1
|
Li HD, Chen SS, Ding J, Zhang CL, Qiu HY, Xia XX, Yang J, Wang XR. Exploration of ETV6::ABL1-positive AML with concurrent NPM1 and FLT3-ITD mutations. Ann Hematol 2024; 103:4295-4304. [PMID: 39105739 DOI: 10.1007/s00277-024-05917-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 07/26/2024] [Indexed: 08/07/2024]
Abstract
ETV6::ABL1 is a rare fusion gene that found in MPN, ALL, and AML. It has a complex and diverse formation mechanism due to the reciprocal orientations of the ETV6 and ABL1 genes relative to the centromeres. NPM1 is frequently mutated in adult AML, often accompanied by FLT3-ITD, which suggests molecular synergisms in AML pathogenesis. Previous reports on ETV6::ABL1 mostly focus on FLT3-ITD. In this study, we present a case of AML with ETV6::ABL1, along with NPM1 and FLT3-ITD. The patient showed a rapid increase in primitive cells at the initial stage, along with the presence of immature granulocytes and erythrocytes. Through cytogenetic analysis, fluorescence in situ hybridization (FISH), and RNA-seq, we elucidated the mechanism behind the formation of the ETV6::ABL1 fusion gene. Despite conventional chemotherapy failure and rapid tumor proliferation, we attempted to add FLT3 inhibitor sorafenib to the treatment, along with chemotherapy bridging to haploidentical transplantation. After haplo-HSCT, a combination of sorafenib and dasatinib was administered as maintenance therapy. The patient achieved complete remission (CR) and maintained it for 11 months. The intricate genetic landscape observed in this case presents diagnostic dilemmas and therapeutic challenges, emphasizing the importance of a comprehensive understanding of its implications for disease classification, risk stratification, and treatment selection.
Collapse
Affiliation(s)
- Hui-Dan Li
- Clinical Laboratory Medicine Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Si-Si Chen
- Clinical Research Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jing Ding
- Clinical Laboratory Medicine Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Chun-Ling Zhang
- Clinical Laboratory Medicine Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Hui-Yin Qiu
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Xin-Xin Xia
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jun Yang
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| | - Xiao-Rui Wang
- Clinical Laboratory Medicine Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200080, China.
| |
Collapse
|
2
|
Zhu P, Wu X, Zhang RY, Hsu CC, Zhang ZY, Tao WA. An Integrated Proteomic Strategy to Identify SHP2 Substrates. J Proteome Res 2022; 21:2515-2525. [PMID: 36103635 PMCID: PMC9597472 DOI: 10.1021/acs.jproteome.2c00481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Protein phosphatases play an essential role in normal cell physiology and the development of diseases such as cancer. The innate challenges associated with studying protein phosphatases have limited our understanding of their substrates, molecular mechanisms, and unique functions within highly coordinated networks. Here, we introduce a novel strategy using substrate-trapping mutants coupled with quantitative proteomics methods to identify physiological substrates of Src homology 2 containing protein tyrosine phosphatase 2 (SHP2) in a high-throughput manner. The technique integrates three parallel mass spectrometry-based proteomics experiments, including affinity isolation of substrate-trapping mutant complex using wild-type and SHP2 KO cells, in vivo global quantitative phosphoproteomics, and in vitro phosphatase reaction. We confidently identified 18 direct substrates of SHP2 in the epidermal growth factor receptor signaling pathways, including both known and novel SHP2 substrates. Docking protein 1 was further validated using biochemical assays as a novel SHP2 substrate, providing a mechanism for SHP2-mediated Ras activation. This advanced workflow improves the systemic identification of direct substrates of protein phosphatases, facilitating our understanding of the equally important roles of protein phosphatases in cellular signaling.
Collapse
Affiliation(s)
- Peipei Zhu
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
- Department of Biochemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Xiaofeng Wu
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ruo-Yu Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Chuan-Chih Hsu
- Department of Biochemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Zhong-Yin Zhang
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, United States
| | - W Andy Tao
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
- Department of Biochemistry, Purdue University, West Lafayette, Indiana 47907, United States
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
3
|
Uemura S, Nishimura N, Hasegawa D, Shono A, Sakaguchi K, Matsumoto H, Nakamachi Y, Saegusa J, Yokoi T, Tahara T, Tamura A, Yamamoto N, Saito A, Kozaki A, Kishimoto K, Ishida T, Nino N, Takafuji S, Mori T, Iijima K, Kosaka Y. ETV6-ABL1 fusion combined with monosomy 7 in childhood B-precursor acute lymphoblastic leukemia. Int J Hematol 2018; 107:604-609. [PMID: 29177615 DOI: 10.1007/s12185-017-2371-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 11/16/2017] [Accepted: 11/21/2017] [Indexed: 01/17/2023]
Abstract
ETV6-ABL1 fusion is a rare but recurrent oncogenic lesion found in childhood B-cell precursor acute lymphoblastic leukemia (BCP-ALL), without an established chromosomal abnormality, and is associated with poor outcome. In ETV6-ABL1-positive cases, an in-frame fusion produced by a complex rearrangement results in constitutive chimeric tyrosine kinase activity. Monosomy 7 is also a rare and unfavorable chromosomal abnormality in childhood BCP-ALL. Here, we report a 14-year-old female BCP-ALL patient with ETV6-ABL1 fusion combined with monosomy 7. She was admitted to our hospital because of persistent fever. Bone marrow nuclear cell count on admission was 855,000/µL with 90.0% blastic cells of lymphoid morphology. Blasts were positive for CD10, CD19, CD20, CD34, cyCD79a, cyTdT, HLA-DR, and CD66c, had a karyotype of 45, XX, - 7 [18/20] and a split signal for ABL1 FISH probe (92.7%), and were sensitive to tyrosine kinase inhibitors, imatinib and dasatinib, in vitro. ETV6-ABL1 fusion transcript was identified by whole transcriptome sequencing and confirmed by RT-PCR. She was treated with the high-risk protocol based on ALL-BFM 95, achieved complete remission (CR) after induction chemotherapy, and maintained CR for 4 months. To our knowledge, this is the first report of ETV6-ABL1 fusion combined with monosomy 7 in childhood BCP-ALL.
Collapse
Affiliation(s)
- Suguru Uemura
- Department of Hematology and Oncology, Children's Cancer Center, Kobe Children's Hospital, Minatojima Minamimachi 1-6-7, Chuo-ku, Kobe, 650-0047, Japan.
- Department of Pediatrics, Kobe University, Graduate School of Medicine, Kobe, Japan.
| | - Noriyuki Nishimura
- Department of Pediatrics, Kobe University, Graduate School of Medicine, Kobe, Japan
| | - Daiichiro Hasegawa
- Department of Hematology and Oncology, Children's Cancer Center, Kobe Children's Hospital, Minatojima Minamimachi 1-6-7, Chuo-ku, Kobe, 650-0047, Japan
| | - Akemi Shono
- Department of Pediatrics, Kobe University, Graduate School of Medicine, Kobe, Japan
| | - Kimiyoshi Sakaguchi
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | | | - Yuji Nakamachi
- Division of Clinical Laboratory, Kobe University Hospital, Kobe, Japan
| | - Jun Saegusa
- Division of Clinical Laboratory, Kobe University Hospital, Kobe, Japan
| | - Takehito Yokoi
- Department of Hematology and Oncology, Children's Cancer Center, Kobe Children's Hospital, Minatojima Minamimachi 1-6-7, Chuo-ku, Kobe, 650-0047, Japan
| | - Teppei Tahara
- Department of Hematology and Oncology, Children's Cancer Center, Kobe Children's Hospital, Minatojima Minamimachi 1-6-7, Chuo-ku, Kobe, 650-0047, Japan
| | - Akihiro Tamura
- Department of Hematology and Oncology, Children's Cancer Center, Kobe Children's Hospital, Minatojima Minamimachi 1-6-7, Chuo-ku, Kobe, 650-0047, Japan
| | - Nobuyuki Yamamoto
- Department of Pediatrics, Kobe University, Graduate School of Medicine, Kobe, Japan
| | - Atsuro Saito
- Department of Hematology and Oncology, Children's Cancer Center, Kobe Children's Hospital, Minatojima Minamimachi 1-6-7, Chuo-ku, Kobe, 650-0047, Japan
| | - Aiko Kozaki
- Department of Hematology and Oncology, Children's Cancer Center, Kobe Children's Hospital, Minatojima Minamimachi 1-6-7, Chuo-ku, Kobe, 650-0047, Japan
| | - Kenji Kishimoto
- Department of Hematology and Oncology, Children's Cancer Center, Kobe Children's Hospital, Minatojima Minamimachi 1-6-7, Chuo-ku, Kobe, 650-0047, Japan
| | - Toshiaki Ishida
- Department of Hematology and Oncology, Children's Cancer Center, Kobe Children's Hospital, Minatojima Minamimachi 1-6-7, Chuo-ku, Kobe, 650-0047, Japan
| | - Nanako Nino
- Department of Pediatrics, Kobe University, Graduate School of Medicine, Kobe, Japan
| | - Satoru Takafuji
- Department of Pediatrics, Kobe University, Graduate School of Medicine, Kobe, Japan
| | - Takeshi Mori
- Department of Pediatrics, Kobe University, Graduate School of Medicine, Kobe, Japan
| | - Kazumoto Iijima
- Department of Pediatrics, Kobe University, Graduate School of Medicine, Kobe, Japan
| | - Yoshiyuki Kosaka
- Department of Hematology and Oncology, Children's Cancer Center, Kobe Children's Hospital, Minatojima Minamimachi 1-6-7, Chuo-ku, Kobe, 650-0047, Japan
| |
Collapse
|
4
|
EML1-ABL1 Is Activated by Coiled-Coil-Mediated Oligomerization and Induces T-Cell Acute Lymphoblastic Leukemia or Myeloproliferative Disease in a Mouse Bone Marrow Transplant Model. Hemasphere 2018; 2:e32. [PMID: 31723760 PMCID: PMC6745902 DOI: 10.1097/hs9.0000000000000032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/09/2018] [Accepted: 01/15/2018] [Indexed: 11/29/2022] Open
Abstract
Supplemental Digital Content is available in the text
Collapse
|
5
|
Yokota A, Hirai H, Shoji T, Maekawa T, Okuda K. Constitutively active ABL family kinases, TEL/ABL and TEL/ARG, harbor distinct leukemogenic activities in vivo. Leukemia 2017; 31:2742-2751. [PMID: 28386107 DOI: 10.1038/leu.2017.114] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 03/27/2017] [Indexed: 01/10/2023]
Abstract
ABL (ABL1) and ARG (ABL2) are highly homologous to each other in overall domain structure and amino-acid sequence, with the exception of their C termini. As with ABL, translocations that fuse ARG to ETV6/TEL have been identified in patients with leukemia. To assess the in vivo leukemogenic activity of constitutively active ABL and ARG, we generated a bone marrow (BM) transplantation model using the chimeric forms TEL/ABL and TEL/ARG, which have comparable kinase activities. TEL/ABL rapidly induced fatal myeloid leukemia in recipient mice, whereas recipients of TEL/ARG-transduced cells did not develop myeloid leukemia, instead, they succumbed to a long-latency infiltrative mastocytosis that could be adoptively transferred to secondary recipients. Swapping of the C termini of ABL and ARG altered disease latency and phenotypes. In a detailed in vitro study, TEL/ARG strongly promoted mast cell differentiation in response to stem cell factor or interleukin-3, whereas TEL/ABL preferentially induced myeloid differentiation of hematopoietic stem/progenitor cells. These results indicate that ABL and ARG kinase activate distinct differentiation pathways to induce specific diseases in vivo, that is, myeloid leukemia and mastocytosis, respectively. Further elucidation of the differences in their properties should provide important insight into the pathogenic mechanisms of oncogenes of the ABL kinase family.
Collapse
Affiliation(s)
- A Yokota
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan
| | - H Hirai
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan
| | - T Shoji
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan
| | - T Maekawa
- Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, Kyoto, Japan
| | - K Okuda
- Department of Molecular Diagnostics and Therapeutics, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| |
Collapse
|
6
|
A novel three-way rearrangement involving ETV6 (12p13) and ABL1 (9q34) with an unknown partner on 3p25 resulting in a possible ETV6-ABL1 fusion in a patient with acute myeloid leukemia: a case report and a review of the literature. Biomark Res 2016; 4:16. [PMID: 27570624 PMCID: PMC5000511 DOI: 10.1186/s40364-016-0070-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 08/02/2016] [Indexed: 11/30/2022] Open
Abstract
Background Acute myeloid leukemia (AML) is commonly characterized by several chromosomal abnormalities resulting in the formation of chimeric genes that play various roles in leukemogenesis. Translocations resulting in the ETV6-ABL1 fusion gene are rare in AML and other hematologic malignancies with only thirty-two previously reported cases in the literature, five of which were AML. Findings Herein, we report the case of a 73-year-old male with acute myeloid leukemia arising from MDS, negative for PDGFRA and PDGFRB, positive for bone marrow eosinophilia, rash, and marked fluid retention, which improved dramatically with imatinib therapy. Conventional cytogenetics revealed a t(3;9)(p25;q34), t(5;18)(q13;p11.2), and additional material of unknown origin at 12p11.2 in 2 out of 10 metaphases analyzed. Interphase FISH studies showed evidence of ETV6 (12p13) and ABL1 (9q34) rearrangements in 41.3 % and 5.7 % of the cells respectively. FISH studies on previously G-banded metaphases showed colocalization of ABL1 and ETV6 signals to the short arm of chromosome 3 at 3p25 suggesting a possible ETV6-ABL1 fusion. Subtelomeric metaphase FISH studies also showed the presence of a subtelomere 3p signal on the long arm of the derivative 9, and no subtelomere 3p signal on the derivative chromosome 12. Conclusions These findings suggest a complex rearrangement involving an insertion of ETV6 into 3p25 followed by a reciprocal translocation involving 3p25 and 9q34, resulting in a possible ETV6-ABL1 fusion. This case highlights the importance of FISH to characterize complex rearrangements in myeloid malignancies, particularly those resulting in clinically significant chimeric genes.
Collapse
|
7
|
Zaliova M, Moorman AV, Cazzaniga G, Stanulla M, Harvey RC, Roberts KG, Heatley SL, Loh ML, Konopleva M, Chen IM, Zimmermannova O, Schwab C, Smith O, Mozziconacci MJ, Chabannon C, Kim M, Frederik Falkenburg JH, Norton A, Marshall K, Haas OA, Starkova J, Stuchly J, Hunger SP, White D, Mullighan CG, Willman CL, Stary J, Trka J, Zuna J. Characterization of leukemias with ETV6-ABL1 fusion. Haematologica 2016; 101:1082-93. [PMID: 27229714 DOI: 10.3324/haematol.2016.144345] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 05/18/2016] [Indexed: 12/26/2022] Open
Abstract
To characterize the incidence, clinical features and genetics of ETV6-ABL1 leukemias, representing targetable kinase-activating lesions, we analyzed 44 new and published cases of ETV6-ABL1-positive hematologic malignancies [22 cases of acute lymphoblastic leukemia (13 children, 9 adults) and 22 myeloid malignancies (18 myeloproliferative neoplasms, 4 acute myeloid leukemias)]. The presence of the ETV6-ABL1 fusion was ascertained by cytogenetics, fluorescence in-situ hybridization, reverse transcriptase-polymerase chain reaction and RNA sequencing. Genomic and gene expression profiling was performed by single nucleotide polymorphism and expression arrays. Systematic screening of more than 4,500 cases revealed that in acute lymphoblastic leukemia ETV6-ABL1 is rare in childhood (0.17% cases) and slightly more common in adults (0.38%). There is no systematic screening of myeloproliferative neoplasms; however, the number of ETV6-ABL1-positive cases and the relative incidence of acute lymphoblastic leukemia and myeloproliferative neoplasms suggest that in adulthood ETV6-ABL1 is more common in BCR-ABL1-negative chronic myeloid leukemia-like myeloproliferations than in acute lymphoblastic leukemia. The genomic profile of ETV6-ABL1 acute lymphoblastic leukemia resembled that of BCR-ABL1 and BCR-ABL1-like cases with 80% of patients having concurrent CDKN2A/B and IKZF1 deletions. In the gene expression profiling all the ETV6-ABL1-positive samples clustered in close vicinity to BCR-ABL1 cases. All but one of the cases of ETV6-ABL1 acute lymphoblastic leukemia were classified as BCR-ABL1-like by a standardized assay. Over 60% of patients died, irrespectively of the disease or age subgroup examined. In conclusion, ETV6-ABL1 fusion occurs in both lymphoid and myeloid leukemias; the genomic profile and clinical behavior resemble BCR-ABL1-positive malignancies, including the unfavorable prognosis, particularly of acute leukemias. The poor outcome suggests that treatment with tyrosine kinase inhibitors should be considered for patients with this fusion.
Collapse
Affiliation(s)
- Marketa Zaliova
- CLIP, Department of Pediatric Hematology and Oncology, 2 Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Anthony V Moorman
- Leukaemia Research Cytogenetics Group, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Giovanni Cazzaniga
- Centro Ricerca Tettamanti, Clinica Pediatrica, Università di Milano-Bicocca, Fondazione MBBM/Ospedale San Gerardo, Monza, Italy
| | - Martin Stanulla
- Pediatric Hematology and Oncology, Hannover Medical School, Germany
| | | | - Kathryn G Roberts
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sue L Heatley
- South Australia Health and Medical Research Institute, Adelaide, Australia
| | - Mignon L Loh
- Department of Pediatrics, Hematology-Oncology, Benioff Children's Hospital, and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Marina Konopleva
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - I-Ming Chen
- University of New Mexico Cancer Center, Albuquerque, NM, USA
| | - Olga Zimmermannova
- CLIP, Department of Pediatric Hematology and Oncology, 2 Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Claire Schwab
- Leukaemia Research Cytogenetics Group, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Owen Smith
- Department of Haematology, Our Lady's Children's Hospital, Dublin, Ireland
| | | | | | - Myungshin Kim
- Department of Laboratory Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | | | - Alice Norton
- Birmingham Children's Hospital, NHS Foundation Trust, UK
| | - Karen Marshall
- Department of Cytogenetics, Leicester Royal Infirmary NHS Trust, UK
| | - Oskar A Haas
- St. Anna Children's Hospital, Childrens Cancer Research Institute, Vienna, Austria
| | - Julia Starkova
- CLIP, Department of Pediatric Hematology and Oncology, 2 Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Jan Stuchly
- CLIP, Department of Pediatric Hematology and Oncology, 2 Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Stephen P Hunger
- Department of Pediatrics and the Center for Childhood Cancer Research, Children's Hospital of Philadelphia and the University of Pennsylvania Perelman School of Medicine, PA, USA
| | - Deborah White
- South Australia Health and Medical Research Institute, Adelaide, Australia
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Jan Stary
- CLIP, Department of Pediatric Hematology and Oncology, 2 Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Jan Trka
- CLIP, Department of Pediatric Hematology and Oncology, 2 Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Jan Zuna
- CLIP, Department of Pediatric Hematology and Oncology, 2 Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
8
|
Gancheva K, Virchis A, Howard-Reeves J, Cross NC, Brazma D, Grace C, Kotzampaltiris P, Partheniou F, Nacheva E. Myeloproliferative neoplasm with ETV6-ABL1 fusion: a case report and literature review. Mol Cytogenet 2013; 6:39. [PMID: 24053143 PMCID: PMC3853649 DOI: 10.1186/1755-8166-6-39] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 08/20/2013] [Indexed: 11/10/2022] Open
Abstract
ETV6-ABL1 is a rare gene fusion with oncogenic properties, reported so far in 28 patients presenting a variety of haematological malignancies associated with clinical outcome, including chronic myeloid leukaemia (CML), acute myeloid leukaemia (AML), acute lymphoblastic leukaemia (ALL) and chronic myeloproliferative neoplasm (cMPN). Here we report on a 46-year-old female who presented with Philadelphia negative CML, positive for the ETV6-ABL1 fusion. Whole genome screening carried out with oligonucleotide arrays showed a subtle loss at 12p13 and cryptic imbalances within the 9q34.3 region in a highly unstable genome. FISH mapping with custom BAC probes identified two breakpoints 5 Mb apart within the 9q34 region, together with a break at 12p13. While FISH with commercial BCR-ABL1 probes failed to detect any ABL1 changes, the ETV6 break-apart probe conclusively identified the ETV6-ABL1 fusion thus determining the probe’s role as the primary diagnostic FISH test for this chimeric oncogene. In addition, we confirm the association of the ETV6-ABL1 fusion with imatinib resistance reported so far in three other patients, while recording excellent response to the 2nd generation tyrosine kinase inhibitor (TKI) nilotinib. In summary, we highlight the value of ETV6 FISH as a diagnostic test and the therapy resistance of ETV6-ABL1 positive disorders to imatinib.
Collapse
Affiliation(s)
- Katya Gancheva
- Leukaemia Cytogenetics, Academic Haematology, UCL Medical School, Royal Free Campus, Rowland Hill Street, London NW3 2PF, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Okuda K, Hirai H. Distinct Transforming Activity of ABL Family Tyrosine Kinase Oncogenes Is Induced by Their C-Terminal Domain*. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/ojbd.2013.33a005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
10
|
Park J, Kim M, Lim J, Kim Y, Han K, Kim JS, Lee S, Kim HJ, Min WS. Variant of ETV6/ABL1 gene is associated with leukemia phenotype. Acta Haematol 2012; 129:78-82. [PMID: 23171811 DOI: 10.1159/000342490] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 08/02/2012] [Indexed: 11/19/2022]
Abstract
The ETV6/ABL1 fusion transcript is thought to be a very rare aberration in hematopoietic malignancies. We describe two new cases of acute leukemia with the ETV6/ABL1 fusion, acute myeloid leukemia with eosinophilia (case 1) and B acute lymphoblastic leukemia (ALL) (case 2), screened by multiplex RT-PCR. The ETV6/ABL1 fusion was also confirmed by fluorescence in situ hybridization using a mixture of BCR/ABL1 and ETV6/RUNX1 probes. A thorough review of all published cases showed that all 7 reported ALL patients possess the type A ETV6/ABL1 fusion transcript, composed of the first 4 exons of ETV6 fused to the second exon of ABL1. The presence of the type A fusion transcript strongly implies ALL manifestation in ETV6/ABL1-positive hematologic malignancies as minor BCR breakpoint in BCR/ABL1-positive ALL.
Collapse
Affiliation(s)
- Joonhong Park
- Department of Laboratory Medicine, Catholic Blood and Marrow Transplantation Center, The Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Gavrilescu LC, Van Etten RA. Murine retroviral bone marrow transplantation models for the study of human myeloproliferative disorders. ACTA ACUST UNITED AC 2012; Chapter 14:Unit14.10. [PMID: 22294220 DOI: 10.1002/0471141755.ph1410s43] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Human myeloproliferative diseases are common hematologic disorders characterized by clonal overproduction of maturing myeloid or erythroid cells, often caused by expression of a mutant, dysregulated tyrosine kinase (TK). These diseases can be accurately modeled in laboratory mice by the retroviral transfer of a mutant TK gene into murine hematopoietic stem and progenitor cells, followed by transplantation of these cells into irradiated recipient mice. This yields a model system for analyzing the molecular pathophysiology of these conditions and provides a platform for testing therapies, particularly molecularly targeted new chemical entities (NCEs). The Basic Protocol in this unit describes the preparation of mouse bone marrow cells to express the relevant human oncogene before transplanting them into irradiated recipient mice. An alternate protocol describes a similar technique that allows specific induction of lymphoproliferative disease by some TKs. Support protocols for generating and titering retroviral stocks are also included.
Collapse
Affiliation(s)
- L Cristina Gavrilescu
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | | |
Collapse
|
12
|
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
|
13
|
Hagemeijer A, Graux C. ABL1 rearrangements in T-cell acute lymphoblastic leukemia. Genes Chromosomes Cancer 2010; 49:299-308. [PMID: 20073070 DOI: 10.1002/gcc.20743] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is the result of multiple oncogenic insults of thymocytes. Recently, new ABL1 fusion genes have been identified that provide proliferation and survival advantage to lymphoblasts. These are the NUP214-ABL1 fusion gene, on amplified episomes, the unique case of EML1-ABL1 fusion due to a cryptic t(9;14)(q34;q32) and the seldom reported BCR-ABL1 and ETV6-ABL1 chimeric genes. The most frequent and strictly associated with T-ALL is the NUP214-ABL1 fusion identified in 6% of cases, in both children and adults. Patients present with classical T-ALL features. Cytogenetically, the fusion is cryptic but seen by FISH on amplified episomes or more rarely as a small hsr. The ABL1 fusion is a late event associated with other genetic alterations like NOTCH1 activating mutation, deletion of CDKN2A locus, and ectopic expression of TLX1 or TLX3. The mechanism of activation of the NUP214-ABL1 protein is unique and requires localization at the nucleopore complex and interaction with other nuclear pore proteins for crossphosphorylation and constitutive kinase activity. The ABL1 fusion proteins are sensitive to tyrosine kinase inhibitors, which can be included in future treatment strategy.
Collapse
Affiliation(s)
- Anne Hagemeijer
- Department of Human Genetics, University of Leuven, Leuven, Belgium.
| | | |
Collapse
|
14
|
Johnson KJ, Griswold IJ, O'Hare T, Corbin AS, Loriaux M, Deininger MW, Druker BJ. A BCR-ABL mutant lacking direct binding sites for the GRB2, CBL and CRKL adapter proteins fails to induce leukemia in mice. PLoS One 2009; 4:e7439. [PMID: 19823681 PMCID: PMC2757918 DOI: 10.1371/journal.pone.0007439] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Accepted: 09/17/2009] [Indexed: 11/19/2022] Open
Abstract
The BCR-ABL tyrosine kinase is the defining feature of chronic myeloid leukemia (CML) and its kinase activity is required for induction of this disease. Current thinking holds that BCR-ABL forms a multi-protein complex that incorporates several substrates and adaptor proteins and is stabilized by multiple direct and indirect interactions. Signaling output from this highly redundant network leads to cellular transformation. Proteins known to be associated with BCR-ABL in this complex include: GRB2, c-CBL, p62(DOK), and CRKL. These proteins in turn, link BCR-ABL to various signaling pathways indicated in cellular transformation. In this study we show that a triple mutant of BCR-ABL with mutations of the direct binding sites for GRB2, CBL, p62(DOK) and CRKL, is defective for transformation of primary hematopoietic cells in vitro and in a murine CML model, while it retains the capacity to induce IL-3 independence in 32D cells. Compared to BCR-ABL, the triple mutant's ability to activate the MAP kinase and PI3-kinase pathways is severely compromised, while STAT5 phosphorylation is maintained, suggesting that the former are crucial for the transformation of primary cells, but dispensable for transformation of factor dependent cell lines. Our data suggest that inhibition of BCR-ABL-induced leukemia by disrupting protein interactions could be possible, but would require blocking of multiple sites.
Collapse
Affiliation(s)
- Kara J Johnson
- Division of Hematology and Medical Oncology, Oregon Health & Science University Knight Cancer Institute, Portland, Oregon, United States of America.
| | | | | | | | | | | | | |
Collapse
|
15
|
Nand R, Bryke C, Kroft SH, Divgi A, Bredeson C, Atallah E. Myeloproliferative disorder with eosinophilia and ETV6–ABL gene rearrangement: Efficacy of second-generation tyrosine kinase inhibitors. Leuk Res 2009; 33:1144-6. [DOI: 10.1016/j.leukres.2009.03.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2009] [Revised: 01/28/2009] [Accepted: 03/09/2009] [Indexed: 10/20/2022]
|
16
|
Kelly JC, Shahbazi N, Scheerle J, Jahn J, Suchen S, Christacos NC, Mowrey PN, Witt MH, Hostetter A, Meloni-Ehrig AM. Insertion (12;9)(p13;q34q34): a cryptic rearrangement involving ABL1/ETV6 fusion in a patient with Philadelphia-negative chronic myeloid leukemia. ACTA ACUST UNITED AC 2009; 192:36-9. [DOI: 10.1016/j.cancergencyto.2009.02.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Revised: 02/12/2009] [Accepted: 02/13/2009] [Indexed: 12/01/2022]
|
17
|
Tefferi A. Molecular drug targets in myeloproliferative neoplasms: mutant ABL1, JAK2, MPL, KIT, PDGFRA, PDGFRB and FGFR1. J Cell Mol Med 2008; 13:215-37. [PMID: 19175693 PMCID: PMC3823350 DOI: 10.1111/j.1582-4934.2008.00559.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Therapeutically validated oncoproteins in myeloproliferative neoplasms (MPN) include BCR-ABL1 and rearranged PDGFR proteins. The latter are products of intra- (e.g. FIP1L1-PDGFRA) or inter-chromosomal (e.g.ETV6-PDGFRB) gene fusions. BCR-ABL1 is associated with chronic myelogenous leukaemia (CML) and mutant PDGFR with an MPN phenotype characterized by eosinophilia and in addition, in case of FIP1L1-PDGFRA, bone marrow mastocytosis. These genotype-phenotype associations have been effectively exploited in the development of highly accurate diagnostic assays and molecular targeted therapy. It is hoped that the same will happen in other MPN with specific genetic alterations: polycythemia vera (JAK2V617F and other JAK2 mutations), essential thrombocythemia (JAK2V617F and MPL515 mutations), primary myelofibrosis (JAK2V617F and MPL515 mutations), systemic mastocytosis (KITD816V and other KIT mutations) and stem cell leukaemia/lymphoma (ZNF198-FGFR1 and other FGFR1 fusion genes). The current review discusses the above-listed mutant molecules in the context of their value as drug targets.
Collapse
Affiliation(s)
- Ayalew Tefferi
- Division of Hematology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
18
|
Ko J, Patel N, Ikawa T, Kawamoto H, Frank O, Rivera RR, Van Etten RA, Murre C. Suppression of E-protein activity interferes with the development of BCR-ABL-mediated myeloproliferative disease. Proc Natl Acad Sci U S A 2008; 105:12967-72. [PMID: 18725623 PMCID: PMC2529058 DOI: 10.1073/pnas.0805073105] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2008] [Indexed: 12/21/2022] Open
Abstract
E-proteins are a class of helix-loop-helix (HLH) proteins, which play multiple roles throughout lymphoid development. The DNA binding activities of the E-proteins are regulated by a distinct class of antagonistic HLH proteins, named Id1-4. Here we demonstrate that Id2 deficient mice in a C57BL/6 genetic background exhibit increased cellularity in the granulocyte/myeloid progenitor compartment and show significantly higher numbers of maturing neutrophils. Within 6 months of age, Id2 deficient mice succumbed from overwhelming granulocytosis. The disease closely mimicked the distinctive features of human chronic myeloid leukemia: leukocytosis with maturing neutrophils, splenomegaly, hepatomegaly, and myeloid infiltration into peripheral tissues, including spleen, liver, and lungs. Strikingly, forced Id2 expression in murine bone marrow cells substantially delayed the onset of myeloproliferative disease (MPD). Collectively, these studies show that suppression of E-protein activity interferes with the development of BCR-ABL-mediated MPD.
Collapse
Affiliation(s)
- Jinkyung Ko
- *Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093
| | - Nihal Patel
- Tufts Medical Center, 800 Washington Street, Boston, MA 02111
| | - Tomokatsu Ikawa
- Laboratory for Lymphocyte Development, RIKEN Research Center for Allergy and Immunology, Yokohama 230-0045, Japan; and
| | - Hiroshi Kawamoto
- Laboratory for Lymphocyte Development, RIKEN Research Center for Allergy and Immunology, Yokohama 230-0045, Japan; and
| | - Oliver Frank
- III Medizinische Klinik, Medizinische Fakultät Mannheim der Universität Heidelberg, Wiesbadener Strasse 7-11, 68305 Mannheim, Germany
| | - Richard R. Rivera
- *Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093
| | | | - Cornelis Murre
- *Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093
| |
Collapse
|
19
|
Baeumler J, Szuhai K, Falkenburg JF, van Schie ML, Ottmann OG, Nijmeijer BA. Establishment and cytogenetic characterization of a human acute lymphoblastic leukemia cell line (ALL-VG) with ETV6/ABL1 rearrangement. ACTA ACUST UNITED AC 2008; 185:37-42. [DOI: 10.1016/j.cancergencyto.2008.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 04/25/2008] [Accepted: 05/02/2008] [Indexed: 11/29/2022]
|
20
|
Walz C, Cross NCP, Van Etten RA, Reiter A. Comparison of mutated ABL1 and JAK2 as oncogenes and drug targets in myeloproliferative disorders. Leukemia 2008; 22:1320-34. [PMID: 18528425 PMCID: PMC3490192 DOI: 10.1038/leu.2008.133] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2008] [Revised: 04/24/2008] [Accepted: 04/25/2008] [Indexed: 01/17/2023]
Abstract
Constitutively activated mutants of the non-receptor tyrosine kinases (TK) ABL1 (Abelson murine leukemia viral (v-abl) homolog (1) protein) and JAK2 (JAnus Kinase 2 or Just Another Kinase 2) play a central role in the pathogenesis of clinically and morphologically distinct chronic myeloproliferative disorders but are also found in some cases of de novo acute leukemia and lymphoma. Ligand-independent activation occurs as a consequence of point mutations or insertions/deletions within functionally relevant regulatory domains (JAK2) or the creation of TK fusion proteins by balanced reciprocal translocations, insertions or episomal amplification (ABL1 and JAK2). Specific abnormalities are correlated with clinical phenotype, although some are broad and encompass several World Health Organization-defined entities. TKs are excellent drug targets as exemplified by the activity of imatinib in BCR-ABL1-positive disease, particularly chronic myeloid leukemia. Resistance to imatinib is seen in a minority of cases and is often associated with the appearance of secondary point mutations within the TK domain of BCR-ABL1. These mutations are highly variable in their sensitivity to increased doses of imatinib or alternative TK inhibitors such as nilotinib or dasatinib. Selective and non-selective inhibitors of JAK2 are currently being developed, and encouraging data from pre-clinical experiments and initial phase-I studies regarding efficacy and potential toxicity of these compounds have already been reported.
Collapse
Affiliation(s)
- Christoph Walz
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Nicholas C. P. Cross
- Wessex Regional Genetics Laboratory, Salisbury and Human Genetics Division, University of Southampton, U.K
| | | | - Andreas Reiter
- III. Medizinische Universitätsklinik, Medizinische Fakultät Mannheim der Universität Heidelberg, Germany
| |
Collapse
|
21
|
Abstract
BCR-ABL is proposed to impair cell-cycle control by disabling p27, a tumor suppressor that inhibits cyclin-dependent kinases. We show that in cell lines p27 expression is inversely correlated with expression of SKP2, the F-box protein of SCF(SKP2) (SKP1/Cul1/F-box), the E3 ubiquitin ligase that promotes proteasomal degradation of p27. Inhibition of BCR-ABL kinase causes G(1) arrest, down-regulation of SKP2, and accumulation of p27. Ectopic expression of wild-type SKP2, but not a mutant unable to recognize p27, partially rescues cell-cycle progression. A similar regulation pattern is seen in cell lines transformed by FLT3-ITD, JAK2(V617F), and TEL-PDGFRbeta, suggesting that the SKP2/p27 conduit may be a universal target for leukemogenic tyrosine kinases. Mice that received transplants of BCR-ABL-infected SKP2(-/-) marrow developed a myeloproliferative syndrome but survival was significantly prolonged compared with recipients of BCR-ABL-expressing SKP2(+/+) marrow. SKP2(-/-) leukemic cells demonstrated higher levels of nuclear p27 than SKP2(+/+) counterparts, suggesting that the attenuation of leukemogenesis depends on increased p27 expression. Our data identify SKP2 as a crucial mediator of BCR-ABL-induced leukemogenesis and provide the first in vivo evidence that SKP2 promotes oncogenesis. Hence, stabilization of p27 by inhibiting its recognition by SCF(SKP2) may be therapeutically useful.
Collapse
|
22
|
Sustained suppression of Bcr-Abl-driven lymphoid leukemia by microRNA mimics. Proc Natl Acad Sci U S A 2007; 104:20501-6. [PMID: 18079287 DOI: 10.1073/pnas.0710532105] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Many cancers and leukemias are associated with strong dominant oncogenic mutations that activate tyrosine kinases and other classes of molecules, including transcription factors and antiapoptotic mechanisms. Some of these events can be targeted with small molecules or antibody-based therapeutics, but many remain intractable. In addition, cancer-related enzyme targets can often mutate, and drug-resistant variants are selected. Therapies directed at the mRNA encoding dominant oncogenes could provide a more global set of technologies for cancer treatment. To test this concept, we have used the model of transformation of hematopoietic cells by the chimeric Bcr-Abl oncogene, a highly activated tyrosine kinase. Our results show that tandem arrays of miRNA mimics, but not single miRNA mimics, directed against the Abl portion of the mRNA and introduced by lentiviral vectors can effectively alter the leukemogenic potency when the degree of suppression of expression of Bcr-Abl is reduced >200-fold from control levels. Only methods capable of such dramatic sustained reduction in the level of expression of highly activated kinase oncogenes are likely to be effective in controlling malignant cell populations.
Collapse
|
23
|
Abstract
Abnormalities of cytokine and growth factor signaling pathways are characteristic of all forms of leukemia: lymphoid and myeloid, acute and chronic. In normal hematopoietic cells, cytokines provide the stimulus for proliferation, survival, self-renewal, differentiation and functional activation. In leukemic cells, these pathways are usurped to subserve critical parts of the malignant program. In this review, our current knowledge of leukemic cell cytokine signaling will be summarized, and some speculations on the significance and implications of these insights will be advanced. A better understanding of aberrant cytokine signaling in leukemia should provide additional targets for the rational therapy of these diseases.
Collapse
Affiliation(s)
- R A Van Etten
- Molecular Oncology Research Institute and Division of Hematology/Oncology, Tufts-New England Medical Center, Boston, MA 02111, USA.
| |
Collapse
|
24
|
DeAngelo DJ. Role of Imatinib-Sensitive Tyrosine Kinases in the Pathogenesis of Chronic Myeloproliferative Disorders. Semin Hematol 2007. [DOI: 10.1053/j.seminhematol.2007.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
25
|
Pecquet C, Nyga R, Penard-Lacronique V, Smithgall TE, Murakami H, Régnier A, Lassoued K, Gouilleux F. The Src tyrosine kinase Hck is required for Tel-Abl- but not for Tel-Jak2-induced cell transformation. Oncogene 2006; 26:1577-85. [PMID: 16953222 DOI: 10.1038/sj.onc.1209949] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Tel-Abl and Tel-Jak2 are fusion proteins associated with human haematologic neoplasms. They possess constitutive tyrosine kinase activity and activate common downstream signalling pathways like Stat-5, PI3-K/Akt, Ras/MapK and NF-kappaB. In this study, we showed the specific requirement of Src family members for the Tel-Abl-mediated cell growth, activation of Stat5, PI3-K/Akt and Ras/MapK while dispensable for Tel-Jak2. Hck was found strongly phosphorylated in Tel-Abl-expressing Ba/F3 cells and sensitive to imatinib mesylate treatment, providing evidence that Hck is a target of Tel-Abl tyrosine kinase activity. Overexpression of a kinase dead form of Hck inhibits the proliferation of Ba/F3 cells expressing Tel-Abl as the phosphorylation of Akt and Erk1/2. These results argue for an important role of Hck in Tel-Abl oncogenic signalling.
Collapse
Affiliation(s)
- C Pecquet
- INSERM, E351, 3 rue des Louvels, Université de Picardie Jules Verne, Amiens, France
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Reciprocal chromosomal translocations may arise as a result of unfaithful repair of spontaneous DNA double-strand breaks, most probably induced by oxidative stress, radiation, genotoxic chemicals and/or replication stress. Genes encoding tyrosine kinases are targeted by these mechanisms resulting in the generation of chimera genes encoding fusion tyrosine kinases (FTKs). FTKs display transforming activity owing to their constitutive kinase activity causing deregulated proliferation, apoptosis, differentiation and adhesion. Moreover, FTKs are able to facilitate DNA repair, prolong activation of G(2)/M and S cell cycle checkpoints, and elevate expression of antiapoptotic protein Bcl-X(L), making malignant cells less responsive to antitumor treatment. FTKs may also stimulate the generation of reactive oxygen species and enhance spontaneous DNA damage in tumor cells. Unfortunately, FTKs compromise the fidelity of DNA repair mechanisms, which contribute to the accumulation of additional genetic abnormalities leading to the resistance to inhibitors such as imatinib mesylate and malignant progression of the disease.
Collapse
Affiliation(s)
- E T P Penserga
- Department of Microbiology and Immunology, School of Medicine, Temple University, Philadelphia, PA 19140, USA.
| | | |
Collapse
|
27
|
Malinge S, Monni R, Bernard O, Penard-Lacronique V. Activation of the NF-kappaB pathway by the leukemogenic TEL-Jak2 and TEL-Abl fusion proteins leads to the accumulation of antiapoptotic IAP proteins and involves IKKalpha. Oncogene 2006; 25:3589-97. [PMID: 16434962 DOI: 10.1038/sj.onc.1209390] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Abnormal activation of tyrosine kinases and of signaling pathways they control plays a critical role in the neoplastic process of human hematopoietic malignancy. The nuclear factor-kappaB (NF-kappaB) pathway is one of the signalings activated by the TEL-Jak2 and TEL-Abl oncoproteins and required for their antiapoptotic activity. To define the signal relay responsible for this activation, we used mouse embryonic fibroblast (MEF) cells and observed that TEL-Jak2- and TEL-Abl-mediated NF-kappaB induction was abolished in cells lacking the IkappaB kinase (IKK)alpha but not in IKKbeta(-/-) cells. Similar observations were performed with oncogenic forms of the FMS-like tyrosine kinase 3 (Flt-3) involved in the pathogenesis of one-third of acute myeloid leukemias. Rescue of TEL-Jak2-mediated NF-kappaB activation was obtained with a kinase-proficient form of IKKalpha in IKKalpha(-/-) MEF. Hematopoietic cells transformed by TEL-Jak2 and TEL-Abl showed sustained IKKalpha activity without promotion of NF-kappaB2/p100 processing, generally associated to IKKalpha functions. Furthermore, IAP1, IAP2 and XIAP, which are central regulators of the NF-kappaB-mediated survival pathway, were highly expressed in cells transformed by these oncoproteins. Our results indicate that these oncogenic tyrosine kinases preferentially use an IKKalpha-dependent mechanism to induce a persistent NF-kappaB activity and allow the production of antiapoptotic effectors that participate to their leukemogenic properties.
Collapse
Affiliation(s)
- S Malinge
- EMI 0210, Hôpital Necker-Enfants Malades, 149 rue de Sèvres, Paris, France
| | | | | | | |
Collapse
|
28
|
Abstract
Alterations of the ets family transcription factor ETV6 (TEL) and the RUNT domain transcription factor RUNX1 (AML1) play pivotal roles in the leukemogenesis of various types of leukemia. While only three fusion partners of RUNX1 namely ETO, ETV6 and MTG16 have been described so far, there is a plethora of ETV6 fusion partners with about 20 partners described so far. Apart from forming fusion genes there are other genetic alterations of ETV6 including deletions, point mutations and possible alterations at the promoter level that might contribute to the malignant phenotype. This review will focus on ETV6 and on the different mechanisms that are used by this gene to cause leukemia.
Collapse
Affiliation(s)
- Stefan K Bohlander
- Department of Medicine III, University Hospital Grosshadern, Marchioninistr. 15, D-81377 Munich, Germany.
| |
Collapse
|
29
|
Meyer-Monard S, Mühlematter D, Streit A, Chase AJ, Gratwohl A, Cross NCP, Jotterand M, Tichelli A. Broad molecular screening of an unclassifiable myeloproliferative disorder reveals an unexpected ETV6/ABL1 fusion transcript. Leukemia 2005; 19:1096-9. [PMID: 15789067 DOI: 10.1038/sj.leu.2403697] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2004] [Accepted: 01/07/2005] [Indexed: 11/08/2022]
|
30
|
Koschmieder S, Göttgens B, Zhang P, Iwasaki-Arai J, Akashi K, Kutok JL, Dayaram T, Geary K, Green AR, Tenen DG, Huettner CS. Inducible chronic phase of myeloid leukemia with expansion of hematopoietic stem cells in a transgenic model of BCR-ABL leukemogenesis. Blood 2004; 105:324-34. [PMID: 15331442 DOI: 10.1182/blood-2003-12-4369] [Citation(s) in RCA: 154] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
To develop murine models of leukemogenesis, a series of transgenic mice expressing BCR-ABL in different hematopoietic cell subsets was generated. Here we describe targeted expression of P210 BCR-ABL in stem and progenitor cells of murine bone marrow using the tet-off system. The transactivator protein tTA was placed under the control of the murine stem cell leukemia (SCL) gene 3' enhancer. Induction of BCR-ABL resulted in neutrophilia and leukocytosis, and the mice became moribund within 29 to 122 days. Autopsy of sick mice demonstrated splenomegaly, myeloid bone marrow hyperplasia, and extramedullary myeloid cell infiltration of multiple organs. BCR-ABL mRNA and protein were detectable in the affected organs. Fluorescence-activated cell sorter (FACS) analysis demonstrated a significant increase in mature and immature myeloid cells in bone marrow and spleen, together with increased bilineal B220+/Mac-1+ cells in the bone marrow. tTA mRNA was expressed in FACS-sorted hematopoietic stem cells expanded 26-fold after BCR-ABL induction. Thirty-one percent of the animals demonstrated a biphasic phenotype, consisting of neutrophilia and subsequent B-cell lymphoblastic disease, reminiscent of blast crisis. In summary, this mouse model recapitulates many characteristics of human chronic myeloid leukemia (CML) and may help elucidate basic leukemogenic mechanisms in CML stem cells during disease initiation and progression.
Collapse
MESH Headings
- Animals
- Bone Marrow/metabolism
- Bone Marrow/pathology
- Cell Transformation, Neoplastic/genetics
- Disease Models, Animal
- Disease Progression
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Leukocytosis/metabolism
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Mice, Transgenic
- Myeloid Cells/metabolism
- Myeloid Cells/pathology
- Neoplasm Invasiveness
- Neutrophils/metabolism
- Neutrophils/pathology
- Phenotype
- Spleen/metabolism
- Spleen/pathology
- Stem Cell Transplantation
- Survival Rate
- Transcriptional Activation/genetics
Collapse
Affiliation(s)
- Steffen Koschmieder
- Department of Hematology/Oncology, Harvard Institutes of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Million RP, Harakawa N, Roumiantsev S, Varticovski L, Van Etten RA. A direct binding site for Grb2 contributes to transformation and leukemogenesis by the Tel-Abl (ETV6-Abl) tyrosine kinase. Mol Cell Biol 2004; 24:4685-95. [PMID: 15143164 PMCID: PMC416425 DOI: 10.1128/mcb.24.11.4685-4695.2004] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2003] [Revised: 02/05/2004] [Accepted: 02/10/2004] [Indexed: 12/31/2022] Open
Abstract
A direct binding site for the Grb2 adapter protein is required for the induction of fatal chronic myeloid leukemia (CML)-like disease in mice by Bcr-Abl. Here, we demonstrate direct binding of Grb2 to the Tel-Abl (ETV6-Abl) fusion protein, the product of complex (9;12) chromosomal translocations in human leukemia, via tyrosine 314 encoded by TEL exon 5. A Tel-Abl point mutant (Y314F) and a splice variant without TEL exon 5 sequences (Deltae5) lacked Grb2 interaction and exhibited decreased binding and phosphorylation of the scaffolding protein Gab2 and impaired activation of phosphatidylinositol 3-kinase, Akt, and extracellular signal-regulated kinase/mitogen-activated protein kinase in hematopoietic cells. Tel-Abl Y314F and Deltae5 were unable to transform fibroblasts to anchorage-independent growth and were defective for B-lymphoid transformation in vitro and lymphoid leukemogenesis in vivo. Previously, we demonstrated that full-length Tel-Abl induced two distinct myeloproliferative diseases in mice: CML-like leukemia similar to that induced by Bcr-Abl and a novel syndrome of small-bowel myeloid infiltration endotoxemia and hepatic and renal failure. Lack of the Grb2 binding site had no effect on development of small bowel syndrome but significantly attenuated the induction of CML-like disease by Tel-Abl. These results suggest that direct binding of Grb2 is a common mechanism contributing to leukemogenesis by oncogenic Abl fusion proteins.
Collapse
Affiliation(s)
- Ryan P Million
- Molecular Oncology Research Institute, Tufts-New England Medical Center, 750 Washington St., Box 5609, Boston, MA 02111, USA
| | | | | | | | | |
Collapse
|
32
|
Roumiantsev S, Krause DS, Neumann CA, Dimitri CA, Asiedu F, Cross NCP, Van Etten RA. Distinct stem cell myeloproliferative/T lymphoma syndromes induced by ZNF198-FGFR1 and BCR-FGFR1 fusion genes from 8p11 translocations. Cancer Cell 2004; 5:287-98. [PMID: 15050920 DOI: 10.1016/s1535-6108(04)00053-4] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2003] [Revised: 01/20/2004] [Accepted: 02/03/2004] [Indexed: 10/26/2022]
Abstract
8p11 myeloproliferative syndrome (EMS) is a hematopoietic stem cell disorder characterized by myeloid hyperplasia and non-Hodgkin's lymphoma with chromosomal translocations fusing several genes, most commonly ZNF198, to fibroblast growth factor receptor-1 (FGFR1). However, patients with BCR-FGFR1 fusion present with typical chronic myeloid leukemia (CML). We demonstrate that ZNF198-FGFR1 induces EMS-like disease in mice, with myeloproliferation and T lymphoma arising from common multipotential progenitors. Mutation of FGFR1 Tyr766 attenuates both myeloid and lymphoid diseases, identifying phospholipase C-gamma1 as a downstream effector. Bcr-FGFR1 binds Grb2 via Bcr Tyr177 and induces CML-like leukemia in mice, whereas Bcr-FGFR1/Y177F lacks Grb2 binding and causes EMS-like disease. These results implicate different signaling pathways originating from both kinase and fusion partner in the pathogenesis of CML and EMS.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Animals
- Bone Marrow/metabolism
- Carrier Proteins/metabolism
- Chromosome Breakage/genetics
- Chromosomes, Human, Pair 8/genetics
- DNA-Binding Proteins/metabolism
- GRB2 Adaptor Protein
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Lymphoma, T-Cell/metabolism
- Mice
- Myeloproliferative Disorders/metabolism
- Oncogene Proteins/metabolism
- Phospholipase C gamma
- Protein-Tyrosine Kinases
- Proteins/metabolism
- Proto-Oncogene Proteins
- Proto-Oncogene Proteins c-bcr
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptor, Fibroblast Growth Factor, Type 1
- Receptors, Fibroblast Growth Factor/metabolism
- Signal Transduction
- Transcription Factors
- Translocation, Genetic/genetics
- Type C Phospholipases/metabolism
Collapse
|
33
|
Chan IT, Kutok JL, Williams IR, Cohen S, Kelly L, Shigematsu H, Johnson L, Akashi K, Tuveson DA, Jacks T, Gilliland DG. Conditional expression of oncogenic K-ras from its endogenous promoter induces a myeloproliferative disease. J Clin Invest 2004; 113:528-38. [PMID: 14966562 PMCID: PMC338267 DOI: 10.1172/jci20476] [Citation(s) in RCA: 219] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2003] [Accepted: 01/07/2004] [Indexed: 12/16/2022] Open
Abstract
Oncogenic ras alleles are among the most common mutations found in patients with acute myeloid leukemia (AML). Previously, the role of oncogenic ras in cancer was assessed in model systems overexpressing oncogenic ras from heterologous promoters. However, there is increasing evidence that subtle differences in gene dosage and regulation of gene expression from endogenous promoters play critical roles in cancer pathogenesis. We characterized the role of oncogenic K-ras expressed from its endogenous promoter in the hematopoietic system using a conditional allele and IFN-inducible, Cre-mediated recombination. Mice developed a completely penetrant myeloproliferative syndrome characterized by leukocytosis with normal maturation of myeloid lineage cells; myeloid hyperplasia in bone marrow; and extramedullary hematopoiesis in the spleen and liver. Flow cytometry confirmed the myeloproliferative phenotype. Genotypic and Western blot analysis demonstrated Cre-mediated excision and expression, respectively, of the oncogenic K-ras allele. Bone marrow cells formed growth factor-independent colonies in methylcellulose cultures, but the myeloproliferative disease was not transplantable into secondary recipients. Thus, oncogenic K-ras induces a myeloproliferative disorder but not AML, indicating that additional mutations are required for AML development. This model system will be useful for assessing the contribution of cooperating mutations in AML and testing ras inhibitors in vivo.
Collapse
Affiliation(s)
- Iris T Chan
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Rawat VPS, Cusan M, Deshpande A, Hiddemann W, Quintanilla-Martinez L, Humphries RK, Bohlander SK, Feuring-Buske M, Buske C. Ectopic expression of the homeobox gene Cdx2 is the transforming event in a mouse model of t(12;13)(p13;q12) acute myeloid leukemia. Proc Natl Acad Sci U S A 2004; 101:817-22. [PMID: 14718672 PMCID: PMC321764 DOI: 10.1073/pnas.0305555101] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Creation of fusion genes by balanced chromosomal translocations is one of the hallmarks of acute myeloid leukemia (AML) and is considered one of the key leukemogenic events in this disease. In t(12;13)(p13;q12) AML, ectopic expression of the homeobox gene CDX2 was detected in addition to expression of the ETV6-CDX2 fusion gene, generated by the chromosomal translocation. Here we show in a murine model of t(12;13)(p13;q12) AML that myeloid leukemogenesis is induced by the ectopic expression of CDX2 and not by the ETV6-CDX2 chimeric gene. Mice transplanted with bone marrow cells retrovirally engineered to express Cdx2 rapidly succumbed to fatal and transplantable AML. The transforming capacity of Cdx2 depended on an intact homeodomain and the N-terminal transactivation domain. Transplantation of bone marrow cells expressing ETV6-CDX2 failed to induce leukemia. Furthermore, coexpression of ETV6-CDX2 and Cdx2 in bone marrow cells did not accelerate the course of disease in transplanted mice compared to Cdx2 alone. These data demonstrate that activation of a protooncogene by a balanced chromosomal translocation can be the pivotal leukemogenic event in AML, characterized by the expression of a leukemia-specific fusion gene. Furthermore, these findings link protooncogene activation to myeloid leukemogenesis, an oncogenic mechanism so far associated mainly with lymphoid leukemias and lymphomas.
Collapse
MESH Headings
- Animals
- Bone Marrow Transplantation
- CDX2 Transcription Factor
- Cell Transformation, Neoplastic/genetics
- Chromosomes, Human, Pair 12/genetics
- Chromosomes, Human, Pair 13/genetics
- Disease Models, Animal
- Gene Expression
- Genes, Homeobox
- Homeodomain Proteins/genetics
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Transgenic
- Myeloid Ecotropic Viral Integration Site 1 Protein
- Neoplasm Proteins/genetics
- Oncogene Proteins, Fusion/genetics
- Trans-Activators
- Translocation, Genetic
Collapse
Affiliation(s)
- Vijay P S Rawat
- GSF-Clinical Cooperative Group Leukemia and Department of Medicine III, Grosshadern, Ludwig Maximilians University, 81377 Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW This review discusses the dosage effects of some oncogenes in leukemogenesis and compares various methods that model human hematologic malignancies in mice by introducing genetic lesions in a cell type-specific, time-controlled, and dosage-relevant manner. RECENT FINDINGS Recent evidence indicates that optimal dosage of cancer-related gene products plays an important role in the induction of mouse tumors that recapitulate their human counterparts. SUMMARY The mouse is a very valuable model system for experimentally dissecting the in vivo pathogenesis of cancer, for identifying pharmacological targets of cancer and for evaluating cancer therapies. In modeling human cancer, it has been shown that both the timing of introducing/activating oncogenic mutation(s) and the cell types into which the genetic lesion(s) is targeted are critical for cancer development. Recent studies also showed that efficient induction of relevant human leukemia in mice by certain oncogenes, such as PML/RARalpha and TEL/ABL, only occurred when they were expressed at a low level or close to pathophysiologically relevant level. These studies stress the importance of studying oncoprotein function at pathophysiologically relevant expression levels. Conditional gene expression systems are powerful tools for developing mouse models for human cancer by introducing genetic lesions in a cell type-specific, time-controlled and dosage-relevant manner. The bone marrow retroviral transduction and transplantation system can also mimic the cell and temporally specific origin of hematological malignancies by targeting oncogenes into sorted hematopoietic cells. This versatile approach is particularly powerful in structure-function analysis of oncogenes in vivo. However, overexpression of a transgene driven by retroviral vectors may alter the biological outcomes of the transgene in vivo. My colleagues and I have shown that generating vectors with modulated transgene expression can overcome this limitation of the retroviral transduction system in modeling human cancer in mice. Conditional gene expression and the modified retroviral transduction systems will be complimentary in studying human cancers in mice.
Collapse
Affiliation(s)
- Ruibao Ren
- Rosenstiel Basic Medical Sciences Research Center, Department of Biology, Brandeis University, Waltham, MA 02454-9110, USA.
| |
Collapse
|
36
|
Barbouti A, Ahlgren T, Johansson B, Höglund M, Lassen C, Turesson I, Mitelman F, Fioretos T. Clinical and genetic studies of ETV6/ABL1-positive chronic myeloid leukaemia in blast crisis treated with imatinib mesylate. Br J Haematol 2003; 122:85-93. [PMID: 12823349 DOI: 10.1046/j.1365-2141.2003.04391.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Most chronic myeloid leukaemia (CML) patients are genetically characterized by the t(9;22)(q34;q11), generating the BCR/ABL1 fusion gene. However, a few CML patients with rearrangements of 9q34 and 12p13, leading to ETV6/ABL1 chimaeras, have also been reported. Here we describe the clinical and genetic response to imatinib mesylate treatment of an ETV6/ABL1-positive CML patient diagnosed in blast crisis (BC). A chronic phase was achieved after acute myeloid leukaemia induction therapy. Then, treatment with imatinib mesylate (600 mg/d) was initiated and the effect was assessed clinically as well as genetically, including by repeated interphase fluorescence in situ hybridization studies. Until d 71 of imatinib mesylate therapy, stable improvements in the clinical and laboratory features were noted, and the frequency of ABL1-rearranged peripheral blood cells decreased from 56% to 11%. At d 92, an additional t(12;13)(p12;q13), with the 12p breakpoint proximal to ETV6, was found. The patient relapsed into BC 126 d after the start of the imatinib mesylate treatment and succumbed to the disease shortly afterwards. No mutations in the tyrosine kinase domain of ABL1 of the ETV6/ABL1 fusion were identified in the second BC. However, whereas the ETV6/ABL1 expression was seemingly the same at diagnosis and at second BC, the expression of ETV6 was markedly lower at the second BC. This decreased expression of wild-type ETV6 may have been a contributory factor for the relapse.
Collapse
MESH Headings
- Adult
- Antineoplastic Agents/therapeutic use
- Benzamides
- Blast Crisis/drug therapy
- Blast Crisis/genetics
- Enzyme Inhibitors/therapeutic use
- Fatal Outcome
- Follow-Up Studies
- Humans
- Imatinib Mesylate
- In Situ Hybridization, Fluorescence/methods
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Male
- Oncogene Proteins, Fusion/genetics
- Piperazines/therapeutic use
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Pyrimidines/therapeutic use
- Recurrence
Collapse
|
37
|
Shim GJ, Wang L, Andersson S, Nagy N, Kis LL, Zhang Q, Mäkelä S, Warner M, Gustafsson JA. Disruption of the estrogen receptor beta gene in mice causes myeloproliferative disease resembling chronic myeloid leukemia with lymphoid blast crisis. Proc Natl Acad Sci U S A 2003; 100:6694-9. [PMID: 12740446 PMCID: PMC164509 DOI: 10.1073/pnas.0731830100] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Proliferation of pluripotent, bone marrow stem cells, which develop to lymphoid and myeloid progenitors, is negatively regulated by estrogen. Although in estrogen deficiency and in estrogen receptor knockout mice there is significant alteration in bone marrow hematopoiesis, the effects of aging on estrogen receptor deficiencies in mice have not been investigated yet. In this study we show that by 1.5 years of age, estrogen receptor beta knockout (ERbeta-/-) mice develop pronounced splenomegaly that is much more severe in females than in males. Further characterization of these mice revealed myelogenous hyperplasia in bone marrow, an increase in the number of granulocytes and B lymphocytes in blood, lymphadenopathy, and infiltration of leukocytes in the liver and lung. Analysis by flow cytometry of the bone marrow cells revealed that the percentage and total number of Gr-1hi/Mac-1hi-positive granulocytes were increased by 15-30% and 100%, respectively. The numbers of B cells in the bone marrow and spleen were significantly higher in ERbeta-/- mice than in WT littermates. Some of the ERbeta-/- mice also had a severe lymphoproliferative phenotype. Thus the absence of ERbeta results in a myeloproliferative disease resembling human chronic myeloid leukemia with lymphoid blast crisis. Our results indicate a previously unknown role for ERbeta in regulating the differentiation of pluripotent hematopoietic progenitor cells and suggest that the ERbeta-/- mouse is a potential model for myeloid and lymphoid leukemia. Furthermore, we suggest that ERbeta agonists might have clinical value in the treatment of leukemia.
Collapse
MESH Headings
- Animals
- Bone Marrow/metabolism
- Estrogen Receptor beta
- Flow Cytometry
- Granulocytes/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Mice
- Mice, Inbred C57BL
- Myeloproliferative Disorders/genetics
- Myeloproliferative Disorders/metabolism
- Myeloproliferative Disorders/pathology
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
Collapse
Affiliation(s)
- Gil-Jin Shim
- Department of Medical Nutrition, Karolinska Institute, NOVUM, S-141 86 Huddinge, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Wertheim JA, Miller JP, Xu L, He Y, Pear WS. The biology of chronic myelogenous leukemia:mouse models and cell adhesion. Oncogene 2002; 21:8612-28. [PMID: 12476308 DOI: 10.1038/sj.onc.1206089] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Chronic myelogenous leukemia (CML) is a biphasic neoplasm of the bone marrow that is precipitated by the Philadelphia chromosome, a t(9;22) balanced translocation that encodes a constitutively activated nonreceptor tyrosine kinase termed P210(BCR-ABL). This oncoprotein has several intracellular functions; however, the most important effect of P210(BCR-ABL) leading to cell transformation is phosphorylation of signaling molecules through a constitutively active tyrosine kinase domain. Despite extensive knowledge of the structure and functional domains of BCR-ABL, its precise function in transformation is not known. Progress has been hampered, in part, by the lack of relevant CML models, as cell culture and in vitro assays do not mimic the pathogenesis of CML. Recently, there has been significant progress toward improving murine models that closely resemble human CML. This has allowed researchers to evaluate critical functions of BCR-ABL and has provided a model to test the efficacy of therapeutic medications that block these pathways. Our laboratory has developed two intersecting research programs to better understand the functioning of P210(BCR-ABL) in leukemogenesis. In one approach, we have developed a murine CML model by transferring HSCs that express BCR-ABL from a retroviral vector. All recipients develop a rapidly fatal MPD that shares several important features with CML. This model has been extremely useful for studying the function of BCR-ABL in the pathogenesis of CML. A second approach utilizes a quantitative cell detachment apparatus capable of measuring small changes in cell adhesion to investigate the mechanism by which P210(BCR-ABL) causes abnormal cell binding. Altered cell adhesion may contribute to the imbalance between proliferation and self-renewal in the hematopoietic progenitor compartment. To better understand the role abnormal adhesion may play in the development of leukemia, we have attempted to correlate the effects of functional P210(BCR-ABL) mutants in regulating adhesion and oncogenicity.
Collapse
Affiliation(s)
- Jason A Wertheim
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6160, USA
| | | | | | | | | |
Collapse
|
39
|
Abstract
Animal models of BCR-ABL+ leukemias have provided important new knowledge about the molecular pathophysiology of these diseases, and answered questions that are difficult or impossible to address using BCR-ABL-expressing cell lines or primary Ph+ leukemia samples from patients. The power of mouse models lies in their ability to recapitulate precisely the phenotypes of BCR-ABL+ leukemias in vivo, but this comes at the price of significant complexity. Here I review recent studies of leukemias induced in mice by BCR-ABL with an emphasis on the intricate nature of these diseases and the need for careful pathological and molecular analysis.
Collapse
Affiliation(s)
- Richard A Van Etten
- The Center for Blood Research and Department of Genetics, Harvard Medical School, Boston, Massachusetts, MA 02115, USA.
| |
Collapse
|