1
|
Tremblay CS, Saw J, Yan F, Boyle JA, Amarasinghe O, Abdollahi S, Vo ANQ, Shields BJ, Mayoh C, McCalmont H, Evans K, Steiner A, Parsons K, McCormack MP, Powell DR, Wong NC, Jane SM, Lock RB, Curtis DJ. Targeting LMO2-induced autocrine FLT3 signaling to overcome chemoresistance in early T-cell precursor acute lymphoblastic leukemia. Leukemia 2025; 39:577-589. [PMID: 39849166 PMCID: PMC11879882 DOI: 10.1038/s41375-024-02491-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 11/25/2024] [Accepted: 11/29/2024] [Indexed: 01/25/2025]
Abstract
Early T-cell Precursor Acute Lymphoblastic Leukemia (ETP-ALL) is an immature subtype of T-cell acute lymphoblastic leukemia (T-ALL) commonly show deregulation of the LMO2-LYL1 stem cell transcription factors, activating mutations of cytokine receptor signaling, and poor early response to intensive chemotherapy. Previously, studies of the Lmo2 transgenic mouse model of ETP-ALL identified a population of stem-like T-cell progenitors with long-term self-renewal capacity and intrinsic chemotherapy resistance linked to cellular quiescence. Here, analyses of Lmo2 transgenic mice, patient-derived xenografts, and single-cell RNA-sequencing data from primary ETP-ALL identified a rare subpopulation of leukemic stem cells expressing high levels of the cytokine receptor FLT3. Despite a highly proliferative state, these FLT3-overexpressing cells had long-term self-renewal capacity and almost complete resistance to chemotherapy. Chromatin immunoprecipitation and assay for transposase-accessible chromatin sequencing demonstrated FLT3 and its ligand may be direct targets of the LMO2 stem-cell complex. Media conditioned by Lmo2 transgenic thymocytes revealed an autocrine FLT3-dependent signaling loop that could be targeted by the FLT3 inhibitor gilteritinib. Consequently, gilteritinib impaired in vivo growth of ETP-ALL and improved the sensitivity to chemotherapy. Furthermore, gilteritinib enhanced response to the BCL2 inhibitor venetoclax, which may enable "chemo-free" treatment of ETP-ALL. Together, these data provide a cellular and molecular explanation for enhanced cytokine signaling in LMO2-driven ETP-ALL beyond activating mutations and a rationale for clinical trials of FLT3 inhibitors in ETP-ALL.
Collapse
Affiliation(s)
- Cedric S Tremblay
- Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.
- Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, MB, Canada.
- Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB, Canada.
- Australian Centre for Blood Diseases (ACBD), School of Translational Medicine, Monash University, Melbourne, VIC, Australia.
| | - Jesslyn Saw
- Australian Centre for Blood Diseases (ACBD), School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Feng Yan
- Australian Centre for Blood Diseases (ACBD), School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Bioinformatics Division, Walter and Eliza Hall Institute (WEHI) of Medical Research, Parkville, VIC, Australia
| | - Jacqueline A Boyle
- Australian Centre for Blood Diseases (ACBD), School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Ovini Amarasinghe
- Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
- Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, MB, Canada
| | - Shokoufeh Abdollahi
- Australian Centre for Blood Diseases (ACBD), School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Anh N Q Vo
- Australian Centre for Blood Diseases (ACBD), School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Benjamin J Shields
- Australian Centre for Blood Diseases (ACBD), School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Chelsea Mayoh
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Hannah McCalmont
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Kathryn Evans
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - Anna Steiner
- Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, MB, Canada
- Community and Researcher Engagement (CaRE) program, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Women in Lymphoma, Lymphoma Australia, Brisbane, QLD, Australia
| | - Kevin Parsons
- Community and Researcher Engagement (CaRE) program, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Matthew P McCormack
- Australian Centre for Blood Diseases (ACBD), School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - David R Powell
- Monash Bioinformatics Platform, Monash University, Clayton, VIC, Australia
| | - Nicholas C Wong
- Australian Centre for Blood Diseases (ACBD), School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Monash Bioinformatics Platform, Monash University, Clayton, VIC, Australia
| | - Stephen M Jane
- Department of Medicine, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Richard B Lock
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, UNSW Medicine & Health, UNSW Centre for Childhood Cancer Research, UNSW Sydney, Sydney, NSW, Australia
| | - David J Curtis
- Australian Centre for Blood Diseases (ACBD), School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Department of Clinical Haematology, Alfred Hospital, Prahran, VIC, Australia
| |
Collapse
|
2
|
De Coninck S, Roels J, Lintermans B, T’Sas S, Taghon T, Curtis DJ, Pieters T, Goossens S, Van Vlierberghe P. Tet2 is a tumor suppressor in the preleukemic phase of T-cell acute lymphoblastic leukemia. Blood Adv 2024; 8:2646-2649. [PMID: 38536906 PMCID: PMC11157202 DOI: 10.1182/bloodadvances.2023011970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/28/2024] [Indexed: 05/30/2024] Open
Affiliation(s)
- Stien De Coninck
- Department of Biomolecular Medicine, Laboratory of Normal and Malignant Hematopoiesis, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Diagnostic Sciences, Unit for Translational Research in Oncology, Ghent University, Ghent, Belgium
| | - Juliette Roels
- Department of Biomolecular Medicine, Laboratory of Normal and Malignant Hematopoiesis, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Diagnostic Sciences, Taghon Laboratory, Ghent University, Ghent, Belgium
| | - Béatrice Lintermans
- Department of Biomolecular Medicine, Laboratory of Normal and Malignant Hematopoiesis, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Sara T’Sas
- Department of Biomolecular Medicine, Laboratory of Normal and Malignant Hematopoiesis, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Diagnostic Sciences, Unit for Translational Research in Oncology, Ghent University, Ghent, Belgium
| | - Tom Taghon
- Department of Diagnostic Sciences, Taghon Laboratory, Ghent University, Ghent, Belgium
| | - David J. Curtis
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Clinical Haematology, Alfred Hospital, Prahran, VIC, Australia
| | - Tim Pieters
- Department of Biomolecular Medicine, Laboratory of Normal and Malignant Hematopoiesis, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Diagnostic Sciences, Unit for Translational Research in Oncology, Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Laboratory for research in oncogenesis and resistance to therapy, Ghent University, Ghent, Belgium
| | - Steven Goossens
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Diagnostic Sciences, Unit for Translational Research in Oncology, Ghent University, Ghent, Belgium
| | - Pieter Van Vlierberghe
- Department of Biomolecular Medicine, Laboratory of Normal and Malignant Hematopoiesis, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
3
|
Tremblay CS, Ting SB, McCluskey A, Robinson PJ, Curtis DJ. Shutting the gate: targeting endocytosis in acute leukemia. Exp Hematol 2021; 104:17-31. [PMID: 34563604 DOI: 10.1016/j.exphem.2021.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/02/2021] [Accepted: 09/15/2021] [Indexed: 10/20/2022]
Abstract
Endocytosis entails selective packaging of cell surface cargos in cytoplasmic vesicles, thereby controlling key intrinsic cellular processes as well as the response of normal and malignant cells to their microenvironment. The purpose of this review is to outline the latest advances in the development of endocytosis-targeting therapeutic strategies in hematological malignancies.
Collapse
Affiliation(s)
- Cedric S Tremblay
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| | - Stephen B Ting
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Clinical Haematology, Eastern Health, Box Hill, Victoria, Australia; Department of Clinical Haematology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Adam McCluskey
- Chemistry, Centre for Chemical Biology, School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales, Australia
| | - Phillip J Robinson
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, New South Wales, Australia; Cell Signalling Unit, Children's Medical Research Institute, Sydney, New South Wales, Australia
| | - David J Curtis
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Clinical Haematology, Alfred Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
4
|
Zhang L, Wu J, Feng Y, Khadka B, Fang Z, Gu J, Tang B, Xiao R, Pan G, Liu JJ. A Regulatory Loop Involving Notch and Wnt Signaling Maintains Leukemia Stem Cells in T-Cell Acute Lymphoblastic Leukemia. Front Cell Dev Biol 2021; 9:678544. [PMID: 34179007 PMCID: PMC8226090 DOI: 10.3389/fcell.2021.678544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/12/2021] [Indexed: 11/13/2022] Open
Abstract
Leukemia-initiating cells play critical role in relapse, resistance to therapies and metastases but the mechanism remains largely elusive. We report that β-catenin is over-expressed in almost all T-ALL patients and flow sorted β-cateninhigh fractions are highly resistant to therapy, leading to liver metastases in nude mice as well as dysregulated lncRNAs. Pharmacological inhibition through XAV-939 as well as si-RNA mediated inhibition of β-catenin is initially effective in re-sensitization to therapy, however, prolonged inhibition shifts dependency from β-catenin to Notch signaling, with particularly high levels of receptors Notch 1 and Notch 2. The results are verifiable in a cohort of T-ALL patients comprising of responders vs. those who have progressed, with β-catenin, Notch 1 and Notch 2 elevated in progressed patients. Further, in patients-derived cells, silencing of Notch 1 or Notch 2 does not counter resistance to β-catenin inhibition, rather pharmacological pan-Notch inhibition is needed to overcome resistance and its effect on in vitro tumor sphere formations as well as in vivo liver metastases. Thus, wnt and Notch signaling are part of a regulatory loop mutually compensating for each other in T-ALL, while ensuring the maintenance of stem cell phenotype.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Hematology and Hematology Institute of Sun Yat-sen University, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jieying Wu
- Department of Hematology and Hematology Institute of Sun Yat-sen University, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yashu Feng
- Department of Hematology and Hematology Institute of Sun Yat-sen University, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bijay Khadka
- Department of Hematology and Hematology Institute of Sun Yat-sen University, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhigang Fang
- Department of Hematology and Hematology Institute of Sun Yat-sen University, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiaming Gu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Baoqiang Tang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ruozhi Xiao
- Department of Hematology and Hematology Institute of Sun Yat-sen University, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Guangjin Pan
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Jia-Jun Liu
- Department of Hematology and Hematology Institute of Sun Yat-sen University, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
5
|
Nguyen TL, Nokin MJ, Terés S, Tomé M, Bodineau C, Galmar O, Pasquet JM, Rousseau B, van Liempd S, Falcon-Perez JM, Richard E, Muzotte E, Rezvani HR, Priault M, Bouchecareilh M, Redonnet-Vernhet I, Calvo J, Uzan B, Pflumio F, Fuentes P, Toribio ML, Khatib AM, Soubeyran P, Murdoch PDS, Durán RV. Downregulation of Glutamine Synthetase, not glutaminolysis, is responsible for glutamine addiction in Notch1-driven acute lymphoblastic leukemia. Mol Oncol 2021; 15:1412-1431. [PMID: 33314742 PMCID: PMC8096784 DOI: 10.1002/1878-0261.12877] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/21/2020] [Accepted: 12/09/2020] [Indexed: 01/03/2023] Open
Abstract
The cellular receptor Notch1 is a central regulator of T-cell development, and as a consequence, Notch1 pathway appears upregulated in > 65% of the cases of T-cell acute lymphoblastic leukemia (T-ALL). However, strategies targeting Notch1 signaling render only modest results in the clinic due to treatment resistance and severe side effects. While many investigations reported the different aspects of tumor cell growth and leukemia progression controlled by Notch1, less is known regarding the modifications of cellular metabolism induced by Notch1 upregulation in T-ALL. Previously, glutaminolysis inhibition has been proposed to synergize with anti-Notch therapies in T-ALL models. In this work, we report that Notch1 upregulation in T-ALL induced a change in the metabolism of the important amino acid glutamine, preventing glutamine synthesis through the downregulation of glutamine synthetase (GS). Downregulation of GS was responsible for glutamine addiction in Notch1-driven T-ALL both in vitro and in vivo. Our results also confirmed an increase in glutaminolysis mediated by Notch1. Increased glutaminolysis resulted in the activation of the mammalian target of rapamycin complex 1 (mTORC1) pathway, a central controller of cell growth. However, glutaminolysis did not play any role in Notch1-induced glutamine addiction. Finally, the combined treatment targeting mTORC1 and limiting glutamine availability had a synergistic effect to induce apoptosis and to prevent Notch1-driven leukemia progression. Our results placed glutamine limitation and mTORC1 inhibition as a potential therapy against Notch1-driven leukemia.
Collapse
Affiliation(s)
- Tra Ly Nguyen
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, Pessac, France
| | - Marie-Julie Nokin
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, Pessac, France
| | - Silvia Terés
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, Pessac, France
| | - Mercedes Tomé
- Centro Andaluz de Biología Molecular y Medicina Regenerativa - CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Seville, Spain.,Angiogenesis and Cancer Microenvironment Laboratory INSERM U1029, Université de Bordeaux, Pessac, France
| | - Clément Bodineau
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, Pessac, France.,Centro Andaluz de Biología Molecular y Medicina Regenerativa - CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Seville, Spain
| | - Oriane Galmar
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, Pessac, France
| | | | - Benoit Rousseau
- Service Commun des Animaleries, University of Bordeaux, France
| | - Sebastian van Liempd
- Exosomes Laboratory and Platform of Metabolomics, CIC bioGUNE, CIBERehd, Derio, Spain
| | - Juan Manuel Falcon-Perez
- Exosomes Laboratory and Platform of Metabolomics, CIC bioGUNE, CIBERehd, Derio, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Elodie Richard
- Institut Bergonié, INSERM U1218, University of Bordeaux, France
| | | | | | - Muriel Priault
- Institut de Biochimie et Génétique Cellulaires, CNRS UMR 5095, Université de Bordeaux, France
| | - Marion Bouchecareilh
- Bordeaux Research in Translational Oncology, INSERM U1053, Université de Bordeaux, France
| | - Isabelle Redonnet-Vernhet
- Maladies Héréditaires du Métabolisme, Laboratoire de Biochimie, Hôpital Pellegrin, CHU Bordeaux, France
| | - Julien Calvo
- UMR967, Inserm, CEA, Université Paris 7, Université Paris 11, Fontenay-aux-Roses, France
| | - Benjamin Uzan
- UMR967, Inserm, CEA, Université Paris 7, Université Paris 11, Fontenay-aux-Roses, France
| | - Françoise Pflumio
- UMR967, Inserm, CEA, Université Paris 7, Université Paris 11, Fontenay-aux-Roses, France
| | - Patricia Fuentes
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Spain
| | - Maria L Toribio
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Spain
| | - Abdel-Majid Khatib
- Angiogenesis and Cancer Microenvironment Laboratory INSERM U1029, Université de Bordeaux, Pessac, France
| | | | - Piedad Del Socorro Murdoch
- Centro Andaluz de Biología Molecular y Medicina Regenerativa - CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Seville, Spain.,Departamento de Bioquímica Vegetal y Biología Molecular, Universidad de Sevilla, Spain
| | - Raúl V Durán
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, Pessac, France.,Centro Andaluz de Biología Molecular y Medicina Regenerativa - CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Seville, Spain
| |
Collapse
|
6
|
Rodrigues GOL, Cramer SD, Winer HY, Hixon JA, Li W, Yunes JA, Durum SK. Mutations that collaborate with IL-7Ra signaling pathways to drive ALL. Adv Biol Regul 2021; 80:100788. [PMID: 33578108 DOI: 10.1016/j.jbior.2021.100788] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/11/2021] [Indexed: 12/30/2022]
Abstract
The IL-7 pathway is required for normal T cell development and survival. In recent years the pathway has been shown to be a major driver of acute lymphoblastic leukemia (ALL), the most common cancer in children. Gain-of-function mutations in the alpha chain of the IL-7 receptor found in ALL patients clearly demonstrated that this pathway was a driver. However mutant IL-7R alone was insufficient to transform primary T cell progenitors, indicating that cooperating mutations were required. Here we review evidence for additional oncogenic mutations in the IL-7 pathway. We discuss several oncogenes, loss of tumor suppressor genes and epigenetic effects that can cooperate with mutant IL-7 receptor. These include NRas, HOXA, TLX3, Notch 1, Arf, PHF6, WT1, PRC, PTPN2 and CK2. As new therapeutics targeting the IL-7 pathway are developed, combination with agents directed to cooperating pathways offer hope for novel therapies for ALL.
Collapse
Affiliation(s)
- Gisele O L Rodrigues
- Cytokines and Immunity Section, Laboratory of Cancer Immunometabolism, National Cancer Institute, National Institutes of Health (NIH), Frederick, MD, USA; Molecular Biology Laboratory, Boldrini Children's Center, Campinas, Brazil; Department of Genetics, Evolution and Bioagents, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Sarah D Cramer
- Cytokines and Immunity Section, Laboratory of Cancer Immunometabolism, National Cancer Institute, National Institutes of Health (NIH), Frederick, MD, USA; Comparative Biomedical Scientist Training Program, NIH, Bethesda, MD, USA; Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Hila Y Winer
- Cytokines and Immunity Section, Laboratory of Cancer Immunometabolism, National Cancer Institute, National Institutes of Health (NIH), Frederick, MD, USA
| | - Julie A Hixon
- Cytokines and Immunity Section, Laboratory of Cancer Immunometabolism, National Cancer Institute, National Institutes of Health (NIH), Frederick, MD, USA
| | - WenQing Li
- Cytokines and Immunity Section, Laboratory of Cancer Immunometabolism, National Cancer Institute, National Institutes of Health (NIH), Frederick, MD, USA
| | - José Andres Yunes
- Department of Genetics, Evolution and Bioagents, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Scott K Durum
- Cytokines and Immunity Section, Laboratory of Cancer Immunometabolism, National Cancer Institute, National Institutes of Health (NIH), Frederick, MD, USA.
| |
Collapse
|
7
|
Tremblay CS, Chiu SK, Saw J, McCalmont H, Litalien V, Boyle J, Sonderegger SE, Chau N, Evans K, Cerruti L, Salmon JM, McCluskey A, Lock RB, Robinson PJ, Jane SM, Curtis DJ. Small molecule inhibition of Dynamin-dependent endocytosis targets multiple niche signals and impairs leukemia stem cells. Nat Commun 2020; 11:6211. [PMID: 33277497 PMCID: PMC7719179 DOI: 10.1038/s41467-020-20091-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 11/09/2020] [Indexed: 02/07/2023] Open
Abstract
Intensive chemotherapy for acute leukemia can usually induce complete remission, but fails in many patients to eradicate the leukemia stem cells responsible for relapse. There is accumulating evidence that these relapse-inducing cells are maintained and protected by signals provided by the microenvironment. Thus, inhibition of niche signals is a proposed strategy to target leukemia stem cells but this requires knowledge of the critical signals and may be subject to compensatory mechanisms. Signals from the niche require receptor-mediated endocytosis, a generic process dependent on the Dynamin family of large GTPases. Here, we show that Dynole 34-2, a potent inhibitor of Dynamin GTPase activity, can block transduction of key signalling pathways and overcome chemoresistance of leukemia stem cells. Our results provide a significant conceptual advance in therapeutic strategies for acute leukemia that may be applicable to other malignancies in which signals from the niche are involved in disease progression and chemoresistance. The tumour microenvironment provides signals to support leukaemic stem cells (LSC) maintenance and chemoresistance. Here, the authors show that disrupting niche-associated signalling by inhibiting receptor-mediated endocytosis with a dynamin GTPase inhibitor overcomes chemoresistance of LSC.
Collapse
Affiliation(s)
- Cedric S Tremblay
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia.
| | - Sung Kai Chiu
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Clinical Haematology, Alfred Health, Melbourne, VIC, Australia
| | - Jesslyn Saw
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Hannah McCalmont
- Lowy Cancer Research Centre, Children's Cancer Institute, University of New South Wales, Sydney, NSW, Australia
| | - Veronique Litalien
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Jacqueline Boyle
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Stefan E Sonderegger
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Ngoc Chau
- Cell Signalling Unit, Children's Medical Research Institute, Sydney, NSW, Australia
| | - Kathryn Evans
- Lowy Cancer Research Centre, Children's Cancer Institute, University of New South Wales, Sydney, NSW, Australia
| | - Loretta Cerruti
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Jessica M Salmon
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Adam McCluskey
- Chemistry, Centre for Chemical Biology, School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| | - Richard B Lock
- Lowy Cancer Research Centre, Children's Cancer Institute, University of New South Wales, Sydney, NSW, Australia
| | - Phillip J Robinson
- Cell Signalling Unit, Children's Medical Research Institute, Sydney, NSW, Australia
| | - Stephen M Jane
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Clinical Haematology, Alfred Health, Melbourne, VIC, Australia
| | - David J Curtis
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Clinical Haematology, Alfred Health, Melbourne, VIC, Australia
| |
Collapse
|
8
|
Milton-Harris L, Jeeves M, Walker SA, Ward SE, Mancini EJ. Small molecule inhibits T-cell acute lymphoblastic leukaemia oncogenic interaction through conformational modulation of LMO2. Oncotarget 2020; 11:1737-1748. [PMID: 32477463 PMCID: PMC7233811 DOI: 10.18632/oncotarget.27580] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 04/03/2020] [Indexed: 01/05/2023] Open
Abstract
Ectopic expression in T-cell precursors of LIM only protein 2 (LMO2), a key factor in hematopoietic development, has been linked to the onset of T-cell acute lymphoblastic leukaemia (T-ALL). In the T-ALL context, LMO2 drives oncogenic progression through binding to erythroid-specific transcription factor SCL/TAL1 and sequestration of E-protein transcription factors, normally required for T-cell differentiation. A key requirement for the formation of this oncogenic protein-protein interaction (PPI) is the conformational flexibility of LMO2. Here we identify a small molecule inhibitor of the SCL-LMO2 PPI, which hinders the interaction in vitro through direct binding to LMO2. Biophysical analysis demonstrates that this inhibitor acts through a mechanism of conformational modulation of LMO2. Importantly, this work has led to the identification of a small molecule inhibitor of the SCL-LMO2 PPI, which can provide a starting point for the development of new agents for the treatment of T-ALL. These results suggest that similar approaches, based on the modulation of protein conformation by small molecules, might be used for therapeutic targeting of other oncogenic PPIs.
Collapse
Affiliation(s)
- Leanne Milton-Harris
- School of Life Sciences, Biochemistry Department, University of Sussex, Falmer, Brighton, BN1 9QG, United Kingdom
| | - Mark Jeeves
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom
| | - Sarah A Walker
- Sussex Drug Discovery Centre, University of Sussex, Brighton, BN1 9QJ, United Kingdom
| | - Simon E Ward
- Medicines Discovery Institute, Cardiff University, Park Place, Cardiff, CF10 3AT, United Kingdom
| | - Erika J Mancini
- School of Life Sciences, Biochemistry Department, University of Sussex, Falmer, Brighton, BN1 9QG, United Kingdom
| |
Collapse
|
9
|
Pais H, Ruggero K, Zhang J, Al-Assar O, Bery N, Bhuller R, Weston V, Kearns PR, Mecucci C, Miller A, Rabbitts TH. Surfaceome interrogation using an RNA-seq approach highlights leukemia initiating cell biomarkers in an LMO2 T cell transgenic model. Sci Rep 2019; 9:5760. [PMID: 30962539 PMCID: PMC6453905 DOI: 10.1038/s41598-019-42214-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/27/2019] [Indexed: 12/27/2022] Open
Abstract
The surfaceome is critical because surface proteins provide a gateway for internal signals and transfer of molecules into cells, and surfaceome differences can influence therapy response. We have used a surfaceome analysis method, based on comparing RNA-seq data between normal and abnormal cells (Surfaceome DataBase Mining or Surfaceome DBM), to identify sets of upregulated cell surface protein mRNAs in an LMO2-mediated T-ALL mouse model and corroborated by protein detection using antibodies. In this model the leukemia initiating cells (LICs) comprise pre-leukaemic, differentiation inhibited thymocytes allowing us to provide a profile of the LIC surfaceome in which GPR56, CD53 and CD59a are co-expressed with CD25. Implementation of cell surface interaction assays demonstrates fluid interaction of surface proteins and CD25 is only internalized when co-localized with other proteins. The Surfaceome DBM approach to analyse cancer cell surfaceomes is a way to find targetable surface biomarkers for clinical conditions where RNA-seq data from normal and abnormal cell are available.
Collapse
Affiliation(s)
- Helio Pais
- Trivago GmbH, Bennigsen-Platz 1, 40474, Düsseldorf, Germany
| | - Katia Ruggero
- Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research (IDIBELL), Gran Via de L'Hospitalet, 199, Barcelona, Spain
| | - Jing Zhang
- MRC Weatherall Institute of Molecular Medicine, MRC Molecular Haematology Unit, University of Oxford, Oxford, OX3 9DS, UK
| | - Osama Al-Assar
- The Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Nicolas Bery
- MRC Weatherall Institute of Molecular Medicine, MRC Molecular Haematology Unit, University of Oxford, Oxford, OX3 9DS, UK
| | - Ravneet Bhuller
- MRC Weatherall Institute of Molecular Medicine, MRC Molecular Haematology Unit, University of Oxford, Oxford, OX3 9DS, UK
| | - Victoria Weston
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Edgbaston, B15 2TT, UK
| | - Pamela R Kearns
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Edgbaston, B15 2TT, UK
| | | | - Ami Miller
- MRC Weatherall Institute of Molecular Medicine, MRC Molecular Haematology Unit, University of Oxford, Oxford, OX3 9DS, UK
| | - Terence H Rabbitts
- MRC Weatherall Institute of Molecular Medicine, MRC Molecular Haematology Unit, University of Oxford, Oxford, OX3 9DS, UK.
| |
Collapse
|
10
|
Downregulation of specific FBXW7 isoforms with differential effects in T-cell lymphoblastic lymphoma. Oncogene 2019; 38:4620-4636. [DOI: 10.1038/s41388-019-0746-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 12/26/2018] [Accepted: 01/29/2019] [Indexed: 12/16/2022]
|
11
|
Kuribayashi W, Takizawa K, Sugata K, Kuramitsu M, Momose H, Sasaki E, Hiradate Y, Furuhata K, Asada Y, Iwama A, Matsuoka M, Mizukami T, Hamaguchi I. Impact of the SCF signaling pathway on leukemia stem cell-mediated ATL initiation and progression in an HBZ transgenic mouse model. Oncotarget 2018; 7:51027-51043. [PMID: 27340921 PMCID: PMC5239456 DOI: 10.18632/oncotarget.10210] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 06/01/2016] [Indexed: 12/31/2022] Open
Abstract
Adult T-cell leukemia (ATL) is a malignant disease caused by human T-lymphotropic virus type 1. In aggressive ATL, the response to chemotherapy is extremely poor. We hypothesized that this poor response is due to the existence of chemotherapy-resistant cells, such as leukemic stem cells. Previously, we successfully identified an ATL stem cell (ATLSC) candidate as the c-kit+/CD38−/CD71− cells in an ATL mouse model using Tax transgenic mice. Here, with a new ATL mouse model using HBZ-transgenic mice, we further discovered that the functional ATLSC candidate, which commonly expresses c-kit, is drug-resistant and has the ability to initiate tumors and reconstitute lymphomatous cells. We characterized the ATLSCs as c-kit+/CD4−/CD8− cells and found that they have a similar gene expression profile as T cell progenitors. Additionally, we found that AP-1 gene family members, including Junb, Jund, and Fosb, were up-regulated in the ATLSC fraction. The results of an in vitro assay showed that ATLSCs cultured with cytokines known to promote stem cell expansion, such as stem cell factor (SCF), showed highly proliferative activity and maintained their stem cell fraction. Inhibition of c-kit–SCF signaling with the neutralizing antibody ACK2 affected ATLSC self-renewal and proliferation. Experiments in Sl/Sld mice, which have a mutation in the membrane-bound c-kit ligand, found that ATL development was completely blocked in these mice. These results clearly suggest that the c-kit–SCF signal plays a key role in ATLSC self-renewal and in ATL initiation and disease progression.
Collapse
Affiliation(s)
- Wakako Kuribayashi
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Disease, Tokyo, Japan.,Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.,Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Kazuya Takizawa
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Disease, Tokyo, Japan
| | - Kenji Sugata
- Laboratory of Virus Control, Institute for Virus Research, Kyoto University, Kyoto, Japan
| | - Madoka Kuramitsu
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Disease, Tokyo, Japan
| | - Haruka Momose
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Disease, Tokyo, Japan
| | - Eita Sasaki
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Disease, Tokyo, Japan
| | - Yuki Hiradate
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Disease, Tokyo, Japan
| | - Keiko Furuhata
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Disease, Tokyo, Japan
| | - Yoshihisa Asada
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Atsushi Iwama
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masao Matsuoka
- Laboratory of Virus Control, Institute for Virus Research, Kyoto University, Kyoto, Japan
| | - Takuo Mizukami
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Disease, Tokyo, Japan
| | - Isao Hamaguchi
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Disease, Tokyo, Japan
| |
Collapse
|
12
|
Novel tumor-suppressor function of KLF4 in pediatric T-cell acute lymphoblastic leukemia. Exp Hematol 2017; 53:16-25. [PMID: 28479419 DOI: 10.1016/j.exphem.2017.04.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 04/21/2017] [Accepted: 04/22/2017] [Indexed: 02/07/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common hematological malignancy in pediatric patients. Despite advances in the treatment of this disease, many children with T-cell ALL (T-ALL) die from disease relapse due to low responses to standard chemotherapy and the lack of a targeted therapy that selectively eradicates the chemoresistant leukemia-initiating cells (LICs) responsible for disease recurrence. We reported recently that the reprogramming factor Krüppel-like factor 4 (KLF4) has a tumor-suppressive function in children with T-ALL. KLF4 silencing by promoter deoxyribonucleic acid (DNA) methylation in patients with T-ALL leads to aberrant activation of the mitogen-activated protein kinase kinase MAP2K7 and the downstream c-Jun NH2-terminal kinase (JNK) pathway that controls the expansion of leukemia cells via c-Jun and activating transcription factor 2. This pathway can be inhibited with small molecules and therefore has the potential to eliminate LICs and eradicate disease in combination with standard therapy for patients with refractory and relapsed disease. The present review summarizes the role of the KLF4-MAP2K7 pathway in T-ALL pathogenesis and the function of JNK and MAP2K7 in carcinogenesis and therapy.
Collapse
|
13
|
Kim JY, Cheng X, Wölfl S. Acidic stress induced G1 cell cycle arrest and intrinsic apoptotic pathway in Jurkat T-lymphocytes. Exp Cell Res 2017; 350:140-146. [DOI: 10.1016/j.yexcr.2016.11.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 10/31/2016] [Accepted: 11/19/2016] [Indexed: 01/19/2023]
|
14
|
Karrman K, Johansson B. Pediatric T-cell acute lymphoblastic leukemia. Genes Chromosomes Cancer 2016; 56:89-116. [PMID: 27636224 DOI: 10.1002/gcc.22416] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/06/2016] [Indexed: 12/29/2022] Open
Abstract
The most common pediatric malignancy is acute lymphoblastic leukemia (ALL), of which T-cell ALL (T-ALL) comprises 10-15% of cases. T-ALL arises in the thymus from an immature thymocyte as a consequence of a stepwise accumulation of genetic and epigenetic aberrations. Crucial biological processes, such as differentiation, self-renewal capacity, proliferation, and apoptosis, are targeted and deranged by several types of neoplasia-associated genetic alteration, for example, translocations, deletions, and mutations of genes that code for proteins involved in signaling transduction, epigenetic regulation, and transcription. Epigenetically, T-ALL is characterized by gene expression changes caused by hypermethylation of tumor suppressor genes, histone modifications, and miRNA and lncRNA abnormalities. Although some genetic and gene expression patterns have been associated with certain clinical features, such as immunophenotypic subtype and outcome, none has of yet generally been implemented in clinical routine for treatment decisions. The recent advent of massive parallel sequencing technologies has dramatically increased our knowledge of the genetic blueprint of T-ALL, revealing numerous fusion genes as well as novel gene mutations. The challenges now are to integrate all genetic and epigenetic data into a coherent understanding of the pathogenesis of T-ALL and to translate the wealth of information gained in the last few years into clinical use in the form of improved risk stratification and targeted therapies. Here, we provide an overview of pediatric T-ALL with an emphasis on the acquired genetic alterations that result in this disease. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kristina Karrman
- Department of Clinical Genetics, Office for Medical Services, Division of Laboratory Medicine, Lund, Sweden.,Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Bertil Johansson
- Department of Clinical Genetics, Office for Medical Services, Division of Laboratory Medicine, Lund, Sweden.,Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|
15
|
Peirs S, Van der Meulen J, Van de Walle I, Taghon T, Speleman F, Poppe B, Van Vlierberghe P. Epigenetics in T-cell acute lymphoblastic leukemia. Immunol Rev 2015; 263:50-67. [PMID: 25510271 DOI: 10.1111/imr.12237] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Normal T-cell development is a strictly regulated process in which hematopoietic progenitor cells migrate from the bone marrow to the thymus and differentiate from early T-cell progenitors toward mature and functional T cells. During this maturation process, cooperation between a variety of oncogenes and tumor suppressors can drive immature thymocytes into uncontrolled clonal expansion and cause T-cell acute lymphoblastic leukemia (T-ALL). Despite improved insights in T-ALL disease biology and comprehensive characterization of its genetic landscape, clinical care remained largely similar over the past decades and still consists of high-dose multi-agent chemotherapy potentially followed by hematopoietic stem cell transplantation. Even with such aggressive treatment regimens, which are often associated with considerable side effects, clinical outcome is still extremely poor in a significant subset of T-ALL patients as a result of therapy resistance or hematological relapses. Recent genetic studies have identified recurrent somatic alterations in genes involved in DNA methylation and post-translational histone modifications in T-ALL, suggesting that epigenetic homeostasis is critically required in restraining tumor development in the T-cell lineage. In this review, we provide an overview of the epigenetic regulators that could be implicated in T-ALL disease biology and speculate how the epigenetic landscape of T-ALL could trigger the development of epigenetic-based therapies to further improve the treatment of human T-ALL.
Collapse
Affiliation(s)
- Sofie Peirs
- Center for Medical Genetics, Ghent University, Ghent, Belgium
| | | | | | | | | | | | | |
Collapse
|
16
|
Durinck K, Goossens S, Peirs S, Wallaert A, Van Loocke W, Matthijssens F, Pieters T, Milani G, Lammens T, Rondou P, Van Roy N, De Moerloose B, Benoit Y, Haigh J, Speleman F, Poppe B, Van Vlierberghe P. Novel biological insights in T-cell acute lymphoblastic leukemia. Exp Hematol 2015; 43:625-39. [PMID: 26123366 DOI: 10.1016/j.exphem.2015.05.017] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 05/24/2015] [Indexed: 01/07/2023]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive type of blood cancer that accounts for about 15% of pediatric and 25% of adult acute lymphoblastic leukemia (ALL) cases. It is considered as a paradigm for the multistep nature of cancer initiation and progression. Genetic and epigenetic reprogramming events, which transform T-cell precursors into malignant T-ALL lymphoblasts, have been extensively characterized over the past decade. Despite our comprehensive understanding of the genomic landscape of human T-ALL, leukemia patients are still treated by high-dose multiagent chemotherapy, potentially followed by hematopoietic stem cell transplantation. Even with such aggressive treatment regimens, which are often associated with considerable acute and long-term side effects, about 15% of pediatric and 40% of adult T-ALL patients still relapse, owing to acquired therapy resistance, and present with very dismal survival perspectives. Unfortunately, the molecular mechanisms by which residual T-ALL tumor cells survive chemotherapy and act as a reservoir for leukemic progression and hematologic relapse remain poorly understood. Nevertheless, it is expected that enhanced molecular understanding of T-ALL disease biology will ultimately facilitate a targeted therapy driven approach that can reduce chemotherapy-associated toxicities and improve survival of refractory T-ALL patients through personalized salvage therapy. In this review, we summarize recent biological insights into the molecular pathogenesis of T-ALL and speculate how the genetic landscape of T-ALL could trigger the development of novel therapeutic strategies for the treatment of human T-ALL.
Collapse
Affiliation(s)
- Kaat Durinck
- Center for Medical Genetics, Department for Pediatrics, Ghent, Belgium
| | - Steven Goossens
- Department for Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Unit for Molecular Oncology, VIB Inflammation Research Center, Ghent, Belgium; Mammalian Functional Genetics Laboratory, Division of Blood Cancers, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Sofie Peirs
- Center for Medical Genetics, Department for Pediatrics, Ghent, Belgium
| | - Annelynn Wallaert
- Center for Medical Genetics, Department for Pediatrics, Ghent, Belgium
| | - Wouter Van Loocke
- Center for Medical Genetics, Department for Pediatrics, Ghent, Belgium
| | | | - Tim Pieters
- Center for Medical Genetics, Department for Pediatrics, Ghent, Belgium; Department for Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Unit for Molecular Oncology, VIB Inflammation Research Center, Ghent, Belgium
| | - Gloria Milani
- Center for Medical Genetics, Department for Pediatrics, Ghent, Belgium
| | - Tim Lammens
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Pieter Rondou
- Center for Medical Genetics, Department for Pediatrics, Ghent, Belgium
| | - Nadine Van Roy
- Center for Medical Genetics, Department for Pediatrics, Ghent, Belgium
| | - Barbara De Moerloose
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Yves Benoit
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Jody Haigh
- Mammalian Functional Genetics Laboratory, Division of Blood Cancers, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Frank Speleman
- Center for Medical Genetics, Department for Pediatrics, Ghent, Belgium
| | - Bruce Poppe
- Center for Medical Genetics, Department for Pediatrics, Ghent, Belgium
| | | |
Collapse
|
17
|
Placing ion channels into a signaling network of T cells: from maturing thymocytes to healthy T lymphocytes or leukemic T lymphoblasts. BIOMED RESEARCH INTERNATIONAL 2015; 2015:750203. [PMID: 25866806 PMCID: PMC4383400 DOI: 10.1155/2015/750203] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 09/19/2014] [Indexed: 12/20/2022]
Abstract
T leukemogenesis is a multistep process, where the genetic errors during T cell maturation cause the healthy progenitor to convert into the leukemic precursor that lost its ability to differentiate but possesses high potential for proliferation, self-renewal, and migration. A new misdirecting "leukemogenic" signaling network appears, composed by three types of participants which are encoded by (1) genes implicated in determined stages of T cell development but deregulated by translocations or mutations, (2) genes which normally do not participate in T cell development but are upregulated, and (3) nondifferentially expressed genes which become highly interconnected with genes expressed differentially. It appears that each of three groups may contain genes coding ion channels. In T cells, ion channels are implicated in regulation of cell cycle progression, differentiation, activation, migration, and cell death. In the present review we are going to reveal a relationship between different genetic defects, which drive the T cell neoplasias, with calcium signaling and ion channels. We suggest that changes in regulation of various ion channels in different types of the T leukemias may provide the intracellular ion microenvironment favorable to maintain self-renewal capacity, arrest differentiation, induce proliferation, and enhance motility.
Collapse
|
18
|
Pottosin I, Delgado-Enciso I, Bonales-Alatorre E, Nieto-Pescador MG, Moreno-Galindo EG, Dobrovinskaya O. Mechanosensitive Ca2+-permeable channels in human leukemic cells: Pharmacological and molecular evidence for TRPV2. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:51-9. [DOI: 10.1016/j.bbamem.2014.09.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 09/12/2014] [Accepted: 09/15/2014] [Indexed: 01/09/2023]
|
19
|
Affiliation(s)
- Steven Goossens
- VIB Inflammation Research Center, Ghent University, Ghent, Belgium
- Department for Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Mammalian Functional Genetics Laboratory, Division of Blood Cancers, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | | |
Collapse
|