear. Here, the authors generate a knock-in murine model to show that activating mutations in IL7Ra can initiate precursor B-cell acute lymphoblastic leukaemia.
- Published
- 2021
- Full Text
- View/download PDF
9. Surprise, surprise: STAT5 is not enough to stop the steroids
- Author
-
Marta B. Fernandes and João T. Barata
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2022
- Full Text
- View/download PDF
10. Human CD4 T Cells From Thymus and Cord Blood Are Convertible Into CD8 T Cells by IL-4
- Author
-
Helena Nunes-Cabaço, Andreia Ramalho-dos-Santos, Ana R. Pires, Leila R. Martins, João T. Barata, and Ana E. Sousa
- Subjects
CD4+ and CD8+ T cell lineage commitment ,IL-4 ,human thymus ,cord blood ,innate-like T cells ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Commitment to the CD4+ or CD8+ T cell lineages is linked to the acquisition of a functional program broadly defined by helper and cytotoxic properties, respectively. The mechanisms underlying these processes in the human thymus remain largely unclear. Moreover, recent thymic emigrants are thought to have some degree of plasticity, which may be important for the shaping of the immune system and adjustment to specific peripheral needs. We show here that IL-4 induces proliferation-independent de novo synthesis of CD8αβ in human CD4 single-positive (SP) thymocytes, generating a stable CD8SP population that features a diverse TCRαβ repertoire, CD4 expression shut-down and ThPOK downregulation. IL-4 also promotes an innate-like program in both CD4SP and CD8SP thymocytes, characterized by Eomes upregulation in the absence of T-bet, in line with its recognized role in the generation of thymic innate-like CD8+ T cells. The clinical relevance of these findings is further supported by the profile of IL-4 production and IL-4 receptor expression that we identified in the human thymus. Importantly, human cord blood CD4+ T cells preserve the ability to generate Eomes+ CD8+ T cells in the presence of IL-4, with implications in neonatal immunity. Our results support a role for IL-4 in the dynamic regulation of human thymocyte plasticity and identify novel strategies to modulate immune responses.
- Published
- 2022
- Full Text
- View/download PDF
11. Optimal interleukin-7 receptor-mediated signaling, cell cycle progression and viability of T-cell acute lymphoblastic leukemia cells rely on casein kinase 2 activity
- Author
-
Alice Melão, Maureen Spit, Bruno A. Cardoso, and João T. Barata
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Interleukin-7 and interleukin-7 receptor are essential for normal T-cell development and homeostasis, whereas excessive interleukin-7/interleukin-7 receptor-mediated signaling promotes leukemogenesis. The protein kinase, casein kinase 2, is overexpressed and hyperactivated in cancer, including T-cell acute lymphoblastic leukemia. Herein, we show that while interleukin-7 had a minor but significant positive effect on casein kinase 2 activity in leukemia T-cells, casein kinase 2 activity was mandatory for optimal interleukin-7/interleukin-7 receptor-mediated signaling. Casein kinase 2 pharmacological inhibition impaired signal transducer and activator of transcription 5 and phosphoinositide 3-kinase/v-Akt murine thymoma viral oncogene homolog 1 pathway activation triggered by interleukin-7 or by mutational activation of interleukin-7 receptor. By contrast, forced expression of casein kinase 2 augmented interleukin-7 signaling in human embryonic kidney 293T cells reconstituted with the interleukin-7 receptor machinery. Casein kinase 2 inactivation prevented interleukin-7-induced B-cell lymphoma 2 upregulation, maintenance of mitochondrial homeostasis and viability of T-cell acute lymphoblastic leukemia cell lines and primary leukemia cells collected from patients at diagnosis. Casein kinase 2 inhibition further abrogated interleukin-7-mediated cell growth and upregulation of the transferrin receptor, and blocked cyclin A and E upregulation and cell cycle progression. Notably, casein kinase 2 was also required for the viability of mutant interleukin-7 receptor expressing leukemia T-cells. Overall, our study identifies casein kinase 2 as a major player in the effects of interleukin-7 and interleukin-7 receptor in T-cell acute lymphoblastic leukemia. This further highlights the potential relevance of targeting casein kinase 2 in this malignancy.
- Published
- 2016
- Full Text
- View/download PDF
12. Adult B-cell acute lymphoblastic leukemia cells display decreased PTEN activity and constitutive hyperactivation of PI3K/Akt pathway despite high PTEN protein levels
- Author
-
A. Margarida Gomes, Maria V. D. Soares, Patrícia Ribeiro, Joana Caldas, Vanda Póvoa, Leila R. Martins, Alice Melão, Ana Serra-Caetano, Aida B. de Sousa, João F. Lacerda, and João T. Barata
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Adult B-cell acute lymphoblastic leukemia remains a major therapeutic challenge, requiring a better characterization of the molecular determinants underlying disease progression and resistance to treatment. Here, using a phospho-flow cytometry approach we show that adult diagnostic B-cell acute lymphoblastic leukemia specimens display PI3K/Akt pathway hyperactivation, irrespective of their BCR-ABL status and despite paradoxically high basal expression of PTEN, the major negative regulator of the pathway. Protein kinase CK2 is known to phosphorylate PTEN thereby driving PTEN protein stabilization and concomitant PTEN functional inactivation. In agreement, we found that adult B-cell acute lymphoblastic leukemia samples show significantly higher CK2 kinase activity and lower PTEN lipid phosphatase activity than healthy controls. Moreover, the clinical-grade CK2 inhibitor CX-4945 (Silmitasertib) reversed PTEN levels in leukemia cells to those observed in healthy controls, and promoted leukemia cell death without significantly affecting normal bone marrow cells. Our studies indicate that CK2-mediated PTEN posttranslational inactivation, associated with PI3K/Akt pathway hyperactivation, are a common event in adult B-cell acute lymphoblastic leukemia and suggest that CK2 inhibition may constitute a valid, novel therapeutic tool in this malignancy.
- Published
- 2014
- Full Text
- View/download PDF
13. Identification of a panel of ten cell surface protein antigens associated with immunotargeting of leukemias and lymphomas by peripheral blood γδ T cells
- Author
-
Anita Q. Gomes, Daniel V. Correia, Ana R. Grosso, Telma Lança, Cristina Ferreira, João F. Lacerda, João T. Barata, Maria Gomes da Silva, and Bruno Silva-Santos
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Background Vγ9Vδ2 T lymphocytes are regarded as promising mediators of cancer immunotherapy due to their capacity to eliminate multiple experimental tumors, particularly within those of hematopoietic origin. However, Vγ9Vδ2 T-cell based lymphoma clinical trials have suffered from the lack of biomarkers that can be used as prognostic of therapeutic success.Design and Methods We have conducted a comprehensive study of gene expression in acute lymphoblastic leukemias and non-Hodgkin’s lymphomas, aimed at identifying markers of susceptibility versus resistance to Vγ9Vδ2 T cell-mediated cytotoxicity. We employed cDNA microarrays and quantitative real-time PCR to screen 20 leukemia and lymphoma cell lines, and 23 primary hematopoietic tumor samples. These data were analyzed using state-of-the-art bioinformatics, and gene expression patterns were correlated with susceptibility to Vγ9Vδ2 T cell mediated cytolysis in vitro.Results We identified a panel of 10 genes encoding cell surface proteins that were statistically differentially expressed between “γδ-susceptible” and “γδ-resistant” hematopoietic tumors. Within this panel, 3 genes (ULBP1, TFR2 and IFITM1) were associated with increased susceptibility to Vγ9Vδ2 T-cell cytotoxicity, whereas the other 7 (CLEC2D, NRP2, SELL, PKD2, KCNK12, ITGA6 and SLAMF1) were enriched in resistant tumors. Furthermore, some of these candidates displayed a striking variance of expression among primary follicular lymphomas and T-cell acute lymphoblastic leukemias.Conclusions Our results suggest that hematopoietic tumors display a highly variable repertoire of surface proteins that can impact on Vγ9Vδ2 cell-mediated immunotargeting. The prognostic value of the proposed markers can now be evaluated in upcoming Vγ9Vδ2 T cell-based lymphoma/leukemia clinical trials.
- Published
- 2010
- Full Text
- View/download PDF
14. Regulation of PTEN by CK2 and Notch1 in primary T-cell acute lymphoblastic leukemia: rationale for combined use of CK2- and γ-secretase inhibitors
- Author
-
Ana Silva, Patrícia Y. Jotta, André B. Silveira, Daniel Ribeiro, Silvia R. Brandalise, J. Andrés Yunes, and João T. Barata
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
T-cell acute lymphoblastic leukemia (T-ALL) patients frequently display NOTCH1 activating mutations and Notch can transcriptionally down-regulate the tumor suppressor PTEN. However, it is not clear whether NOTCH1 mutations associate with decreased PTEN expression in primary T-ALL. Here, we compared patients with or without NOTCH1 mutations and report that the former presented higher MYC transcript levels and decreased PTEN mRNA expression. We recently showed that T-ALL cells frequently display CK2-mediated PTEN phosphorylation, resulting in PTEN protein stabilization and concomitant functional inactivation. Accordingly, the T-ALL samples analyzed, irrespectively of their NOTCH1 mutational status, expressed significantly higher PTEN protein levels than normal controls. To evaluate the integrated functional impact of Notch transcriptional and CK2 post-translational inactivation of PTEN, we treated T-ALL cells with both the gamma-secretase inhibitor DAPT and the CK2 inhibitors DRB/TBB. Our data suggest that combined use of gamma-secretase and CK2 inhibitors may have therapeutic potential in T-ALL.
- Published
- 2010
- Full Text
- View/download PDF
15. Supplementary Figures 1-4 from IL-7 Contributes to the Progression of Human T-cell Acute Lymphoblastic Leukemias
- Author
-
João T. Barata, Benedict Seddon, J. Andrés Yunes, Jocelyne Demengeot, Bruno A. Cardoso, Leila R. Martins, Angelo B.A. Laranjeira, and Ana Silva
- Abstract
Supplementary Figures 1-4 from IL-7 Contributes to the Progression of Human T-cell Acute Lymphoblastic Leukemias
- Published
- 2023
- Full Text
- View/download PDF
16. Intratumoral heterogeneity of MYC drives medulloblastoma metastasis and angiogenesis
- Author
-
Nan Qin, Eunice Paisana, Maike Langini, Daniel Picard, Bastian Malzkorn, Carlos Custódia, Rita Cascão, Frauke-Dorothee Meyer, Lena Blümel, Sarah Göbbels, Kübra Taban, Jasmin Bartl, Nicole Bechmann, Catleen Conrad, Jan Gravemeyer, Jürgen C Becker, Anja Stefanski, Stéphanie Puget, João T Barata, Kai Stühler, Ute Fischer, Jörg Felsberg, Olivier Ayrault, Guido Reifenberger, Arndt Borkhardt, Graeme Eisenhofer, Claudia C Faria, and Marc Remke
- Subjects
Proto-Oncogene Proteins c-myc ,Cancer Research ,Oncology ,Basic and Translational Investigations ,Medizin ,Humans ,Neurology (clinical) ,Cerebellar Neoplasms ,Prognosis ,In Situ Hybridization, Fluorescence ,Medulloblastoma - Abstract
Background Intratumoral heterogeneity is crucially involved in metastasis, resistance to therapy, and cancer relapse. Amplifications of the proto-oncogene MYC display notable heterogeneity at the single-cell level and are associated with a particularly dismal prognosis in high-risk medulloblastomas (MBs). The aim of this study was to establish the relevance of interclonal cross-talk between MYC-driven and non-MYC-driven MB cells. Methods We used fluorescence in situ hybridization, single-cell transcriptomics, and immunohistochemistry, in vitro isogenic cell models, non-targeted proteomics, mass spectrometry-based metabolite quantification, HUVECs tube formation assay, and orthotopic in vivo experiments to investigate interclonal cross-talk in MB. Results We found that the release of lactate dehydrogenase A (LDHA) from MYC-driven cells facilitates metastatic seeding and outgrowth, while secretion of dickkopf WNT signaling pathway inhibitor 3 from non-MYC-driven cells promotes tumor angiogenesis. This tumor-supporting interaction between both subclones was abrogated by targeting the secretome through pharmacological and genetic inhibition of LDHA, which significantly suppressed tumor cell migration. Conclusion Our study reveals the functional relevance of clonal diversity and highlights the therapeutic potential of targeting the secretome to interrupt interclonal communication and progression in high-risk MB.
- Published
- 2023
17. INPP5K controls the dynamic structure and signaling of wild-type and mutated, leukemia-associated IL-7 receptors
- Author
-
Bastien Moës, Hua Li, Patricia Molina-Ortiz, Coraline Radermecker, Adeline Rosu, Charles-Andrew Vande Catsyne, Sufyan Ali Sayyed, João Fontela, Mafalda Duque, Alice Mostafa, Abdelhalim Azzi, João T. Barata, Ramon Merino, Chenqi Xu, Christophe J. Desmet, Stéphane Schurmans, and Repositório da Universidade de Lisboa
- Subjects
Lymphoid neoplasia ,Immunology ,Cell Biology ,Hematology ,Immunotherapy ,Biochemistry ,Immunobiology - Abstract
© 2023 by The American Society of Hematology., The downstream signaling of the interleukin-7 (IL-7) receptor (IL-7R) plays important physiological and pathological roles, including the differentiation of lymphoid cells and proliferation of acute lymphoblastic leukemia cells. Gain-of-function mutations in the IL-7Rα chain, the specific component of the receptor for IL-7, result in constitutive, IL-7-independent signaling and trigger acute lymphoblastic leukemia. Here, we show that the loss of the phosphoinositide 5-phosphatase INPP5K is associated with increased levels of the INPP5K substrate phosphatidylinositol 4,5-bisphosphate (PtdIns[4,5]P2) and causes an altered dynamic structure of the IL-7 receptor. We discovered that the IL-7Rα chain contains a very conserved positively charged polybasic amino acid sequence in its cytoplasmic juxtamembrane region; this region establish stronger ionic interactions with negatively charged PtdIns(4,5)P2 in the absence of INPP5K, freezing the IL-7Rα chain structure. This dynamic structural alteration causes defects in IL-7R signaling, culminating in decreased expressions of EBF1 and PAX5 transcription factors, in microdomain formation, cytoskeletal reorganization, and bone marrow B-cell differentiation. Similar alterations after the reduced INPP5K expression also affected mutated, constitutively activated IL-7Rα chains that trigger leukemia development, leading to reduced cell proliferation. Altogether, our results indicate that the lipid 5-phosphatase INPP5K hydrolyzes PtdIns(4,5)P2, allowing the requisite conformational changes of the IL-7Rα chain for optimal signaling.
- Published
- 2023
18. IL-7 and IL-7R in health and disease: An update through COVID times
- Author
-
Marta B. Fernandes and João T. Barata
- Subjects
Cancer Research ,Genetics ,Molecular Medicine ,Molecular Biology - Abstract
The role of IL-7 and IL-7R for normal lymphoid development and an adequately functioning immune system has been recognized for long, with severe immune deficiency and lymphoid leukemia as extreme examples of the consequences of deregulation of the IL-7-IL-7R axis. In this review, we provide an update (focusing on the past couple of years) on IL-7 and IL-7R in health and disease. We highlight the findings on IL-7/IL-7R signaling mechanisms and the, sometimes controversial, impact of IL-7 and its receptor on leukocyte biology, COVID-19, acute lymphoblastic leukemia, and different solid tumors, as well as their relevance as therapeutic tools or targets.
- Published
- 2022
19. BSCI-04 TARGETING THE PI3K-MTOR PATHWAY TO TREAT UBE2C-DRIVEN BRAIN METASTASES
- Author
-
Eunice Paisana, Rita Cascão, Carlos Custódia, Nan Qin, Daniel Picard, David Pauck, Tânia Carvalho, Pedro Ruivo, Rafael Roque, José Pimentel, Marc Remke, João T Barata, and Cláudia C Faria
- Subjects
General Medicine - Abstract
Brain metastases (BMs) are a devastating complication of advanced cancers associated with poor prognosis. Contrarily to the current improvement in systemic therapies, BMs are still incurable and one of the main causes of death in cancer patients. We analyzed BMs from thirty patients with various primary tumor origins by RNA sequencing and identified the upregulation of UBE2C, a gene involved in the correct transition from metaphase to anaphase. Using an independent cohort of patients with BMs, we demonstrated that high protein expression of UBE2C was associated with worse survival. UBE2C-driven cancer cells promoted migration and invasion in vitro and induced an aggressive phenotype and decreased survival in mouse orthotopic xenografts. PI3K/mTOR inhibition effectively blocked cancer cell signaling and prevented the development of leptomeningeal metastases. Therefore, we have identified UBE2C as a molecular marker of worse outcome in BMs patients and pre-clinically validated an effective therapy against UBE2C-driven brain metastatic disease.
- Published
- 2022
- Full Text
- View/download PDF
20. Mutant IL7R collaborates with MYC to induce T-cell acute lymphoblastic leukemia
- Author
-
Mariana L. Oliveira, Alexandra Veloso, Elaine G. Garcia, Sowmya Iyer, Clara Pereira, Vasco M. Barreto, David M. Langenau, João T. Barata, Repositório da Universidade de Lisboa, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), and Centro de Estudos de Doenças Crónicas (CEDOC)
- Subjects
EXPRESSION ,Cancer Research ,Carcinogenesis ,T-Lymphocytes ,BIOLOGY ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,ACTIVATION ,Animals, Genetically Modified ,Interleukin-7 Receptor alpha Subunit ,SDG 3 - Good Health and Well-being ,NOTCH1 ,OF-FUNCTION MUTATIONS ,C-MYC ,KINASE ,Animals ,Humans ,Child ,Zebrafish ,Science & Technology ,IL-7 ,Receptors, Interleukin-7 ,Hematology ,GENE ,Oncology ,GROWTH ,Life Sciences & Biomedicine ,Signal Transduction - Abstract
© The Author(s) 2022. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/., T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive pediatric cancer. Amongst the wide array of driver mutations, 10% of T-ALL patients display gain-of-function mutations in the IL-7 receptor α chain (IL-7Rα, encoded by IL7R), which occur in different molecular subtypes of this disease. However, it is still unclear whether IL-7R mutational activation is sufficient to transform T-cell precursors. Also, which genes cooperate with IL7R to drive leukemogenesis remain poorly defined. Here, we demonstrate that mutant IL7R alone is capable of inducing T-ALL with long-latency in stable transgenic zebrafish and transformation is associated with MYC transcriptional activation. Additionally, we find that mutant IL7R collaborates with Myc to induce early onset T-ALL in transgenic zebrafish, supporting a model where these pathways collaborate to drive leukemogenesis. T-ALLs co-expressing mutant IL7R and Myc activate STAT5 and AKT pathways, harbor reduced numbers of apoptotic cells and remake tumors in transplanted zebrafish faster than T-ALLs expressing Myc alone. Moreover, limiting-dilution cell transplantation experiments reveal that activated IL-7R signaling increases the overall frequency of leukemia propagating cells. Our work highlights a synergy between mutant IL7R and Myc in inducing T-ALL and demonstrates that mutant IL7R enriches for leukemia propagating potential., This work was supported by NIH grant R01CA211734 (DML), the MGH Research Scholar Award (DML), the ERC consolidator CoG-648455 and proof-of-concept PoC-862545 grants from the European Research Council, under the European Union’s Horizon 2020 research and innovation programme (JTB), and the FCT grants FAPESP/ 20015/2014, PTDC/MEC-HEM/31588/2017 and PTDC/MEC-ONC/4606/2021 (JTB)
- Published
- 2021
21. The Bone Marrow-Mediated Protection of Myeloproliferative Neoplastic Cells to Vorinostat and Ruxolitinib Relies on the Activation of JNK and PI3K Signalling Pathways.
- Author
-
Bruno A Cardoso, Hélio Belo, João T Barata, and António M Almeida
- Subjects
Medicine ,Science - Abstract
The classical BCR-ABL-negative Myeloproliferative Neoplasms (MPN) are a group of heterogeneous haematological diseases characterized by constitutive JAK-STAT pathway activation. Targeted therapy with Ruxolitinib, a JAK1/2-specific inhibitor, achieves symptomatic improvement but does not eliminate the neoplastic clone. Similar effects are seen with histone deacetylase inhibitors (HDACi), albeit with poorer tolerance. Here, we show that bone marrow (BM) stromal cells (HS-5) protected MPN-derived cell lines (SET-2; HEL and UKE-1) and MPN patient-derived BM cells from the cytotoxic effects of Ruxolitinib and the HDACi Vorinostat. This protective effect was mediated, at least in part, by the secretion of soluble factors from the BM stroma. In addition, it correlated with the activation of signalling pathways important for cellular homeostasis, such as JAK-STAT, PI3K, JNK, MEK-ERK and NF-κB. Importantly, the pharmacological inhibition of JNK and PI3K pathways completely abrogated the BM protective effect on MPN cell lines and MPN patient samples. Our findings shed light on mechanisms of tumour survival and may indicate novel therapeutic approaches for the treatment of MPN.
- Published
- 2015
- Full Text
- View/download PDF
22. A fully human anti-IL-7Rα antibody promotes antitumor activity against T-cell acute lymphoblastic leukemia
- Author
-
Rita Fragoso, Mattia Matasci, Julie A. Hixon, Gonçalo J. L. Bernardes, Scott K. Durum, Padma Akkapeddi, João T. Barata, Francisco Corzana, Mariana L. Oliveira, Dario Neri, Ana Sofia Ramalho, Tânia Carvalho, Andreas Gloger, Lopes Bernardes, Goncalo [0000-0001-6594-8917], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Cancer Research ,medicine.drug_class ,T-Lymphocytes ,T cell ,Antineoplastic Agents ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Monoclonal antibody ,Article ,Cell Line ,Mice ,03 medical and health sciences ,Targeted therapies ,0302 clinical medicine ,Animals ,Humans ,Cytotoxic T cell ,Medicine ,Receptors, Interleukin-7 ,Acute lymphocytic leukaemia ,biology ,business.industry ,Interleukin-7 ,Antibodies, Monoclonal ,Interleukin ,Cancer ,Hematology ,medicine.disease ,3. Good health ,Leukemia ,030104 developmental biology ,Cell killing ,medicine.anatomical_structure ,Oncology ,Immunoglobulin G ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Antibody ,business ,Signal Transduction - Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological cancer for which treatment options often result in incomplete therapeutic efficacy and long-term side-effects. Interleukin 7 (IL-7) and its receptor IL-7Rα promote T-ALL development and mutational activation of IL-7Rα associates with very high risk in relapsed disease. Using combinatorial phage-display libraries and antibody reformatting, we generated a fully human IgG1 monoclonal antibody (named B12) against both wild-type and mutant human IL-7Rα, predicted to form a stable complex with IL-7Rα at a different site from IL-7. B12 impairs IL-7/IL-7R-mediated signaling, sensitizes T-ALL cells to treatment with dexamethasone and can induce cell death per se. The antibody also promotes antibody-dependent natural killer-mediated leukemia cytotoxicity in vitro and delays T-cell leukemia development in vivo, reducing tumor burden and promoting mouse survival. B12 is rapidly internalized and traffics to the lysosome, rendering it an attractive vehicle for targeted intracellular delivery of cytotoxic cargo. Consequently, we engineered a B12–MMAE antibody–drug conjugate and provide proof-of-concept evidence that it has increased leukemia cell killing abilities as compared with the naked antibody. Our studies serve as a stepping stone for the development of novel targeted therapies in T-ALL and other diseases where IL-7Rα has a pathological role. ISSN:1476-5551 ISSN:0887-6924
- Published
- 2019
- Full Text
- View/download PDF
23. IL-7R-mediated signaling in T-cell acute lymphoblastic leukemia: An update
- Author
-
João T. Barata, Daniel Dias Ribeiro, Padma Akkapeddi, Mariana L. Oliveira, Alice Melão, and Repositório da Universidade de Lisboa
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Cancer Research ,Cell Survival ,T cell ,Interleukin 7 ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Article ,B-cell acute lymphoblastic leukemia ,Interleukin-7 Receptor alpha Subunit ,03 medical and health sciences ,0302 clinical medicine ,JAK/STAT pathway ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Severe combined immunodeficiency ,Cell Cycle ,JAK-STAT signaling pathway ,IL-7R ,medicine.disease ,PI3K/Akt/mTOR pathway ,Neoplasm Proteins ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine ,T-ALL ,T-cell acute lymphoblastic leukemia ,Signal Transduction ,Lymphoid leukemia - Abstract
© 2018 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/BY-NC-ND/4.0/), Interleukin 7 (IL-7) and its receptor (IL-7R, a heterodimer of IL-7Rα and γc) are essential for normal lymphoid development. In their absence, severe combined immunodeficiency occurs. By contrast, excessive IL-7/IL-7R-mediated signaling can drive lymphoid leukemia development, disease acceleration and resistance to chemotherapy. IL-7 and IL-7R activate three main pathways: STAT5, PI3K/Akt/mTOR and MEK/Erk, ultimately leading to the promotion of leukemia cell viability, cell cycle progression and growth. However, the contribution of each of these pathways towards particular functional outcomes is still not completely known and appears to differ between normal and malignant states. For example, IL-7 upregulates Bcl-2 in a PI3K/Akt/mTOR-dependent and STAT5-independent manner in T-ALL cells. This is a 'symmetric image' of what apparently happens in normal lymphoid cells, where PI3K/Akt/mTOR does not impact on Bcl-2 and regulates proliferation rather than survival. In this review, we provide an updated summary of the knowledge on IL-7/IL-7R-mediated signaling in the context of cancer, focusing mainly on T-cell acute lymphoblastic leukemia, where this axis has been more extensively studied., Publication costs were supported by LISBOA-01-0145-FEDER-007391, project cofunded by FEDER, through POR Lisboa2020 Programa Operacional Regional de Lisboa, PORTUGAL 2020, and Fundação para a Ciência e a Tecnologia (FCT, Portugal). The research work in JTB's lab related to the present review was supported by the grants FAPESP/20015/2014 and PTDC/MEC-HEM/31588/2017, from FCT; and by the consolidator grant ERC CoG-648455 from the European Research Council, under the European Union's Horizon 2020 research and innovation programme. JTB is an FCT investigator (consolidator). MLO is a LisbonBioMed PhD student and received a fellowship from FCT. PA received a PhD fellowship from the EU Marie Sklodowska-Curie ITN Protein Conjugates.
- Published
- 2019
- Full Text
- View/download PDF
24. Patient-derived models of brain metastases recapitulate human disseminated disease
- Author
-
Claudia C. Faria, Rita Cascão, Carlos Custódia, Eunice Paisana, Tânia Carvalho, Pedro Pereira, Rafael Roque, José Pimentel, José Miguéns, Isidro Cortes-Ciriano, and João T. Barata
- Subjects
Disease Models, Animal ,Mice ,Phosphatidylinositol 3-Kinases ,Brain Neoplasms ,Animals ,Heterografts ,Humans ,Precision Medicine ,General Biochemistry, Genetics and Molecular Biology ,Phosphoinositide-3 Kinase Inhibitors - Abstract
Dissemination of cancer cells from primary tumors to the brain occurs in many cancer patients, increasing morbidity and death. There is an unmet medical need to develop translational platforms to evaluate therapeutic responses. Toward this goal, we established a library of 23 patient-derived xenografts (PDXs) of brain metastases (BMs) from eight distinct primary tumors. In vivo tumor formation correlates with patients' poor survival. Mouse subcutaneous xenografts develop spontaneous metastases and intracardiac PDXs increase dissemination to the CNS, both models mimicking the dissemination pattern of the donor patient. We test the FDA-approved drugs buparlisib (pan-PI3K inhibitor) and everolimus (mTOR inhibitor) and show their efficacy in treating our models. Finally, we show by RNA sequencing that human BMs and their matched PDXs have similar transcriptional profiles. Overall, these models of BMs recapitulate the biology of human metastatic disease and can be valuable translational platforms for precision medicine.
- Published
- 2021
25. Immunophenotype of Gastric Tumors Unveils a Pleiotropic Role of Regulatory T Cells in Tumor Development
- Author
-
Joaquín J Maqueda, Sara Pinto Teles, Irene Gullo, Maria M. Azevedo, Carla Oliveira, Marieke E. Ijsselsteijn, Marta I. Oliveira, Joana Carvalho, Afonso P Basto, Gabriela M. Almeida, Noel F C C de Miranda, Luis Graca, João T. Barata, Sara Rocha, Gilza Gonçalves, Fátima Carneiro, Rocha, Sara [0000-0002-5390-3807], Teles, Sara P [0000-0002-7409-9651], Gullo, Irene [0000-0003-3377-0530], Almeida, Gabriela M [0000-0001-8712-7394], Maqueda, Joaquín J [0000-0002-0800-4167], Carneiro, Fátima [0000-0002-1964-1006], Oliveira, Carla [0000-0001-8340-2264], Apollo - University of Cambridge Repository, Repositório da Universidade de Lisboa, and Instituto de Investigação e Inovação em Saúde
- Subjects
0301 basic medicine ,Cancer Research ,Regulatory T cell ,Perineural invasion ,Context (language use) ,Biology ,Immunofluorescence ,lcsh:RC254-282 ,Article ,Tumor-infiltrating CD4 T cells ,regulatory T cells ,molecular regulation ,03 medical and health sciences ,0302 clinical medicine ,Immunophenotyping ,Immune system ,medicine ,medicine.diagnostic_test ,gastric cancer ,Cancer ,Regulatory T cells ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,3. Good health ,Blot ,030104 developmental biology ,medicine.anatomical_structure ,Molecular regulation ,Oncology ,tumor-infiltrating CD4 T cells ,030220 oncology & carcinogenesis ,Cancer research ,Gastric cancer - Abstract
Copyright: © 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/)., Gastric cancer (GC) patients display increased regulatory T cell (Tregs) numbers in peripheral blood and among tumor-infiltrating lymphocytes. Nevertheless, the role of Tregs in GC progression remains controversial. Here, we sought to explore the impact of Tregs in GCs with distinct histology, and whether Tregs can directly influence tumor cell behavior and GC development. We performed a comprehensive immunophenotyping of 82 human GC cases, through an integrated analysis of multispectral immunofluorescence detection of T cells markers and patient clinicopathological data. Moreover, we developed 3D in vitro co-cultures with Tregs and tumor cells that were followed by high-throughput and light-sheet imaging, and their biological features studied with conventional/imaging flow cytometry and Western blotting. We showed that Tregs located at the tumor nest were frequent in intestinal-type GCs but did not associate with increased levels of effector T cells. Our in vitro results suggested that Tregs preferentially infiltrated intestinal-type GC spheroids, induced the expression of IL2Rα and activation of MAPK signaling pathway in tumor cells, and promoted spheroid growth. Accumulation of Tregs in intestinal-type GCs was increased at early stages of the stomach wall invasion and in the absence of vascular and perineural invasion. In this study, we proposed a non-immunosuppressive mechanism through which Tregs might directly modulate GC cells and thereby promote tumor growth. Our findings hold insightful implications for therapeutic strategies targeting intestinal-type GCs and other tumors with similar immune context., This work was supported by grants: (1) “Institute for Research and Innovation in Health Sciences” (POCI-01-0145-FEDER-007274), by FEDER—Fundo Europeu de Desenvolvimento Regional funds through the COMPETE 2020— Operacional Program for Competitiveness and Internationalisation (POCI), PORTUGAL 2020, Portuguese funds through Portuguese Science & Technology Foundation (FCT)/Ministério da Ciência, Tecnologia e Inovação; (2) Project CANCER Ref. NORTE-01-0145-FEDER-000029 and DOCnet Ref. NORTE-01-0145-FEDER-000003), by Norte Portugal Regional Program (NORTE 2020), under the PORTUGAL 2020 Partnership Agreement, through the European Regional Development Fund (ERDF); (3) Project Ref. PTDC/BBB-ECT/2518/2014, by FCT;(4) FCT PhD Programs and by Programa Operacional Potencial Humano (POCH), specifically by the Biotech Health Programe (Doctoral Program on Cellular and Molecular Biotechnology Applied to Health Sciences); (5) “The role of gastric cancer cell-derived extracellular vesicles”, by IPATIMUP Board of Directors; (6) GenomePT project (POCI-01-0145-FEDER-022184), by COMPETE 2020—POCI, Lisboa Portugal Regional Operational Program (Lisboa2020), Algarve Portugal Regional Operational Program (CRESC Algarve2020), under the PORTUGAL 2020 Partnership Agreement, through ERDF, and by FCT; (7) Marie Skłodowska-Curie grant agreement No. 722148 to Train-EV project by Euro-pean Union’s Horizon 2020 research and innovation program; 8) Project CANCERSTEM funded by ERDF, POCI and FCT; 9) FCT fellowship PD/BD/128406/2017 to SR, and Junior Research contractto JC (regulated by the decree-law 57/2016 emended by law 57/2017). IPATIMUP integrates the i3S Research Unit, which is partially supported by FCT. NFCCdM has received funding from the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation program (grant agreement No. 852832). The authors acknowledgethe support of the Advanced Light Microscopy, Bioimaging, BioSciences Screening and Histologyand Electron Microscopy i3S Scientific Platforms members of the national infrastructure PPBI—Portuguese Platform of Bioimaging (PPBI-POCI-01-0145-FEDER-022122), and the support providedby the Translational Cytometry i3S Scientific Platforms.
- Published
- 2021
- Full Text
- View/download PDF
26. Patient-derived models of brain metastases recapitulate the histopathology and biology of human metastatic cancers
- Author
-
Carlos Custódia, Eunice Paisana, Rita Cascão, José Miguéns, Claudia C. Faria, Tânia Carvalho, José Pimentel, Rafael Roque, João T. Barata, and Pedro M. Pereira
- Subjects
medicine.medical_specialty ,In vivo ,Cancer cell ,medicine ,Cancer research ,Cancer ,Histopathology ,Disease ,Biology ,medicine.disease ,Precision medicine ,Primary tumor ,Intracardiac injection - Abstract
PurposeDissemination of cancer cells from primary tumors to the brain is observed in the great majority of cancer patients, contributing to increased morbidity and being the main cause of death. Most mechanistic and preclinical studies have relied on aggressive cancer cell lines, which fail to represent tumor heterogeneity and are unsuitable to validate therapies due to fast cancer progression in vivo.Experimental designWe established a unique library of subcutaneous and intracardiac patient-derived xenografts (PDXs) of brain metastases (BMs) from eight distinct primary tumor origins. Cancer progression in mice was compared to the matched patient clinical outcome, metastatic dissemination pattern and histopathological features. Preclinical studies with FDA approved drugs were performed.ResultsIn vivo tumor formation of flank-implanted BMs correlated with patients’ poor survival and serial passaging increased tumor aggressiveness. Subcutaneous xenografts originated spontaneous metastases in 61% of the cases, including in the leptomeningeal space (21%). The intracardiac model increased the tropism to the brain and leptomeninges (46%). Strikingly, 62% of intracardiac PDXs shared metastatic sites with the donor patients, including the primary cancer organ and the central nervous system (CNS). Of therapeutic relevance, PDX-derived cultures and corresponding mouse xenografts can be effectively treated with targeted anticancer drugs.ConclusionsPatient-derived models of BMs recapitulate the biology of human metastatic disease and can be a valuable translational platform for precision medicine.TRANSLATIONAL RELEVANCESubcutaneous and intracardiac mouse xenografts of human brain metastases exhibit a spontaneous dissemination pattern that resembles patients’ metastatic disease. The preclinical testing of targeted anticancer drugs using patient-derived cultures and patient-derived xenografts of brain metastasis showed an effective therapeutic response. These translational models represent an outstanding tool to advance the understanding of the biology of brain metastases and to foster the rapid discovery of novel therapeutics.
- Published
- 2020
- Full Text
- View/download PDF
27. STAT5 is essential for IL-7–mediated viability, growth, and proliferation of T-cell acute lymphoblastic leukemia cells
- Author
-
Ruben van Boxtel, Paul J. Coffer, Bruno A. Cardoso, Alice Melão, Milene Costa da Silva, Daniel Dias Ribeiro, Cristina Santos, Ana Elisa Bauer de Camargo Silva, and João T. Barata
- Subjects
0301 basic medicine ,Transcription, Genetic ,Cell Survival ,T cell ,T-cell leukemia ,PIM1 ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,03 medical and health sciences ,Proto-Oncogene Proteins c-pim-1 ,Downregulation and upregulation ,hemic and lymphatic diseases ,STAT5 Transcription Factor ,Tumor Cells, Cultured ,medicine ,Humans ,Protein kinase B ,Mechanistic target of rapamycin ,Cell Proliferation ,Lymphoid Neoplasia ,biology ,Gene Expression Regulation, Leukemic ,Chemistry ,Interleukin-7 ,JAK-STAT signaling pathway ,Hematology ,medicine.disease ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Cancer research ,biology.protein - Abstract
T-cell acute lymphoblastic leukemia (T-ALL) constitutes an aggressive subset of ALL, the most frequent childhood malignancy. Whereas interleukin-7 (IL-7) is essential for normal T-cell development, it can also accelerate T-ALL development in vivo and leukemia cell survival and proliferation by activating phosphatidylinositol 3-kinase/protein kinase B/mechanistic target of rapamycin signaling. Here, we investigated whether STAT5 could also mediate IL-7 T-ALL-promoting effects. We show that IL-7 induces STAT pathway activation in T-ALL cells and that STAT5 inactivation prevents IL-7-mediated T-ALL cell viability, growth, and proliferation. At the molecular level, STAT5 is required for IL-7-induced downregulation of p27kip1 and upregulation of the transferrin receptor, CD71. Surprisingly, STAT5 inhibition does not significantly affect IL-7-mediated Bcl-2 upregulation, suggesting that, contrary to normal T-cells, STAT5 promotes leukemia cell survival through a Bcl-2-independent mechanism. STAT5 chromatin immunoprecipitation sequencing and RNA sequencing reveal a diverse IL-7-driven STAT5-dependent transcriptional program in T-ALL cells, which includes BCL6 inactivation by alternative transcription and upregulation of the oncogenic serine/threonine kinase PIM1 Pharmacological inhibition of PIM1 abrogates IL-7-mediated proliferation on T-ALL cells, indicating that strategies involving the use of PIM kinase small-molecule inhibitors may have therapeutic potential against a majority of leukemias that rely on IL-7 receptor (IL-7R) signaling. Overall, our results demonstrate that STAT5, in part by upregulating PIM1 activity, plays a major role in mediating the leukemia-promoting effects of IL-7/IL-7R.
- Published
- 2018
- Full Text
- View/download PDF
28. LMD-12. Ubiquitin Conjugating Enzymes promote leptomeningeal dissemination and decrease survival in patients with brain metastatic disease
- Author
-
Tânia Carvalho, Eunice Paisana, Pedro Pereira, Rita Cascão, Marc Remke, Rafael Roque, David Pauck, Daniel Picard, João T. Barata, Pedro Ruivo, Claudia C. Faria, José Pimentel, Nan Qin, José Miguéns, and Carlos Custódia
- Subjects
Leptomeningeal Disease ,business.industry ,Cancer research ,Medicine ,AcademicSubjects/MED00300 ,In patient ,AcademicSubjects/MED00310 ,Disease ,Ubiquitin-conjugating enzyme ,business ,Supplement Abstracts - Abstract
The dissemination of cancer cells to the brain parenchyma (brain metastases - BMs) and to the leptomeninges (leptomeningeal dissemination – LD) are a late-stage complication of systemic cancers, with a poor survival. Despite advances in radiation and chemotherapy, including intrathecal administration of anticancer agents, these forms of advanced cancer are incurable. Therefore, there is an unmet clinical need for novel effective therapies that target both parenchymal and leptomeningeal disease. We analysed the transcriptomic profile of BMs from patients with diverse primary tumors, treated at CHULN, to identify genetic drivers of cancer cell dissemination to the brain and potential novel targets for therapy. The most differentially expressed gene codifies a ubiquitin conjugating enzyme (UCE). UCE levels were evaluated in tissue microarrays of BMs from an independent cohort of patients and correlated with clinical data. UCE functional role was assessed in vitro and in vivo using modulated lung and breast cancer cell lines. A high-throughput drug screening was performed to find UCE-targeting compounds. High protein levels of the UCE were associated with decreased survival in patients with BMs, independently of the primary tumor origin. High levels of UCE led to increased migration and invasion abilities in cancer cell lines in vitro, with no effect in proliferation. In vivo, high levels of UCE increased leptomeningeal dissemination and decreased survival in orthotopic models of breast cancer BMs. Leptomeningeal disease promoted by UCE was prevented by oral administration of inhibitor A, identified in our high-throughput drug screening. In conclusion, we have identified UCE as a prognostic marker in patients with BMs from different primary tumors. In orthotopic mouse models of the disease, UCE led to a worse survival and promoted leptomeningeal dissemination. Strikingly, this aggressive disease phenotype was prevented by oral therapy with inhibitor A.
- Published
- 2021
29. Flip the coin: IL-7 and IL-7R in health and disease
- Author
-
João T, Barata, Scott K, Durum, and Benedict, Seddon
- Subjects
Receptors, Interleukin-7 ,Cell Survival ,Interleukin-7 ,Neoplasms ,T-Lymphocytes ,Animals ,Homeostasis ,Humans ,Cell Differentiation ,Immunotherapy - Abstract
The cytokine IL-7 and its receptor, IL-7R, are critical for T cell and, in the mouse, B cell development, as well as differentiation and survival of naive T cells, and generation and maintenance of memory T cells. They are also required for innate lymphoid cell (ILC) development and maintenance, and consequently for generation of lymphoid structures and barrier defense. Here we discuss the central role of IL-7 and IL-7R in the lymphoid system and highlight the impact of their deregulation, placing a particular emphasis on their 'dark side' as promoters of cancer development. We also explore therapeutic implications and opportunities associated with either positive or negative modulation of the IL-7-IL-7R signaling axis.
- Published
- 2019
30. Overexpression of wild-type IL-7Rα promotes T-cell acute lymphoblastic leukemia/lymphoma
- Author
-
Thea Hogan, João Luís Neto, Benedict Seddon, Sofie Demeyer, Ana Patricia Silva, Yunlei Li, Mafalda Matos, Ana Rita Grosso, Afonso R. M. Almeida, Jules P.P. Meijerink, João T. Barata, Jan Cools, Ana Cachucho, Pathology, DCV - Departamento de Ciências da Vida, UCIBIO - Applied Molecular Biosciences Unit, and Repositório da Universidade de Lisboa
- Subjects
Oncology ,medicine.medical_specialty ,Carcinogenesis ,T cell ,Immunology ,Mice, Transgenic ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Biochemistry ,Interleukin-7 Receptor alpha Subunit ,03 medical and health sciences ,Mice ,0302 clinical medicine ,SDG 3 - Good Health and Well-being ,Internal medicine ,medicine ,media_common.cataloged_instance ,Animals ,Humans ,European union ,030304 developmental biology ,media_common ,0303 health sciences ,Acute lymphoblastic leukemia-lymphoma ,Lymphoid Neoplasia ,Receptors, Interleukin-7 ,Thymocytes ,business.industry ,Gene Expression Regulation, Leukemic ,European research ,Cell Biology ,Hematology ,Neoplasms, Experimental ,3. Good health ,Neoplasm Proteins ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Mutation ,business ,Protein overexpression ,Signal Transduction - Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is often not cured by frontline chemotherapy, and efforts to improve treatment by targeting oncogenes such as NOTCH1 have been hampered by toxicity. Silva and colleagues studied primary patient samples to show that high-level interleukin 7 receptor α (IL7Rα) gene expression correlates with ongoing, oncogenic IL7R-mediated signaling. Using new in vivo models, they characterized the impact of IL7Rα expression on the pathogenesis of T-ALL and its response to various targeted therapies that reduce IL7-related signaling., Key Points Mice overexpressing IL-7Rα develop leukemia with features of human T-ALL and sensitivity to ruxolitinib, dactolisib, and venetoclax.T-ALL patients with high levels of wild-type IL7R present with evidence of ongoing, oncogenic-like IL-7R–mediated activation of signaling., Visual Abstract, Tight regulation of IL-7Rα expression is essential for normal T-cell development. IL-7Rα gain-of-function mutations are known drivers of T-cell acute lymphoblastic leukemia (T-ALL). Although a subset of patients with T-ALL display high IL7R messenger RNA levels and cases with IL7R gains have been reported, the impact of IL-7Rα overexpression, rather than mutational activation, during leukemogenesis remains unclear. In this study, overexpressed IL-7Rα in tetracycline-inducible Il7r transgenic and Rosa26 IL7R knockin mice drove potential thymocyte self-renewal, and thymus hyperplasia related to increased proliferation of T-cell precursors, which subsequently infiltrated lymph nodes, spleen, and bone marrow, ultimately leading to fatal leukemia. The tumors mimicked key features of human T-ALL, including heterogeneity in immunophenotype and genetic subtype between cases, frequent hyperactivation of the PI3K/Akt pathway paralleled by downregulation of p27Kip1 and upregulation of Bcl-2, and gene expression signatures evidencing activation of JAK/STAT, PI3K/Akt/mTOR and Notch signaling. Notably, we also found that established tumors may no longer require high levels of IL-7R expression upon secondary transplantation and progressed in the absence of IL-7, but remain sensitive to inhibitors of IL-7R–mediated signaling ruxolitinib (Jak1), AZD1208 (Pim), dactolisib (PI3K/mTOR), palbociclib (Cdk4/6), and venetoclax (Bcl-2). The relevance of these findings for human disease are highlighted by the fact that samples from patients with T-ALL with high wild-type IL7R expression display a transcriptional signature resembling that of IL-7–stimulated pro-T cells and, critically, of IL7R-mutant cases of T-ALL. Overall, our study demonstrates that high expression of IL-7Rα can promote T-cell tumorigenesis, even in the absence of IL-7Rα mutational activation.
- Published
- 2019
31. Introduction to the special issue on T-cell acute lymphoblastic leukemia genetics, biology and therapeutics
- Author
-
João T. Barata
- Subjects
0303 health sciences ,Cancer Research ,Lymphoblastic Leukemia ,T cell ,Computational biology ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Genetics ,medicine ,Molecular Medicine ,Humans ,Periodicals as Topic ,Molecular Biology ,030304 developmental biology - Published
- 2019
- Full Text
- View/download PDF
32. Stem Cell Leukemia: how a TALented actor can go awry on the hematopoietic stage
- Author
-
João T. Barata, Nádia C. Correia, Marie-Laure Arcangeli, and Françoise Pflumio
- Subjects
0301 basic medicine ,Cancer Research ,Leukemia, T-Cell ,Carcinogenesis ,T-Cell Acute Lymphocytic Leukemia Protein 1 ,Biology ,03 medical and health sciences ,Proto-Oncogene Proteins ,Basic Helix-Loop-Helix Transcription Factors ,medicine ,Humans ,Transcription factor ,Hematopoietic stem cell ,Hematology ,medicine.disease ,3. Good health ,Haematopoiesis ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Immunology ,Neoplastic Stem Cells ,Ectopic expression ,Stem cell ,Neuroscience ,TAL1 - Abstract
TAL1/SCL/TCL5 is a critical transcription factor for hematopoietic stem cell maintenance and regulation of early hematopoiesis. However, aberrant expression of TAL1 in committed T-cell precursors is also directly implicated in the development of T-cell leukemia. Roughly 25 years ago TAL1 was identified in early hematopoietic cells and involved in leukemia. Here, we review the wealth of knowledge gained since then on its physiological roles and mechanisms by which TAL1 ectopic expression contributes to leukemogenesis. We emphasize recent findings that shed light into the intricacies of TAL1 (epi)genetic regulation and the transcription network orchestrated by this major T-cell oncogene. Importantly, an exciting time is coming when data using the mechanistic knowledge accumulated on TAL1 may be used to develop novel anti-leukemia targeted therapies.
- Published
- 2016
- Full Text
- View/download PDF
33. CHK1 and replicative stress in T-cell leukemia: Can an irreverent tumor suppressor end up playing the oncogene?
- Author
-
João T. Barata and Leonor M. Sarmento
- Subjects
DNA Replication ,0301 basic medicine ,Cancer Research ,Leukemia, T-Cell ,DNA repair ,DNA damage ,T-cell leukemia ,Biology ,law.invention ,Mice ,03 medical and health sciences ,law ,Genetics ,medicine ,Animals ,Humans ,Genes, Tumor Suppressor ,CHEK1 ,Molecular Biology ,Oncogene ,DNA replication ,Cancer ,Oncogenes ,medicine.disease ,3. Good health ,030104 developmental biology ,Checkpoint Kinase 1 ,Immunology ,Cancer research ,Molecular Medicine ,Suppressor ,biological phenomena, cell phenomena, and immunity - Abstract
Replicative stress (RS) is a cell-intrinsic phenomenon enhanced by oncogenic transformation. Checkpoint kinase 1 (CHK1) is a key component of the ATR-dependent DNA damage response pathway that protects cells from RS by preventing replication fork collapse and activating homologous DNA repair. Taking this knowledge into account, one would predict CHK1 behaves strictly as a tumor suppressor. However, the reality seems far more complex. CHEK1 loss-of-function mutations have not been found in human tumors, and transgenic expression of Chek1 in mice promotes oncogene-induced transformation through RS inhibition. Moreover, CHK1 is overexpressed in various human cancers and CHK1 inhibitors have been developed as sensitizers to enhance the cytotoxicity of DNA damage-inducing chemotherapies. Here, we summarize the literature on the involvement of CHK1 in cancer progression, including our recent observation that CHK1 sustains T-cell acute lymphoblastic leukemia (T-ALL) cell viability. We also debate the importance of identifying patients that could benefit the most from treatment with CHK1 inhibitors, taking T-ALL as a model, and propose possible markers of therapeutic response.
- Published
- 2016
- Full Text
- View/download PDF
34. PRL3 enhances T cell acute lymphoblastic leukemia growth through suppressing T cell signaling pathways and apoptosis
- Author
-
Jessica S. Blackburn, D Do, Mariana L. Oliveira, P. Van Vlierberghe, Sara P. Garcia, C Yan, João T. Barata, Filip Matthijssens, Tom Taghon, J R Allen, W Van Loocke, David M. Langenau, Franki Speleman, Elaine G. Garcia, Sowmya Iyer, A Veloso, R Morris, Siebe Loontiens, Alejandro Gutierrez, Karin M. McCarthy, N Iftimia, Wilhelm Haas, and Dalton C. Brunson
- Subjects
0301 basic medicine ,Cancer Research ,VAV1 ,T cell ,T-Lymphocytes ,Phosphatase ,Apoptosis ,Protein tyrosine phosphatase ,Mice, SCID ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Mice, Inbred NOD ,medicine ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Animals ,Humans ,Zebrafish ,Cell Proliferation ,Hematology ,medicine.disease ,Prognosis ,Xenograft Model Antitumor Assays ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Phosphorylation ,Female ,Signal transduction ,Protein Tyrosine Phosphatases ,Tyrosine kinase - Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy of thymocytes and is largely driven by the NOTCH/MYC pathway. Yet, additional oncogenic drivers are required for transformation. Here, we identify protein tyrosine phosphatase type 4 A3 (PRL3) as a collaborating oncogenic driver in T-ALL. PRL3 is expressed in a large fraction of primary human T-ALLs and is commonly co-amplified with MYC. PRL3 also synergized with MYC to initiate early-onset ALL in transgenic zebrafish and was required for human T-ALL growth and maintenance. Mass-spectrometry phosphoproteomic analysis and mechanistic studies uncovered that PRL3 suppresses downstream T-cell phosphorylation signaling pathways, including those modulated by VAV1, and subsequently suppresses apoptosis in leukemia cells. Taken together, our studies have identified new roles for PRL3 as a collaborating oncogenic driver in human T-ALL and suggest that therapeutic targeting of the PRL3 phosphatase will likely be a useful treatment strategy for T-ALL.
- Published
- 2020
35. PS920 PHF6 LOSS DRIVES IL7R ONCOGENE ADDICTION IN TLX1 DRIVEN T-ALL
- Author
-
Franki Speleman, Tom Taghon, P. Van Vlierberghe, Givani Dewyn, David M. Langenau, João T. Barata, Suzanne Vanhauwaert, Mariana L. Oliveira, Kaat Durinck, Lisa Depestel, Jan Cools, Siebe Loontiens, and C E de Bock
- Subjects
Cancer research ,Hematology ,Interleukin-7 receptor ,Oncogene Addiction ,Psychology - Published
- 2019
- Full Text
- View/download PDF
36. CHK1 overexpression in T-cell acute lymphoblastic leukemia is essential for proliferation and survival by preventing excessive replication stress
- Author
-
Gonçalo Real, Paula M. Alves, Miguel Abecasis, Leila R. Martins, R. Nascimento, R. M. E. Parkhouse, Vanda Póvoa, Jules P.P. Meijerink, João T. Barata, Catarina Moita, Luis F. Moita, Leonor M. Sarmento, I Antunes, and Pediatrics
- Subjects
DNA Replication ,Cancer Research ,Programmed cell death ,Cell Survival ,DNA damage ,T cell ,Ataxia Telangiectasia Mutated Proteins ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Mice ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,CHEK1 ,Molecular Biology ,Cell Proliferation ,Benzodiazepinones ,Thymocytes ,Caspase 3 ,Cell growth ,Cell Cycle ,DNA replication ,Cell cycle ,medicine.disease ,Leukemia ,medicine.anatomical_structure ,Gene Knockdown Techniques ,Checkpoint Kinase 1 ,Immunology ,Cancer research ,Pyrazoles ,biological phenomena, cell phenomena, and immunity ,Protein Kinases ,Neoplasm Transplantation ,DNA Damage - Abstract
Checkpoint kinase 1 (CHK1) is a key component of the ATR (ataxia telangiectasia-mutated and Rad3-related)-dependent DNA damage response pathway that protect cells from replication stress, a cell intrinsic phenomenon enhanced by oncogenic transformation. Here, we show that CHK1 is overexpressed and hyperactivated in T-cell acute lymphoblastic leukemia (T-ALL). CHEK1 mRNA is highly abundant in patients of the proliferative T-ALL subgroup and leukemia cells exhibit constitutively elevated levels of the replication stress marker phospho-RPA32 and the DNA damage marker γH2AX. Importantly, pharmacologic inhibition of CHK1 using PF-004777736 or CHK1 short hairpin RNA-mediated silencing impairs T-ALL cell proliferation and viability. CHK1 inactivation results in the accumulation of cells with incompletely replicated DNA, ensuing DNA damage, ATM/CHK2 activation and subsequent ATM- and caspase-3-dependent apoptosis. In contrast to normal thymocytes, primary T-ALL cells are sensitive to therapeutic doses of PF-004777736, even in the presence of stromal or interleukin-7 survival signals. Moreover, CHK1 inhibition significantly delays in vivo growth of xenotransplanted T-ALL tumors. We conclude that CHK1 is critical for T-ALL proliferation and viability by downmodulating replication stress and preventing ATM/caspase-3-dependent cell death. Pharmacologic inhibition of CHK1 may be a promising therapeutic alternative for T-ALL treatment.
- Published
- 2015
37. Multifaceted CK2 in malignant and healthy T cells
- Author
-
Bruno Silva-Santos, Sérgio T. Ribeiro, João T. Barata, and Repositório da Universidade de Lisboa
- Subjects
0303 health sciences ,Information retrieval ,Computer science ,γδ T lymphocytes ,Creative commons ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,Editorial ,Oncology ,Protein kinase CK2 ,License ,T-cell acute lymphoblastic leukemia ,030304 developmental biology ,030215 immunology - Abstract
Copyright © Ribeiro et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License 3.0 (CC BY 3.0), which permits unrestricted use, distribution, and re-production in any medium, provided the original author and source are credited., Among kinases that support the survival and turnover of tumor cells, the serine/threonine protein kinase CK2 has been shown to be frequently overexpressed or hyperactivated in solid and hematological malignancies. Our previous work on T-cell acute lymphoblastic leukemia (T-ALL) showed that CK2 maintains leukemia cell viability by phosphorylating and thereby inactivating the tumor suppressor PTEN, which results in hyperactivation of PI3K/AKT signaling. We also demonstrated the potential of using the clinical-grade CK2-specific chemical inhibitor, CX-4945 (Silmitasertib), against primary T-ALL cells. We have now extended these findings to the rare (
- Published
- 2017
38. From the outside, from within: Biological and therapeutic relevance of signal transduction in T-cell acute lymphoblastic leukemia
- Author
-
Bruno A. Cardoso, Isabel Alcobia, João T. Barata, Teresa L. Serafim, Padma Akkapeddi, Rita Fragoso, Mariana L. Oliveira, and Afonso R. M. Almeida
- Subjects
0301 basic medicine ,Kinase ,T cell ,Cancer ,Cell Biology ,Biology ,medicine.disease ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Models, Biological ,3. Good health ,03 medical and health sciences ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Drug development ,Cancer research ,medicine ,Animals ,Humans ,Signal transduction ,Receptor ,Interleukin-7 receptor ,Signal Transduction - Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological cancer that arises from clonal expansion of transformed T-cell precursors. In this review we summarize the current knowledge on the external stimuli and cell-intrinsic lesions that drive aberrant activation of pivotal, pro-tumoral intracellular signaling pathways in T-cell precursors, driving transformation, leukemia expansion, spread or resistance to therapy. In addition to their pathophysiological relevance, receptors and kinases involved in signal transduction are often attractive candidates for targeted drug development. As such, we discuss also the potential of T-ALL signaling players as targets for therapeutic intervention.
- Published
- 2017
39. Abstract 3696: PHF6loss drives IL7R oncogene addiction in TLX1 driven T-ALL
- Author
-
Charles E. de Bock, Givani Dewyn, Jan Cools, João T. Barata, Kaat Durinck, Lisa Depestel, Pieter Van Vlierberghe, David M. Langenau, Tom Taghon, Franki Speleman, Suzanne Vanhauwaert, Siebe Loontiens, and Mariana L Oliveira
- Subjects
Regulation of gene expression ,Cancer Research ,Gene knockdown ,Oncogene ,Lymphoblast ,Biology ,Oncogene Addiction ,medicine.disease ,biology.organism_classification ,Leukemia ,Oncology ,medicine ,Cancer research ,Interleukin-7 receptor ,Zebrafish - Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a genetically heterogeneous disease. The PHF6 gene is frequently targeted by loss-of-function mutations or deletions, with the highest prevalence in TLX1 or TLX3 rearranged T-ALLs. To gain insights into the putative function of PHF6 as a tumor suppressor in the T-cell lineage, we investigated the effects of PHF6 knock down during normal and malignant thymocytes. Notably, we observed broad effects on the investigated transcriptomes suggesting an important role for PHF6 in gene regulation. Furthermore, IL7R was identified as a common transcriptional target that was significantly upregulated upon PHF6 knockdown in both normal and malignant T cells. IL7R encodes a cytokine receptor critically involved in normal thymic development and which also acts as a bona fide oncogene in subset of primary T-ALLs. Thus, loss of PHF6 might further boost oncogenic addiction of leukemic T-cell lymphoblast to IL7-induced JAK-STAT signaling. To further explore the role of PHF6 inactivation in TLX1 driven leukemogenesis in vivo, we performed zebrafish modeling. For this, we generated a stable tg(rag2:TLX1, rag2:GFP) overexpressing as well as a phf6 knock out zebrafish line. These lines were crossed and offspring was monitored for T-ALL formation. Interestingly, three fish out of a cohort of 80 animals developed leukemia between 10 to 18 months of age. These leukemias originated from the thymus, spreaded throughout the whole body and were transplantable. Thus far, no leukemia was detected in PHF6 mutated or TLX1 overexpressing only zebrafish. Leukemic cells obtained from tumors that developed in the PHF6null TLX1rag2-TLX1/GFP animals were subjected to RNA-, ATAC- and H3K27ac ChIP-sequencing to assess the epigenetic status of the IL7R locus. In addition, exome-, and CNV-sequencing was performed to identify somatic lesions that cooperated loss of Phf6 during TLX1 driven T-cell transformation in zebrafish. Furthermore, additional injections of TLX1 in combination with an activating IL7R mutant into phf6 mutant zebrafish are currently ongoing to monitor additional effects on accelerated tumor formation. In conclusion, our data suggest that loss of PHF6 drives TLX1 mediated leukemogenesis, at least in part, by increasing surface IL7R expression. Therefore, we believe that increased addiction to oncogenic JAK-STAT signaling may render PHF6 mutant leukemic cells more sensitive to JAK inhibitors, a notion that we are currently investigating in our TLX1/PHF6 and TLX1/PHF6/IL7R zebrafish models. Citation Format: Siebe Loontiens, Kaat Durinck, Suzanne Vanhauwaert, Lisa Depestel, Mariana L. Oliveira, Givani Dewyn, Charles De Bock, João T. Barata, David Langenau, Jan Cools, Tom Taghon, Pieter Van Vlierberghe, Frank Speleman. PHF6loss drives IL7R oncogene addiction in TLX1 driven T-ALL [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 3696.
- Published
- 2019
- Full Text
- View/download PDF
40. Novel TAL1 targets beyond protein-coding genes: identification of TAL1-regulated microRNAs in T-cell acute lymphoblastic leukemia
- Author
-
Franki Speleman, Maté Ongenaert, João T. Barata, Nádia C. Correia, Kaat Durinck, Francisco J. Enguita, Pieter Rondou, and Ana Paula Leite
- Subjects
Cancer Research ,T-Cell Acute Lymphocytic Leukemia Protein 1 ,T cell ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Proto-Oncogene Proteins ,hemic and lymphatic diseases ,microRNA ,Basic Helix-Loop-Helix Transcription Factors ,medicine ,Humans ,Gene ,030304 developmental biology ,Regulation of gene expression ,Genetics ,0303 health sciences ,hemic and immune systems ,Hematology ,3. Good health ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,medicine.anatomical_structure ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,Identification (biology) ,TAL1 - Abstract
Novel TAL1 targets beyond protein-coding genes: identification of TAL1-regulated microRNAs in T-cell acute lymphoblastic leukemia
- Published
- 2013
- Full Text
- View/download PDF
41. Optimal interleukin-7 receptor-mediated signaling, cell cycle progression and viability of T-cell acute lymphoblastic leukemia cells rely on casein kinase 2 activity
- Author
-
Bruno A. Cardoso, João T. Barata, Alice Melão, and Maureen Spit
- Subjects
0301 basic medicine ,Cell Survival ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Article ,Interleukin-7 Receptor alpha Subunit ,03 medical and health sciences ,Downregulation and upregulation ,medicine ,Humans ,Interleukin-7 receptor ,Protein kinase A ,Casein Kinase II ,Cells, Cultured ,Cell growth ,Interleukin-7 ,Cell Cycle ,Hematology ,Cell cycle ,medicine.disease ,Cell biology ,Leukemia ,030104 developmental biology ,HEK293 Cells ,Casein kinase 2 ,Signal transduction ,Signal Transduction - Abstract
Interleukin-7 and interleukin-7 receptor are essential for normal T-cell development and homeostasis, whereas excessive interleukin-7/interleukin-7 receptor-mediated signaling promotes leukemogenesis. The protein kinase, casein kinase 2, is overexpressed and hyperactivated in cancer, including T-cell acute lymphoblastic leukemia. Herein, we show that while interleukin-7 had a minor but significant positive effect on casein kinase 2 activity in leukemia T-cells, casein kinase 2 activity was mandatory for optimal interleukin-7/interleukin-7 receptor-mediated signaling. Casein kinase 2 pharmacological inhibition impaired signal transducer and activator of transcription 5 and phosphoinositide 3-kinase/v-Akt murine thymoma viral oncogene homolog 1 pathway activation triggered by interleukin-7 or by mutational activation of interleukin-7 receptor. By contrast, forced expression of casein kinase 2 augmented interleukin-7 signaling in human embryonic kidney 293T cells reconstituted with the interleukin-7 receptor machinery. Casein kinase 2 inactivation prevented interleukin-7-induced B-cell lymphoma 2 upregulation, maintenance of mitochondrial homeostasis and viability of T-cell acute lymphoblastic leukemia cell lines and primary leukemia cells collected from patients at diagnosis. Casein kinase 2 inhibition further abrogated interleukin-7-mediated cell growth and upregulation of the transferrin receptor, and blocked cyclin A and E upregulation and cell cycle progression. Notably, casein kinase 2 was also required for the viability of mutant interleukin-7 receptor expressing leukemia T-cells. Overall, our study identifies casein kinase 2 as a major player in the effects of interleukin-7 and interleukin-7 receptor in T-cell acute lymphoblastic leukemia. This further highlights the potential relevance of targeting casein kinase 2 in this malignancy.
- Published
- 2016
- Full Text
- View/download PDF
42. FOXP3 can modulate TAL1 transcriptional activity through interaction with LMO2
- Author
-
Veerle Fleskens, Cornelieke E.G.M. Pals, S G J M Coenen, Michal Mokry, Bruno A. Cardoso, Janneke G. C. Peeters, Paul J. Coffer, S Huppelschoten, A M van der Leun, João T. Barata, J van Loosdregt, and Repositório da Universidade de Lisboa
- Subjects
0301 basic medicine ,LMO2 ,Cancer Research ,T-Cell Acute Lymphocytic Leukemia Protein 1 ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,03 medical and health sciences ,Proto-Oncogene Proteins ,hemic and lymphatic diseases ,Basic Helix-Loop-Helix Transcription Factors ,Genetics ,Journal Article ,Humans ,Viability assay ,Transcription factor ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Tumor Suppressor Proteins ,Cell Cycle ,FOXP3 ,Forkhead Transcription Factors ,hemic and immune systems ,LIM Domain Proteins ,Cell cycle ,030104 developmental biology ,Cell culture ,Cancer research ,TAL1 - Abstract
© 2015 Macmillan Publishers Limited. All rights reserved., T-cell acute lymphoblastic leukemia (T-ALL) frequently involves aberrant expression of TAL1 (T-cell acute lymphocytic leukemia 1) and LMO2, oncogenic members of the TAL1 transcriptional complex. Transcriptional activity of the TAL1-complex is thought to have a pivotal role in the transformation of thymocytes and is associated with a differentiation block and self-renewal. The transcription factor Forkhead Box P3 (FOXP3) was recently described to be expressed in a variety of malignancies including T-ALL. Here we show that increased FOXP3 levels negatively correlate with expression of genes regulated by the oncogenic TAL1-complex in human T-ALL patient samples as well as a T-ALL cell line ectopically expressing FOXP3. In these cells, FOXP3 expression results in altered regulation of cell cycle progression and reduced cell viability. Finally, we demonstrate that FOXP3 binds LMO2 in vitro, resulting in decreased interaction between LMO2 and TAL1, providing a molecular mechanism for FOXP3-mediated transcriptional modulation in T-ALL. Collectively, our findings provide initial evidence for a novel role of FOXP3 as a tumor suppressor in T-ALL through modulation of TAL1 transcriptional activity., VF and JvL were supported by a grant from the Dutch Arthritis Foundation (Rheumafonds), and BAC was supported by a fellowship from Fundação para a Ciência e a Tecnologia. This work was supported by grant from the Dutch Arthritis Foundation (Rheumafonds), and a fellowship from Fundação para a Ciência e a Tecnologia.
- Published
- 2016
43. MiR-146b negatively regulates migration and delays progression of T-cell acute lymphoblastic leukemia
- Author
-
Nádia C. Correia, Francisco J. Enguita, Rita Fragoso, João T. Barata, Tânia Carvalho, and Repositório da Universidade de Lisboa
- Subjects
Male ,0301 basic medicine ,T cell ,Chemokinesis ,Kaplan-Meier Estimate ,Mice, SCID ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Filamentous actin ,Article ,03 medical and health sciences ,Cell Movement ,Mice, Inbred NOD ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Gene silencing ,Progenitor cell ,T-Cell Acute Lymphocytic Leukemia Protein 1 ,Multidisciplinary ,Gene Expression Regulation, Leukemic ,medicine.disease ,Xenograft Model Antitumor Assays ,MicroRNAs ,Haematopoiesis ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Gene Knockdown Techniques ,Immunology ,Disease Progression ,Cancer research ,Female ,TAL1 - Abstract
© The Author(s) 2016. This work is licensed under a Creative Commons Attribution 4.0 International License. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in the credit line; if the material is not included under the Creative Commons license, users will need to obtain permission from the license holder to reproduce the material. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/, Previous results indicated that miR-146b-5p is downregulated by TAL1, a transcription factor critical for early hematopoiesis that is frequently overexpressed in T-cell acute lymphoblastic leukemia (T-ALL) where it has an oncogenic role. Here, we confirmed that miR-146b-5p expression is lower in TAL1-positive patient samples than in other T-ALL cases. Furthermore, leukemia T-cells display decreased levels of miR-146b-5p as compared to normal T-cells, thymocytes and other hematopoietic progenitors. MiR-146b-5p silencing enhances the in vitro migration and invasion of T-ALL cells, associated with increased levels of filamentous actin and chemokinesis. In vivo, miR-146b overexpression in a TAL1-positive cell line extends mouse survival in a xenotransplant model of human T-ALL. In contrast, knockdown of miR-146b-5p results in leukemia acceleration and decreased mouse overall survival, paralleled by faster tumor infiltration of the central nervous system. Our results suggest that miR-146b-5p is a functionally relevant microRNA gene in the context of T-ALL, whose negative regulation by TAL1 and possibly other oncogenes contributes to disease progression by modulating leukemia cell motility and disease aggressiveness., These studies were supported by Liga Portuguesa Contra o Cancro (Terry Fox Award) and by Fundação para a Ciência e a Tecnologia (project PTDC/BIM-ONC/1548/2012). N.C.C. received an FCT-SFRH PhD fellowship. R.F. and J.T.B. are supported by FCT investigator Starting and Consolidation grants, respectively.
- Published
- 2016
44. Human naïve regulatory T-cells feature high steady-state turnover and are maintained by IL-7
- Author
-
Rui M. M. Victorino, João Sette Whitaker Ferreira, Paula Matoso, Adriana S. Albuquerque, Susana M. Fernandes, Íris Caramalho, Ana R. Pires, Ana Serra-Caetano, Rémi Cheynier, João T. Barata, Russell B. Foxall, Ana E. Sousa, Susana L. Silva, and Repositório da Universidade de Lisboa
- Subjects
Adult ,0301 basic medicine ,Adolescent ,Cell Survival ,Clinical settings ,chemical and pharmacologic phenomena ,T-Lymphocytes, Regulatory ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Regulatory T-cell homeostasis ,Microbiology section ,Humans ,Medicine ,Immune response ,Human regulatory T-cells ,Cell survival ,Thymic involution ,IL-7 ,business.industry ,Interleukin-7 ,Research Paper: Immunology ,Immunity ,Forkhead Transcription Factors ,hemic and immune systems ,Naïve regulatory T-cells ,Thymectomy ,030104 developmental biology ,Oncology ,Healthy individuals ,Immunology ,Immunology and Microbiology Section ,business ,030215 immunology - Abstract
Naïve FoxP3-expressing regulatory T-cells (Tregs) are essential to control immune responses via continuous replenishment of the activated-Treg pool with thymus-committed suppressor cells. The mechanisms underlying naïve-Treg maintenance throughout life in face of the age-associated thymic involution remain unclear. We found that in adults thymectomized early in infancy the naïve-Treg pool is remarkably well preserved, in contrast to conventional naïve CD4 T-cells. Naïve-Tregs featured high levels of cycling and pro-survival markers, even in healthy individuals, and contrasted with other circulating naïve/memory CD4 T-cell subsets in terms of their strong γc-cytokine-dependent signaling, particularly in response to IL-7. Accordingly, ex-vivo stimulation of naïve-Tregs with IL-7 induced robust cytokine-dependent signaling, Bcl-2 expression, and phosphatidylinositol 3-kinase (PI3K)-dependent proliferation, whilst preserving naïve phenotype and suppressive capacity. Altogether, our data strongly implicate IL-7 in the thymus-independent long-term survival of functional naïve-Tregs, and highlight the potential of targeting the IL-7 pathway to modulate Tregs in different clinical settings., This work was supported by Fundação para a Ciência e Tecnologia (FCT; POCI2010/IC/83068/2007 to RMMV; PTDC/SAU-MIC/109786/2009 to AES), and Gulbenkian Foundation (96526/2009 to JF; P132532/2013 to AES). SLS, ASA, RBF, ARP, PM and SMF received FCT scholarships.
- Published
- 2016
- Full Text
- View/download PDF
45. TAL1/SCL is downregulated upon histone deacetylase inhibition in T-cell acute lymphoblastic leukemia cells
- Author
-
P J Coffer, José Andrés Yunes, Maria Carmo-Fonseca, Bruno A. Cardoso, Angelo Brunelli Albertoni Laranjeira, S F de Almeida, João T. Barata, and University of Groningen
- Subjects
EXPRESSION ,Chromatin Immunoprecipitation ,Cancer Research ,T-Cell Acute Lymphocytic Leukemia Protein 1 ,Down-Regulation ,TAL1/SCL ,SCL GENE ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,ACTIVATION ,HDAC inhibitors ,Proto-Oncogene Proteins ,TAL-1 ,Basic Helix-Loop-Helix Transcription Factors ,Tumor Cells, Cultured ,medicine ,Humans ,TRANSCRIPTION FACTOR ,CANCER-CELLS ,Histone deacetylase 5 ,HDAC11 ,LOOP-HELIX PROTEIN ,HDAC8 ,Hematology ,medicine.disease ,Molecular biology ,HDAC1 ,Up-Regulation ,APOPTOSIS ,TRANSLOCATION ,Histone Deacetylase Inhibitors ,Leukemia ,Oncology ,Cancer research ,Histone deacetylase ,T-ALL ,TAL1 - Abstract
The transcription factor T-cell acute lymphocytic leukemia (TAL)-1 is a major T-cell oncogene associated with poor prognosis in T-cell acute lymphoblastic leukemia (T-ALL). TAL1 binds histone deacetylase 1 and incubation with histone deacetylase inhibitors (HDACis) promotes apoptosis of leukemia cells obtained from TAL1 transgenic mice. Here, we show for the first time that TAL1 protein expression is strikingly downregulated upon histone deacetylase inhibition in T-ALL cells. This is due to decreased TAL1 gene transcription in cells with native TAL1 promoter, and due to impaired TAL1 mRNA translation in cells that harbor the TAL1(d) microdeletion and consequently express TAL1 under the control of the SCL/TAL1 interrupting locus (SIL) promoter. Notably, HDACi-triggered apoptosis of T-ALL cells is significantly reversed by TAL1 forced overexpression. Our results indicate that the HDACi-mediated apoptotic program in T-ALL cells is partially dependent on their capacity to downregulate TAL1 and provide support for the therapeutic use of HDACi in T-ALL. Leukemia (2011) 25, 1578-1586; doi:10.1038/leu.2011.140; published online 7 June 2011
- Published
- 2011
- Full Text
- View/download PDF
46. Therapeutic potential of Notch inhibition in T-cell acute lymphoblastic leukemia: rationale, caveats and promises
- Author
-
Leonor M. Sarmento and João T. Barata
- Subjects
Poor prognosis ,business.industry ,Lymphoblastic Leukemia ,T cell ,Notch signaling pathway ,Antineoplastic Agents ,Pharmacology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Malignancy ,medicine.disease ,Molecular targeting ,medicine.anatomical_structure ,Oncology ,Intestinal toxicity ,Cancer research ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Molecular Targeted Therapy ,Amyloid Precursor Protein Secretases ,Enzyme Inhibitors ,Receptor, Notch1 ,business - Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a malignancy that presents with poor prognosis. Treatment relies on the application of aggressive therapies that produce deleterious side-effects, justifying the quest for novel, more efficient and selective molecular targeting agents. Mutations leading to abnormal Notch-1 activity are present in more than half of the T-ALL patients, underscoring the potential therapeutic relevance of targeting Notch-1 inhibition and further reinforcing the need to better comprehend the mechanisms by which Notch-1 drives T cell leukemogenesis. Clinical application of γ-secretase inhibitors to block Notch signaling in T-ALL revealed new challenges that involve improvement of the therapeutic benefit and reduction of intestinal toxicity. Here, we review the latest advances in the development and use of Notch antagonists and summarize the current knowledge on Notch function in T-ALL to understand how it may translate into novel therapeutic strategies that increment the efficiency of Notch inhibition.
- Published
- 2011
- Full Text
- View/download PDF
47. Intracellular reactive oxygen species are essential for PI3K/Akt/mTOR-dependent IL-7-mediated viability of T-cell acute lymphoblastic leukemia cells
- Author
-
Cristina Santos, Fernando Antunes, Inês Cebola, João T. Barata, Ashiley Sarmento da Silva, and Ana Girio
- Subjects
Cancer Research ,Cell Survival ,Glucose uptake ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Phosphatidylinositol 3-Kinases ,Downregulation and upregulation ,Tumor Cells, Cultured ,Humans ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Akt/PKB signaling pathway ,Interleukin-7 ,TOR Serine-Threonine Kinases ,Glucose transporter ,Receptor Cross-Talk ,Hematology ,Up-Regulation ,Cell biology ,Mitochondrial respiratory chain ,Oncology ,Biochemistry ,biology.protein ,GLUT1 ,Reactive Oxygen Species ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Interleukin-7 (IL-7) activates phosphoinositide 3-kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway, thereby mediating viability, proliferation and growth of T-cell acute lymphoblastic leukemia (T-ALL) cells. Reactive oxygen species (ROS) can be upregulated by growth factors and are known to regulate proliferation and viability. Here, we show that IL-7 upregulates ROS in T-ALL cells in a manner that is dependent on PI3K/Akt/mTOR pathway activity and that relies on both NADPH oxidase and mitochondrial respiratory chain. Conversely, IL-7-induced activation of PI3K signaling pathway requires mitochondrial respiration and ROS. We have previously shown that IL-7-mediated activation of PI3K pathway drives the upregulation of the glucose transporter Glut1, promoting glucose uptake in T-ALL cells. Using phloretin to inhibit Glut function, we demonstrate that glucose uptake is mandatory for ROS upregulation in IL-7-treated T-ALL cells, suggesting that IL-7 stimulation leads to increased ROS via PI3K pathway activation and consequent upregulation of Glut1 and glucose uptake. Overall, our data reveal the existence of a critical crosstalk between PI3K/Akt signaling pathway and ROS that is essential for IL-7-mediated T-ALL cell survival, and that may constitute a novel target for therapeutic intervention.
- Published
- 2011
- Full Text
- View/download PDF
48. The impact of PTEN regulation by CK2 on PI3K-dependent signaling and leukemia cell survival
- Author
-
João T. Barata
- Subjects
Cancer Research ,Cell Survival ,medicine.disease_cause ,law.invention ,Phosphatidylinositol 3-Kinases ,law ,Neoplasms ,Genetics ,medicine ,Animals ,Humans ,PTEN ,Casein Kinase II ,Molecular Biology ,PI3K/AKT/mTOR pathway ,Mutation ,Leukemia ,biology ,Mechanism (biology) ,PTEN Phosphohydrolase ,Cancer ,medicine.disease ,Cancer research ,biology.protein ,Molecular Medicine ,Phosphorylation ,Suppressor ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Gene alterations affecting elements of PI3K signaling pathway do not appear to be sufficient to explain the extremely high frequency of PI3K signaling hyperactivation in leukemia. It has been known for long that PTEN phosphorylation at the C-terminal tail, in particular by CK2, contributes to the stabilization and simultaneous inhibition of this critical tumor suppressor. However, direct evidence of the involvement of this mechanism in cancer has been gathered only recently. It is now known that CK2-mediated posttranslational, non-deleting, inactivation of PTEN occurs in T-ALL, CLL and probably other leukemias and solid tumors. To explore this knowledge for therapeutic purposes remains one of the challenges ahead.
- Published
- 2011
- Full Text
- View/download PDF
49. IL-7 Either Promotes or Inhibits Autophagy, Depending on the Stress Context, to Promote the Viability of T-Cell Acute Lymphoblastic Leukemia Cells
- Author
-
Inês P Lopes, Daniel Dias Ribeiro, Carlos Custódia, Marta Abreu, and João T. Barata
- Subjects
MAPK/ERK pathway ,Programmed cell death ,Cell growth ,Chemistry ,Immunology ,Autophagy ,Cell Biology ,Hematology ,Biochemistry ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,030212 general & internal medicine ,Viability assay ,Signal transduction ,Protein kinase B ,PI3K/AKT/mTOR pathway - Abstract
T-cell acute lymphoblastic leukemia (T-ALL), an aggressive and common childhood hematological malignancy, arises from clonal expansion of T-cell progenitors. Autophagy is a homeostatic process characterized by the sequestration of cytoplasmic compartments within double-membrane vesicles (autophagosomes) to promote their degradation. Importantly, autophagy is upregulated during starvation and cellular stress, as a compensatory mechanism to provide nutrients and stress relief. By mitigating stress and allowing cell survival, autophagy may serve as a pro-tumoral mechanism. On the other hand, persistent autophagy can lead to cell death and thereby prevent tumor growth. Interleukin-7 (IL-7), a cytokine produced by the bone marrow and thymic stroma, is essential for normal T-cell development. However, IL-7-mediated signaling can also contribute to leukemogenesis. A majority of T-ALL patients (~70%) expresses the IL-7 receptor and IL-7 accelerates T-ALL progression in vivo and promotes T-ALL cell proliferation, survival and metabolic activation in vitro via PI3K/Akt/mTOR pathway (a master negative regulator of autophagy). IL-7 can also activate MEK/Erk pathway (which has been implicated in promotion of autophagy). Because IL-7 has the ability to activate signaling pathways with potentially opposing roles in autophagy regulation, we explored whether IL-7 impacted on the autophagic process in T-ALL cells and sought to elucidate the molecular mechanisms and functional consequences of IL-7-mediated autophagy regulation under different culture conditions: a) medium with 10% serum - a scenario where cells have optimal growth conditions, b) medium with low (1%) serum - to mimic a milieu with nutrient stress (which may happen in densely populated leukemic niches in vivo), and c) regular medium plus asparaginase (ASNase) - to induce treatment-imposed stress. Using IL-7-responsive T-ALL cell lines and patient-derived xenograft (PDX) samples, we show that, in optimal culture conditions, IL-7 leads to a decrease in LC3-I/-II conversion and a reduction in both LC3 puncta and autophagosome/autolysosome formation, as determined by immunoblot, confocal microscopy, flow cytometry and electron microscopy - indicating that IL-7 inhibits autophagy in T-ALL. Using signaling-specific small molecule inhibitors (UO126 for MEK/Erk; LY294002 and rapamycin for PI3K/Akt/mTOR) we found that IL-7-mediated regulation of autophagy occurs in a complex manner that involves concomitant triggering of both pro- (via MEK/Erk) and anti- (via PI3K/Akt/mTOR) autophagic signaling, with the effects of the latter prevailing over the former. In this scenario, IL-7-mediated viability relies on PI3K/Akt/mTOR pathway and, as expected, autophagy inhibition (using MRT68921) does not prevent the ability of IL-7 to promote leukemia cell survival. In contrast, under serum starvation IL-7 promotes autophagy, and IL-7-mediated leukemia cell viability partially relies on autophagy activation, and strictly requires MEK/Erk activation. Mechanistically, we provide evidence that depending on the culture conditions, IL-7 can balance the relative activation of PI3K/Akt/mTOR and MEK/Erk pathways, inhibiting or facilitating autophagy, in order to consistently promote T-ALL cell viability. We further extended our studies to a therapy-related scenario and found that under ASNase treatment, IL-7 still promotes increased survival of leukemic cells, a possible mechanism of treatment resistance. Functionally, we demonstrate that the IL-7-mediated increase in survival under ASNase treatment is, in part, mediated via activation of autophagy, suggesting that combining ASNase administration with autophagy inhibitors may be an attractive strategy to prevent resistance. In summary, our results indicate that IL-7 makes use of a 'flexible strategy' to promote T-ALL cell viability by activating both pro- and anti-autophagic pathways, which are differentially recruited, depending on the microenvironmental conditions, to prevent tumor cell death. Moreover, our findings strengthen the notion that combination therapies against PI3K/Akt/mTOR and MEK/Erk pathways may constitute a valid therapeutic avenue and highlight the potential of using autophagy inhibitors to prevent microenvironment-induced chemotherapy resistance in T-ALL. Disclosures Barata: Instituto de Medicina Molecular João Lobo Antunes: Patents & Royalties: Patents.
- Published
- 2018
- Full Text
- View/download PDF
50. IL-7 Activates a STAT5/PIM1 Axis to Promote T-Cell Acute Lymphoblastic Leukemia Proliferation and Viability in a Bcl-2-Independent Manner
- Author
-
Ruben van Boxtel, Ribeiro Daniel, Alice Melão, Ana Elisa Bauer de Camargo Silva, Milene Costa da Silva, Bruno A. Cardoso, João T. Barata, Paul J. Coffer, and Cristina Santos
- Subjects
biology ,Cell growth ,Immunology ,Cyclin A ,PIM1 ,Cell Biology ,Hematology ,Cell cycle ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,hemic and lymphatic diseases ,biology.protein ,Cancer research ,030212 general & internal medicine ,Signal transduction ,Protein kinase B ,PI3K/AKT/mTOR pathway - Abstract
Background: T-cell acute lymphoblastic leukemia (T-ALL) constitutes an aggressive subset of ALL, the most frequent childhood malignancy. Although risk-adjusted chemotherapeutic regimens are currently extremely effective, their efficacy is associated with significant long-term side effects. Moreover, a significant fraction of the patients still relapse despite intensive chemotherapy, prompting the need for a deeper understanding of T-ALL biology in order to develop novel therapies. Interleukin-7 is a cytokine essential for normal T-cell development, where it has a pivotal role in promoting thymocyte survival via Bcl-2 upregulation. In this normal setting, Bcl-2 is under the control of STAT5 mediated signaling. Previously, we have shown that IL-7 promotes T-ALL expansion in vivo and leukemia cell survival and proliferation in vitro by activating PI3K/Akt/mTOR signaling pathway, consequently leading to p27kip1 downregulation and Bcl-2 upregulation. However, it is also known that T-cell lymphomas arising spontaneously in IL-7 transgenic mice depend on STAT5 activity and that leukemias displaying IL7R gain-of-function mutations are sensitive to JAK and STAT5 inhibitors. Thus, we investigated whether STAT5 could also be involved in the IL-7 pro-leukemia effects in human T-ALL cells. Methods: We used an IL-7-dependent leukemia T-cell line (TAIL7), an IL-7-responsive T-ALL cell line (HPB-ALL, with or without shRNA-mediated STAT5 silencing), primary T-ALL samples collected at diagnosis and patient-derived xenografts (PDX) and treated them with inhibitors of STAT5 (N-((4-Oxo-4H-chromen-3-yl) methylene) nicotinohydrazide) and PIM (AZD1208). Analysis of viability, cell size, cell cycle, surface CD71 and Bcl-2 expression was performed by flow cytometry. Signaling pathway activation, STAT5, PIM1, Bcl-xL, Mcl-1 and cell cycle protein expression was performed by immunoblot analysis. Proliferation was assessed by 3H-Thymidine incorporation. STAT5 ChIP-seq and RNA-seq were performed on TAIL7 cells. ChIP-PCR of histone marks H3K4me3, H3K27me, H3K27ac was performed in TAIL7 and HPB-ALL. Results: IL-7 induces JAK/STAT5 pathway activation in T-ALL cells and STAT5 genetic or pharmacological inactivation prevents IL-7-mediated T-ALL cell viability, growth and proliferation. At the molecular level, STAT5 is required for IL-7-induced downregulation of p27kip1, and upregulation of Cyclin A and TfR/CD71. Surprisingly, STAT5 inhibition does not significantly affect IL-7-mediated Bcl-2 upregulation, suggesting that, contrary to normal T-cells, STAT5 promotes leukemia cell survival via a Bcl-2-independent mechanism. In addition, IL-7-mediated increase in transcription of BCL2, BCL2L1 (Bcl-xL) and MCL1 is not affected by STAT5 silencing. To understand how STAT5 mediates the survival effects of IL-7 in T-ALL cells without affecting BCL2 transcription, we performed STAT5 ChIP-seq together with RNA-seq. Data cross-analysis reveal a diverse IL-7-driven STAT5-dependent transcriptional program in T-ALL cells, which includes transcription of the serine/threonine kinase PIM1. PIM1 is involved in cell cycle regulation and apoptosis, thereby constituting a possible alternative to Bcl-2-dependent prevention of apoptosis. We show that STAT5 silencing prevents IL-7-mediated PIM1 expression and the upregulation of active chromatin marks, H3K4me3 and H3K27ac, at the STAT5 binding region in the PIM1 gene. Notably, pharmacological inhibition of PIM kinase abrogates IL-7-mediated T-ALL cell growth and proliferation, however, without affecting cell survival. In agreement, PIM inhibition does not affect expression of Bcl-2 or Bcl-2 family anti-apoptotic members Bcl-xL and MCL1. Conclusion: Here we present evidence that T-ALL cells may have an alternative wiring of signaling networks downstream of IL-7 to that present in normal T-cells. In contrast to healthy lymphoid cells, IL-7-mediated control of survival of T-ALL cells via STAT5 does not rely on modulation of Bcl-2. Moreover, exploration of STAT5 downstream signaling reveals that PIM1 is required for IL-7-mediated proliferation of human T-ALL cells, indicating that strategies involving the use of PIM kinase small molecule inhibitors may have therapeutic potential against leukemias that rely on IL-7R and STAT5 signaling. Disclosures Barata: Instituto de Medicina Molecular João Lobo Antunes: Patents & Royalties: Patents.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.