1
|
Peramangalam PS, Surapally S, Veltri AJ, Zheng S, Burns R, Zhu N, Rao S, Muller-Tidow C, Bushweller JH, Pulikkan JA. N-MYC regulates cell survival via eIF4G1 in inv(16) acute myeloid leukemia. SCIENCE ADVANCES 2024; 10:eadh8493. [PMID: 38416825 PMCID: PMC10901375 DOI: 10.1126/sciadv.adh8493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 01/24/2024] [Indexed: 03/01/2024]
Abstract
N-MYC (encoded by MYCN) is a critical regulator of hematopoietic stem cell function. While the role of N-MYC deregulation is well established in neuroblastoma, the importance of N-MYC deregulation in leukemogenesis remains elusive. Here, we demonstrate that N-MYC is overexpressed in acute myeloid leukemia (AML) cells with chromosome inversion inv(16) and contributes to the survival and maintenance of inv(16) leukemia. We identified a previously unknown MYCN enhancer, active in multiple AML subtypes, essential for MYCN mRNA levels and survival in inv(16) AML cells. We also identified eukaryotic translation initiation factor 4 gamma 1 (eIF4G1) as a key N-MYC target that sustains leukemic survival in inv(16) AML cells. The oncogenic role of eIF4G1 in AML has not been reported before. Our results reveal a mechanism whereby N-MYC drives a leukemic transcriptional program and provides a rationale for the therapeutic targeting of the N-MYC/eIF4G1 axis in myeloid leukemia.
Collapse
Affiliation(s)
| | - Sridevi Surapally
- Program in Stem Cell Biology and Hematopoiesis, Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Anthony J. Veltri
- Program in Stem Cell Biology and Hematopoiesis, Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Shikan Zheng
- Program in Stem Cell Biology and Hematopoiesis, Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Robert Burns
- Program in Stem Cell Biology and Hematopoiesis, Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Nan Zhu
- Program in Stem Cell Biology and Hematopoiesis, Versiti Blood Research Institute, Milwaukee, WI, USA
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sridhar Rao
- Program in Stem Cell Biology and Hematopoiesis, Versiti Blood Research Institute, Milwaukee, WI, USA
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, Division of Hematology, Oncology, and Transplantation, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Carsten Muller-Tidow
- Department of Medicine, Hematology, Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - John H. Bushweller
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - John A. Pulikkan
- Program in Stem Cell Biology and Hematopoiesis, Versiti Blood Research Institute, Milwaukee, WI, USA
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
2
|
Lamba JK, Cao X, Raimondi S, Downing J, Ribeiro R, Gruber TA, Rubnitz J, Pounds S. DNA Methylation Clusters and Their Relation to Cytogenetic Features in Pediatric AML. Cancers (Basel) 2020; 12:cancers12103024. [PMID: 33080932 PMCID: PMC7603219 DOI: 10.3390/cancers12103024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 10/15/2020] [Indexed: 12/13/2022] Open
Abstract
Acute Myeloid Leukemia (AML) is characterized by recurrent genetic and cytogenetic lesions that are utilized for risk stratification and for making treatment decisions. In recent years, methylation dysregulation has been extensively studied and associated with risk groups and prognosis in adult AML, however, such studies in pediatric AML are limited. Moreover, the mutations in epigenetic genes such as DNMT3A, IDH1 or IDH2 are almost absent or rare in pediatric patients as compared to their abundance in adult AML. In the current study, we evaluated methylation patterns that occur with or independent of the well-defined cytogenetic features in pediatric AML patients enrolled on multi-site AML02 clinical trial (NCT00136084). Our results demonstrate that unlike adult AML, cytosine DNA methylation does not result in significant unique clusters in pediatric AML, however, DNA methylation signatures correlated significantly with the most common and recurrent cytogenetic features. Paired evaluation of DNA methylation and expression identified genes and pathways of biological relevance that hold promise for novel therapeutic strategies. Our results further demonstrate that epigenetic signatures occur complimentary to the well-established chromosomal/mutational landscape, implying that dysregulation of oncogenes or tumor suppressors might be leveraging both genetic and epigenetic mechanisms to impact biological pathways critical for leukemogenesis.
Collapse
Affiliation(s)
- Jatinder K. Lamba
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL 32608, USA
- Correspondence:
| | - Xueyuan Cao
- Department of Acute and Tertiary Care, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Susana Raimondi
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (S.R.); (J.D.)
| | - James Downing
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (S.R.); (J.D.)
| | - Raul Ribeiro
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.R.); (J.R.)
| | - Tanja A. Gruber
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Jeffrey Rubnitz
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.R.); (J.R.)
| | - Stanley Pounds
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| |
Collapse
|
3
|
Pieters T, T'Sas S, Demoen L, Almeida A, Haenebalcke L, Matthijssens F, Lemeire K, D'Hont J, Van Rockeghem F, Hochepied T, Lintermans B, Reunes L, Lammens T, Berx G, Haigh JJ, Goossens S, Van Vlierberghe P. Novel strategy for rapid functional in vivo validation of oncogenic drivers in haematological malignancies. Sci Rep 2019; 9:10577. [PMID: 31332244 PMCID: PMC6646380 DOI: 10.1038/s41598-019-46853-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 07/05/2019] [Indexed: 12/17/2022] Open
Abstract
In cancer research, it remains challenging to functionally validate putative novel oncogenic drivers and to establish relevant preclinical models for evaluation of novel therapeutic strategies. Here, we describe an optimized and efficient pipeline for the generation of novel conditional overexpression mouse models in which putative oncogenes, along with an eGFP/Luciferase dual reporter, are expressed from the endogenous ROSA26 (R26) promoter. The efficiency of this approach was demonstrated by the generation and validation of novel R26 knock-in (KI) mice that allow conditional overexpression of Jarid2, Runx2, MN1 and a dominant negative allele of ETV6. As proof of concept, we confirm that MN1 overexpression in the hematopoietic lineage is sufficient to drive myeloid leukemia. In addition, we show that T-cell specific activation of MN1 in combination with loss of Pten increases tumour penetrance and stimulates the formation of Lyl1+ murine T-cell lymphoblastic leukemias or lymphomas (T-ALL/T-LBL). Finally, we demonstrate that these luciferase-positive murine AML and T-ALL/T-LBL cells are transplantable into immunocompromised mice allowing preclinical evaluation of novel anti-leukemic drugs in vivo.
Collapse
Affiliation(s)
- Tim Pieters
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,VIB Inflammation Research Center, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Sara T'Sas
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,VIB Inflammation Research Center, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Lisa Demoen
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - André Almeida
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Lieven Haenebalcke
- VIB Inflammation Research Center, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Filip Matthijssens
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Kelly Lemeire
- VIB Inflammation Research Center, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Jinke D'Hont
- VIB Inflammation Research Center, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Frederique Van Rockeghem
- VIB Inflammation Research Center, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Tino Hochepied
- VIB Inflammation Research Center, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Beatrice Lintermans
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Lindy Reunes
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Tim Lammens
- Cancer Research Institute Ghent, Ghent, Belgium.,Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Geert Berx
- VIB Inflammation Research Center, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Jody J Haigh
- Mammalian Functional Genetics Group, Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.,Research Institute in Oncology and Hematology, Cancer Care Manitoba, Winnipeg, Manitoba, Canada
| | - Steven Goossens
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium. .,VIB Inflammation Research Center, Ghent, Belgium. .,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium. .,Cancer Research Institute Ghent, Ghent, Belgium.
| | - Pieter Van Vlierberghe
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium. .,Cancer Research Institute Ghent, Ghent, Belgium.
| |
Collapse
|
4
|
Berger A, Brady NJ, Bareja R, Robinson B, Conteduca V, Augello MA, Puca L, Ahmed A, Dardenne E, Lu X, Hwang I, Bagadion AM, Sboner A, Elemento O, Paik J, Yu J, Barbieri CE, Dephoure N, Beltran H, Rickman DS. N-Myc-mediated epigenetic reprogramming drives lineage plasticity in advanced prostate cancer. J Clin Invest 2019; 129:3924-3940. [PMID: 31260412 DOI: 10.1172/jci127961] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Despite recent therapeutic advances, prostate cancer remains a leading cause of cancer-related death. A subset of castration resistant prostate cancers become androgen receptor (AR) signaling-independent and develop neuroendocrine prostate cancer (NEPC) features through lineage plasticity. These NEPC tumors, associated with aggressive disease and poor prognosis, are driven, in part, by aberrant expression of N-Myc, through mechanisms that remain unclear. Integrative analysis of the N-Myc transcriptome, cistrome and interactome using in vivo, in vitro and ex vivo models (including patient-derived organoids) identified a lineage switch towards a neural identity associated with epigenetic reprogramming. N-Myc and known AR-co-factors (e.g., FOXA1 and HOXB13) overlapped, independently of AR, at genomic loci implicated in neural lineage specification. Moreover, histone marks specifically associated with lineage-defining genes were reprogrammed by N-Myc. We also demonstrated that the N-Myc-induced molecular program accurately classifies our cohort of patients with advanced prostate cancer. Finally, we revealed the potential for EZH2 inhibition to reverse the N-Myc-induced suppression of epithelial lineage genes. Altogether, our data provide insights on how N-Myc regulates lineage plasticity and epigenetic reprogramming associated with lineage-specification. The N-Myc signature we defined could also help predict the evolution of prostate cancer and thus better guide the choice of future therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Rohan Bareja
- Caryl and Israel Englander Institute for Precision Medicine, NewYork-Presbyterian Hospital
| | - Brian Robinson
- Department of Pathology and Laboratory Medicine.,Caryl and Israel Englander Institute for Precision Medicine, NewYork-Presbyterian Hospital
| | | | | | | | - Adnan Ahmed
- Department of Biochemistry, Weill Cornell Medicine, New York, New York, USA
| | | | - Xiaodong Lu
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Inah Hwang
- Department of Pathology and Laboratory Medicine
| | | | - Andrea Sboner
- Department of Pathology and Laboratory Medicine.,Caryl and Israel Englander Institute for Precision Medicine, NewYork-Presbyterian Hospital.,Department of Physiology and Biophysics, Institute for Computational Biomedicine, and.,Meyer Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, NewYork-Presbyterian Hospital.,Department of Physiology and Biophysics, Institute for Computational Biomedicine, and.,Meyer Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | - Jihye Paik
- Department of Pathology and Laboratory Medicine.,Meyer Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | - Jindan Yu
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Christopher E Barbieri
- Caryl and Israel Englander Institute for Precision Medicine, NewYork-Presbyterian Hospital.,Department of Urology, and.,Meyer Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | - Noah Dephoure
- Department of Biochemistry, Weill Cornell Medicine, New York, New York, USA.,Meyer Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | - Himisha Beltran
- Caryl and Israel Englander Institute for Precision Medicine, NewYork-Presbyterian Hospital.,Department of Medicine.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - David S Rickman
- Department of Pathology and Laboratory Medicine.,Caryl and Israel Englander Institute for Precision Medicine, NewYork-Presbyterian Hospital.,Meyer Cancer Center, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
5
|
Gong S, Guo M, Tang G, Yang J, Qiu H. Overexpression of TEL-MN1 Fusion Enhances Resistance of HL-60 Cells to Idarubicin. Chemotherapy 2019; 63:308-314. [PMID: 30840968 DOI: 10.1159/000495073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 11/02/2018] [Indexed: 12/17/2023]
Abstract
BACKGROUND The translocation t(12; 22) (p13;q12) is a recurrent but infrequent chromosome abnormality in human myeloid malignancies. To date, the role of TEL-MN1 fusion in leukemogenic process and drug resistance is still largely unknown. METHODS In the present study, the TEL-MN1 fusion was transfected into HL-60 cells to upregulate TEL-MN1 expression via a retroviral vector. MTT assay was employed to examine cell viability and flow cytometry was performed to evaluate cell apoptosis. Idarubicin was used to treat HL-60 cells for estimating the effect of TEL-MN1 fusion on the chemotherapy resistance. RESULTS The results showed that overexpression of TEL-MN1 in HL-60 cells could promote cell proliferation, suggesting that TEL-MN1 may be involved in the leukemogenesis process. HL-60 cells treated with idarubicin showed a weakened cell viability, whereas TEL-MN1 overexpression attenuated the idarubicin-induced inhibition of cell viability and acceleration of cell apoptosis of HL-60 cells. CONCLUSION Taken together, our results indicated that TEL-MN1 fusion is an oncogene involved in the leukemogenesis process and TEL-MN1 overexpression enhanced resistance of HL-60 cells to idarubicin, which may provide a useful tool for studying the mechanism of leukemogenesis and drug resistance.
Collapse
Affiliation(s)
- Shenglan Gong
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Mengqiao Guo
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Gusheng Tang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jianmin Yang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Huiying Qiu
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China,
| |
Collapse
|
6
|
Shao H, Cen J, Chen S, Qiu H, Pan J. Myeloid neoplasms with t(12;22)(p13;q12)/MN1-EVT6: a systematic review of 12 cases. Ann Hematol 2018; 97:417-424. [PMID: 29273914 DOI: 10.1007/s00277-017-3208-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 12/13/2017] [Indexed: 02/06/2023]
Abstract
t(12;22)(p13;q12) is a rare but recurrent chromosomal abnormality involving the ETS transcription factor ETV6 and meningioma 1 (MN1) genes. In this study, we analyzed the clinical, cytogenetic, and molecular features of five new patients with the t(12;22)/MN1-EVT6 who presented with acute myeloid leukemia or chronic myelomonocytic leukemia. We subsequently reviewed the literature and identified seven additional cases reported with t(12;22)/MN1-EVT6. Our data suggest that neoplasms carrying the t(12;22)/MN1-ETV6, although rare, can commonly present as myeloid neoplasms at the initial diagnosis, including acute myeloid leukemia (n = 8), myelodysplastic syndrome (n = 2), and myelodysplastic/myeloproliferative neoplasms (n = 2). There were five men and seven women with a median age of 43 years (range, 15-63 years) at initial diagnosis. Cytogenetics revealed t(12;22) as the sole abnormality in five patients, with the remaining seven patients harboring additional chromosomal aberrations. Of the five patients who received known therapy regimens, all of them had poor response to the idarubicin/mitoxantrone + cytarabine regimen. Of the seven patients with follow-up information, six patients died with a median overall survival time of only 5 months (range, 1-12 months) after the emergence of t(12;22). In summary, patients with t(12;22) are frequently associated with myeloid neoplasms, poor response to chemotherapy, and inferior outcome.
Collapse
Affiliation(s)
- Haigang Shao
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Jiannong Cen
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Suning Chen
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Huiying Qiu
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Jinlan Pan
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China.
| |
Collapse
|
7
|
Rickman DS, Schulte JH, Eilers M. The Expanding World of N-MYC–Driven Tumors. Cancer Discov 2018; 8:150-163. [DOI: 10.1158/2159-8290.cd-17-0273] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 08/04/2017] [Accepted: 10/18/2017] [Indexed: 11/16/2022]
|
8
|
Aref S, Ibrahim L, Morkes H, Azmy E, Ebrahim M. Meningioma 1 (MN1) expression: refined risk stratification in acute myeloid leukemia with normal cytogenetics (CN-AML). ACTA ACUST UNITED AC 2013; 18:277-83. [PMID: 23394438 DOI: 10.1179/1607845412y.0000000065] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Prognostic stratification of cytogenetic normal acute myeloid leukemia (CN-AML) is an area of active research. The aim of this study was to determine the prognostic importance of the meningioma 1 (MN1) gene expression levels in CN-AML. One hundred patients with CN-AML were diagnosed; MN1 expressions were analyzed using quantitative real-time polymerase chain reaction. High expressions were detected in 48 (48%) patients (expression range: 2.35-31.99, mean: 13.9 ± 8.49) in comparison with 52 (52%) patients with low expression (expression range: 0.02-2.3, mean: 0.68 ± 0.77). The course of the disease in patients with high MN1 expression was unfavorable. Patients with high MN1 expression was associated with significant low complete remission rate (62.5 vs. 8.4%, high vs. low MN1, P = 0.001) and high mortality rate (75% vs. 46.1, P = 0.03). AML patients with high MN1 expression tended to be refractory (37.5 vs. 19.2%, P = 0.00) and relapse risk (54.1 vs. 23%, P = 0.02). Multivariable analysis confirmed high MN1 expression as an independent risk factor for disease-free survival and overall survival. In conclusion, MN1 overexpression independently predicts bad clinical outcome in CN-AML patients.
Collapse
Affiliation(s)
- Salah Aref
- Mansoura Faculty of Medicine, Mansoura, Egypt
| | | | | | | | | |
Collapse
|
9
|
ter Haar WM, Meester-Smoor MA, van Wely KHM, Schot CCMM, Janssen MJFW, Geverts B, Bonten J, Grosveld GC, Houtsmuller AB, Zwarthoff EC. The leukemia-associated fusion protein MN1-TEL blocks TEL-specific recognition sequences. PLoS One 2012; 7:e46085. [PMID: 23049943 PMCID: PMC3458806 DOI: 10.1371/journal.pone.0046085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 08/28/2012] [Indexed: 11/18/2022] Open
Abstract
The leukemia-associated fusion protein MN1-TEL combines the transcription-activating domains of MN1 with the DNA-binding domain of the transcriptional repressor TEL. Quantitative photobleaching experiments revealed that ∼20% of GFP-tagged MN1 and TEL is transiently immobilised, likely due to indirect or direct DNA binding, since transcription inhibition abolished immobilisation. Interestingly, ∼50% of the MN1-TEL fusion protein was immobile with much longer binding times than unfused MN1 and TEL. MN1-TEL immobilisation was not observed when the TEL DNA-binding domain was disrupted, suggesting that MN1-TEL stably occupies TEL recognition sequences, preventing binding of factors required for proper transcription regulation, which may contribute to leukemogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | - Bart Geverts
- Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Jacqueline Bonten
- Department of Genetics and Tumor Cell Biology, St Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Gerard C. Grosveld
- Department of Genetics and Tumor Cell Biology, St Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | | | | |
Collapse
|
10
|
Pardee TS. Overexpression of MN1 confers resistance to chemotherapy, accelerates leukemia onset, and suppresses p53 and Bim induction. PLoS One 2012; 7:e43185. [PMID: 22905229 PMCID: PMC3419213 DOI: 10.1371/journal.pone.0043185] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 07/20/2012] [Indexed: 11/18/2022] Open
Abstract
Background The transcriptional co-activator MN1 confers a worse prognosis for patients with acute myeloid leukemia (AML) when highly expressed; however, the mechanisms involved are unknown. We sought to model the effects of high MN1 expression in AML models to explore the underlying mechanisms. Methodology/Principal Findings We used cell lines and a genetically defined mouse model of AML to examine the effects of MN1 overexpression on prognosis and response to cytarabine and doxorubicin in vitro and in vivo. Murine AML that was engineered to overexpress MN1 became more aggressive in vivo, leading to shortened survival in both treated and control groups. In vitro murine AML cells that overexpressed MN1 became resistant to treatment with cytarabine and highly resistant to doxorubicin. This resistant phenotype was also seen in vivo, where treatment with the combination of cytarabine and doxorubicin selected for cells expressing MN1. When therapy-induced DNA damage levels were assessed by γH2AX foci, no reduction was seen in MN1 expressing cells arguing against a drug efflux mechanism. Despite no reduction in DNA damage, MN1-expressing cells showed less apoptosis as assessed by annexin V and propidium iodide staining. Following treatment, p53 and BIM induction were markedly reduced in cells expressing MN1. Pharmacologic inhibition of the p53 E3 ligase MDM2 resulted in increased p53 levels and improved response to doxorubicin in vitro. Conclusions/Significance MN1 overexpression accelerates an already aggressive leukemia, confers resistance to chemotherapy, and suppresses p53 and BIM induction, resulting in decreased apoptosis. This provides a mechanistic explanation of the poor prognosis observed with high MN1 expression and suggests that therapies directed at increasing p53 function may be useful for these patients.
Collapse
Affiliation(s)
- Timothy S Pardee
- Wake Forest University Health Sciences, Department of Internal Medicine, Section on Hematology and Oncology, Winston-Salem, North Carolina, United States of America.
| |
Collapse
|
11
|
Gotou M, Hanamura I, Nagoshi H, Wakabayashi M, Sakamoto N, Tsunekawa N, Horio T, Goto M, Mizuno S, Takahashi M, Suganuma K, Yamamoto H, Hiramatsu A, Watarai M, Shikami M, Imamura A, Mihara H, Taki T, Miwa H, Taniwaki M, Nitta M. Establishment of a novel human myeloid leukemia cell line, AMU-AML1, carrying t(12;22)(p13;q11) without chimeric MN1-TEL and with high expression of MN1. Genes Chromosomes Cancer 2011; 51:42-53. [DOI: 10.1002/gcc.20929] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 08/09/2011] [Accepted: 08/12/2011] [Indexed: 11/10/2022] Open
|
12
|
MN1-ETV6 fusion gene arising from MDS With 5q−. Leuk Res 2011; 35:e123-6. [DOI: 10.1016/j.leukres.2011.03.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 01/16/2011] [Accepted: 03/16/2011] [Indexed: 11/20/2022]
|
13
|
Noor SM, Bell R, Ward AC. Shooting the messenger: Targeting signal transduction pathways in leukemia and related disorders. Crit Rev Oncol Hematol 2011; 78:33-44. [DOI: 10.1016/j.critrevonc.2010.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2009] [Revised: 04/04/2010] [Accepted: 05/05/2010] [Indexed: 01/12/2023] Open
|
14
|
Nagel S, Venturini L, Meyer C, Kaufmann M, Scherr M, Drexler HG, Macleod RAF. Transcriptional deregulation of oncogenic myocyte enhancer factor 2C in T-cell acute lymphoblastic leukemia. Leuk Lymphoma 2011; 52:290-7. [PMID: 21261500 DOI: 10.3109/10428194.2010.537003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Myocyte enhancer factor 2C (MEF2C) encodes a transcription factor which is ectopically expressed in T-cell acute lymphoblastic leukemia (T-ALL) cell lines, deregulated directly by ectopically expressed homeodomain protein NKX2-5 or by loss of promoter regions via del(5)(q14). Here, we analyzed the MEF2C 5'-region, thus identifying potential regulatory binding sites for GFI1B, basic helix-loop-helix proteins, STAT5, and HOXA9/HOXA10. Chromatin immunoprecipitation and overexpression analyses demonstrated direct activation by GFI1B and LYL1 and inhibition by STAT5. HOXA9/HOXA10 activated expression of NMYC which in turn mediated MEF2C repression, indicating an indirect mode of regulation via NMYC interactor (NMI) and STAT5. Lacking comma: Chromosomal deletion of the STAT5 binding site in LOUCY cells reduced protein levels of STAT5 in some MEF2C-positve T-ALL cell lines, and the presence of inhibitory IL7-JAK-STAT5 signaling highlighted the repressive impact of this factor in MEF2C regulation. Taken together, our results indicate that the expression of MEF2C in T-ALL cells is principally deregulated via activating leukemic transcription factors GFI1B or NKX2-5 and by escaping inhibitory developmental STAT5 signaling.
Collapse
Affiliation(s)
- Stefan Nagel
- Department of Human and Animal Cell Lines, DSMZ - German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany.
| | | | | | | | | | | | | |
Collapse
|
15
|
Zaidi SZ, Owaidah T, Al Sharif F, Ahmed SY, Chaudhri N, Aljurf M. The challenge of risk stratification in acute myeloid leukemia with normal karyotype. Hematol Oncol Stem Cell Ther 2010; 1:141-58. [PMID: 20063545 DOI: 10.1016/s1658-3876(08)50023-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Cytogenetic aberrations have long been recognized as the most important prognostic variable in acute myeloid leukemia (AML) and are now a major stratification tool for post-remission therapy. Cytogenetics-based stratification improves survival. Patients with AML and normal cytogenetics, the largest single subgroup, have had a very heterogeneous outcome with standard chemotherapy in multiple clinical trials. Hence it is difficult to recommend a "one size fits all" kind of treatment for this heterogeneous population of AML patients. New emerging data from preclinical, retrospective, and large, randomized controlled studies indicate that in addition to cytogenetic abnormalities, many other molecular aberrations are operative in the response to treatment as well as in the risk of relapse. Such molecular markers are being tested for developing targeted therapies and may help in improved stratification of patients in the selection of post-remission therapy. Emerging evidence reveals that at the submicroscopic level, AML with normal cytogenetics may carry poor prognostic genetic lesions or "molecular signatures" as is the case with FLT3 mutations and overexpression of BAALC, ERG or MN1, or may have aberrations that predict better risk as is the case with isolated NPM1 or CEBPA mutations. Later studies have tried to explore the interaction of various prognostically important genes in this group of AML patients. The utility of the evolving data for bedside management of such patients is expected to improve with the wider application of modern tools, using the proposed clinical outcome models, and probably by development of a risk-scoring system based on the relative risk associated with each molecular aberration. The goals include identifying those patients most likely to benefit from upfront allogeneic HSCT and sparing good-prognosis patients from unnecessary transplant-related morbidity. The following is an outline of the most common molecular changes, their impact on the outcome of AML patients with normal cytogenetics and challenges in their wide scale application in risk stratification.
Collapse
Affiliation(s)
- Syed Z Zaidi
- Adult Hematology & SCT Department, Prince Sultan Hematology Oncology Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | | | | | | | | | | |
Collapse
|
16
|
Liu T, Jankovic D, Brault L, Ehret S, Baty F, Stavropoulou V, Rossi V, Biondi A, Schwaller J. Functional characterization of high levels of meningioma 1 as collaborating oncogene in acute leukemia. Leukemia 2010; 24:601-12. [PMID: 20072157 DOI: 10.1038/leu.2009.272] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Retroviral expression of leukemogenic oncogenes in the murine hematopoietic system is essential but not sufficient to induce acute leukemia. Proviral integration-mediated elevated expression of the meningioma 1 (MN1) oncogene suggested MN1 acting as cooperating event in mixed-lineage leukemia 1 (MLL) and eleven nineteen leukemia (ENL)-induced murine leukemia. Indeed, co-expression of MN1 with MLL-ENL enhanced transformation in vivo, and resulted in a significantly reduced latency for induction of an aggressive acute leukemia when compared with MN1 or MLL-ENL alone. In addition, co-expression of MN1 increased the granulocyte macrophage progenitor cell population with leukemia-initiating properties as shown in secondary transplantation experiments. Gene expression profiling experiments identified putative downstream MN1 targets, of which FMS-like tyrosine kinase 3 (FLT3) and CD34 were upregulated in both MN1-overexpressing murine leukemias and in pediatric acute leukemias with high MN1 levels. Interestingly, small interfering RNA (siRNA)-mediated MN1 knockdown resulted in cell cycle arrest and impaired clonogenic growth of human leukemia cell lines with high MN1 levels. Our work shows for the first time that high MN1 levels are important for the growth of leukemic cells, and that increased MN1 expression can synergize with MLL-ENL and probably other transforming fusion genes in leukemia induction through a distinct gene expression program that is able to expand the leukemia-initiating cell population.
Collapse
Affiliation(s)
- T Liu
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Retroviral insertional mutagenesis identifies Zeb2 activation as a novel leukemogenic collaborating event in CALM-AF10 transgenic mice. Blood 2009; 115:1194-203. [PMID: 20007546 DOI: 10.1182/blood-2009-04-216184] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The t(10;11) translocation results in a CALM-AF10 fusion gene in a subset of leukemia patients. Expression of a CALM-AF10 transgene results in leukemia, with prolonged latency and incomplete penetrance, suggesting that additional events are necessary for leukemic transformation. CALM-AF10 mice infected with the MOL4070LTR retrovirus developed acute leukemia, and ligation-mediated polymerase chain reaction was used to identify retroviral insertions at 19 common insertion sites, including Zeb2, Nf1, Mn1, Evi1, Ift57, Mpl, Plag1, Kras, Erg, Vav1, and Gata1. A total of 26% (11 of 42) of the mice had retroviral integrations near Zeb2, a transcriptional corepressor leading to overexpression of the Zeb2-transcript. A total of 91% (10 of 11) of mice with Zeb2 insertions developed B-lineage acute lymphoblastic leukemia, suggesting that Zeb2 activation promotes the transformation of CALM-AF10 hematopoietic precursors toward B-lineage leukemias. More than half of the mice with Zeb2 integrations also had Nf1 integrations, suggesting cooperativity among CALM-AF10, Zeb2, and Ras pathway mutations. We searched for Nras, Kras, and Ptpn11 point mutations in the CALM-AF10 leukemic mice. Three mutations were identified, all of which occurred in mice with Zeb2 integrations, consistent with the hypothesis that Zeb2 and Ras pathway activation promotes B-lineage leukemic transformation in concert with CALM-AF10.
Collapse
|
18
|
Reintroduction of CEBPA in MN1-overexpressing hematopoietic cells prevents their hyperproliferation and restores myeloid differentiation. Blood 2009; 114:1596-606. [PMID: 19561324 DOI: 10.1182/blood-2009-02-205443] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Forced expression of MN1 in primitive mouse hematopoietic cells causes acute myeloid leukemia and impairs all-trans retinoic acid-induced granulocytic differentiation. Here, we studied the effects of MN1 on myeloid differentiation and proliferation using primary human CD34(+) hematopoietic cells, lineage-depleted mouse bone marrow cells, and bipotential (granulocytic/monocytic) human acute myeloid leukemia cell lines. We show that exogenous MN1 stimulated the growth of CD34(+) cells, which was accompanied by enhanced survival and increased cell cycle traverse in cultures supporting progenitor cell growth. Forced MN1 expression impaired both granulocytic and monocytic differentiation in vitro in primary hematopoietic cells and acute myeloid leukemia cell lines. Endogenous MN1 expression was higher in human CD34(+) cells compared with both primary and in vitro-differentiated monocytes and granulocytes. Microarray and real-time reverse-transcribed polymerase chain reaction analysis of MN1-overexpressing CD34(+) cells showed down-regulation of CEBPA and its downstream target genes. Reintroduction of conditional and constitutive CEBPA overcame the effects of MN1 on myeloid differentiation and inhibited MN1-induced proliferation in vitro. These results indicate that down-regulation of CEBPA activity contributes to MN1-modulated proliferation and impaired myeloid differentiation of hematopoietic cells.
Collapse
|
19
|
Gregory TK, Wald D, Chen Y, Vermaat JM, Xiong Y, Tse W. Molecular prognostic markers for adult acute myeloid leukemia with normal cytogenetics. J Hematol Oncol 2009; 2:23. [PMID: 19490647 PMCID: PMC2700131 DOI: 10.1186/1756-8722-2-23] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Accepted: 06/02/2009] [Indexed: 12/23/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogenous disorder that results from a block in the differentiation of hematopoietic progenitor cells along with uncontrolled proliferation. In approximately 60% of cases, specific recurrent chromosomal aberrations can be identified by modern cytogenetic techniques. This cytogenetic information is the single most important tool to classify patients at their initial diagnosis into three prognostic categories: favorable, intermediate, and poor risk. Currently, favorable risk AML patients are usually treated with contemporary chemotherapy while poor risk AML patients receive allogeneic stem cell transplantation if suitable stem cell donors exist. The largest subgroup of AML patients (aproximately 40%) have no identifiable cytogenetic abnormalities and are classified as intermediate risk. The optimal therapeutic strategies for these patients are still largely unclear. Recently, it is becoming increasingly evident that it is possible to identify a subgroup of poorer risk patients among those with normal cytogenic AML (NC-AML). Molecular risk stratification for NC-AML patients may be possible due to mutations of NPM1, FLT3, MLL, and CEBPalpha as well as alterations in expression levels of BAALC, MN1, ERG, and AF1q. Further prospective studies are needed to confirm if poorer risk NC-AML patients have improved clinical outcomes after more aggressive therapy.
Collapse
MESH Headings
- Adult
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Cytogenetics
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic
- Genetic Markers/physiology
- Humans
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Neoplasm, Residual
- Nuclear Proteins/genetics
- Nuclear Proteins/physiology
- Nucleophosmin
- Prognosis
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/physiology
Collapse
Affiliation(s)
- Tara K Gregory
- Division of Medical Oncology, University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - David Wald
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Yichu Chen
- Division of Medical Oncology, University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Johanna M Vermaat
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Yin Xiong
- Division of Medical Oncology, University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - William Tse
- Division of Medical Oncology, University of Colorado Cancer Center, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
20
|
Zhang X, Dowd DR, Moore MC, Kranenburg TA, Meester-Smoor MA, Zwarthoff EC, MacDonald PN. Meningioma 1 is required for appropriate osteoblast proliferation, motility, differentiation, and function. J Biol Chem 2009; 284:18174-83. [PMID: 19386590 DOI: 10.1074/jbc.m109.001354] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The vitamin D endocrine system is essential for calcium and phosphate homeostasis and skeletal mineralization. The 1,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)) hormone binds to the vitamin D receptor (VDR) to regulate gene expression. These gene products in turn mediate the actions of 1,25(OH)(2)D(3) in mineral-regulating target cells such as the osteoblast. We showed previously that meningioma 1 (MN1) is a novel target of 1,25(OH)(2)D(3) in MG-63 osteoblastic cells and that it is a coactivator for VDR-mediated transcription (Sutton, A. L., Zhang, X., Ellison, T. I., and MacDonald, P. N. (2005) Mol. Endocrinol. 19, 2234-2244). However, the functional significance of MN1 in osteoblastic cell biology is largely unknown. Here, we demonstrate that MN1 expression is increased dramatically during differentiation of primary osteoblastic cells. Using calvarial osteoblasts derived from wild-type and MN1 knock-out mice, we provide data supporting an essential role of MN1 in maintaining appropriate osteoblast proliferation, differentiation, and function. MN1 knock-out osteoblasts displayed altered morphology, decreased growth rate, impaired motility, and attenuated 1,25(OH)(2)D(3)/VDR-mediated transcription as well as reduced alkaline phosphatase activity and mineralized nodule formation. MN1 null osteoblasts were also impaired in supporting osteoclastogenesis in co-culture studies presumably because of marked reduction in the RANKL:OPG ratio in the MN1 null cells. Mechanistic studies supported a transcriptional role for MN1 in controlling RANKL gene expression through activation of the RANKL promoter. Cumulatively, these studies indicate an important role for MN1 in maintaining the appropriate maturation and function of calvarial osteoblasts.
Collapse
Affiliation(s)
- Xiaoxue Zhang
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Liu W, Lan Y, Pauws E, Meester-Smoor MA, Stanier P, Zwarthoff EC, Jiang R. The Mn1 transcription factor acts upstream of Tbx22 and preferentially regulates posterior palate growth in mice. Development 2008; 135:3959-68. [PMID: 18948418 DOI: 10.1242/dev.025304] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The mammalian secondary palate exhibits morphological, pathological and molecular heterogeneity along the anteroposterior axis. Although the cell proliferation rates are similar in the anterior and posterior regions during palatal outgrowth, previous studies have identified several signaling pathways and transcription factors that specifically regulate the growth of the anterior palate. By contrast, no factor has been shown to preferentially regulate posterior palatal growth. Here, we show that mice lacking the transcription factor Mn1 have defects in posterior but not anterior palatal growth. We show that Mn1 mRNA exhibits differential expression along the anteroposterior axis of the developing secondary palate, with preferential expression in the middle and posterior regions during palatal outgrowth. Extensive analyses of palatal gene expression in wild-type and Mn1(-/-) mutant mice identified Tbx22, the mouse homolog of the human X-linked cleft palate gene, as a putative downstream target of Mn1 transcriptional activation. Tbx22 exhibits a similar pattern of expression with that of Mn1 along the anteroposterior axis of the developing palatal shelves and its expression is specifically downregulated in Mn1(-/-) mutants. Moreover, we show that Mn1 activated reporter gene expression driven by either the human or mouse Tbx22 gene promoters in co-transfected NIH3T3 cells. Overexpression of Mn1 in NIH3T3 cells also increased endogenous Tbx22 mRNA expression in a dose-dependent manner. These data indicate that Mn1 and Tbx22 function in a novel molecular pathway regulating mammalian palate development.
Collapse
Affiliation(s)
- Wenjin Liu
- Department of Biomedical Genetics and Center for Oral Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Kawagoe H, Kandilci A, Kranenburg TA, Grosveld GC. Overexpression of N-Myc rapidly causes acute myeloid leukemia in mice. Cancer Res 2007; 67:10677-85. [PMID: 18006809 DOI: 10.1158/0008-5472.can-07-1118] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
N-MYC encodes a basic helix-loop-helix/leucine zipper (bHLH/LZ) transcription factor that is frequently overexpressed in human neuroblastoma. N-MYC overexpression has also been reported in human acute myeloid leukemias (AML), which we show here is a frequent event. Myeloid cells in N-Myc-overexpressing mouse bone marrow hyperproliferate but those in c-MYC-overexpressing bone marrow do not. The NH(2)-terminal transactivation domain, nuclear localization signal, and bHLH/LZ domain of N-Myc are essential for this effect. Microarray analysis revealed 969 differentially expressed genes between N-Myc- and c-MYC-overexpressing myeloid cells. N-Myc-overexpressing cells showed decreased transforming growth factor beta signaling and increased c-Jun-NH(2)-kinase signaling, both of which are associated with proliferation and leukemic transformation of myeloid cells. Mice transplanted with bone marrow expressing wild-type N-Myc developed clonal and transplantable AML after approximately 1 month; those transplanted with bone marrow expressing mutant N-Myc did not. Twist, a known suppressor of the p19Arf/p53 pathway, was up-regulated in all tumors. These results show that N-Myc overexpression is highly oncogenic in mouse myeloid cells and suggest that N-MYC up-regulation contributes to human myeloid leukemogenesis.
Collapse
Affiliation(s)
- Hiroyuki Kawagoe
- Department of Genetics and Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | | | | | | |
Collapse
|
23
|
Kasner MT, Laury A, Kasner SE, Carroll M, Luger SM. Increased cerebral blood flow after leukapheresis for acute myelogenous leukemia. Am J Hematol 2007; 82:1110-2. [PMID: 17654677 DOI: 10.1002/ajh.21006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Leukapheresis is often considered in the management of acute myelogenous leukemia (AML) with hyperleukocytosis and its sequelae, including myocardial infarction, pulmonary complications, and stroke. It is utilized on the assumption that leukapheresis improves blood rheology. We present a woman with AML and a history of meningioma encasing her left internal carotid artery. She presented with hyperleukocytosis and symptoms of ischemia. As her white blood cell continued to rise despite initiation of hydroxyurea therapy, she underwent leukapheresis emergently. Transcranial Doppler ultrasound demonstrated increased flow velocities in the left internal carotid and the right middle cerebral arteries, which normalized after leukapheresis. This is the first documentation that leukapheresis, in combination with hydroxyurea, improves cerebral hemodynamics in a patient with AML.
Collapse
Affiliation(s)
- Margaret T Kasner
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
| | | | | | | | | |
Collapse
|
24
|
Keeshan K, Shestova O, Ussin L, Pear WS. Tribbles homolog 2 (Trib2) and HoxA9 cooperate to accelerate acute myelogenous leukemia. Blood Cells Mol Dis 2007; 40:119-21. [PMID: 17988908 DOI: 10.1016/j.bcmd.2007.06.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2007] [Accepted: 06/07/2007] [Indexed: 01/26/2023]
Abstract
Trib2 is a member of the Trib family of serine/threonine kinase-like proteins (Trib1, Trib2, Trib3). Mice reconstituted with hematopoietic stem cells (HSC) retrovirally expressing Trib2 uniformly developed fatal transplantable acute myelogenous leukemia (AML). Trib2-induced AML was clonal and we sought to identify cooperating genes in Trib2-induced AML. Using Splinkerette PCR techniques, we identified proviral insertion near HoxA9 in a Trib2 monoclonal tumor, which resulted in greatly elevated HoxA9 expression. Mice reconstituted with HSC cotransduced with HoxA9 and Trib2 had accelerated onset of AML compared to either gene alone. These data identify Trib2 and HoxA9 as cooperating genes in AML.
Collapse
Affiliation(s)
- Karen Keeshan
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104-6160, USA
| | | | | | | |
Collapse
|
25
|
Heuser M, Argiropoulos B, Kuchenbauer F, Yung E, Piper J, Fung S, Schlenk RF, Dohner K, Hinrichsen T, Rudolph C, Schambach A, Baum C, Schlegelberger B, Dohner H, Ganser A, Humphries RK. MN1 overexpression induces acute myeloid leukemia in mice and predicts ATRA resistance in patients with AML. Blood 2007; 110:1639-47. [PMID: 17494859 DOI: 10.1182/blood-2007-03-080523] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
AbstractOverexpression of wild-type MN1 is a negative prognostic factor in patients with acute myeloid leukemia (AML) with normal cytogenetics. We evaluated whether MN1 plays a functional role in leukemogenesis. We demonstrate using retroviral gene transfer and bone marrow (BM) transplantation that MN1 overexpression rapidly induces lethal AML in mice. Insertional mutagenesis and chromosomal instability were ruled out as secondary aberrations. MN1 increased resistance to all-trans retinoic acid (ATRA)–induced cell-cycle arrest and differentiation by more than 3000-fold in vitro. The differentiation block could be released by fusion of a transcriptional activator (VP16) to MN1 without affecting the ability to immortalize BM cells, suggesting that MN1 blocks differentiation by transcriptional repression. We then evaluated whether MN1 expression levels in patients with AML (excluding M3-AML) correlated with resistance to ATRA treatment in elderly patients uniformly treated within treatment protocol AMLHD98-B. Strikingly, patients with low MN1 expression who received ATRA had a significantly prolonged event-free (P = .008) and overall (P = .04) survival compared with patients with either low MN1 expression and no ATRA, or high MN1 expression with or without ATRA. MN1 is a unique oncogene in hematopoiesis that both promotes proliferation/self-renewal and blocks differentiation, and may become useful as a predictive marker in AML treatment.
Collapse
MESH Headings
- Aged
- Animals
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/pharmacology
- Biomarkers, Tumor/biosynthesis
- Biomarkers, Tumor/genetics
- Bone Marrow Cells/metabolism
- Cell Cycle/drug effects
- Cell Cycle/genetics
- Cell Differentiation/drug effects
- Cell Differentiation/genetics
- Cell Transformation, Viral/drug effects
- Cell Transformation, Viral/genetics
- Chromosomal Instability/genetics
- Disease-Free Survival
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Leukemic/drug effects
- Gene Expression Regulation, Leukemic/genetics
- Hematopoiesis/drug effects
- Hematopoiesis/genetics
- Herpes Simplex Virus Protein Vmw65/biosynthesis
- Herpes Simplex Virus Protein Vmw65/genetics
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/pathology
- Male
- Middle Aged
- Mutagenesis, Insertional/drug effects
- Mutagenesis, Insertional/genetics
- Predictive Value of Tests
- Recombinant Fusion Proteins/biosynthesis
- Recombinant Fusion Proteins/genetics
- Repressor Proteins/biosynthesis
- Repressor Proteins/genetics
- Retroviridae
- Risk Factors
- Survival Rate
- Trans-Activators
- Transduction, Genetic
- Tretinoin/administration & dosage
- Tretinoin/pharmacology
- Tumor Suppressor Proteins/biosynthesis
- Tumor Suppressor Proteins/genetics
Collapse
Affiliation(s)
- Michael Heuser
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
|
27
|
Abstract
The transcriptional coactivator MN1 has been identified as a gene overexpressed in certain types of human acute myeloid leukemia. Upregulation is invariantly associated with inv(16) AML but is also found in other AML subtypes. Overexpression of this gene is also associated with a worse prognosis and a shorter survival in AML patients with a normal karyotype. In this short review, I will discuss the role of MN1 in myeloid leukemia.
Collapse
Affiliation(s)
- Gerard C Grosveld
- Department of Genetics and Tumor Cell Biology, St Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
28
|
Slape C, Hartung H, Lin YW, Bies J, Wolff L, Aplan PD. Retroviral insertional mutagenesis identifies genes that collaborate with NUP98-HOXD13 during leukemic transformation. Cancer Res 2007; 67:5148-55. [PMID: 17545593 PMCID: PMC1950322 DOI: 10.1158/0008-5472.can-07-0075] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The t(2;11)(q31;p15) chromosomal translocation results in a fusion between the NUP98 and HOXD13 genes and has been observed in patients with myelodysplastic syndrome (MDS) or acute myelogenous leukemia. We previously showed that expression of the NUP98-HOXD13 (NHD13) fusion gene in transgenic mice results in an invariably fatal MDS; approximately one third of mice die due to complications of severe pancytopenia, and about two thirds progress to a fatal acute leukemia. In the present study, we used retroviral insertional mutagenesis to identify genes that might collaborate with NHD13 as the MDS transformed to an acute leukemia. Newborn NHD13 transgenic mice and littermate controls were infected with the MOL4070LTR retrovirus. The onset of leukemia was accelerated, suggesting a synergistic effect between the NHD13 transgene and the genes neighboring retroviral insertion events. We identified numerous common insertion sites located near protein-coding genes and confirmed dysregulation of a subset of these by expression analyses. Among these genes were Meis1, a known collaborator of HOX and NUP98-HOX fusion genes, and Mn1, a transcriptional coactivator involved in human leukemia through fusion with the TEL gene. Other putative collaborators included Gata2, Erg, and Epor. Of note, we identified a common insertion site that was >100 kb from the nearest coding gene, but within 20 kb of the miR29a/miR29b1 microRNA locus. Both of these miRNA were up-regulated, demonstrating that retroviral insertional mutagenesis can target miRNA loci as well as protein-coding loci. Our data provide new insights into NHD13-mediated leukemogenesis as well as retroviral insertional mutagenesis mechanisms.
Collapse
Affiliation(s)
- Christopher Slape
- Genetics Branch, Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland 20889-5105, USA
| | | | | | | | | | | |
Collapse
|
29
|
Carella C, Bonten J, Sirma S, Kranenburg TA, Terranova S, Klein-Geltink R, Shurtleff S, Downing JR, Zwarthoff EC, Liu PP, Grosveld GC. MN1 overexpression is an important step in the development of inv(16) AML. Leukemia 2007; 21:1679-90. [PMID: 17525718 DOI: 10.1038/sj.leu.2404778] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The gene encoding the transcriptional co-activator MN1 is the target of the reciprocal chromosome translocation (12;22)(p13;q12) in some patients with acute myeloid leukemia (AML). In addition, expression array analysis showed that MN1 was overexpressed in AML specified by inv(16), in some AML overexpressing ecotropic viral integration 1 site (EVI1) and in some AML without karyotypic abnormalities. Here we describe that mice receiving transplants of bone marrow (BM) overexpressing MN1 rapidly developed myeloproliferative disease (MPD). This BM also generated myeloid cell lines in culture. By mimicking the situation in human inv(16) AML, forced coexpression of MN1 and Cbfbeta-SMMHC rapidly caused AML in mice. These findings identify MN1 as a highly effective hematopoietic oncogene and suggest that MN1 overexpression is an important cooperative event in human inv(16) AML.
Collapse
MESH Headings
- Acute Disease
- Animals
- Bone Marrow Transplantation
- Cells, Cultured
- Chromosome Inversion
- Chromosomes, Human, Pair 16/genetics
- Female
- Flow Cytometry
- Gene Expression Regulation, Neoplastic/physiology
- Humans
- Leukemia, Myeloid/etiology
- Leukemia, Myeloid/metabolism
- Leukemia, Myeloid/pathology
- Mice
- Mice, Transgenic
- Myeloproliferative Disorders/etiology
- Myeloproliferative Disorders/metabolism
- Myeloproliferative Disorders/pathology
- Oncogene Proteins/genetics
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Survival Rate
- Trans-Activators
- Translocation, Genetic/genetics
- Tumor Suppressor Proteins
Collapse
Affiliation(s)
- C Carella
- Department of Genetics and Tumor Cell Biology, St Jude Children's Research Hospital, Memphis, TN 38105-0318, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Gutiérrez L, Nikolic T, van Dijk TB, Hammad H, Vos N, Willart M, Grosveld F, Philipsen S, Lambrecht BN. Gata1 regulates dendritic-cell development and survival. Blood 2007; 110:1933-41. [PMID: 17505015 PMCID: PMC1975828 DOI: 10.1182/blood-2006-09-048322] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Dendritic cells are key initiators and regulators of the immune response. Dendritic cell commitment and function require orchestrated regulation of transcription. Gata1 is a transcription factor expressed in several hematopoietic lineages. However, Gata1 function has not been explored in the monocytic or dendritic cell compartment. Here, we show that Gata1 is expressed in myeloid and plasmacytoid dendritic cells and that Gata1 ablation affects the survival of dendritic cells. Furthermore, lipopolysaccharide (LPS) stimulation of dendritic cells prompts Gata1 up-regulation, which is accompanied by increased levels of BclX and Ifng. Our findings show that Gata1 is a transcriptional regulator of dendritic cell differentiation and suggest that Gata1 is involved in the dendritic cell and macrophage lineage separation.
Collapse
Affiliation(s)
- Laura Gutiérrez
- Department of Cell Biology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
van Wely KHM, Meester-Smoor MA, Janssen MJFW, Aarnoudse AJ, Grosveld GC, Zwarthoff EC. The MN1-TEL myeloid leukemia-associated fusion protein has a dominant-negative effect on RAR-RXR-mediated transcription. Oncogene 2007; 26:5733-40. [PMID: 17369854 DOI: 10.1038/sj.onc.1210382] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The translocation t(12;22)(p13;q11) creates an MN1-TEL fusion gene leading to acute myeloid leukemia. MN1 is a transcription coactivator of the retinoic acid and vitamin D receptors, and TEL (ETV6) is a member of the E26-transformation-specific family of transcription factors. In MN1-TEL, the transactivating domains of MN1 are combined with the DNA-binding domain of TEL. We show that MN1-TEL inhibits retinoic acid receptor (RAR)-mediated transcription, counteracts coactivators such as p160 and p300, and acts as a dominant-negative mutant of MN1. Compared to MN1, the same transactivation domains in MN1-TEL are poorly stimulated by p160, p300 or histone deacetylase inhibitors, indicating that the block of RAR-mediated transcription by MN1-TEL is caused by dysfunctional transactivation domains rather than by recruitment of corepressors. The mechanism leading to myeloid leukemia in t(12;22) thus differs from the translocations that involve RAR itself.
Collapse
MESH Headings
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Chromosomes, Human, Pair 12/genetics
- Chromosomes, Human, Pair 22/genetics
- DNA-Binding Proteins
- E1A-Associated p300 Protein/physiology
- Enzyme Inhibitors
- Genes, Dominant
- Histone Deacetylase Inhibitors
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Mutation
- Nuclear Proteins/physiology
- Nucleocytoplasmic Transport Proteins/physiology
- Oncogene Proteins, Fusion/physiology
- RNA-Binding Proteins
- Receptors, Retinoic Acid/genetics
- Retinoid X Receptors/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Transcription Factors/physiology
- Transcription, Genetic
- Transcriptional Activation
- Transfection
- Translocation, Genetic
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- K H M van Wely
- Department of Pathology, Josephine Nefkens Institute, Erasmus MC, CA Rotterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
32
|
Keeshan K, He Y, Wouters BJ, Shestova O, Xu L, Sai H, Rodriguez CG, Maillard I, Tobias JW, Valk P, Carroll M, Aster JC, Delwel R, Pear WS. Tribbles homolog 2 inactivates C/EBPalpha and causes acute myelogenous leukemia. Cancer Cell 2006; 10:401-11. [PMID: 17097562 PMCID: PMC2839500 DOI: 10.1016/j.ccr.2006.09.012] [Citation(s) in RCA: 191] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2005] [Revised: 06/23/2006] [Accepted: 09/01/2006] [Indexed: 01/14/2023]
Abstract
Tribbles homolog 2 (Trib2) was identified as a downregulated transcript in leukemic cells undergoing growth arrest. To investigate the effects of Trib2 in hematopoietic progenitors, mice were reconstituted with hematopoietic stem cells retrovirally expressing Trib2. Trib2-transduced bone marrow cells exhibited a growth advantage ex vivo and readily established factor-dependent cell lines. In vivo, Trib2-reconstituted mice uniformly developed fatal transplantable acute myelogenous leukemia (AML). In mechanistic studies, we found that Trib2 associated with and inhibited C/EBPalpha. Furthermore, Trib2 expression was elevated in a subset of human AML patient samples. Together, our data identify Trib2 as an oncogene that induces AML through a mechanism involving inactivation of C/EBPalpha.
Collapse
Affiliation(s)
- Karen Keeshan
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Institute for Medicine & Engineering University of Pennsylvania, Philadelphia, PA 19104-6160, USA
| | - Yiping He
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Institute for Medicine & Engineering University of Pennsylvania, Philadelphia, PA 19104-6160, USA
| | - Bas J. Wouters
- Department of Hematology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Olga Shestova
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Institute for Medicine & Engineering University of Pennsylvania, Philadelphia, PA 19104-6160, USA
| | - Lanwei Xu
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Institute for Medicine & Engineering University of Pennsylvania, Philadelphia, PA 19104-6160, USA
| | - Hong Sai
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Institute for Medicine & Engineering University of Pennsylvania, Philadelphia, PA 19104-6160, USA
| | - Carlos G. Rodriguez
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Institute for Medicine & Engineering University of Pennsylvania, Philadelphia, PA 19104-6160, USA
| | - Ivan Maillard
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Institute for Medicine & Engineering University of Pennsylvania, Philadelphia, PA 19104-6160, USA
- Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John W. Tobias
- University of Pennsylvania Bioinformatics Core, University of Pennsylvania, Philadelphia, PA 19104
| | - Peter Valk
- Department of Hematology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Martin Carroll
- Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jon C. Aster
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Ruud Delwel
- Department of Hematology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Warren S. Pear
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Institute for Medicine & Engineering University of Pennsylvania, Philadelphia, PA 19104-6160, USA
| |
Collapse
|
33
|
Heuser M, Beutel G, Krauter J, Döhner K, von Neuhoff N, Schlegelberger B, Ganser A. High meningioma 1 (MN1) expression as a predictor for poor outcome in acute myeloid leukemia with normal cytogenetics. Blood 2006; 108:3898-905. [PMID: 16912223 DOI: 10.1182/blood-2006-04-014845] [Citation(s) in RCA: 187] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The translocation t(12;22) involves MN1 and TEL and is rarely found in acute myeloid leukemia (AML). Recently, it has been shown in a mouse model that the fusion protein MN1-TEL can promote growth of primitive hematopoietic progenitor cells (HPCs) and, in cooperation with HOXA9, induce AML. We quantified MN1 expression by real-time reverse transcriptase-polymerase chain reaction (RT-PCR) in 142 adult patients with AML with normal cytogenetics treated uniformly in trial AML-SHG 01/99. AML samples were dichotomized at the median MN1 expression. High MN1 expression was significantly correlated with unmutated NPM1 (P < .001), poor response to the first course of induction treatment (P = .02), a higher relapse rate (P = .03), and shorter relapse-free (P = .002) and overall survivals (P = .03). In multivariate analysis, MN1 expression was an independent prognostic marker (P = .02) in addition to age and Eastern Cooperative Oncology Group (ECOG) performance status. Excluding patients with NPM1(mutated)/FLT3ITD(negative), high MN1 expression was associated with shorter relapse-free survival (P = .057). MN1 was highly expressed in some patients with acute lymphoblastic but not chronic lymphocytic or myeloid leukemia. MN1 was highly expressed in HPCs compared with differentiated cells and was down-regulated during in vitro differentiation of CD34(+) cells, suggesting a functional role in HPCs. In conclusion, our data suggest MN1 overexpression as a new prognostic marker in AML with normal cytogenetics.
Collapse
MESH Headings
- Adolescent
- Adult
- Animals
- Biomarkers, Tumor/biosynthesis
- Biomarkers, Tumor/genetics
- Cytogenetic Analysis/methods
- Disease-Free Survival
- Female
- Gene Expression Regulation, Leukemic
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/therapy
- Male
- Mice
- Middle Aged
- Nucleophosmin
- Oncogene Proteins, Fusion/biosynthesis
- Oncogene Proteins, Fusion/genetics
- Predictive Value of Tests
- Reverse Transcriptase Polymerase Chain Reaction
- Survival Rate
- Trans-Activators
- Transcription Factors/biosynthesis
- Transcription Factors/genetics
- Tumor Suppressor Proteins/biosynthesis
- Tumor Suppressor Proteins/genetics
Collapse
Affiliation(s)
- Michael Heuser
- British Columbia Cancer Research Centre, 675 West 10th Ave, Vancouver, BC, Canada.
| | | | | | | | | | | | | |
Collapse
|
34
|
Carella C, Bonten J, Rehg J, Grosveld GC. MN1-TEL, the product of the t(12;22) in human myeloid leukemia, immortalizes murine myeloid cells and causes myeloid malignancy in mice. Leukemia 2006; 20:1582-92. [PMID: 16810199 DOI: 10.1038/sj.leu.2404298] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
MN1-TEL is the product of the recurrent t(12;22)(p12;q11) associated with human myeloid malignancies. MN1-TEL functions as an activated transcription factor, exhibiting weak transforming activity in NIH3T3 fibroblasts that depends on the presence of a functional TEL DNA-binding domain, the N-terminal transactivating sequences of MN1 and C-terminal sequences of MN1. We determined the transforming activity of MN1-TEL in mouse bone marrow (BM) by using retroviral transfer. MN1-TEL-transduced BM showed increased self-renewal capacity of primitive progenitors in vitro, and prolonged in vitro culture of MN1-TEL-expressing BM produced immortalized myeloid, interleukin (IL)-3/stem cell factor-dependent cell lines with a primitive morphology. Transplantation of such cell lines into lethally irradiated mice rescued them from irradiation-induced death and resulted in the contribution of MN1-TEL-expressing cells to all hematopoietic lineages, underscoring the primitive nature of these cells and their capacity to differentiate in vivo. Three months after transplantation, all mice succumbed to promonocytic leukemia. Transplantation of freshly MN1-TEL-transduced BM into lethally irradiated mice also caused acute myeloid leukemia within 3 months of transplantation. We infer that MN1-TEL is a hematopoietic oncogene that stimulates the growth of hematopoietic cells, but depends on secondary mutations to cause leukemia in mice.
Collapse
MESH Headings
- Animals
- Cell Proliferation
- Cell Transformation, Neoplastic
- Cell Transplantation
- Chromosomes, Human, Pair 12
- Chromosomes, Human, Pair 22
- Hematopoietic Stem Cells/pathology
- Humans
- Leukemia, Myeloid/genetics
- Leukemia, Myeloid/pathology
- Mice
- Mice, Inbred C57BL
- Oncogene Proteins, Fusion/genetics
- Transcription Factors/genetics
- Translocation, Genetic
Collapse
Affiliation(s)
- C Carella
- Department of Genetics and Tumor Cell Biology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | | | |
Collapse
|
35
|
Kawagoe H, Grosveld GC. MN1-TEL myeloid oncoprotein expressed in multipotent progenitors perturbs both myeloid and lymphoid growth and causes T-lymphoid tumors in mice. Blood 2005; 106:4278-86. [PMID: 16081688 PMCID: PMC1895241 DOI: 10.1182/blood-2005-04-1674] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The MN1-TEL (meningioma 1-translocation-ETS-leukemia) fusion oncoprotein is the product of the t(12;22)(p13;q11) in human myeloid leukemia consisting of N-terminal MN1 sequences, a transcriptional coactivator, fused to C-terminal TEL sequences, an E26-transformation-specific (ETS) transcription factor. To analyze the role of MN1-TEL in leukemogenesis, we created a site-directed transgenic (knock-in) mouse model carrying a conditional MN1-TEL transgene under the control of the Aml1 regulatory sequences. After induction, MN1-TEL expression was detected in both myeloid and lymphoid cells. Activation of MN1-TEL expression enhanced the repopulation ability of myeloid progenitors in vitro as well as partially inhibited their differentiation in vivo. MN1-TEL also promoted the proliferation of thymocytes while it blocked their differentiation from CD4-/CD8- to CD4+/CD8+ in vivo. After long latency, 30% of the MN1-TEL-positive mice developed T-lymphoid tumors. This process was accelerated by N-ethyl-N-nitrosourea-induced mutations. MN1-TEL-positive T-lymphoid tumors showed elevated expression of the Notch-1, Hes-1, c-Myc, and Lmo-2 genes while their Ink4a/pRB and Arf/p53 pathways were impaired, suggesting that these alterations cooperatively transform T progenitors. We conclude that MN1-TEL exerts its nonlineage-specific leukemogenic effects by promoting the growth of primitive progenitors and blocking their differentiation, but cooperative mutations are necessary to fully induce leukemic transformation.
Collapse
Affiliation(s)
- Hiroyuki Kawagoe
- Department of Genetics and Tumor Cell Biology, St Jude Children's Research Hospital, 332 North Lauderdale, Memphis, TN 38105, USA
| | | |
Collapse
|