51
|
IL-7R is essential for leukemia-initiating cell activity of T-cell acute lymphoblastic leukemia. Blood 2020; 134:2171-2182. [PMID: 31530562 DOI: 10.1182/blood.2019000982] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 09/05/2019] [Indexed: 12/13/2022] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy resulting from the dysregulation of signaling pathways that control intrathymic T-cell development. Relapse rates are still significant, and prognosis is particularly bleak for relapsed patients. Therefore, development of novel therapies specifically targeting pathways controlling leukemia-initiating cell (LIC) activity is mandatory for fighting refractory T-ALL. The interleukin-7 receptor (IL-7R) is a crucial T-cell developmental pathway that is commonly expressed in T-ALL and has been implicated in leukemia progression; however, the significance of IL-7R/IL-7 signaling in T-ALL pathogenesis and its contribution to disease relapse remain unknown. To directly explore whether IL-7R targeting may be therapeutically efficient against T-ALL relapse, we focused on a known Notch1-induced T-ALL model, because a majority of T-ALL patients harbor activating mutations in NOTCH1, which is a transcriptional regulator of IL-7R expression. Using loss-of-function approaches, we show that Il7r-deficient, but not wild-type, mouse hematopoietic progenitors transduced with constitutively active Notch1 failed to generate leukemia upon transplantation into immunodeficient mice, thus providing formal evidence that IL-7R function is essential for Notch1-induced T-cell leukemogenesis. Moreover, we demonstrate that IL-7R expression is an early functional biomarker of T-ALL cells with LIC potential and report that impaired IL-7R signaling hampers engraftment and progression of patient-derived T-ALL xenografts. Notably, we show that IL-7R-dependent LIC activity and leukemia progression can be extended to human B-cell acute lymphoblastic leukemia (B-ALL). These results have important therapeutic implications, highlighting the relevance that targeting normal IL-7R signaling may have in future therapeutic interventions, particularly for preventing T-ALL (and B-ALL) relapse.
Collapse
|
52
|
Mouse acute leukemia develops independent of self-renewal and differentiation potentials in hematopoietic stem and progenitor cells. Blood Adv 2020; 3:419-431. [PMID: 30733302 DOI: 10.1182/bloodadvances.2018022400] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 01/03/2019] [Indexed: 12/17/2022] Open
Abstract
The cell of origin, defined as the normal cell in which the transformation event first occurs, is poorly identified in leukemia, despite its importance in understanding of leukemogenesis and improving leukemia therapy. Although hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) were used for leukemia models, whether their self-renewal and differentiation potentials influence the initiation and development of leukemia is largely unknown. In this study, the self-renewal and differentiation potentials in 2 distinct types of HSCs (HSC1 [CD150+CD41-CD34-Lineage-Sca-1+c-Kit+ cells] and HSC2 [CD150-CD41-CD34-Lineage-Sca-1+c-Kit+ cells]) and 3 distinct types of HPCs (HPC1 [CD150+CD41+CD34-Lineage-Sca-1+c-Kit+ cells], HPC2 [CD150+CD41+CD34+Lineage-Sca-1+c-Kit+ cells], and HPC3 [CD150-CD41-CD34+Lineage-Sca-1+c-Kit+ cells]) were isolated from adult mouse bone marrow, and examined by competitive repopulation assay. Then, cells from each population were retrovirally transduced to initiate MLL-AF9 acute myelogenous leukemia (AML) and the intracellular domain of NOTCH-1 T-cell acute lymphoblastic leukemia (T-ALL). AML and T-ALL similarly developed from all HSC and HPC populations, suggesting multiple cellular origins of leukemia. New leukemic stem cells (LSCs) were also identified in these AML and T-ALL models. Notably, switching between immunophenotypical immature and mature LSCs was observed, suggesting that heterogeneous LSCs play a role in the expansion and maintenance of leukemia. Based on this mouse model study, we propose that acute leukemia arises from multiple cells of origin independent of the self-renewal and differentiation potentials in hematopoietic stem and progenitor cells and is amplified by LSC switchover.
Collapse
|
53
|
Gianni F, Belver L, Ferrando A. The Genetics and Mechanisms of T-Cell Acute Lymphoblastic Leukemia. Cold Spring Harb Perspect Med 2020; 10:a035246. [PMID: 31570389 PMCID: PMC7050584 DOI: 10.1101/cshperspect.a035246] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic malignancy derived from early T-cell progenitors. The recognition of clinical, genetic, transcriptional, and biological heterogeneity in this disease has already translated into new prognostic biomarkers, improved leukemia animal models, and emerging targeted therapies. This work reviews our current understanding of the molecular mechanisms of T-ALL.
Collapse
Affiliation(s)
- Francesca Gianni
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York 10032, USA
| | - Laura Belver
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York 10032, USA
| | - Adolfo Ferrando
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York 10032, USA
- Department of Pathology, Columbia University Medical Center, New York, New York 10032, USA
- Department of Pediatrics, Columbia University Medical Center, New York, New York 10032, USA
| |
Collapse
|
54
|
Shu Y, Wang Y, Lv WQ, Peng DY, Li J, Zhang H, Jiang GJ, Yang BJ, Liu S, Zhang J, Chen YH, Tang S, Wan KX, Yuan JT, Guo W, Fu G, Qi XK, Liu ZD, Liu HY, Yang C, Zhang LH, Liu FJ, Yu J, Zhang PH, Qu B, Zhao H, He TC, Zou L. ARRB1-Promoted NOTCH1 Degradation Is Suppressed by OncomiR miR-223 in T-cell Acute Lymphoblastic Leukemia. Cancer Res 2020; 80:988-998. [PMID: 31822496 PMCID: PMC7056567 DOI: 10.1158/0008-5472.can-19-1471] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/24/2019] [Accepted: 12/04/2019] [Indexed: 12/17/2022]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a type of aggressive leukemia with inferior prognosis. Although activating mutations of NOTCH1 are observed in most T-ALL cases, these mutations alone are not sufficient to drive the full development of T-ALL. β-Arrestins (ARRB) are versatile and multifunctional adapter proteins that regulate diverse cellular functions, including promoting the development of cancer. However, the role of ARRBs in T-ALL has largely remained elusive. In this study, we showed that ARRB1 is expressed at low levels in assayed T-ALL clinical samples and cell lines. Exogenous ARRB1 expression inhibited T-ALL proliferation and improved the survival of T-ALL xenograft animals. ARRB1 facilitated NOTCH1 ubiquitination and degradation through interactions with NOTCH1 and DTX1. Mechanistically, the oncogenic miRNA (oncomiR) miR-223 targets the 3'-UTR of ARRB1 (BUTR) and inhibits its expression in T-ALL. Furthermore, overexpression of the ARRB1-derived miR-223 sponge suppressed T-ALL cell proliferation and induced apoptosis. Collectively, these results demonstrate that ARRB1 acts as a tumor suppressor in T-ALL by promoting NOTCH1 degradation, which is inhibited by elevated miR-223, suggesting that ARRB1 may serve as a valid drug target in the development of novel T-ALL therapeutics.Significance: These findings highlight a novel tumor suppressive function of the adaptor protein β-arrestin1 in T-ALL.
Collapse
Affiliation(s)
- Yi Shu
- Center for Clinical Molecular Medicine, The Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
- Molecular Oncology Laboratory, Departments of Surgery and Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois
| | - Yi Wang
- Center for Clinical Molecular Medicine, The Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
| | - Wen-Qiong Lv
- Center for Clinical Molecular Medicine, The Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
| | - Dan-Yi Peng
- Center for Clinical Molecular Medicine, The Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
| | - Juan Li
- Institute of Biochemistry and Cell Biochemistry, Shanghai Institute of Biomedical Sciences, Shanghai, China
| | - Hang Zhang
- Center for Clinical Molecular Medicine, The Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
| | - Guang-Jie Jiang
- Center for Clinical Molecular Medicine, The Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
| | - Bi-Jie Yang
- Center for Clinical Molecular Medicine, The Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
| | - Shan Liu
- Center for Clinical Molecular Medicine, The Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
| | - Jia Zhang
- Center for Clinical Molecular Medicine, The Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
| | - Yan-Hua Chen
- Center for Clinical Molecular Medicine, The Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
| | - Shi Tang
- Center for Clinical Molecular Medicine, The Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
| | - Ke-Xing Wan
- Center for Clinical Molecular Medicine, The Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
| | - Jun-Tao Yuan
- Center for Clinical Molecular Medicine, The Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
| | - Wei Guo
- Center for Clinical Molecular Medicine, The Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
| | - Guo Fu
- Center for Clinical Molecular Medicine, The Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
| | - Xin-Kun Qi
- Center for Clinical Molecular Medicine, The Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
| | - Zhi-Dai Liu
- Center for Clinical Molecular Medicine, The Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
| | - Hai-Yan Liu
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
- Department of Hematology, The Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Chao Yang
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
- Molecular Oncology Laboratory, Departments of Surgery and Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois
| | - Ling-Huan Zhang
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
- Molecular Oncology Laboratory, Departments of Surgery and Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois
| | - Fang-Jie Liu
- Center for Clinical Molecular Medicine, The Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
| | - Jie Yu
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
- Department of Hematology, The Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Peng-Hui Zhang
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
- Clinical Laboratory Center, The Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Bin Qu
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Homburg, Germany
| | - Hui Zhao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Departments of Surgery and Orthopaedic Surgery, The University of Chicago Medical Center, Chicago, Illinois.
| | - Lin Zou
- Center for Clinical Molecular Medicine, The Children's Hospital of Chongqing Medical University, Chongqing, China.
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing Stem Cell Therapy Engineering Center, Chongqing, China
| |
Collapse
|
55
|
Cardoso BA. The Bone Marrow Niche - The Tumor Microenvironment That Ensures Leukemia Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:259-293. [PMID: 32130704 DOI: 10.1007/978-3-030-34025-4_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The human body requires a constant delivery of fresh blood cells that are needed to maintain body homeostasis. Hematopoiesis is the process that drives the formation of new blood cells from a single stem cell. This is a complex, orchestrated and tightly regulated process that occurs within the bone marrow. When such process is faulty or deregulated, leukemia arises, develops and thrives by subverting normal hematopoiesis and availing the supplies of this rich milieu.In this book chapter we will describe and characterize the bone marrow microenvironment and its key importance for leukemia expansion. The several components of the bone marrow niche, their interaction with the leukemic cells and the cellular pathways activated within the malignant cells will be emphasized. Finally, novel therapeutic strategies to target this sibling interaction will also be discussed.
Collapse
Affiliation(s)
- Bruno António Cardoso
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
56
|
Bigas A, Guillén Y, Schoch L, Arambilet D. Revisiting β-Catenin Signaling in T-Cell Development and T-Cell Acute Lymphoblastic Leukemia. Bioessays 2019; 42:e1900099. [PMID: 31854474 DOI: 10.1002/bies.201900099] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 11/28/2019] [Indexed: 12/25/2022]
Abstract
β-Catenin/CTNNB1 is critical for leukemia initiation or the stem cell capacity of several hematological malignancies. This review focuses on a general evaluation of β-catenin function in normal T-cell development and T-cell acute lymphoblastic leukemia (T-ALL). The integration of the existing literature offers a state-of-the-art dissection of the complexity of β-catenin function in leukemia initiation and maintenance in both Notch-dependent and independent contexts. In addition, β-catenin mutations are screened for in T-ALL primary samples, and it is found that they are rare and with little clinical relevance. Transcriptional analysis of Wnt family members (Ctnnb1, Axin2, Tcf7, and Lef1) and Myc in different publicly available T-ALL cohorts indicates that the expression of these genes may correlate with T-ALL subtypes and/or therapy outcomes.
Collapse
Affiliation(s)
- Anna Bigas
- Cancer Research Program, CIBERONC, Institut Mar d'Investigacions Mèdiques (IMIM), Doctor Aiguader 88, 08003, Barcelona, Spain
| | - Yolanda Guillén
- Cancer Research Program, CIBERONC, Institut Mar d'Investigacions Mèdiques (IMIM), Doctor Aiguader 88, 08003, Barcelona, Spain
| | - Leonie Schoch
- Cancer Research Program, CIBERONC, Institut Mar d'Investigacions Mèdiques (IMIM), Doctor Aiguader 88, 08003, Barcelona, Spain
| | - David Arambilet
- Cancer Research Program, CIBERONC, Institut Mar d'Investigacions Mèdiques (IMIM), Doctor Aiguader 88, 08003, Barcelona, Spain
| |
Collapse
|
57
|
The effect of co-occurring lesions on leukaemogenesis and drug response in T-ALL and ETP-ALL. Br J Cancer 2019; 122:455-464. [PMID: 31792348 PMCID: PMC7028932 DOI: 10.1038/s41416-019-0647-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 10/14/2019] [Accepted: 10/30/2019] [Indexed: 01/27/2023] Open
Abstract
Despite advances in the management of acute lymphoblastic leukaemia (ALL), current regimens fail to significantly transform outcomes for patients with high-risk subtypes. Advances in genomic analyses have identified novel lesions including mutations in genes that encode chromatin modifiers and those that influence cytokine and kinase signalling, rendering many of these alterations potentially targetable by tyrosine kinase and epigenetic inhibitors currently in clinical use. Although specific genomic lesions, gene expression patterns, and immunophenotypic profiles have been associated with specific clinical outcomes in some cancers, the application of precision medicine approaches based on these data has been slow. This approach is complicated by the reality that patients often harbour multiple mutations, and in many cases, the precise functional significance and interaction of these mutations in driving leukaemia and drug responsiveness/resistance remains unknown. Given that signalling pathways driving leukaemic pathogenesis could plausibly result from the co-existence of specific lesions and the resultant perturbation of protein interactions, the use of combined therapeutics that target multiple aberrant pathways, according to an individual’s mutational profile, might improve outcomes and lower a patient’s risk of relapse. Here we outline the genomic alterations that occur in T cell ALL (T-ALL) and early T cell precursor (ETP)-ALL and review studies highlighting the possible effects of co-occurring lesions on leukaemogenesis and drug response.
Collapse
|
58
|
Abstract
The search for oncogenic mutations in haematological malignancies has largely focused on coding sequence variants. These variants have been critical in understanding these complex cancers in greater detail, ultimately leading to better disease monitoring, subtyping and prognostication. In contrast, the search for oncogenic variants in the noncoding genome has proven to be challenging given the vastness of the search space, the intrinsic difficulty in assessing the impact of variants that do not code for functional proteins, and our still primitive understanding of the function harboured by large parts of the noncoding genome. Recent studies have broken ground on this quest, identifying somatically acquired and recurrent mutations in the noncoding genome that activate the expression of proto-oncogenes. In this Review, we explore some of the best-characterised examples of noncoding mutations in haematological malignancies, and highlight how a significant majority of these variants impinge on gene regulation through the formation of aberrant enhancers and promoters. We delve into the challenges faced by those that embark on a search for noncoding driver mutations, and provide a framework distilled from studies that have successfully identified such variants to overcome some of the most salient hurdles. Finally, we discuss the current therapeutic strategies being explored to target the oncogenic mechanism supported by recurrent noncoding variants. We postulate that the continued discovery and functional characterisation of somatic variants in the noncoding genome will not only advance our understanding of haematological malignancies, but offer novel therapeutic avenues and provide important insights into transcriptional regulation on a broader scale. Summary: This Review highlights the challenging but rewarding search for somatic mutations in the noncoding genome, and how such variants nucleate aberrant enhancers and promoters that drive the expression of proto-oncogenes.
Collapse
Affiliation(s)
- Sunniyat Rahman
- Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Marc R Mansour
- Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| |
Collapse
|
59
|
Rodriguez S, Abundis C, Boccalatte F, Mehrotra P, Chiang MY, Yui MA, Wang L, Zhang H, Zollman A, Bonfim-Silva R, Kloetgen A, Palmer J, Sandusky G, Wunderlich M, Kaplan MH, Mulloy JC, Marcucci G, Aifantis I, Cardoso AA, Carlesso N. Therapeutic targeting of the E3 ubiquitin ligase SKP2 in T-ALL. Leukemia 2019; 34:1241-1252. [PMID: 31772299 PMCID: PMC7192844 DOI: 10.1038/s41375-019-0653-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 10/18/2019] [Accepted: 11/13/2019] [Indexed: 12/15/2022]
Abstract
Timed degradation of the cyclin-dependent kinase inhibitor p27Kip1 by the E3 ubiquitin ligase F-box protein SKP2 is critical for T-cell progression into cell cycle, coordinating proliferation and differentiation processes. SKP2 expression is regulated by mitogenic stimuli and by Notch signaling, a key pathway in T-cell development and in T-cell acute lymphoblastic leukemia (T-ALL); however, it is not known whether SKP2 plays a role in the development of T-ALL. Here, we determined that SKP2 function is relevant for T-ALL leukemogenesis, whereas is dispensable for T-cell development. Targeted inhibition of SKP2 by genetic deletion or pharmacological blockade markedly inhibited proliferation of human T-ALL cells in vitro and antagonized disease in vivo in murine and xenograft leukemia models, with little effect on normal tissues. We also demonstrate a novel feed forward feedback loop by which Notch and IL-7 signaling cooperatively converge on SKP2 induction and cell cycle activation. These studies show that the Notch/SKP2/p27Kip1 pathway plays a unique role in T-ALL development and provide a proof-of-concept for the use of SKP2 as a new therapeutic target in T-cell acute lymphoblastic leukemia (T-ALL).
Collapse
Affiliation(s)
- Sonia Rodriguez
- Beckman Research Institute, Gehr Leukemia Center, City of Hope, Duarte, CA, 91010, USA.,Herman B Wells Center, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Christina Abundis
- Beckman Research Institute, Gehr Leukemia Center, City of Hope, Duarte, CA, 91010, USA
| | - Francesco Boccalatte
- Department of Pathology and Perlmutter Cancer Center, NYU Langone Medical Center, New York, NY, 10016, USA
| | - Purvi Mehrotra
- Herman B Wells Center, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Mark Y Chiang
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA
| | - Mary A Yui
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Lin Wang
- Herman B Wells Center, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Huajia Zhang
- Herman B Wells Center, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Amy Zollman
- Herman B Wells Center, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Ricardo Bonfim-Silva
- Herman B Wells Center, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Riberão Preto, São Paulo, 14049-900, Brazil
| | - Andreas Kloetgen
- Department of Pathology and Perlmutter Cancer Center, NYU Langone Medical Center, New York, NY, 10016, USA
| | - Joycelynne Palmer
- Beckman Research Institute, Gehr Leukemia Center, City of Hope, Duarte, CA, 91010, USA
| | - George Sandusky
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Mark Wunderlich
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Mark H Kaplan
- Herman B Wells Center, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - James C Mulloy
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Guido Marcucci
- Beckman Research Institute, Gehr Leukemia Center, City of Hope, Duarte, CA, 91010, USA
| | - Iannis Aifantis
- Department of Pathology and Perlmutter Cancer Center, NYU Langone Medical Center, New York, NY, 10016, USA
| | - Angelo A Cardoso
- Beckman Research Institute, Gehr Leukemia Center, City of Hope, Duarte, CA, 91010, USA
| | - Nadia Carlesso
- Beckman Research Institute, Gehr Leukemia Center, City of Hope, Duarte, CA, 91010, USA. .,Herman B Wells Center, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
60
|
Zhang Y, Gundelach J, Lindquist LD, Baker DJ, van Deursen J, Bram RJ. Chemotherapy-induced cellular senescence suppresses progression of Notch-driven T-ALL. PLoS One 2019; 14:e0224172. [PMID: 31661505 PMCID: PMC6818774 DOI: 10.1371/journal.pone.0224172] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 10/06/2019] [Indexed: 12/31/2022] Open
Abstract
T cell acute lymphoblastic leukemia (T-ALL) is a serious hematologic malignancy that occurs in children and young adults. Current therapies include intensive chemotherapy and ionizing radiation that preferentially kill malignant cells. Unfortunately, they are frequently accompanied by unintended negative impacts, including the induction of cellular senescence and long-term toxicities in normal host tissues. Whether these senescent cells resulting from therapy increase the susceptibility to relapse or secondary cancers is unknown. Using transgenic and pharmacological approaches to eliminate doxorubicin-induced senescent cells in a Notch-driven T-ALL relapse mouse model, we find that these cells inhibit tumor recurrence, suggesting that senescence in response to treatment suppresses tumorigenesis. This finding, together with extensive evidence from others demonstrating that age-associated health problems develop dramatically earlier among childhood cancer survivors compared to age-matched counterparts, suggests a relationship between therapy-induced senescence and tumorigenesis. Although cancer risk is increased through accelerated premature-aging in the long run, therapy-induced senescence appears to protect survivors from recurrence, at least in the short run.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Justin Gundelach
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Lonnie D. Lindquist
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Darren J. Baker
- Department of Pediatric and Adolescent Medicine, and Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States of America
| | - Jan van Deursen
- Department of Pediatric and Adolescent Medicine, and Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States of America
| | - Richard J. Bram
- Department of Immunology, and Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, United States of America
- * E-mail:
| |
Collapse
|
61
|
Gao J, Van Meter M, Hernandez Lopez S, Chen G, Huang Y, Ren S, Zhao Q, Rojas J, Gurer C, Thurston G, Kuhnert F. Therapeutic targeting of Notch signaling and immune checkpoint blockade in a spontaneous, genetically heterogeneous mouse model of T-cell acute lymphoblastic leukemia. Dis Model Mech 2019; 12:dmm.040931. [PMID: 31399482 PMCID: PMC6765191 DOI: 10.1242/dmm.040931] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 07/29/2019] [Indexed: 01/05/2023] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic cancer derived from the malignant transformation of T-cell progenitors. Outcomes remain poor for T-ALL patients who have either primary resistance to standard-of-care chemotherapy or disease relapse. Notably, there are currently no targeted therapies available in T-ALL. This lack of next-generation therapies highlights the need for relevant preclinical disease modeling to identify and validate new targets and treatment approaches. Here, we adapted a spontaneously arising, genetically heterogeneous, thymic transplantation-based murine model of T-ALL, recapitulating key histopathological and genetic features of the human disease, to the preclinical testing of targeted and immune-directed therapies. Genetic engineering of the murine Notch1 locus aligned the spectrum of Notch1 mutations in the mouse model to that of human T-ALL and confirmed aberrant, recombination-activating gene (RAG)-mediated 5′ Notch1 recombination events as the preferred pathway in murine T-ALL development. Testing of Notch1-targeting therapeutic antibodies demonstrated T-ALL sensitivity to different classes of Notch1 blockers based on Notch1 mutational status. In contrast, genetic ablation of Notch3 did not impact T-ALL development. The T-ALL model was further applied to the testing of immunotherapeutic agents in fully immunocompetent, syngeneic mice. In line with recent clinical experience in T-cell malignancies, programmed cell death 1 (PD-1) blockade alone lacked anti-tumor activity against murine T-ALL tumors. Overall, the unique features of the spontaneous T-ALL model coupled with genetic manipulations and the application to therapeutic testing in immunocompetent backgrounds will be of great utility for the preclinical evaluation of novel therapies against T-ALL. Summary: Adapting a spontaneous, genetically heterogenous T-ALL model to preclinical testing demonstrated that response to therapeutic anti-Notch1 antibodies was determined by Notch1 mutational status and that PD-1 immune checkpoint blockade alone lacked anti-tumor activity.
Collapse
Affiliation(s)
- Jie Gao
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY 10591, USA
| | | | | | - Guoying Chen
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY 10591, USA
| | - Ying Huang
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY 10591, USA
| | - Shumei Ren
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY 10591, USA
| | - Qi Zhao
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY 10591, USA
| | - Jose Rojas
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY 10591, USA
| | - Cagan Gurer
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY 10591, USA
| | - Gavin Thurston
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY 10591, USA
| | - Frank Kuhnert
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY 10591, USA
| |
Collapse
|
62
|
Habets RA, de Bock CE, Serneels L, Lodewijckx I, Verbeke D, Nittner D, Narlawar R, Demeyer S, Dooley J, Liston A, Taghon T, Cools J, de Strooper B. Safe targeting of T cell acute lymphoblastic leukemia by pathology-specific NOTCH inhibition. Sci Transl Med 2019; 11:11/494/eaau6246. [DOI: 10.1126/scitranslmed.aau6246] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 01/18/2019] [Accepted: 04/29/2019] [Indexed: 12/15/2022]
Abstract
Given the high frequency of activating NOTCH1 mutations in T cell acute lymphoblastic leukemia (T-ALL), inhibition of the γ-secretase complex remains an attractive target to prevent ligand-independent release of the cytoplasmic tail and oncogenic NOTCH1 signaling. However, four different γ-secretase complexes exist, and available inhibitors block all complexes equally. As a result, these cause severe “on-target” gastrointestinal tract, skin, and thymus toxicity, limiting their therapeutic application. Here, we demonstrate that genetic deletion or pharmacologic inhibition of the presenilin-1 (PSEN1) subclass of γ-secretase complexes is highly effective in decreasing leukemia while avoiding dose-limiting toxicities. Clinically, T-ALL samples were found to selectively express only PSEN1-containing γ-secretase complexes. The conditional knockout of Psen1 in developing T cells attenuated the development of a mutant NOTCH1-driven leukemia in mice in vivo but did not abrogate normal T cell development. Treatment of T-ALL cell lines with the selective PSEN1 inhibitor MRK-560 effectively decreased mutant NOTCH1 processing and led to cell cycle arrest. These observations were extended to T-ALL patient-derived xenografts in vivo, demonstrating that MRK-560 treatment decreases leukemia burden and increased overall survival without any associated gut toxicity. Therefore, PSEN1-selective compounds provide a potential therapeutic strategy for safe and effective targeting of T-ALL and possibly also for other diseases in which NOTCH signaling plays a role.
Collapse
|
63
|
Mallaney C, Ostrander EL, Celik H, Kramer AC, Martens A, Kothari A, Koh WK, Haussler E, Iwamori N, Gontarz P, Zhang B, Challen GA. Kdm6b regulates context-dependent hematopoietic stem cell self-renewal and leukemogenesis. Leukemia 2019; 33:2506-2521. [PMID: 30936419 PMCID: PMC6773521 DOI: 10.1038/s41375-019-0462-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/28/2019] [Accepted: 03/18/2019] [Indexed: 12/20/2022]
Abstract
The histone demethylase KDM6B (JMJD3) is upregulated in blood disorders, suggesting it may have important pathogenic functions. Here we examined the function of Kdm6b in hematopoietic stem cells (HSC) to evaluate its potential as a therapeutic target. Loss of Kdm6b lead to depletion of phenotypic and functional HSCs in adult mice, and Kdm6b is necessary for HSC self-renewal in response to inflammatory and proliferative stress. Loss of Kdm6b leads to a pro-differentiation poised state in HSCs due to the increased expression of the AP-1 transcription factor complex (Fos and Jun) and immediate early response (IER) genes. These gene expression changes occurred independently of chromatin modifications. Targeting AP-1 restored function of Kdm6b-deficient HSCs, suggesting Kdm6b regulates this complex during HSC stress response. We also show Kdm6b supports developmental context-dependent leukemogenesis for T-cell acute lymphoblastic leukemia (T-ALL) and M5 acute myeloid leukemia (AML). Kdm6b is required for effective fetal-derived T-ALL and adult-derived AML, but not vice versa. These studies identify a crucial role for Kdm6b in regulating HSC self-renewal in different contexts, and highlight the potential of KDM6B as a therapeutic target in different hematopoietic malignancies.
Collapse
Affiliation(s)
- Cates Mallaney
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Elizabeth L Ostrander
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hamza Celik
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ashley C Kramer
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Andrew Martens
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Alok Kothari
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Won Kyun Koh
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Emily Haussler
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Naoki Iwamori
- Laboratory of Biomedicine, Division of Pathobiology, Department of Basic Medicine, Faculty of Medicine, Kyushu University, Fukuoka, 812-8582, Japan
| | - Paul Gontarz
- Center of Regenerative Medicine, Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Bo Zhang
- Center of Regenerative Medicine, Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Grant A Challen
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
64
|
Ke N, Liu Q, Pi L, Fang J, Chen L, Chen X. The antitumor function of arctigenin in human retinoblastoma cells is mediated by jagged‑1. Mol Med Rep 2019; 19:3642-3648. [PMID: 30896794 PMCID: PMC6470923 DOI: 10.3892/mmr.2019.10026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 02/19/2019] [Indexed: 02/07/2023] Open
Abstract
Retinoblastoma is an intraocular malignant tumor that may severely affect vision and represents a life‑threatening disease in children. Arctigenin (ATG) is an active compound that exhibits numerous pharmacological activities, which is isolated from the seeds of greater burdock (Arctium lappa Linnaeus), a plant used in traditional Chinese herbal medicine. The present study aimed to investigate the effects of ATG on cancer progression by analyzing the retinoblastoma cell line Y79. ATG exhibited a significant inhibitory effect on the viability of Y79 cells in a dose‑dependent manner. Furthermore, treatment with ATG promoted apoptosis, and increased the protein expression levels of B‑cell lymphoma 2 (BCL‑2)‑associated X protein and decreased the protein expression levels of BCL‑2. Cell migration was suppressed following treatment with ATG, as assessed by Transwell migration assay. Furthermore, the protein expression levels of jagged‑1 (JAG1) were decreased, and various factors involved in the Notch signaling pathway, including the Notch intracellular domain (NICD), transcription factor HES (HES)5 and HES1 were downregulated following treatment with ATG. The decreased expression levels of JAG1 were restored in response to JAG1 overexpression, alongside increases in the protein expression levels of NICD, HES5 and HES1. Furthermore, overexpression of JAG1 partly restored the cell viability and migration suppressed following treatment with ATG. In addition, ATG‑induced apoptosis was reduced by JAG1 overexpression. Collectively, the present results suggested that ATG may serve as an antitumor compound by suppressing the proliferation and migration of retinoblastoma cells, inducing apoptosis, downregulating the protein expression levels of JAG1, and decreasing the activity of the Notch signaling pathway.
Collapse
Affiliation(s)
- Ning Ke
- Department of Ophthalmology, Children's Hospital, Chongqing Medical University, Chongqing 400014, P.R. China
| | - Qing Liu
- Department of Ophthalmology, Children's Hospital, Chongqing Medical University, Chongqing 400014, P.R. China
| | - Lianhong Pi
- Department of Ophthalmology, Children's Hospital, Chongqing Medical University, Chongqing 400014, P.R. China
| | - Jing Fang
- Department of Ophthalmology, Children's Hospital, Chongqing Medical University, Chongqing 400014, P.R. China
| | - Lin Chen
- Department of Ophthalmology, Children's Hospital, Chongqing Medical University, Chongqing 400014, P.R. China
| | - Xinke Chen
- Department of Ophthalmology, Children's Hospital, Chongqing Medical University, Chongqing 400014, P.R. China
| |
Collapse
|
65
|
Sulima SO, Kampen KR, Vereecke S, Pepe D, Fancello L, Verbeeck J, Dinman JD, De Keersmaecker K. Ribosomal Lesions Promote Oncogenic Mutagenesis. Cancer Res 2018; 79:320-327. [PMID: 30482776 DOI: 10.1158/0008-5472.can-18-1987] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/17/2018] [Accepted: 11/20/2018] [Indexed: 12/22/2022]
Abstract
Ribosomopathies are congenital disorders caused by mutations in ribosomal proteins (RP) or assembly factors and are characterized by cellular hypoproliferation at an early stage. Paradoxically, many of these disorders have an elevated risk to progress to hyperproliferative cancer at a later stage. In addition, somatic RP mutations have recently been identified in various cancer types, for example, the recurrent RPL10-R98S mutation in T-cell acute lymphoblastic leukemia (T-ALL) and RPS15 mutations in chronic lymphocytic leukemia (CLL). We previously showed that RPL10-R98S promotes expression of oncogenes, but also induces a proliferative defect due to elevated oxidative stress. In this study, we demonstrate that this proliferation defect is eventually rescued by RPL10-R98S mouse lymphoid cells that acquire 5-fold more secondary mutations than RPL10-WT cells. The presence of RPL10-R98S and other RP mutations also correlated with a higher mutational load in patients with T-ALL, with an enrichment in NOTCH1-activating lesions. RPL10-R98S-associated cellular oxidative stress promoted DNA damage and impaired cell growth. Expression of NOTCH1 eliminated these phenotypes in RPL10-R98S cells, in part via downregulation of PKC-θ, with no effect on RPL10-WT cells. Patients with RP-mutant CLL also demonstrated a higher mutational burden, enriched for mutations that may diminish oxidative stress. We propose that oxidative stress due to ribosome dysfunction causes hypoproliferation and cellular insufficiency in ribosomopathies and RP-mutant cancer. This drives surviving cells, potentiated by genomic instability, to acquire rescuing mutations, which ultimately promote transition to hyperproliferation. SIGNIFICANCE: Ribosomal lesions cause oxidative stress and increase mutagenesis, promoting acquisition of rescuing mutations that stimulate proliferation.
Collapse
Affiliation(s)
- Sergey O Sulima
- Department of Oncology, KU Leuven, LKI - Leuven Cancer Institute, Leuven, Belgium
| | - Kim R Kampen
- Department of Oncology, KU Leuven, LKI - Leuven Cancer Institute, Leuven, Belgium
| | - Stijn Vereecke
- Department of Oncology, KU Leuven, LKI - Leuven Cancer Institute, Leuven, Belgium
| | - Daniele Pepe
- Department of Oncology, KU Leuven, LKI - Leuven Cancer Institute, Leuven, Belgium
| | - Laura Fancello
- Department of Oncology, KU Leuven, LKI - Leuven Cancer Institute, Leuven, Belgium
| | - Jelle Verbeeck
- Department of Oncology, KU Leuven, LKI - Leuven Cancer Institute, Leuven, Belgium
| | - Jonathan D Dinman
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland
| | - Kim De Keersmaecker
- Department of Oncology, KU Leuven, LKI - Leuven Cancer Institute, Leuven, Belgium.
| |
Collapse
|
66
|
Su H, Hu J, Huang L, Yang Y, Thenoz M, Kuchmiy A, Hu Y, Li P, Feng H, Zhou Y, Taghon T, Van Vlierberghe P, Qing G, Chen Z, Liu H. SHQ1 regulation of RNA splicing is required for T-lymphoblastic leukemia cell survival. Nat Commun 2018; 9:4281. [PMID: 30323192 PMCID: PMC6189109 DOI: 10.1038/s41467-018-06523-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 09/06/2018] [Indexed: 12/21/2022] Open
Abstract
T-acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic malignancy with complicated heterogeneity. Although expression profiling reveals common elevated genes in distinct T-ALL subtypes, little is known about their functional role(s) and regulatory mechanism(s). We here show that SHQ1, an H/ACA snoRNP assembly factor involved in snRNA pseudouridylation, is highly expressed in T-ALL. Mechanistically, oncogenic NOTCH1 directly binds to the SHQ1 promoter and activates its transcription. SHQ1 depletion induces T-ALL cell death in vitro and prolongs animal survival in murine T-ALL models. RNA-Seq reveals that SHQ1 depletion impairs widespread RNA splicing, and MYC is one of the most prominently downregulated genes due to inefficient splicing. MYC overexpression significantly rescues T-ALL cell death resulted from SHQ1 inactivation. We herein report a mechanism of NOTCH1–SHQ1–MYC axis in T-cell leukemogenesis. These findings not only shed light on the role of SHQ1 in RNA splicing and tumorigenesis, but also provide additional insight into MYC regulation. T-acute lymphoblastic leukemia is an aggressive cancer. Here the authors provide insights into the functional role of SHQ1, an H/ACA snoRNP assembly factor involved in snRNA pseudouridylation, in T-lymphoblastic leukemia cell survival through regulating the maturation of MYC mRNA.
Collapse
Affiliation(s)
- Hexiu Su
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Medical Research Institute, Wuhan University, Wuhan, 430071, China
| | - Juncheng Hu
- Medical Research Institute, Wuhan University, Wuhan, 430071, China
| | - Liang Huang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yang Yang
- College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Morgan Thenoz
- Department of Biomolecular Medicine, Ghent University, Ghent, 9000, Belgium
| | - Anna Kuchmiy
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, 9000, Belgium
| | - Yufeng Hu
- Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Peng Li
- South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Hui Feng
- Department of Pharmacology and Department of Medicine, Cancer Research Center, Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Yu Zhou
- College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Tom Taghon
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, 9000, Belgium
| | | | - Guoliang Qing
- Medical Research Institute, Wuhan University, Wuhan, 430071, China.,Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zhichao Chen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Hudan Liu
- Medical Research Institute, Wuhan University, Wuhan, 430071, China. .,Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
67
|
Gachet S, El-Chaar T, Avran D, Genesca E, Catez F, Quentin S, Delord M, Thérizols G, Briot D, Meunier G, Hernandez L, Pla M, Smits WK, Buijs-Gladdines JG, Van Loocke W, Menschaert G, André-Schmutz I, Taghon T, Van Vlierberghe P, Meijerink JP, Baruchel A, Dombret H, Clappier E, Diaz JJ, Gazin C, de Thé H, Sigaux F, Soulier J. Deletion 6q Drives T-cell Leukemia Progression by Ribosome Modulation. Cancer Discov 2018; 8:1614-1631. [PMID: 30266814 DOI: 10.1158/2159-8290.cd-17-0831] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 02/12/2018] [Accepted: 09/25/2018] [Indexed: 11/16/2022]
Abstract
Deletion of chromosome 6q is a well-recognized abnormality found in poor-prognosis T-cell acute lymphoblastic leukemia (T-ALL). Using integrated genomic approaches, we identified two candidate haploinsufficient genes contiguous at 6q14, SYNCRIP (encoding hnRNP-Q) and SNHG5 (that hosts snoRNAs), both involved in regulating RNA maturation and translation. Combined silencing of both genes, but not of either gene alone, accelerated leukemogeneis in a Tal1/Lmo1/Notch1-driven mouse model, demonstrating the tumor-suppressive nature of the two-gene region. Proteomic and translational profiling of cells in which we engineered a short 6q deletion by CRISPR/Cas9 genome editing indicated decreased ribosome and mitochondrial activities, suggesting that the resulting metabolic changes may regulate tumor progression. Indeed, xenograft experiments showed an increased leukemia-initiating cell activity of primary human leukemic cells upon coextinction of SYNCRIP and SNHG5. Our findings not only elucidate the nature of 6q deletion but also highlight the role of ribosomes and mitochondria in T-ALL tumor progression. SIGNIFICANCE: The oncogenic role of 6q deletion in T-ALL has remained elusive since this chromosomal abnormality was first identified more than 40 years ago. We combined genomic analysis and functional models to show that the codeletion of two contiguous genes at 6q14 enhances malignancy through deregulation of a ribosome-mitochondria axis, suggesting the potential for therapeutic intervention.This article is highlighted in the In This Issue feature, p. 1494.
Collapse
Affiliation(s)
- Stéphanie Gachet
- INSERM UMR944 and CNRS UMR7212, Hôpital Saint-Louis, Paris, France.,Institute of Hematology (IUH), Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Tiama El-Chaar
- INSERM UMR944 and CNRS UMR7212, Hôpital Saint-Louis, Paris, France.,Institute of Hematology (IUH), Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - David Avran
- INSERM UMR944 and CNRS UMR7212, Hôpital Saint-Louis, Paris, France.,Institute of Hematology (IUH), Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Hematology Laboratory APHP, Hôpital Saint-Louis, Paris, France
| | - Eulalia Genesca
- INSERM UMR944 and CNRS UMR7212, Hôpital Saint-Louis, Paris, France.,Institute of Hematology (IUH), Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Frédéric Catez
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon Bérard; Université Lyon 1, Lyon, France
| | - Samuel Quentin
- INSERM UMR944 and CNRS UMR7212, Hôpital Saint-Louis, Paris, France.,Institute of Hematology (IUH), Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Hematology Laboratory APHP, Hôpital Saint-Louis, Paris, France
| | - Marc Delord
- Institute of Hematology (IUH), Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Gabriel Thérizols
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon Bérard; Université Lyon 1, Lyon, France
| | - Delphine Briot
- INSERM UMR944 and CNRS UMR7212, Hôpital Saint-Louis, Paris, France.,Institute of Hematology (IUH), Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Hematology Laboratory APHP, Hôpital Saint-Louis, Paris, France
| | - Godelieve Meunier
- INSERM UMR944 and CNRS UMR7212, Hôpital Saint-Louis, Paris, France.,Institute of Hematology (IUH), Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Lucie Hernandez
- INSERM UMR944 and CNRS UMR7212, Hôpital Saint-Louis, Paris, France.,Institute of Hematology (IUH), Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Marika Pla
- Institute of Hematology (IUH), Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,INSERM UMRS 940, Hôpital Saint-Louis, Paris, France
| | - Willem K Smits
- Department of Pediatric Oncology/Hematology, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Jessica G Buijs-Gladdines
- Department of Pediatric Oncology/Hematology, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | | | - Isabelle André-Schmutz
- U1163 INSERM, Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Tom Taghon
- Cancer Research Institute, Ghent University, Ghent, Belgium
| | | | - Jules P Meijerink
- Department of Pediatric Oncology/Hematology, Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - André Baruchel
- Institute of Hematology (IUH), Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Hematology Pediatry Department, Robert Debré Hospital, Paris, France
| | - Hervé Dombret
- Institute of Hematology (IUH), Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Hematology Department, Hôpital Saint-Louis, Paris, France
| | - Emmanuelle Clappier
- INSERM UMR944 and CNRS UMR7212, Hôpital Saint-Louis, Paris, France.,Institute of Hematology (IUH), Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Hematology Laboratory APHP, Hôpital Saint-Louis, Paris, France
| | - Jean-Jacques Diaz
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS 5286, Centre Léon Bérard; Université Lyon 1, Lyon, France
| | - Claude Gazin
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, Direction de La Recherche Fondamentale, CEA, Evry, France
| | - Hugues de Thé
- INSERM UMR944 and CNRS UMR7212, Hôpital Saint-Louis, Paris, France.,Institute of Hematology (IUH), Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - François Sigaux
- INSERM UMR944 and CNRS UMR7212, Hôpital Saint-Louis, Paris, France.,Institute of Hematology (IUH), Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Hematology Laboratory APHP, Hôpital Saint-Louis, Paris, France
| | - Jean Soulier
- INSERM UMR944 and CNRS UMR7212, Hôpital Saint-Louis, Paris, France. .,Institute of Hematology (IUH), Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Hematology Laboratory APHP, Hôpital Saint-Louis, Paris, France
| |
Collapse
|
68
|
Giambra V, Gusscott S, Gracias D, Song R, Lam SH, Panelli P, Tyshchenko K, Jenkins CE, Hoofd C, Lorzadeh A, Carles A, Hirst M, Eaves CJ, Weng AP. Epigenetic Restoration of Fetal-like IGF1 Signaling Inhibits Leukemia Stem Cell Activity. Cell Stem Cell 2018; 23:714-726.e7. [PMID: 30269902 DOI: 10.1016/j.stem.2018.08.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 06/15/2018] [Accepted: 08/30/2018] [Indexed: 12/13/2022]
Abstract
Acute leukemias are aggressive malignancies of developmentally arrested hematopoietic progenitors. We sought here to explore the possibility that changes in hematopoietic stem/progenitor cells during development might alter the biology of leukemias arising from this tissue compartment. Using a mouse model of acute T cell leukemia, we found that leukemias generated from fetal liver (FL) and adult bone marrow (BM) differed dramatically in their leukemia stem cell activity with FL leukemias showing markedly reduced serial transplantability as compared to BM leukemias. We present evidence that this difference is due to NOTCH1-driven autocrine IGF1 signaling, which is active in FL cells but restrained in BM cells by EZH2-dependent H3K27 trimethylation. Further, we confirmed this mechanism is operative in human disease and show that enforced IGF1 signaling effectively limits leukemia stem cell activity. These findings demonstrate that resurrecting dormant fetal programs in adult cells may represent an alternate therapeutic approach in human cancer.
Collapse
Affiliation(s)
- Vincenzo Giambra
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada; Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo (FG), Italy.
| | - Samuel Gusscott
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Deanne Gracias
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Raymond Song
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Sonya H Lam
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Patrizio Panelli
- Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo (FG), Italy
| | | | | | - Catherine Hoofd
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Alireza Lorzadeh
- Michael Smith Laboratories and Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Annaick Carles
- Michael Smith Laboratories and Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Martin Hirst
- Michael Smith Laboratories and Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Connie J Eaves
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Andrew P Weng
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada.
| |
Collapse
|
69
|
RUNX1 promotes cell growth in human T-cell acute lymphoblastic leukemia by transcriptional regulation of key target genes. Exp Hematol 2018; 64:84-96. [DOI: 10.1016/j.exphem.2018.04.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 04/19/2018] [Accepted: 04/27/2018] [Indexed: 11/23/2022]
|
70
|
Intrathymic Notch3 and CXCR4 combinatorial interplay facilitates T-cell leukemia propagation. Oncogene 2018; 37:6285-6298. [PMID: 30038265 PMCID: PMC6284016 DOI: 10.1038/s41388-018-0401-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 05/20/2018] [Accepted: 06/10/2018] [Indexed: 12/27/2022]
Abstract
Notch hyperactivation dominates T-cell acute lymphoblastic leukemia development, but the mechanisms underlying “pre-leukemic” cell dissemination are still unclear. Here we describe how deregulated Notch3 signaling enhances CXCR4 cell-surface expression and migratory ability of CD4+CD8+ thymocytes, possibly contributing to “pre-leukemic” cell propagation, early in disease progression. In transgenic mice overexpressing the constitutively active Notch3 intracellular domain, we detect the progressive increase in circulating blood and bone marrow of CD4+CD8+ cells, characterized by high and combined surface expression of Notch3 and CXCR4. We report for the first time that transplantation of such CD4+CD8+ cells reveals their competence in infiltrating spleen and bone marrow of immunocompromised recipient mice. We also show that CXCR4 surface expression is central to the migratory ability of CD4+CD8+ cells and such an expression is regulated by Notch3 through β-arrestin in human leukemia cells. De novo, we propose that hyperactive Notch3 signaling by boosting CXCR4-dependent migration promotes anomalous egression of CD4+CD8+ cells from the thymus in early leukemia stages. In fact, in vivo CXCR4 antagonism prevents bone marrow colonization by such CD4+CD8+ cells in young Notch3 transgenic mice. Therefore, our data suggest that combined therapies precociously counteracting intrathymic Notch3/CXCR4 crosstalk may prevent dissemination of “pre-leukemic” CD4+CD8+ cells, by a “thymus-autonomous” mechanism.
Collapse
|
71
|
Giaimo BD, Borggrefe T. Introduction to Molecular Mechanisms in Notch Signal Transduction and Disease Pathogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:3-30. [DOI: 10.1007/978-3-319-89512-3_1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
72
|
Animal models of T-cell acute lymphoblastic leukemia: mimicking the human disease. JOURNAL OF BIO-X RESEARCH 2018. [DOI: 10.1097/jbr.0000000000000001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
73
|
Steinbuck MP, Winandy S. A Review of Notch Processing With New Insights Into Ligand-Independent Notch Signaling in T-Cells. Front Immunol 2018; 9:1230. [PMID: 29910816 PMCID: PMC5992298 DOI: 10.3389/fimmu.2018.01230] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/16/2018] [Indexed: 12/12/2022] Open
Abstract
The Notch receptor is an evolutionarily highly conserved transmembrane protein essential to a wide spectrum of cellular systems, and its deregulation has been linked to a vast number of developmental disorders and malignancies. Regulated Notch function is critical for the generation of T-cells, in which abnormal Notch signaling results in leukemia. Notch activation through trans-activation of the receptor by one of its ligands expressed on adjacent cells has been well defined. In this canonical ligand-dependent pathway, Notch receptor undergoes conformational changes upon ligand engagement, stimulated by a pulling-force on the extracellular fragment of Notch that results from endocytosis of the receptor-bound ligand into the ligand-expressing cell. These conformational changes in the receptor allow for two consecutive proteolytic cleavage events to occur, which release the intracellular region of the receptor into the cytoplasm. It can then travel to the nucleus, where it induces gene transcription. However, there is accumulating evidence that other pathways may induce Notch signaling. A ligand-independent mechanism of Notch activation has been described in which receptor processing is initiated via cell-internal signals. These signals result in the internalization of Notch into endosomal compartments, where chemical changes existing in this microenvironment result in the conformational modifications required for receptor processing. This review will present mechanisms underlying both canonical ligand-dependent and non-canonical ligand-independent Notch activation pathways and discuss the latter in the context of Notch signaling in T-cells.
Collapse
Affiliation(s)
- Martin Peter Steinbuck
- Immunology Training Program, Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Susan Winandy
- Immunology Training Program, Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
74
|
García-Peydró M, Fuentes P, Mosquera M, García-León MJ, Alcain J, Rodríguez A, García de Miguel P, Menéndez P, Weijer K, Spits H, Scadden DT, Cuesta-Mateos C, Muñoz-Calleja C, Sánchez-Madrid F, Toribio ML. The NOTCH1/CD44 axis drives pathogenesis in a T cell acute lymphoblastic leukemia model. J Clin Invest 2018; 128:2802-2818. [PMID: 29781813 DOI: 10.1172/jci92981] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 04/10/2018] [Indexed: 12/14/2022] Open
Abstract
NOTCH1 is a prevalent signaling pathway in T cell acute lymphoblastic leukemia (T-ALL), but crucial NOTCH1 downstream signals and target genes contributing to T-ALL pathogenesis cannot be retrospectively analyzed in patients and thus remain ill defined. This information is clinically relevant, as initiating lesions that lead to cell transformation and leukemia-initiating cell (LIC) activity are promising therapeutic targets against the major hurdle of T-ALL relapse. Here, we describe the generation in vivo of a human T cell leukemia that recapitulates T-ALL in patients, which arises de novo in immunodeficient mice reconstituted with human hematopoietic progenitors ectopically expressing active NOTCH1. This T-ALL model allowed us to identify CD44 as a direct NOTCH1 transcriptional target and to recognize CD44 overexpression as an early hallmark of preleukemic cells that engraft the BM and finally develop a clonal transplantable T-ALL that infiltrates lymphoid organs and brain. Notably, CD44 is shown to support crucial BM niche interactions necessary for LIC activity of human T-ALL xenografts and disease progression, highlighting the importance of the NOTCH1/CD44 axis in T-ALL pathogenesis. The observed therapeutic benefit of anti-CD44 antibody administration in xenotransplanted mice holds great promise for therapeutic purposes against T-ALL relapse.
Collapse
Affiliation(s)
- Marina García-Peydró
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, and
| | - Patricia Fuentes
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, and
| | - Marta Mosquera
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, and
| | - María J García-León
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, and
| | - Juan Alcain
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, and
| | - Antonio Rodríguez
- Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Pablo Menéndez
- Josep Carreras Leukemia Research Institute and Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBER-ONC), Barcelona, ISCIII, Spain.,Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Kees Weijer
- Department of Cell Biology and Histology, Academic Medical Center, and
| | - Hergen Spits
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - David T Scadden
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Stem Cell Institute and Harvard University Department of Stem Cell and Regenerative Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Carlos Cuesta-Mateos
- Servicio de Inmunología, Hospital Universitario de la Princesa, UAM, IIS-IP, Madrid, Spain
| | - Cecilia Muñoz-Calleja
- Servicio de Inmunología, Hospital Universitario de la Princesa, UAM, IIS-IP, Madrid, Spain
| | - Francisco Sánchez-Madrid
- Servicio de Inmunología, Hospital Universitario de la Princesa, UAM, IIS-IP, Madrid, Spain.,Department of Vascular Biology and Inflammation, Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, Madrid, Spain
| | - María L Toribio
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, CSIC-UAM, and
| |
Collapse
|
75
|
Canonical Notch Signaling Directs the Fate of Differentiating Neurocompetent Progenitors in the Mammalian Olfactory Epithelium. J Neurosci 2018; 38:5022-5037. [PMID: 29739871 DOI: 10.1523/jneurosci.0484-17.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/31/2018] [Accepted: 05/04/2018] [Indexed: 12/24/2022] Open
Abstract
The adult olfactory epithelium (OE) has the remarkable capacity to regenerate fully both neurosensory and non-neuronal cell types after severe epithelial injury. Lifelong persistence of two stem cell populations supports OE regeneration when damaged: the horizontal basal cells (HBCs), dormant and held in reserve; and globose basal cells, a heterogeneous population most of which are actively dividing. Both populations regenerate all cell types of the OE after injury, but the mechanisms underlying neuronal versus non-neuronal lineage commitment after recruitment of the stem cell pools remains unknown. We used both retroviral transduction and mouse lines that permit conditional cell-specific genetic manipulation as well as the tracing of progeny to study the role of canonical Notch signaling in the determination of neuronal versus non-neuronal lineages in the regenerating adult OE. Excision of either Notch1 or Notch2 genes alone in HBCs did not alter progenitor fate during recovery from epithelial injury, whereas conditional knock-out of both Notch1 and Notch2 together, retroviral transduction of progenitors with a dominant-negative form of MAML (mastermind-like), or excision of the downstream cofactor RBPJ caused progeny to adopt a neuronal fate exclusively. Conversely, we show that overexpressing the Notch1-intracellular domain (N1ICD) either genetically or by transduction blocks neuronal differentiation completely. However, N1ICD overexpression requires both alleles of the canonical cofactor RBPJ to specify downstream lineage. Together, our results suggest that canonical RBPJ-dependent Notch signaling through redundant Notch1 and Notch2 receptors is both necessary and sufficient for determining neuronal versus non-neuronal differentiation in the regenerating adult OE.SIGNIFICANCE STATEMENT Despite the substantial reconstitution of the olfactory epithelium and its population of sensory neurons after injury, disruption and exhaustion of neurogenesis is a consequence of aging and a cause of olfactory dysfunction. Understanding the mechanisms underlying the generation of replacement neurons and non-neuronal cells is critical to any therapeutic strategy aimed at rebuilding a functional neuroepithelium. The results shown here demonstrate that canonical Notch signaling determines the balance between neurons and non-neuronal cells during restoration of the epithelium after injury. Moreover, the complexities of the multiple Notch pathways impinging on that decision are dissected in detail. Finally, RBPJ, the canonical Notch transcriptional cofactor, exhibits a heretofore unreported haploinsufficiency in setting the balance among the regenerating populations.
Collapse
|
76
|
Chen X, Cai Y, Liu Q, Pan L, Shi S, Liu X, Chen Y, Li J, Wang J, Li Y, Li X, Wang S. ETS1 and SP1 drive DHX15 expression in acute lymphoblastic leukaemia. J Cell Mol Med 2018; 22:2612-2621. [PMID: 29512921 PMCID: PMC5908128 DOI: 10.1111/jcmm.13525] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 12/10/2017] [Indexed: 12/19/2022] Open
Abstract
DHX15 plays a role in leukaemogenesis and leukaemia relapse. However, the mechanism underlying the transcriptional regulation of DHX15 in ALL has not been elucidated. Our present study aimed to explore the functional promoter region of DHX15 and to investigate the transcription factors controlling the transcription of this gene. A luciferase assay performed with several truncated constructs identified a 501-bp region as the core promoter region of DHX15. Site-directed mutagenesis, electrophoretic mobility shift and chromatin immunoprecipitation assays showed that ETS1 and SP1 occupied the DHX15 promoter. Furthermore, knockdown of ETS1 and SP1 resulted in suppression of DHX15, whereas the overexpression of these genes led to up-regulation of DHX15. Interestingly, in samples obtained from patients with ALL at diagnosis, both ETS1 and SP1 correlated positively with DHX15 expression. Additionally, differences in methylation of the DHX15 core promoter region were not observed between the patients and controls. In conclusion, we identified the core promoter region of DHX15 and demonstrated that ETS1 and SP1 regulated DHX15 expression in ALL.
Collapse
Affiliation(s)
- Xiang‐Lei Chen
- Department of HematologyFujian Institute of HematologyFujian Provincial Key Laboratory on HematologyFujian Medical University Union HospitalFuzhouChina
- Union Clinical Medical CollegesFujian Medical UniversityFuzhouChina
| | - Yuan‐Hua Cai
- Department of HematologyFujian Institute of HematologyFujian Provincial Key Laboratory on HematologyFujian Medical University Union HospitalFuzhouChina
- Union Clinical Medical CollegesFujian Medical UniversityFuzhouChina
| | - Qiao Liu
- Department of HematologyFujian Institute of HematologyFujian Provincial Key Laboratory on HematologyFujian Medical University Union HospitalFuzhouChina
- Union Clinical Medical CollegesFujian Medical UniversityFuzhouChina
| | - Li‐Li Pan
- Department of HematologyFujian Institute of HematologyFujian Provincial Key Laboratory on HematologyFujian Medical University Union HospitalFuzhouChina
- Union Clinical Medical CollegesFujian Medical UniversityFuzhouChina
| | - Shui‐Ling Shi
- Union Clinical Medical CollegesFujian Medical UniversityFuzhouChina
| | - Xiao‐Li Liu
- Union Clinical Medical CollegesFujian Medical UniversityFuzhouChina
| | - Yuan Chen
- Department of HematologyFujian Institute of HematologyFujian Provincial Key Laboratory on HematologyFujian Medical University Union HospitalFuzhouChina
- Union Clinical Medical CollegesFujian Medical UniversityFuzhouChina
| | - Jing‐Gang Li
- Department of HematologyFujian Institute of HematologyFujian Provincial Key Laboratory on HematologyFujian Medical University Union HospitalFuzhouChina
- Union Clinical Medical CollegesFujian Medical UniversityFuzhouChina
| | - Jing Wang
- Union Clinical Medical CollegesFujian Medical UniversityFuzhouChina
| | - Yang Li
- Department of HematologyFujian Institute of HematologyFujian Provincial Key Laboratory on HematologyFujian Medical University Union HospitalFuzhouChina
- Union Clinical Medical CollegesFujian Medical UniversityFuzhouChina
| | - Xiao‐Fan Li
- Union Clinical Medical CollegesFujian Medical UniversityFuzhouChina
| | - Shao‐Yuan Wang
- Department of HematologyFujian Institute of HematologyFujian Provincial Key Laboratory on HematologyFujian Medical University Union HospitalFuzhouChina
- Union Clinical Medical CollegesFujian Medical UniversityFuzhouChina
| |
Collapse
|
77
|
Högstrand K, Darmanin S, Forshell TP, Grandien A. Transformation of mouse T cells requires MYC and AKT activity in conjunction with inhibition of intrinsic apoptosis. Oncotarget 2018; 9:21396-21410. [PMID: 29765548 PMCID: PMC5940390 DOI: 10.18632/oncotarget.25113] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 03/15/2018] [Indexed: 12/13/2022] Open
Abstract
Peripheral T-cell lymphoma is an aggressive non-Hodgkin's lymphoma characterized by excessive proliferation of transformed mature T cells. The number and nature of genetic aberrations required and sufficient for transformation of normal T cells into lymphomas is unknown. Here, using a combinatorial in vitro-approach, we demonstrate that overexpression of MYC together with activated AKT in conditions of inhibition of intrinsic apoptosis rapidly resulted in transformation of mature mouse T cells with a frequency approaching 100%. Injection of transformed cells into mice resulted in rapid development of aggressive T cell lymphoma, characterized by spread to several organs, destruction of tissue architecture and rapid death of the animals. TcR-sequencing revealed a polyclonal repertoire of tumor cells indicating that co-expression of MYC, activated AKT and BCLXL is sufficient for tumor transformation and do not require acquisition of additional genetic events. When analyzing cells with inducible expression we found that proliferation of transformed T cells required sustained expression of both MYC and AKT. AKT exerted a dual function as it inhibited induction of, and promoted exit from, cellular quiescence and contributed to inhibion of apoptosis. Downregulation of AKT and/or MYC together with BCLXL resulted in rapid and complete elimination of cells through induction of apoptotic cell death.
Collapse
Affiliation(s)
- Kari Högstrand
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital-Huddinge, 141 57 Stockholm, Sweden
| | - Stephanie Darmanin
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital-Huddinge, 141 57 Stockholm, Sweden
| | - TachaZi Plym Forshell
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital-Huddinge, 141 57 Stockholm, Sweden
| | - Alf Grandien
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital-Huddinge, 141 57 Stockholm, Sweden
| |
Collapse
|
78
|
Piovan E, Tosello V, Amadori A, Zanovello P. Chemotactic Cues for NOTCH1-Dependent Leukemia. Front Immunol 2018; 9:633. [PMID: 29666622 PMCID: PMC5891592 DOI: 10.3389/fimmu.2018.00633] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/14/2018] [Indexed: 12/12/2022] Open
Abstract
The NOTCH signaling pathway is a conserved signaling cascade that regulates many aspects of development and homeostasis in multiple organ systems. Aberrant activity of this signaling pathway is linked to the initiation and progression of several hematological malignancies, exemplified by T-cell acute lymphoblastic leukemia (T-ALL). Interestingly, frequent non-mutational activation of NOTCH1 signaling has recently been demonstrated in B-cell chronic lymphocytic leukemia (B-CLL), significantly extending the pathogenic significance of this pathway in B-CLL. Leukemia patients often present with high-blood cell counts, diffuse disease with infiltration of the bone marrow, secondary lymphoid organs, and diffusion to the central nervous system (CNS). Chemokines are chemotactic cytokines that regulate migration of cells between tissues and the positioning and interactions of cells within tissue. Homeostatic chemokines and their receptors have been implicated in regulating organ-specific infiltration, but may also directly and indirectly modulate tumor growth. Recently, oncogenic NOTCH1 has been shown to regulate infiltration of leukemic cells into the CNS hijacking the CC-chemokine ligand 19/CC-chemokine receptor 7 chemokine axis. In addition, a crucial role for the homing receptor axis CXC-chemokine ligand 12/CXC-chemokine receptor 4 has been demonstrated in leukemia maintenance and progression. Moreover, the CCL25/CCR9 axis has been implicated in the homing of leukemic cells into the gut, particularly in the presence of phosphatase and tensin homolog tumor suppressor loss. In this review, we summarize the latest developments regarding the role of NOTCH signaling in regulating the chemotactic microenvironmental cues involved in the generation and progression of T-ALL and compare these findings to B-CLL.
Collapse
Affiliation(s)
- Erich Piovan
- UOC Immunologia e Diagnostica Molecolare Oncologica, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy.,Dipartimento di Scienze Chirurgiche, Oncologiche e Gastroenterologiche, Università di Padova, Padova, Italy
| | - Valeria Tosello
- UOC Immunologia e Diagnostica Molecolare Oncologica, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - Alberto Amadori
- UOC Immunologia e Diagnostica Molecolare Oncologica, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy.,Dipartimento di Scienze Chirurgiche, Oncologiche e Gastroenterologiche, Università di Padova, Padova, Italy
| | - Paola Zanovello
- UOC Immunologia e Diagnostica Molecolare Oncologica, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy.,Dipartimento di Scienze Chirurgiche, Oncologiche e Gastroenterologiche, Università di Padova, Padova, Italy
| |
Collapse
|
79
|
Cattaneo M, Orlandi R, Ronchini C, Granelli P, Malferrari G, Menard S, Biunno I. The Expression of Sel1L and Tan-1 in Normal and Neoplastic Cells. Int J Biol Markers 2018; 15:26-32. [PMID: 10763137 DOI: 10.1177/172460080001500105] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We have previously reported on the isolation and chromosomal mapping of a novel human gene (SEL1L), which shows sequence similarity to sel-1, an extragenic suppressor of C. elegans. sel-1 functions as a negative regulator of lin-12 activity, the latter being implicated in the control of diverse cellular differentiation events. In the present study we compare the expression patterns of SEL1L and TAN-1, the human ortholog of lin-12 in normal and neoplastic cells. We found that, whereas both genes are expressed in fetal tissues at similar levels, they are differentially expressed in normal adult and neoplastic cells. In normal adult cells SEL1L is generally present at very low levels; only in the cells of the pancreas does it show maximum expression. By contrast, SEL1L is generally well represented in most neoplastic cells but not in those of pancreatic and gastric carcinomas, where transcription is either downregulated or completely repressed. TAN-1 on the other hand is well represented in almost all normal and neoplastic cells, with very few exceptions. Our observations suggest that SEL1L is presumably implicated in pancreatic and gastric carcinogenesis and that, along with TAN-1, it is very important for normal cell function. Alterations in the expression of SEL1L may be used as a prognostic marker for gastric and pancreatic cancers.
Collapse
Affiliation(s)
- M Cattaneo
- Institute of Advanced Biomedical Technologies, National Research Council, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
80
|
Steinbuck MP, Arakcheeva K, Winandy S. Novel TCR-Mediated Mechanisms of Notch Activation and Signaling. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:997-1007. [PMID: 29288204 PMCID: PMC5854196 DOI: 10.4049/jimmunol.1700070] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 11/22/2017] [Indexed: 01/04/2023]
Abstract
The Notch receptor is an evolutionarily highly conserved transmembrane protein that is essential to a wide spectrum of cellular systems. Notch signaling is especially important to T cell development, and its deregulation leads to leukemia. Although not well characterized, it continues to play an integral role in peripheral T cells, in which a unique mode of Notch activation can occur. In contrast to canonical Notch activation initiated by adjacent ligand-expressing cells, TCR stimulation is sufficient to induce Notch signaling. However, the interactions between these two pathways have not been defined. In this article, we show that Notch activation occurs in peripheral T cells within a few hours post-TCR stimulation and is required for optimal T cell activation. Using a panel of inhibitors against components of the TCR signaling cascade, we demonstrate that Notch activation is facilitated through initiation of protein kinase C-induced ADAM activity. Moreover, our data suggest that internalization of Notch via endocytosis plays a role in this process. Although ligand-mediated Notch stimulation relies on mechanical pulling forces that disrupt the autoinhibitory domain of Notch, we hypothesized that, in T cells in the absence of ligands, these conformational changes are induced through chemical adjustments in the endosome, causing alleviation of autoinhibition and receptor activation. Thus, T cells may have evolved a unique method of Notch receptor activation, which is described for the first time, to our knowledge, in this article.
Collapse
Affiliation(s)
- Martin Peter Steinbuck
- Department of Pathology and Laboratory Medicine, Immunology Training Program, Boston University School of Medicine, Boston, MA 02118
| | - Ksenia Arakcheeva
- Department of Pathology and Laboratory Medicine, Immunology Training Program, Boston University School of Medicine, Boston, MA 02118
| | - Susan Winandy
- Department of Pathology and Laboratory Medicine, Immunology Training Program, Boston University School of Medicine, Boston, MA 02118
| |
Collapse
|
81
|
Huo L, Wei W, Wu S, Zhao X, Zhao C, Zhao H, Sun L. Effect of dihydroarteminin combined with siRNA targeting Notch1 on Notch1/c-Myc signaling in T-cell lymphoma cells. Exp Ther Med 2018; 15:3059-3065. [PMID: 29599840 DOI: 10.3892/etm.2018.5784] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/16/2018] [Indexed: 02/07/2023] Open
Abstract
The effectiveness of therapy combining dihydroartemisinin (DHA) and small interfering RNA targeting Notch1 (siNotch1) in T-cell lymphoma remains unknown. The present study explored the potential and possible mechanisms of combined dihydroarteminin, and siNotch1 therapy for T-cell lymphoma. It was demonstrated that the viability rates of siRNA-DHA-treated cells was significantly suppressed in comparison with those in control cells, control siRNA cells, siRNA-treated cells and DHA-treated cells (P<0.01). Additionally, there was a significant increase in cell apoptosis of siRNA-DHA-treated cells in comparison with those of control cells, control siRNA cells, siRNA-treated cells, DHA-treated cells (P<0.05). Furthermore, Notch1 and c-Myc mRNA and protein expression were decreased in siRNA-DHA-treated cells (P<0.05). The present study demonstrated that DHA combined with siNotch1 is able to suppress proliferation and promote apoptosis, and downregulate the expression of Notch1 and c-Myc mRNA and protein in T-cell lymphoma cells. Targeting Notch1/c-Myc signaling with siRNA and DHA may represent a novel strategy for treating human T-cell lymphoma.
Collapse
Affiliation(s)
- Lanfen Huo
- Department of Hematology, Affiliated Hospital, Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Wenwen Wei
- Department of Hematology, Affiliated Hospital, Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Shaoling Wu
- Department of Hematology, Affiliated Hospital, Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Xindong Zhao
- Department of Hematology, School of Medicine, Qingdao University, Qingdao, Shandong 266021, P.R. China
| | - Chunting Zhao
- Department of Hematology, Affiliated Hospital, Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Hongguo Zhao
- Department of Hematology, Affiliated Hospital, Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Lingjie Sun
- Department of Hematology, Affiliated Hospital, Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
82
|
Notch signaling: its roles and therapeutic potential in hematological malignancies. Oncotarget 2018; 7:29804-23. [PMID: 26934331 PMCID: PMC5045435 DOI: 10.18632/oncotarget.7772] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 02/11/2016] [Indexed: 01/07/2023] Open
Abstract
Notch is a highly conserved signaling system that allows neighboring cells to communicate, thereby controlling their differentiation, proliferation and apoptosis, with the outcome of its activation being highly dependent on signal strength and cell type. As such, there is growing evidence that disturbances in physiological Notch signaling contribute to cancer development and growth through various mechanisms. Notch was first reported to contribute to tumorigenesis in the early 90s, through identification of the involvement of the Notch1 gene in the chromosomal translocation t(7;9)(q34;q34.3), found in a small subset of T-cell acute lymphoblastic leukemia. Since then, Notch mutations and aberrant Notch signaling have been reported in numerous other precursor and mature hematological malignancies, of both myeloid and lymphoid origin, as well as many epithelial tumor types. Of note, Notch has been reported to have both oncogenic and tumor suppressor roles, dependent on the cancer cell type. In this review, we will first give a general description of the Notch signaling pathway, and its physiologic role in hematopoiesis. Next, we will review the role of aberrant Notch signaling in several hematological malignancies. Finally, we will discuss current and potential future therapeutic approaches targeting this pathway.
Collapse
|
83
|
Abstract
Notch is commonly activated in lymphoid malignancies through ligand-independent and ligand-dependent mechanisms. In T-cell acute lymphoblastic leukemia/lymphoma (T-ALL), ligand-independent activation predominates. Negative Regulatory Region (NRR) mutations trigger supraphysiological Notch1 activation by exposing the S2 site to proteolytic cleavage in the absence of ligand. Subsequently, cleavage at the S3 site generates the activated form of Notch, intracellular Notch (ICN). In contrast to T-ALL, in mature lymphoid neoplasms such as chronic lymphocytic leukemia (CLL), the S2 cleavage site is exposed through ligand-receptor interactions. Thus, agents that disrupt ligand-receptor interactions might be useful for treating these malignancies. Notch activation can be enhanced by mutations that delete the C-terminal proline (P), glutamic acid (E), serine (S), and threonine (T) (PEST) domain. These mutations do not activate the Notch pathway per se, but rather impair degradation of ICN. In this chapter, we review the mechanisms of Notch activation and the importance of Notch for the genesis and maintenance of lymphoid malignancies. Unfortunately, targeting the Notch pathway with pan-Notch inhibitors in clinical trials has proven challenging. These clinical trials have encountered dose-limiting on-target toxicities and primary resistance. Strategies to overcome these challenges have emerged from the identification and improved understanding of direct oncogenic Notch target genes. Other strategies have arisen from new insights into the "nuclear context" that selectively directs Notch functions in lymphoid cancers. This nuclear context is created by factors that co-bind ICN at cell-type specific transcriptional regulatory elements. Disrupting the functions of these proteins or inhibiting downstream oncogenic pathways might combat cancer without the intolerable side effects of pan-Notch inhibition.
Collapse
|
84
|
Adamowicz M. Breaking up with ATM. JOURNAL OF IMMUNOLOGICAL SCIENCES 2018; 2:26-31. [PMID: 29652413 PMCID: PMC5892715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
ATM kinase is a master regulator of the DNA damage response (DDR). A recently published report from the d'Adda di Fagagna laboratory1 sheds a light onto our understanding of ATM activation. In this short-commentary we will expand on this and other work to perceive better some of the aspects of ATM regulation.
Collapse
Affiliation(s)
- Marek Adamowicz
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9RH, UK,Correspondence: Dr. Marek Adamowicz, Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9RH, UK;
| |
Collapse
|
85
|
Ye F. MicroRNA expression and activity in T-cell acute lymphoblastic leukemia. Oncotarget 2017; 9:5445-5458. [PMID: 29435192 PMCID: PMC5797063 DOI: 10.18632/oncotarget.23539] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 12/01/2017] [Indexed: 12/21/2022] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a lymphoid malignancy caused by the oncogenic transformation of immature T-cell progenitors. Many biologically relevant genetic and epigenetic alterations have been identified as driving factors for this transformation. Recently, microRNAs (miRNAs) have been shown to influence various leukemias, including T-ALL. Aberrant expression of miRNAs can function as either oncogenes or tumor suppressors in T-ALL through the regulation of cell migration, invasion, proliferation, apoptosis, and chemoresistance. This occurs by targeting key signaling pathways or transcriptional factors that play a critical role in T-ALL pathology and progression. Different miRNA expression profiles have been linked to specific genetic subtypes of human T-ALL. Furthermore, miRNAs can also act as independent prognostic factors to predict clinical outcomes for T-ALL patients. In the current review, we will focus on the role of miRNAs in the development and progression of T-ALL.
Collapse
Affiliation(s)
- Fang Ye
- Department of Hematology, Beijing Chuiyangliu Hospital Affiliated to Tsinghua University, Beijing, China
| |
Collapse
|
86
|
Pajcini KV, Xu L, Shao L, Petrovic J, Palasiewicz K, Ohtani Y, Bailis W, Lee C, Wertheim GB, Mani R, Muthusamy N, Li Y, Meijerink JPP, Blacklow SC, Faryabi RB, Cherry S, Pear WS. MAFB enhances oncogenic Notch signaling in T cell acute lymphoblastic leukemia. Sci Signal 2017; 10:10/505/eaam6846. [PMID: 29138297 DOI: 10.1126/scisignal.aam6846] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Activating mutations in the gene encoding the cell-cell contact signaling protein Notch1 are common in human T cell acute lymphoblastic leukemias (T-ALLs). However, expressing Notch1 mutant alleles in mice fails to efficiently induce the development of leukemia. We performed a gain-of-function screen to identify proteins that enhanced signaling by leukemia-associated Notch1 mutants. The transcription factors MAFB and ETS2 emerged as candidates that individually enhanced Notch1 signaling, and when coexpressed, they synergistically increased signaling to an extent similar to that induced by core components of the Notch transcriptional complex. In mouse models of T-ALL, MAFB enhanced leukemogenesis by the naturally occurring Notch1 mutants, decreased disease latency, and increased disease penetrance. Decreasing MAFB abundance in mouse and human T-ALL cells reduced the expression of Notch1 target genes, including MYC and HES1, and sustained MAFB knockdown impaired T-ALL growth in a competitive setting. MAFB bound to ETS2 and interacted with the acetyltransferases PCAF and P300, highlighting its importance in recruiting coactivators that enhance Notch1 signaling. Together, these data identify a mechanism for enhancing the oncogenic potential of weak Notch1 mutants in leukemia models, and they reveal the MAFB-ETS2 transcriptional axis as a potential therapeutic target in T-ALL.
Collapse
Affiliation(s)
- Kostandin V Pajcini
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA. .,Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lanwei Xu
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lijian Shao
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jelena Petrovic
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karol Palasiewicz
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Yumi Ohtani
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Will Bailis
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Curtis Lee
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gerald B Wertheim
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rajeswaran Mani
- The James, Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Natarajan Muthusamy
- The James, Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Yunlei Li
- Department of Pediatric Oncology/Hematology, Erasmus Medical Center, Rotterdam, Netherlands
| | | | - Stephen C Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Robert B Faryabi
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sara Cherry
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Warren S Pear
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA. .,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
87
|
Li J, Qin Y, Zhang H. Identification of key miRNA-gene pairs in chronic lymphocytic leukemia through integrated analysis of mRNA and miRNA microarray. Oncol Lett 2017; 15:361-367. [PMID: 29285196 PMCID: PMC5738675 DOI: 10.3892/ol.2017.7287] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 10/17/2017] [Indexed: 12/17/2022] Open
Abstract
The aim of the present study was to explore the miRNA-Gene regulatory mechanism in chronic lymphocytic leukemia (CLL), and identify new targets for the therapy of CLL. The miRNA expression dataset GSE62137 and mRNA expression dataset GSE22529 were downloaded from National Center of Biotechnology Information Gene Expression Omnibus database. In CLL samples compared with normal B cell samples, differentially expressed miRNAs (DEMs) were identified via the GEO2R instrument of GEO and differentially expressed genes (DEGs) were obtained via the limma package of R. Functional enrichment analysis of the DEGs was performed via the Database for Annotation, Visualization and Integrated Discovery. The targets of the DEMs were identified based on the miRNAWalk platform. The overlaps between the DEGs and the targets of the DEMs were selected, and the miRNA-Gene regulatory network was constructed based on the overlaps and the corresponding DEMs. A total of 63 DEMs and 504 DEGs were identified in CLL samples compared with normal B cell samples. Eleven enriched functional clusters of the DEGs were obtained. 405 miRNA-Gene regulatory pairs were identified. The miRNA-Gene regulatory pairs contained 351 target genes of the DEMs, including 9 overlaps with the DEGs. A miRNA-Gene regulatory network was constructed. Bioinformatics methods could help us develop a better understanding of the molecular mechanism of CLL. MiRNAs may play a critical role in regulating the process of CLL. They may affect CLL by regulating the processes of immunoreactivity and protein degradation. Genes such as Neurogenic Locus Notch Homolog Protein 2, PR/SET domain 4 and A-kinase anchoring protein 12 may be their regulating targets in CLL.
Collapse
Affiliation(s)
- Jie Li
- Department of Transfusion Medicine, The Fifth Central Hospital of Tianjin, Tianjin 300450, P.R. China
| | - Yi Qin
- Institute of Medical Laboratory, Tianjin Medical University, Tianjin 300072, P.R. China
| | - Haiyan Zhang
- Department of Medical Record Management, The Fifth Central Hospital of Tianjin, Tianjin 300450, P.R. China
| |
Collapse
|
88
|
Choi SH, Severson E, Pear WS, Liu XS, Aster JC, Blacklow SC. The common oncogenomic program of NOTCH1 and NOTCH3 signaling in T-cell acute lymphoblastic leukemia. PLoS One 2017; 12:e0185762. [PMID: 29023469 PMCID: PMC5638296 DOI: 10.1371/journal.pone.0185762] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 09/19/2017] [Indexed: 11/24/2022] Open
Abstract
Notch is a major oncogenic driver in T cell acute lymphoblastic leukemia (T-ALL), in part because it binds to an enhancer that increases expression of MYC. Here, we exploit the capacity of activated NOTCH1 and NOTCH3 to induce T-ALL, despite substantial divergence in their intracellular regions, as a means to elucidate a broad, common Notch-dependent oncogenomic program through systematic comparison of the transcriptomes and Notch-bound genomic regulatory elements of NOTCH1- and NOTCH3-dependent T-ALL cells. ChIP-seq studies show a high concordance of functional NOTCH1 and NOTCH3 genomic binding sites that are enriched in binding motifs for RBPJ, the transcription factor that recruits activated Notch to DNA. The interchangeability of NOTCH1 and NOTCH3 was confirmed by rescue of NOTCH1-dependent T-ALL cells with activated NOTCH3 and vice versa. Despite remarkable overall similarity, there are nuanced differences in chromatin landscapes near critical common Notch target genes, most notably at a Notch-dependent enhancer that regulates MYC, which correlates with responsiveness to Notch pathway inhibitors. Overall, a common oncogenomic program driven by binding of either Notch is sufficient to maintain T-ALL cell growth, whereas cell-context specific differences appear to influence the response of T-ALL cells to Notch inhibition.
Collapse
Affiliation(s)
- Sung Hee Choi
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, United States of America
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA, United States of America
| | - Eric Severson
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States of America
- Departments of Biostatistics and Computational Biology, Dana Farber Cancer Institute, and Harvard School of Public Health, Boston, MA, United States of America
| | - Warren S. Pear
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States of America
| | - Xiaole S. Liu
- Departments of Biostatistics and Computational Biology, Dana Farber Cancer Institute, and Harvard School of Public Health, Boston, MA, United States of America
| | - Jon C. Aster
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States of America
- * E-mail: (SCB); (JCA)
| | - Stephen C. Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, United States of America
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA, United States of America
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States of America
- * E-mail: (SCB); (JCA)
| |
Collapse
|
89
|
Tan SH, Bertulfo FC, Sanda T. Leukemia-Initiating Cells in T-Cell Acute Lymphoblastic Leukemia. Front Oncol 2017; 7:218. [PMID: 29034206 PMCID: PMC5627022 DOI: 10.3389/fonc.2017.00218] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 09/01/2017] [Indexed: 12/26/2022] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a hematological malignancy characterized by the clonal proliferation of immature T-cell precursors. T-ALL has many similar pathophysiological features to acute myeloid leukemia, which has been extensively studied in the establishment of the cancer stem cell (CSC) theory, but the CSC concept in T-ALL is still debatable. Although leukemia-initiating cells (LICs), which can generate leukemia in a xenograft setting, have been found in both human T-ALL patients and animal models, the nature and origin of LICs are largely unknown. In this review, we discuss recent studies on LICs in T-ALL and the potential mechanisms of LIC emergence in this disease. We focus on the oncogenic transcription factors TAL1, LMO2, and NOTCH1 and highlight the significance of the transcriptional regulatory programs in normal hematopoietic stem cells and T-ALL.
Collapse
Affiliation(s)
- Shi Hao Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Fatima Carla Bertulfo
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Takaomi Sanda
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
90
|
Liu Y, Easton J, Shao Y, Maciaszek J, Wang Z, Wilkinson MR, McCastlain K, Edmonson M, Pounds SB, Shi L, Zhou X, Ma X, Sioson E, Li Y, Rusch M, Gupta P, Pei D, Cheng C, Smith MA, Auvil JG, Gerhard DS, Relling MV, Winick NJ, Carroll AJ, Heerema NA, Raetz E, Devidas M, Willman CL, Harvey RC, Carroll WL, Dunsmore KP, Winter SS, Wood BL, Sorrentino BP, Downing JR, Loh ML, Hunger SP, Zhang J, Mullighan CG. The genomic landscape of pediatric and young adult T-lineage acute lymphoblastic leukemia. Nat Genet 2017; 49. [PMID: 28671688 PMCID: PMC5535770 DOI: 10.1038/ng.3909 10.1182/ng.3909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Genetic alterations that activate NOTCH1 signaling and T cell transcription factors, coupled with inactivation of the INK4/ARF tumor suppressors, are hallmarks of T-lineage acute lymphoblastic leukemia (T-ALL), but detailed genome-wide sequencing of large T-ALL cohorts has not been carried out. Using integrated genomic analysis of 264 T-ALL cases, we identified 106 putative driver genes, half of which had not previously been described in childhood T-ALL (for example, CCND3, CTCF, MYB, SMARCA4, ZFP36L2 and MYCN). We describe new mechanisms of coding and noncoding alteration and identify ten recurrently altered pathways, with associations between mutated genes and pathways, and stage or subtype of T-ALL. For example, NRAS/FLT3 mutations were associated with immature T-ALL, JAK3/STAT5B mutations in HOXA1 deregulated ALL, PTPN2 mutations in TLX1 deregulated T-ALL, and PIK3R1/PTEN mutations in TAL1 deregulated ALL, which suggests that different signaling pathways have distinct roles according to maturational stage. This genomic landscape provides a logical framework for the development of faithful genetic models and new therapeutic approaches.
Collapse
Affiliation(s)
- Yu Liu
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - John Easton
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Ying Shao
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States,Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Jamie Maciaszek
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Zhaoming Wang
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Mark R. Wilkinson
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Kelly McCastlain
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Michael Edmonson
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Stanley B. Pounds
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Lei Shi
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Xin Zhou
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Xiaotu Ma
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Edgar Sioson
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Yongjin Li
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Michael Rusch
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Pankaj Gupta
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Deqing Pei
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Cheng Cheng
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Malcolm A. Smith
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland, United States
| | - Jaime Guidry Auvil
- Office of Cancer Genomics, National Cancer Institute, Bethesda, Maryland United States
| | - Daniela S. Gerhard
- Office of Cancer Genomics, National Cancer Institute, Bethesda, Maryland United States
| | - Mary V. Relling
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Naomi J. Winick
- University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Andrew J. Carroll
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Nyla A. Heerema
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Elizabeth Raetz
- Department of Pediatrics, Huntsman Cancer Institute and Primary Children’s Hospital, University of Utah, Salt Lake City, Utah, United States
| | - Meenakshi Devidas
- Department of Biostatistics, Colleges of Medicine, Public Health & Health Profession, University of Florida, Gainesville, Florida, United States
| | - Cheryl L. Willman
- Department of Pathology, The Cancer Research and Treatment Center, University of New Mexico, Albuquerque, New Mexico, United States
| | - Richard C. Harvey
- Department of Pathology, The Cancer Research and Treatment Center, University of New Mexico, Albuquerque, New Mexico, United States
| | - William L. Carroll
- Department of Pediatrics, Perlmutter Cancer Center, New York University Medical Center, New York, New York, United States
| | - Kimberly P. Dunsmore
- Health Sciences Center, University of Virginia, Charlottesville, Virginia, United States
| | - Stuart S. Winter
- Department of Pediatrics, University of New Mexico, Albuquerque, New Mexico, United States
| | - Brent L Wood
- Seattle Cancer Care Alliance, Seattle, Washington, United States
| | - Brian P. Sorrentino
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - James R. Downing
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Mignon L. Loh
- Department of Pediatrics, Benioff Children’s Hospital, University of California at San Francisco, San Francisco, California, United States
| | - Stephen P Hunger
- Department of Pediatrics and the Center for Childhood Cancer Research, Children’s Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States,Address for correspondence: Stephen P. Hunger, Children’s Hospital of Philadelphia, CTRB #3060, 3501 Civic Center Boulevard, Philadelphia, PA 19104, ; Jinghui Zhang, St. Jude Children’s Research Hospital, Department of Computational Biology, 262 Danny Thomas Place, Mail Stop 1135, Memphis, TN 38105, T: 1-901-595- 6829, ; Charles G. Mullighan, St. Jude Children’s Research Hospital, Department of Pathology, Mail Stop 342, 262 Danny Thomas Place, Memphis, TN 38105, T: 1-901-595-3387, F: 1-901-595-5947,
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States,Address for correspondence: Stephen P. Hunger, Children’s Hospital of Philadelphia, CTRB #3060, 3501 Civic Center Boulevard, Philadelphia, PA 19104, ; Jinghui Zhang, St. Jude Children’s Research Hospital, Department of Computational Biology, 262 Danny Thomas Place, Mail Stop 1135, Memphis, TN 38105, T: 1-901-595- 6829, ; Charles G. Mullighan, St. Jude Children’s Research Hospital, Department of Pathology, Mail Stop 342, 262 Danny Thomas Place, Memphis, TN 38105, T: 1-901-595-3387, F: 1-901-595-5947,
| | - Charles G. Mullighan
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States,Address for correspondence: Stephen P. Hunger, Children’s Hospital of Philadelphia, CTRB #3060, 3501 Civic Center Boulevard, Philadelphia, PA 19104, ; Jinghui Zhang, St. Jude Children’s Research Hospital, Department of Computational Biology, 262 Danny Thomas Place, Mail Stop 1135, Memphis, TN 38105, T: 1-901-595- 6829, ; Charles G. Mullighan, St. Jude Children’s Research Hospital, Department of Pathology, Mail Stop 342, 262 Danny Thomas Place, Memphis, TN 38105, T: 1-901-595-3387, F: 1-901-595-5947,
| |
Collapse
|
91
|
Liu Y, Easton J, Shao Y, Maciaszek J, Wang Z, Wilkinson MR, McCastlain K, Edmonson M, Pounds SB, Shi L, Zhou X, Ma X, Sioson E, Li Y, Rusch M, Gupta P, Pei D, Cheng C, Smith MA, Auvil JG, Gerhard DS, Relling MV, Winick NJ, Carroll AJ, Heerema NA, Raetz E, Devidas M, Willman CL, Harvey RC, Carroll WL, Dunsmore KP, Winter SS, Wood BL, Sorrentino BP, Downing JR, Loh ML, Hunger SP, Zhang J, Mullighan CG. The genomic landscape of pediatric and young adult T-lineage acute lymphoblastic leukemia. Nat Genet 2017; 49:1211-1218. [PMID: 28671688 PMCID: PMC5535770 DOI: 10.1038/ng.3909] [Citation(s) in RCA: 679] [Impact Index Per Article: 84.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 06/09/2017] [Indexed: 12/11/2022]
Abstract
Genetic alterations that activate NOTCH1 signaling and T cell transcription factors, coupled with inactivation of the INK4/ARF tumor suppressors, are hallmarks of T-lineage acute lymphoblastic leukemia (T-ALL), but detailed genome-wide sequencing of large T-ALL cohorts has not been carried out. Using integrated genomic analysis of 264 T-ALL cases, we identified 106 putative driver genes, half of which had not previously been described in childhood T-ALL (for example, CCND3, CTCF, MYB, SMARCA4, ZFP36L2 and MYCN). We describe new mechanisms of coding and noncoding alteration and identify ten recurrently altered pathways, with associations between mutated genes and pathways, and stage or subtype of T-ALL. For example, NRAS/FLT3 mutations were associated with immature T-ALL, JAK3/STAT5B mutations in HOXA1 deregulated ALL, PTPN2 mutations in TLX1 deregulated T-ALL, and PIK3R1/PTEN mutations in TAL1 deregulated ALL, which suggests that different signaling pathways have distinct roles according to maturational stage. This genomic landscape provides a logical framework for the development of faithful genetic models and new therapeutic approaches.
Collapse
Affiliation(s)
- Yu Liu
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - John Easton
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Ying Shao
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Jamie Maciaszek
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Zhaoming Wang
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Mark R. Wilkinson
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Kelly McCastlain
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Michael Edmonson
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Stanley B. Pounds
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Lei Shi
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Xin Zhou
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Xiaotu Ma
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Edgar Sioson
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Yongjin Li
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Michael Rusch
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Pankaj Gupta
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Deqing Pei
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Cheng Cheng
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Malcolm A. Smith
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland, United States
| | - Jaime Guidry Auvil
- Office of Cancer Genomics, National Cancer Institute, Bethesda, Maryland United States
| | - Daniela S. Gerhard
- Office of Cancer Genomics, National Cancer Institute, Bethesda, Maryland United States
| | - Mary V. Relling
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Naomi J. Winick
- University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Andrew J. Carroll
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Nyla A. Heerema
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Elizabeth Raetz
- Department of Pediatrics, Huntsman Cancer Institute and Primary Children’s Hospital, University of Utah, Salt Lake City, Utah, United States
| | - Meenakshi Devidas
- Department of Biostatistics, Colleges of Medicine, Public Health & Health Profession, University of Florida, Gainesville, Florida, United States
| | - Cheryl L. Willman
- Department of Pathology, The Cancer Research and Treatment Center, University of New Mexico, Albuquerque, New Mexico, United States
| | - Richard C. Harvey
- Department of Pathology, The Cancer Research and Treatment Center, University of New Mexico, Albuquerque, New Mexico, United States
| | - William L. Carroll
- Department of Pediatrics, Perlmutter Cancer Center, New York University Medical Center, New York, New York, United States
| | - Kimberly P. Dunsmore
- Health Sciences Center, University of Virginia, Charlottesville, Virginia, United States
| | - Stuart S. Winter
- Department of Pediatrics, University of New Mexico, Albuquerque, New Mexico, United States
| | - Brent L Wood
- Seattle Cancer Care Alliance, Seattle, Washington, United States
| | - Brian P. Sorrentino
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - James R. Downing
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Mignon L. Loh
- Department of Pediatrics, Benioff Children’s Hospital, University of California at San Francisco, San Francisco, California, United States
| | - Stephen P Hunger
- Department of Pediatrics and the Center for Childhood Cancer Research, Children’s Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Charles G. Mullighan
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
| |
Collapse
|
92
|
Sanda T, Leong WZ. TAL1 as a master oncogenic transcription factor in T-cell acute lymphoblastic leukemia. Exp Hematol 2017; 53:7-15. [PMID: 28652130 DOI: 10.1016/j.exphem.2017.06.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 06/11/2017] [Indexed: 11/29/2022]
Abstract
In hematopoietic cell development, the transcriptional program is strictly regulated in a lineage- and stage-specific manner that requires a number of transcription factors to work in a cascade or in a loop, in addition to interactions with nonhematopoietic cells in the microenvironment. Disruption of the transcriptional program alters the cellular state and may predispose cells to the acquisition of genetic abnormalities. Early studies have shown that proteins that promote cell differentiation often serve as tumor suppressors, whereas inhibitors of those proteins act as oncogenes in the context of acute leukemia. A prime example is T-cell acute lymphoblastic leukemia (T-ALL), a malignant disorder characterized by clonal proliferation of immature stage thymocytes. Although a relatively small number of genetic abnormalities are observed in T-ALL, these abnormalities are crucial for leukemogenesis. Many oncogenes and tumor suppressors in T-ALL are transcription factors that are required for normal hematopoiesis. The transformation process in T-ALL is efficient and orchestrated; the oncogene disrupts the transcriptional program directing T-cell differentiation and also uses its native ability as a master transcription factor in hematopoiesis. This imbalance in the transcriptional program is a primary determinant underlying the molecular pathogenesis of T-ALL. In this review, we focus on the oncogenic transcription factor TAL1 and the tumor-suppressor E-proteins and discuss the malignant cell state, the transcriptional circuit, and the consequence of molecular abnormalities in T-ALL.
Collapse
Affiliation(s)
- Takaomi Sanda
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Wei Zhong Leong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| |
Collapse
|
93
|
Oliveira ML, Akkapeddi P, Alcobia I, Almeida AR, Cardoso BA, Fragoso R, Serafim TL, Barata JT. From the outside, from within: Biological and therapeutic relevance of signal transduction in T-cell acute lymphoblastic leukemia. Cell Signal 2017. [PMID: 28645565 DOI: 10.1016/j.cellsig.2017.06.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
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.
Collapse
Affiliation(s)
- Mariana L Oliveira
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Padma Akkapeddi
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Isabel Alcobia
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal; Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Afonso R Almeida
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Bruno A Cardoso
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Rita Fragoso
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Teresa L Serafim
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - João T Barata
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal.
| |
Collapse
|
94
|
Ding J, Fishel ML, Reed AM, McAdams E, Czader MB, Cardoso AA, Kelley MR. Ref-1/APE1 as a Transcriptional Regulator and Novel Therapeutic Target in Pediatric T-cell Leukemia. Mol Cancer Ther 2017; 16:1401-1411. [PMID: 28446640 DOI: 10.1158/1535-7163.mct-17-0099] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/23/2017] [Accepted: 04/14/2017] [Indexed: 12/14/2022]
Abstract
The increasing characterization of childhood acute lymphoblastic leukemia (ALL) has led to the identification of multiple molecular targets but has yet to translate into more effective targeted therapies, particularly for high-risk, relapsed T-cell ALL. Searching for master regulators controlling multiple signaling pathways in T-ALL, we investigated the multifunctional protein redox factor-1 (Ref-1/APE1), which acts as a signaling "node" by exerting redox regulatory control of transcription factors important in leukemia. Leukemia patients' transcriptome databases showed increased expression in T-ALL of Ref-1 and other genes of the Ref-1/SET interactome. Validation studies demonstrated that Ref-1 is expressed in high-risk leukemia T cells, including in patient biopsies. Ref-1 redox function is active in leukemia T cells, regulating the Ref-1 target NF-κB, and inhibited by the redox-selective Ref-1 inhibitor E3330. Ref-1 expression is not regulated by Notch signaling, but is upregulated by glucocorticoid treatment. E3330 disrupted Ref-1 redox activity in functional studies and resulted in marked inhibition of leukemia cell viability, including T-ALL lines representing different genotypes and risk groups. Potent leukemia cell inhibition was seen in primary cells from ALL patients, relapsed and glucocorticoid-resistant T-ALL cells, and cells from a murine model of Notch-induced leukemia. Ref-1 redox inhibition triggered leukemia cell apoptosis and downregulation of survival genes regulated by Ref-1 targets. For the first time, this work identifies Ref-1 as a novel molecular effector in T-ALL and demonstrates that Ref-1 redox inhibition results in potent inhibition of leukemia T cells, including relapsed T-ALL. These data also support E3330 as a specific Ref-1 small-molecule inhibitor for leukemia. Mol Cancer Ther; 16(7); 1401-11. ©2017 AACR.
Collapse
Affiliation(s)
- Jixin Ding
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Melissa L Fishel
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana.,Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana
| | - April M Reed
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Erin McAdams
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Magdalena B Czader
- Department of Pathology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Angelo A Cardoso
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Mark R Kelley
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana. .,Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
95
|
Dnmt3a regulates T-cell development and suppresses T-ALL transformation. Leukemia 2017; 31:2479-2490. [PMID: 28321121 PMCID: PMC5636646 DOI: 10.1038/leu.2017.89] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 03/13/2017] [Accepted: 03/16/2017] [Indexed: 12/30/2022]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematopoietic neoplasm resulting from the malignant transformation of T-cell progenitors, and comprises approximately 15% and 25% of pediatric and adult ALL cases respectively. It is well-established that activating NOTCH1 mutations are the major genetic lesions driving T-ALL in most patients, but efforts to develop targeted therapies against this pathway have produced limited success in decreasing leukemic burden and come with significant clinical side effects. A finer detailed understanding of the genetic and molecular mechanisms underlying T-ALL is required identify patients at increased risk for treatment failure and the development of precision medicine strategies. Generation of genetic models that more accurately reflect the normal developmental history of T-ALL are necessary to identify new avenues for treatment. The DNA methyltransferase enzyme DNMT3A is also recurrently mutated in T-ALL patients, and we show here that inactivation of Dnmt3a combined with Notch1 gain-of-function leads to an aggressive T-ALL in mouse models. Moreover, conditional inactivation of Dnmt3a in mouse hematopoietic cells leads to an accumulation of immature progenitors in the thymus which are less apoptotic. These data demonstrate that Dnmt3a is required for normal T-cell development, and acts as a T-ALL tumor suppressor.
Collapse
|
96
|
Wallaert A, Durinck K, Taghon T, Van Vlierberghe P, Speleman F. T-ALL and thymocytes: a message of noncoding RNAs. J Hematol Oncol 2017; 10:66. [PMID: 28270163 PMCID: PMC5341419 DOI: 10.1186/s13045-017-0432-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 02/24/2017] [Indexed: 02/06/2023] Open
Abstract
In the last decade, the role for noncoding RNAs in disease was clearly established, starting with microRNAs and later expanded towards long noncoding RNAs. This was also the case for T cell acute lymphoblastic leukemia, which is a malignant blood disorder arising from oncogenic events during normal T cell development in the thymus. By studying the transcriptomic profile of protein-coding genes, several oncogenic events leading to T cell acute lymphoblastic leukemia (T-ALL) could be identified. In recent years, it became apparent that several of these oncogenes function via microRNAs and long noncoding RNAs. In this review, we give a detailed overview of the studies that describe the noncoding RNAome in T-ALL oncogenesis and normal T cell development.
Collapse
Affiliation(s)
- Annelynn Wallaert
- Center for Medical Genetics, Ghent University, Ghent, Belgium. .,Cancer Research Institute Ghent, Ghent, Belgium.
| | - Kaat Durinck
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Tom Taghon
- Cancer Research Institute Ghent, Ghent, Belgium.,Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Ghent, Belgium
| | - Pieter Van Vlierberghe
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Frank Speleman
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
97
|
Abstract
The Notch signalling cascade is an evolutionarily conserved pathway that has a crucial role in regulating development and homeostasis in various tissues. The cellular processes and events that it controls are diverse, and continued investigation over recent decades has revealed how the role of Notch signalling is multifaceted and highly context dependent. Consistent with the far-reaching impact that Notch has on development and homeostasis, aberrant activity of the pathway is also linked to the initiation and progression of several malignancies, and Notch can in fact be either oncogenic or tumour suppressive depending on the tissue and cellular context. The Notch pathway therefore represents an important target for therapeutic agents designed to treat many types of cancer. In this Review, we focus on the latest developments relating specifically to the tumour-suppressor activity of Notch signalling and discuss the potential mechanisms by which Notch can inhibit carcinogenesis in various tissues. Potential therapeutic strategies aimed at restoring or augmenting Notch-mediated tumour suppression will also be highlighted.
Collapse
Affiliation(s)
- Craig S Nowell
- CMU, Department for Pathology and Immunology, University of Geneva, Rue Michel Servet, 1211 Geneva 4, Switzerland
| | - Freddy Radtke
- Ecole Polytechnique Fédérale de Lausanne, School of Life Sciences, Swiss Institute for Experimental Cancer Research, Lausanne, Vaud 1015, Switzerland
| |
Collapse
|
98
|
Tan Y, Sementino E, Xu J, Pei J, Liu Z, Ito TK, Cai KQ, Peri S, Klein-Szanto AJP, Wiest DL, Testa JR. The homeoprotein Dlx5 drives murine T-cell lymphomagenesis by directly transactivating Notch and upregulating Akt signaling. Oncotarget 2017; 8:14941-14956. [PMID: 28122332 PMCID: PMC5362456 DOI: 10.18632/oncotarget.14784] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/11/2017] [Indexed: 12/01/2022] Open
Abstract
Homeobox genes play a critical role in embryonic development, but they have also been implicated in cancer through mechanisms that are largely unknown. While not expressed during normal T-cell development, homeobox transcription factor genes can be reactivated via recurrent chromosomal rearrangements in human T-cell acute leukemia/lymphoma (T-ALL), a malignancy often associated with activated Notch and Akt signaling. To address how epigenetic reprogramming via an activated homeobox gene might contribute to T-lymphomagenesis, we investigated a transgenic mouse model with thymocyte-specific overexpression of the Dlx5 homeobox gene. We demonstrate for the first time that Dlx5 induces T-cell lymphomas with high penetrance. Integrated ChIP-seq and mRNA microarray analyses identified Notch1/3 and Irs2 as direct transcriptional targets of Dlx5, a gene signature unique to lymphomas from Lck-Dlx5 mice as compared to T-cell lymphomas from Lck-MyrAkt2 mice, which were previously reported by our group. Moreover, promoter/enhancer studies confirmed that Dlx5 directly transactivates Notch expression. Notch1/3 expression and Irs2-induced Akt signaling were upregulated throughout early stages of T-cell development, which promoted cell survival during β-selection of T lymphocytes. Dlx5 was required for tumor maintenance via its activation of Notch and Akt, as tumor cells were highly sensitive to Notch and Akt inhibitors. Together, these findings provide unbiased genetic and mechanistic evidence that Dlx5 acts as an oncogene when aberrantly expressed in T cells, and that it is a novel discovery that Notch is a direct target of Dlx5. These experimental findings provide mechanistic insights about how reactivation of the Dlx5 gene can drive T-ALL by aberrant epigenetic reprogramming of the T-cell genome.
Collapse
Affiliation(s)
- Yinfei Tan
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Eleonora Sementino
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Jinfei Xu
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Jianming Pei
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Zemin Liu
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Timothy K Ito
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Kathy Q Cai
- Histopathology Facility, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Suraj Peri
- Department of Biostatistics and Bioinformatics, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Andres J P Klein-Szanto
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
- Histopathology Facility, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - David L Wiest
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Joseph R Testa
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| |
Collapse
|
99
|
Nwaeburu CC, Abukiwan A, Zhao Z, Herr I. Quercetin-induced miR-200b-3p regulates the mode of self-renewing divisions in pancreatic cancer. Mol Cancer 2017; 16:23. [PMID: 28137273 PMCID: PMC5282715 DOI: 10.1186/s12943-017-0589-8] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 01/17/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Cancer stem cells are suggested to contribute to the extremely poor prognosis of pancreatic ductal adenocarcinoma and dysregulation of symmetric and asymmetric stem cell division may be involved. Anticancer benefits of phytochemicals like the polyphenol quercetin, present in many fruits, nuts and vegetables, could be expedited by microRNAs, which orchestrate cell-fate decisions and tissue homeostasis. The mechanisms regulating the division mode of cancer stem cells in relation to phytochemical-induced microRNAs are poorly understood. METHODS Patient-derived pancreas tissue and 3 established pancreatic cancer cell lines were examined by immunofluorescence and time-lapse microscopy, microRNA microarray analysis, bioinformatics and computational analysis, qRT-PCR, Western blot analysis, self-renewal and differentiation assays. RESULTS We show that symmetric and asymmetric division occurred in patient tissues and in vitro, whereas symmetric divisions were more extensive. By microarray analysis, bioinformatics prediction and qRT-PCR, we identified and validated quercetin-induced microRNAs involved in Notch signaling/cell-fate determination. Further computational analysis distinguished miR-200b-3p as strong candidate for cell-fate determinant. Mechanistically, miR-200b-3p switched symmetric to asymmetric cell division by reversing the Notch/Numb ratio, inhibition of the self-renewal and activation of the potential to differentiate to adipocytes, osteocytes and chondrocytes. Low miR-200b-3p levels fostered Notch signaling and promoted daughter cells to become symmetric while high miR-200b-3p levels lessened Notch signaling and promoted daughter cells to become asymmetric. CONCLUSIONS Our findings provide a better understanding of the cross talk between phytochemicals, microRNAs and Notch signaling in the regulation of self-renewing cancer stem cell divisions.
Collapse
Affiliation(s)
- Clifford C Nwaeburu
- Department of General, Molecular OncoSurgery, Section Surgical Research, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 365, 69120, Heidelberg, Germany
| | - Alia Abukiwan
- Department of General, Molecular OncoSurgery, Section Surgical Research, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 365, 69120, Heidelberg, Germany
| | - Zhefu Zhao
- Department of General, Molecular OncoSurgery, Section Surgical Research, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 365, 69120, Heidelberg, Germany
| | - Ingrid Herr
- Department of General, Molecular OncoSurgery, Section Surgical Research, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 365, 69120, Heidelberg, Germany.
| |
Collapse
|
100
|
The NOTCH1-MYC highway toward T-cell acute lymphoblastic leukemia. Blood 2017; 129:1124-1133. [PMID: 28115368 DOI: 10.1182/blood-2016-09-692582] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 11/14/2016] [Indexed: 12/21/2022] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a highly proliferative hematologic malignancy that results from the transformation of immature T-cell progenitors. Aberrant cell growth and proliferation in T-ALL lymphoblasts are sustained by activation of strong oncogenic drivers promoting cell anabolism and cell cycle progression. Oncogenic NOTCH signaling, which is activated in more than 65% of T-ALL patients by activating mutations in the NOTCH1 gene, has emerged as a major regulator of leukemia cell growth and metabolism. T-ALL NOTCH1 mutations result in ligand-independent and sustained NOTCH1-receptor signaling, which translates into activation of a broad transcriptional program dominated by upregulation of genes involved in anabolic pathways. Among these, the MYC oncogene plays a major role in NOTCH1-induced transformation. As result, the oncogenic activity of NOTCH1 in T-ALL is strictly dependent on MYC upregulation, which makes the NOTCH1-MYC regulatory circuit an attractive therapeutic target for the treatment of T-ALL.
Collapse
|