The Effect of Chromosomal Translocations in Acute Leukemias: the LMO2 Paradigm in Transcription and Development I

Total Page:16

File Type:pdf, Size:1020Kb

The Effect of Chromosomal Translocations in Acute Leukemias: the LMO2 Paradigm in Transcription and Development I [CANCER RESEARCH (SUPPL.) 59, 1794s-1798s, April 1, 1999] The Effect of Chromosomal Translocations in Acute Leukemias: The LMO2 Paradigm in Transcription and Development I Terence H. Rabbitts, 2 Katharina Bucher, Grace Chung, Gerald Grutz, Alan Warren, and Yoshi Yamada Medical Research Council Laboratory of Molecular Biology, Division of Protein and Nucleic Acid Chemistry, Hills Road, Cambridge, CB2 2QH United Kingdom Abstract proto-oncogene from a Burkitt's lymphoma translocation breakpoint t(8;14)(q24;q32) revealed that, while the translocated gene was intact, Two general features have emerged about genes that are activated after there were many mutations in both the noncoding first exon and chromosomal translocations in acute forms of cancer. The protein prod- within the coding region itself (4-11). In T-cell acute leukemias, there ucts of these genes are transcription regulators and are involved in are several different chromosomal translocations found in individual developmental processes, and it seems that the subversion of these normal functions accounts for their role in tumorigenesis. The features of the tumors, and the analysis of these has led to the discovery of many LMO family of genes, which encode LIM-domain proteins involved in different, novel genes that can contribute to T-cell tumorigenesis (1). T-cell acute leukemia through chromosomal translocations, typify these These and many other studies have led to two conclusions: abnormal functions in tumorigenesis. For example, the LMO2 protein is (a) many of the chromosomal translocation-activated oncogenes are involved in the formation of multimeric DNA-binding complexes, which transcription regulators in their normal sites of expression, and it is may vary in composition at different stages of hematopoiesis and function this property that is instrumental in their involvement in tumor etiol- to control differentiation of specific lineages. In T cells, enforced expres- ogy after the chromosomal translocation. This was suggested previ- sion of Lmo2 causes aberrant protein complex formation that primarily ously from the early studies on hematopoietic tumor translocations seems to hinder the T-ceU differentiation program. These observations and later, in addition, from the very large range of changes found in underscore the conclusion that protein-protein interaction (in this case, hematopoietic and mesenchymal tumors (reviewed in Ref. 1); and through the LIM domain) is a key determinant in tumorigenesis. Further- (b) the biological role of many chromosomal translocation-acti- more, the study of chromosomal translocations as naturally occurring mutations has been informative about mechanisms in hematopoiesis as vated genes is normally in developmental processes leading to the well as in tumor etiology. notion that subversion of development may be their crucial biological role in tumorigenesis, perhaps explaining why so few of the chromo- Introduction somal translocation-activated genes were previously identified as on- cogenes from other experimental approaches. The LMO 1 and LMO2 The cytogenetic analysis of tumors, particularly those of hemato- LIM-only proteins are involved in T-cell leukemia because of their poietic origin, have revealed that reciprocal chromosomal transloca- ability to interact with other transcription factors. This enhances the tions are recurring features of these tumors (reviewed in Ref. (1). conclusion that protein-protein interaction is a common component of Furthermore, it has become clear that particular chromosomal trans- oncogene function. locations are consistently found in specific tumor subtypes. Through the cloning of the chromosomal breakpoints and identification of Results and Discussion oncogenes at many different breakpoints, followed by transgenic (2) and homologous recombination knock-in analysis (3), it has become The LMO Family Genes. The LMO family of genes was uncov- clear that these abnormal tumor-associated chromosomes are impor- ered (Fig. 1) by the association of LM01 (previously called RBTN1 or tant in the etiology of tumors. The scientific challenge of the last TTG1) with the chromosomal translocation t(11;14)(p15;ql 1). The decade has been to define the contribution of the genes activated by transcription unit was first observed in a T-cell line (12), and the translocations to the course of tumor development and to ascertain mRNA sequence was obtained from its cDNA sequence (13, 14) and whether any general principles can be discerned about these "trans- shown to encode a protein essentially consisting of two zinc-binding location" genes. LIM domains (15). Using an LMO1 probe, the two related genes There are a variety of chromosomal translocations in leukemias and LM02 and LM03 were isolated (previously called RBTN2 or TTG2 in solid tumors of mesenchymal origin (sarcomas), and two main and RBTN3, respectively; Refs. (16, and 17), of which LM02 was outcomes are apparent (1). One of these is confined to lymphoid found located at the junction of the chromosomal translocation t(11; tumors in which the process of antigen receptor rearrangement (im- 14)(p13;ql 1) in T-ALL 3 (16, 18). The LMO-associated chromosomal munoglobulin and T-cell receptor) occurs and which occasionally translocations seem to have occurred by an error of the usual RAG- aberrantly mediates chromosomal translocation. This type of chromo- mediated variable diversity joining recombinase process inasmuch as somal translocation causes oncogene activation resulting from the sequence analysis of the breakpoints on chromosome 1 lp13 detected new chromosomal environment of the rearranged gene, and, in gen- recombinase signal sequences at the junctions and because the joins eral, this means inappropriate gene expression. In this first category of on chromosome 14 in the TCR~ locus or on chromosome 7 in the chromosomal translocation, the B- and T-cell tumors exemplify many TCR~ locus occur precisely at the end of D-segments (19). consistently occurring features. For instance, the cloning of the CMYC The unique feature of the LMO-derived protein sequences is that they are small proteins comprising two tandem LIM domains. These Received 11/11/98; accepted 2/4/99. zinc-binding finger-like structures have structural similarities to the 1 Presented at the "General Motors Cancer Research Foundation Twentieth Annual DNA-binding GATA fingers (20, 21) but as yet no case of a specific Scientific Conference: Developmental Biology and Cancer," June 9-10, 1998, Bethesda, LIM-DNA interaction has been reported; rather the function of this MD. This work was supported by the Medical Research Council and by Grants from the Leukaemia Research Fund (United Kingdom) and the National Foundation for Cancer domain seems to be restricted to protein-protein interaction (see Research (United States). K.B. is supported by the Roche Foundation; G.G. was "LMO2 Functions by Protein Interaction"). supported by an EMBO Fellowship, and Y. Y. by Kyoto University. 2 To whom requests for reprints should be addressed, at MRC Laboratory of Molecular Biology, Division of Protein and Nucleic Acid Chemistry, Hills Road, Cambridge, CB2 3 The abbreviations used are: T-ALL, T-cell acute lymphocytic leukemia; bHLH, 2QH United Kingdom. basic-helix-loop-helix; DN, double negative; Tall, Tall/Scl. 1794s Downloaded from cancerres.aacrjournals.org on September 28, 2021. © 1999 American Association for Cancer Research. LMO GENES IN LEUKEMIA LMO2 Is a Regulator of Mouse Hematopoiesis. To gain insights into the function of the LMO genes in tumorigenesis, an integrated approach has been adopted to attempt to understand the normal Pluri-potent cell Myeloid function at the biological level as well as the function of LMO proteins and to use this information to explore the function in tumors. Mes od~erm-'~~ lineage As a first step, gene targeting was used to introduce null mutations in Lymphoid mouse Lmo2 gene, which showed that Lmo2 is necessary for yolk sac Self-renewa/j lineage erythropoiesis in mouse embryogenesis (22). Furthermore, the use of embryonic stem cells with null mutations of both alleles of Lmo2 in Lmo2 null defect chimeric mice has shown that adult hematopoiesis, including lympho- poiesis and myelopoiesis, fails completely in the absence of Lmo2 Fig. 2. Lmo2 is necessary for adult hematopoiesis in mice. The tissue contribution of embryonic stem cells with homozygous null mutation of Lmo2 showed that hematopoiesis (23). These data show that Lmo2 must function at early stages of in adult mice is dependent on this gene (23). Thus, the gene product is required for all of hematopoiesis (Fig. 2) either at the level of the pluripotent stem cell the stages of adult hematopoiesis, functioning at least before the bifurcation of myeloid and lymphoid lineages. The function may be restricted to the bone marrow stem cell or even perhaps before this when ventral mesoderm gives rise to these (either at the self-renewal stage or at the proliferative stages that produce committed precursor cells. Remarkably similar results have been shown for progenitors) or in the ventral mesoderm bone marrow precursor cells. another T-cell oncogene, Tall/Scl (herein called Tall; Refs. (24-27). The notion of genes that function in developmental processes is clearly delineated by the Lmo2 gene. LDB1 interact in vivo in T-ALL and in neuroblastoma cell lines (41). LMO2 Functions by Protein Interaction: The Role of the LIM This array of interactions led to the observation that Lmo2 is found in Domain. The concept that oncogenes can interfere with developmen- an oligomeric complex in erythroid cells that involves--in addition to tal processes because they are themselves normally performing this Lmo2--Tall, E47, Ldbl and Gata-I (40). This complex is able to function has been suggested to explain their role in tumor etiology. bind DNA at least in vitro and in reporter assays, with the Lmo2 and The finding that genes that encode proteins such as Lmo2 control Ldbl components seeming to bridge a bipartite DNA-binding com- development of hematopoietic lineages led to clear tests of molecular plex (Fig. 3A). These findings suggest that Lmo2 is part of a tran- function in both normal and tumor contexts.
Recommended publications
  • Characterization of a Pediatric T-Cell Acute Lymphoblastic Leukemia Patient with Simultaneous LYL1 and LMO2 Rearrangements
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Erasmus University Digital Repository Articles and Brief Reports Acute Lymphoblastic Leukemia Characterization of a pediatric T-cell acute lymphoblastic leukemia patient with simultaneous LYL1 and LMO2 rearrangements Irene Homminga, 1 Maartje J. Vuerhard, 1 Anton W. Langerak, 2 Jessica Buijs-Gladdines, 1 Rob Pieters, 1 and Jules P.P. Meijerink 1 1Department of Pediatric Oncology/Hematology, Erasmus MC/Sophia Children’s Hospital, Rotterdam; and 2Department of Immunology, Erasmus MC, Rotterdam, The Netherlands ABSTRACT Translocation of the LYL1 oncogene are rare in T-cell acute consistently clustered along with cases having TAL1 or lymphoblastic leukemia, whereas the homologous TAL1 LMO2 rearrangements. Therefore, LYL1 -rearranged cases are gene is rearranged in approximately 20% of patients. not necessarily associated with immature T-cell develop - Previous gene-expression studies have identified an imma - ment, despite high LYL1 levels, but elicit a TALLMO expres - ture T-cell acute lymphoblastic leukemia subgroup with high sion signature. LYL1 expression in the absence of chromosomal aberrations. Molecular characterization of a t(7;19)(q34;p13) in a pediatric Key words: T-ALL, pediatric, LYL1, LMO2, rearrangements. T-cell acute lymphoblastic leukemia patient led to the identi - fication of a translocation between the TRB@ and LYL1 loci. Citation: Homminga I, Vuerhard MJ, Langerak AW, Buijs- Similar to incidental T-cell acute lymphoblastic leukemia Gladdines J, Pieters R, and Meijerink JPP. Characterization of a cases with synergistic, double translocations affecting pediatric T-cell acute lymphoblastic leukemia patient with simul - TAL1/2 and LMO1/2 oncogenes, this LYL1 -translocated taneous LYL1 and LMO2 rearrangements.
    [Show full text]
  • The Role of Noncoding Mutations in Blood Cancers Sunniyat Rahman and Marc R
    © 2019. Published by The Company of Biologists Ltd | Disease Models & Mechanisms (2019) 12, dmm041988. doi:10.1242/dmm.041988 REVIEW The role of noncoding mutations in blood cancers Sunniyat Rahman and Marc R. Mansour* ABSTRACT survival in adult acute myeloid leukaemia (AML) and ALL are The search for oncogenic mutations in haematological malignancies still less than 50%, with significant treatment challenges including has largely focused on coding sequence variants. These variants treatment-resistant disease, clonal heterogeneity of the underlying have been critical in understanding these complex cancers in greater disease, treatment-associated toxicities and poor tolerance for detail, ultimately leading to better disease monitoring, subtyping and intensive treatment regimens, particularly in older patients with prognostication. In contrast, the search for oncogenic variants in the comorbidities (Kansagra et al., 2018). Because of this, there is a noncoding genome has proven to be challenging given the vastness significant impetus to discover new genetic aberrations, including of the search space, the intrinsic difficulty in assessing the impact of driver mutations (see Box 1 for a glossary of terms), that may variants that do not code for functional proteins, and our still primitive provide insight into the mechanisms of malignant haematopoiesis, understanding of the function harboured by large parts of the as well as offering novel therapeutic opportunities. noncoding genome. Recent studies have broken ground on this Detailed genetic characterisation of haematological malignancies quest, identifying somatically acquired and recurrent mutations in the has already identified alterations that are now being used for better noncoding genome that activate the expression of proto-oncogenes. diagnosis, prognostication, subtype identification and to inform In this Review, we explore some of the best-characterised examples therapeutic decisions (Taylor et al., 2017).
    [Show full text]
  • LMO T-Cell Translocation Oncogenes Typify Genes Activated by Chromosomal Translocations That Alter Transcription and Developmental Processes
    Downloaded from genesdev.cshlp.org on September 25, 2021 - Published by Cold Spring Harbor Laboratory Press PERSPECTIVE LMO T-cell translocation oncogenes typify genes activated by chromosomal translocations that alter transcription and developmental processes Terence H. Rabbitts1 Medical Research Council (MRC) Laboratory of Molecular Biology, Division of Protein and Nucleic Acid Chemistry, Cambridge CB2 2QH, UK The cytogenetic analysis of tumors, particularly those of ment (immunoglobulin or T-cell receptor) occurs and oc- hematopoietic origin, has revealed that reciprocal chro- casionally mediates chromosomal translocation. This mosomal translocations are recurring features of these type of translocation causes oncogene activation result- tumors. Further from the initial recognition of the trans- ing from the new chromosomal environment of the re- location t(9;22) (Nowell and Hungerford 1960; Rowley arranged gene. In general, this means inappropriate gene 1973), it has become clear that particular chromosomal expression. The second, and probably the most common translocations are found consistently in specific tumor outcome of chromosomal translocations, is gene fusion subtypes. A principle example of this is the translocation in which exons from a gene on each of the involved chro- t(8;14)(q24;q32.1) invariably found in the human B cell mosomes are linked after the chromosomal transloca- tumor Burkitt’s lymphoma (Manolov and Manolov 1972; tion, resulting in a fusion mRNA and protein. This type Zech et al. 1976). The link with the immunoglobulin of event is found in many cases of hematopoietic tumors H-chain locus on chromosome 14, band q32.1 (Croce et and in the sarcomas. al. 1979; Hobart et al.
    [Show full text]
  • A Previously Unrecognized Promoter of LMO2 Forms Part of A
    Oncogene (2010) 29, 5796–5808 & 2010 Macmillan Publishers Limited All rights reserved 0950-9232/10 www.nature.com/onc ORIGINAL ARTICLE A previously unrecognized promoter of LMO2 forms part of a transcriptional regulatory circuit mediating LMO2 expression in a subset of T-acute lymphoblastic leukaemia patients SH Oram1, JAI Thoms2, C Pridans1, ME Janes2, SJ Kinston1, S Anand1, J-R Landry3, RB Lock4, P-S Jayaraman5, BJ Huntly1, JE Pimanda2 and B Go¨ttgens1 1Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK; 2Lowy Cancer Research Centre and The Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales, Australia; 3Institute for Research in Immunology and Cancer, Universite´ de Montre´al, Montre´al, Quebec, Canada; 4Children’s Cancer Institute Australia, University of New South Wales, Sydney, New South Wales, Australia and 5Birmingham University Stem Cell Centre, University of Birmingham, Edgbaston, Birmingham, UK The T-cell oncogene Lim-only 2 (LMO2) critically Introduction influences both normal and malignant haematopoiesis. LMO2 is not normally expressed in T cells, yet ectopic Lim-only 2 (LMO2), also known as TTG2 or RBTN2, expression is seen in the majority of T-acute lympho- was originally reported as a T-cell oncogene where it blastic leukaemia (T-ALL) patients with specific translo- was recurrently rearranged by chromosomal transloca- cations involving LMO2 in only a subset of these patients. tion in T-cell malignancies (Boehm et al., 1991; Royer- Ectopic lmo2 expression in thymocytes of transgenic mice Pokora et al., 1991). LMO2 encodes a 156 amino-acid causes T-ALL, and retroviral vector integration into the transcriptional co-factor containing two LIM domain LMO2 locus was implicated in the development of clonal zinc-fingers (Kadrmas and Beckerle, 2004) that do not T-cell disease in patients undergoing gene therapy.
    [Show full text]
  • Letters to the Editor
    LETTERS TO THE EDITOR The significant upregulation of Gata1 and EpoR mRNA Mice over-expressing human erythropoietin expression in hEPO over-expressing tg6 mice was con - indicate that erythropoietin enhances expression firmed by analyzing the spleen as a major source of of its receptor via up-regulated Gata1 and Tal1 hematopoiesis (Figure 2A and B). To further dissect the complexity of changes in the transcriptional network, the analysis of Myb mRNA expression served as marker for The development of medullary hematopoiesis is char - adult definitive erythroblasts, 10 showing significantly acterized by a specific expression profile of hematopoiet - ic transcription factors, including GATA transcription fac - tors. At mid-gestation, when hematopoiesis is newly A GATA 1 2 established in the bone marrow of human fetuses, initial - wt ly high GATA2 expression becomes subsequently down- regulated, while GATA1 expression increases in parallel. 1 1.5 Both transcription factors bind to overlapping sets of tg6 hematopoietic downstream target genes, often at distinct 1 sites, to regulate the balance between proliferation and differentiation. Chromatin occupancy by GATA1 and 0.5 GATA2 can change in the course of hematopoietic differ - entiation, leading to the so-called GATA switch. 2 Thus, a 0 n d7 d21 d49 i spatio-temporal regulation of GATA1 or GATA2 activities t c is required within lineage-specific differentiation. During a - erythroid differentiation GATA1 expression peaks at the b B GATA 2 3 o level of colony-forming units (CFU-E), where erythro - t 2 e poietin (Epo) exerts most specifically its effects, but v i t blocks terminal maturation if constitutively over- a 1.5 l 4 e expressed.
    [Show full text]
  • Rahman Et Al 2017 Somatically Acquired Activation of LMO2 in T-ALL
    Activation of the LMO2 oncogene through a somatically acquired neomorphic promoter in T-cell acute lymphoblastic leukemia The MIT Faculty has made this article openly available. Please share how this access benefits you. Your story matters. Citation Rahman, Sunniyat, Michael Magnussen, Theresa E. León, Nadine Farah, Zhaodong Li, Brian J. Abraham, Krisztina Z. Alapi, et al. “Activation of the LMO2 Oncogene through a Somatically Acquired Neomorphic Promoter in T-Cell Acute Lymphoblastic Leukemia.” Blood 129, no. 24 (March 7, 2017): 3221–3226. As Published https://doi.org/10.1182/blood-2016-09-742148 Publisher American Society of Hematology Version Author's final manuscript Citable link http://hdl.handle.net/1721.1/117283 Terms of Use Creative Commons Attribution-Noncommercial-Share Alike Detailed Terms http://creativecommons.org/licenses/by-nc-sa/4.0/ 1 Activation of the LMO2 oncogene through a somatically acquired 2 neomorphic promoter in T-Cell Acute Lymphoblastic Leukemia 3 4 Sunniyat Rahman1, Michael Magnussen1, Theresa E. León1, Nadine Farah1, Zhaodong Li2, 5 Brian J Abraham3, Krisztina Z. Alapi1, Rachel J. Mitchell1, Tom Naughton1, Adele K. 6 Fielding1, Arnold Pizzey1, Sophia Bustraan1, Christopher Allen1, Teodora Popa , Karin Pike- 7 Overzet5, Laura Garcia-Perez5, Rosemary E. Gale1, David C. Linch1, Frank J.T. Staal5, 8 Richard A. Young3,4, A. Thomas Look2,6, Marc R. Mansour1 9 10 1. University College London Cancer Institute, Department of Haematology, 72 Huntley 11 Street, London. WC1E 6DD. United Kingdom. 12 2. Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical 13 School, Boston, MA 02215 14 3. Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 15 02142 16 4.
    [Show full text]
  • Kobe University Repository : Thesis
    Kobe University Repository : Thesis Afadin Regulates Puncta Adherentia Junction Formation and 学位論文題目 Presynaptic Differentiation in Hippocampal Neurons(アファディンは海 Title 馬ニューロンにおいてアドへレンスジャンクションの形成と前シナプ スの分化を調節している) 氏名 Toyoshima, Daisaku Author 専攻分野 博士(医学) Degree 学位授与の日付 2014-03-25 Date of Degree 公開日 2015-03-01 Date of Publication 資源タイプ Thesis or Dissertation / 学位論文 Resource Type 報告番号 甲第6188号 Report Number 権利 Rights JaLCDOI URL http://www.lib.kobe-u.ac.jp/handle_kernel/D1006188 ※当コンテンツは神戸大学の学術成果です。無断複製・不正使用等を禁じます。著作権法で認められている範囲内で、適切にご利用ください。 PDF issue: 2021-09-26 Afadin Regulates Puncta Adherentia Junction Formation and Presynaptic Differentiation in Hippocampal Neurons アファディンは海馬ニューロンにおいてアドへレンスジャンクション の形成と前シナプスの分化を調節している 豊嶋大作, 萬代研二, 丸尾知彦、Irwan Supriyanto、 富樫英、井上貴仁、森正弘、高井義美 神戸大学大学院医学研究科医科学専攻 小児科学 (指導教員:飯島一誠 教授) 豊嶋大作 Key words: Afadin, puncta adherentia junction, presynapse, hippocampal neuron Manuscript Click here to download Manuscript: Toyoshima_text_revised_2nd_final.pdf 1 Afadin Regulates Puncta Adherentia Junction Formation and 2 Presynaptic Differentiation in Hippocampal Neurons 3 4 Daisaku Toyoshima1,3,4, Kenji Mandai1,3, Tomohiko Maruo1,3, Irwan Supriyanto2,3, 5 Hideru Togashi1,3, Takahito Inoue1,3, Masahiro Mori2,3 & Yoshimi Takai1,3 6 7 1. Department of Biochemistry and Molecular Biology, Kobe University Graduate 8 School of Medicine, Kobe, Hyogo 650-0047, Japan. 9 2. Faculty of Health Sciences, Kobe University Graduate School of Health Sciences, 10 Kobe, Hyogo 654-0142, Japan. 11 3. CREST, Japan Science and Technology Agency, Kobe, Hyogo 650-0047, Japan. 12 4. Present address: Department of Pediatrics, Kobe University Graduate School of 13 Medicine, Kobe, Hyogo 650-0017, Japan. 14 15 Correspondence should be addressed to Y.T. ([email protected]), K.M. 16 ([email protected]).
    [Show full text]
  • T-Cell Lymphoblastic Lymphoma Arising in the Setting of Myeloid/Lymphoid Neoplasms with Eosinophilia
    cancers Article T-Cell Lymphoblastic Lymphoma Arising in the Setting of Myeloid/Lymphoid Neoplasms with Eosinophilia: LMO2 Immunohistochemistry as a Potentially Useful Diagnostic Marker Magda Zanelli 1 , Giuseppe G. Loscocco 2,3 , Elena Sabattini 4, Maurizio Zizzo 5,6,* , Francesca Sanguedolce 7, Luigi Panico 8, Daniela Fanni 9, Raffaella Santi 10, Cecilia Caprera 11, Cristiana Rossi 12, Alessandra Soriano 13,14, Alberto Cavazza 1, Alessandro Giunta 5, Cristina Mecucci 15, Alessandro M. Vannucchi 2,3, Stefano A. Pileri 16 and Stefano Ascani 11,15 1 Pathology Unit, Azienda Unità Sanitaria Locale—IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; [email protected] (M.Z.); [email protected] (A.C.) 2 Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; gloscocco@unifi.it (G.G.L.); a.vannucchi@unifi.it (A.M.V.) 3 Center of Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Azienda Ospedaliero-Universitaria Careggi, 50139 Florence, Italy 4 Citation: Zanelli, M.; Loscocco, G.G.; Haematopathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; Sabattini, E.; Zizzo, M.; [email protected] 5 Sanguedolce, F.; Panico, L.; Fanni, D.; Surgical Oncology Unit, Azienda Unità Sanitaria Locale—IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; [email protected] Santi, R.; Caprera, C.; Rossi, C.; et al. 6 Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, T-Cell Lymphoblastic Lymphoma 41121 Modena, Italy Arising in the Setting of 7 Pathology Unit, Azienda Ospedaliero-Universitaria—Ospedali Riuniti di Foggia, 71122 Foggia, Italy; Myeloid/Lymphoid Neoplasms with [email protected] Eosinophilia: LMO2 8 Pathology Unit Azienda Ospedaliera dei Colli Monaldi-Cotugno-CTO, P.O.
    [Show full text]
  • Letters to the Editor
    LETTERS TO THE EDITOR routine karyotypic analysis at a rate of 4.5% (12 of 264). High frequency of cryptic chromosomal LMO2 mRNA expression levels were measured on 10 T- rearrangements involving the LMO2 gene in T-cell ALL patients with and 39 without LMO2 rearrangements. acute lymphoblastic leukemia The qRT-PCR results showed that LMO2 transcripts were significantly higher in cases with LMO2 rearrangements (P=0.02) than without (Figure 1A). Meanwhile, the mRNA T-cell acute lymphoblastic leukemia (T-ALL) is an aggres- expression levels of LEF1, LYL1, MEF2C, STAG2, SEPT1, sive malignancy of thymocytes resulting from the transfor- TLX1, and TLX3 were also measured by qRT-PCR in these mation of T-cell progenitors. Around half of T-ALL patients patients, showing no differences between patients with harbor recurrent cytogenetic alterations, including juxtapo- and without LMO2 rearrangements, with the exception of sition of strong promoters and enhancers located in the LEF1. Our findings show that patients with LMO2 TCRB (chr. 7q34) or TCRA-TCRD (chr. 14q11) loci with a rearrangements had higher LEF1 transcripts (P=0.015) variety of oncogenic transcription factors, such as LIM-only (Figure 1A), which raises the possibility that LMO2 interac- domain (LMO) genes, LMO1 and LMO2 resulting in their tome includes LEF1 in T-ALL, as reported in B-cell lym- 1 aberrant expression. The LIM-only gene LMO2 encodes a phomas.9 protein that participates in a transcription factor complex, Furthermore, to explore undetectable cytogenetic abnor- which includes E2A, GATA1, and LDB1, TAL1. LMO2 was malities, we performed integrative genomic and transcrip- reportedly activated in 4 cases of T-ALL arising via retrovi- tional analyses on these 24 T-ALL patients with LMO2 ral insertion mutagenesis in a gene therapy trial for X- rearrangements.
    [Show full text]
  • SUPPLEMENTARY INFORMATION in Format Provided by Yui & Rothenberg (AUGUST 2014)
    SUPPLEMENTARY INFORMATION In format provided by Yui & Rothenberg (AUGUST 2014) Table 2 | Critical phase 1‑specific transcription factor genes in murine early T cells and their progenitors Gene (protein) Protein family Knockout phenotype Overexpression phenotype Selected References Lmo2 LIM • Severe stem cell defects T‑ALL 1–4 • No T cell effects if deleted after DN2 stage Gata2 GATA ZnF Early stem cell defects Unknown 5,6 Mef2c MADS-box T cell, B cell and NK cell defects • AML 7,8 • T‑ALL association Meis1 MEIS homeodomain Early stem cell defects AML association 9–11 Hoxa9 HOX homeobox • Defect in HSC proliferation AML 11–14 • Partial DN2 stage block Tal1 TAL bHLH Early stem cell defects T‑ALL 15,16 Gfi1b • SNAG HSC proliferative expansion Unknown 17 • C2H2‑like ZnF Lyl1 TAL bHLH Defects in LMPPs, ETPs and DN2a cells • B-ALL 18–20 • T-ALL Spi1 ETS • Absence of T cell and NK cell • Diversion to DC or myeloid 21–25 (PU.1) development cell lineages (reviewed in 26) • AML • T cell lymphoma Bcl11a ZnF C2H2‑like Required for B, T and NK cell Myeloid and B cell 6,27–30 development malignancies Hhex Homeobox Impaired monocyte development T‑ALL 2,3,32 Mycn MYC bHLH Defects in HSC survival and proliferation AML 33,34 (with C‑MYC) Erg ETS Early stem cell defects T‑ALL and other leukaemias 35,36 AML, acute myeloid leukaemia; B-ALL, B cell acute lymphoblastic leukaemia; bHLH, basic helix–loop–helix; Bcl11a, B cell lymphoma–leukaemia 11A; C2H2, a form of zinc finger (Cys2His2); DC, dendritic cell; DN, double negative; ETP, early thymic progenitor; ETS,
    [Show full text]
  • Interplay Between Cofactors and Transcription Factors in Hematopoiesis and Hematological Malignancies
    Signal Transduction and Targeted Therapy www.nature.com/sigtrans REVIEW ARTICLE OPEN Interplay between cofactors and transcription factors in hematopoiesis and hematological malignancies Zi Wang 1,2, Pan Wang2, Yanan Li2, Hongling Peng1, Yu Zhu2, Narla Mohandas3 and Jing Liu2 Hematopoiesis requires finely tuned regulation of gene expression at each stage of development. The regulation of gene transcription involves not only individual transcription factors (TFs) but also transcription complexes (TCs) composed of transcription factor(s) and multisubunit cofactors. In their normal compositions, TCs orchestrate lineage-specific patterns of gene expression and ensure the production of the correct proportions of individual cell lineages during hematopoiesis. The integration of posttranslational and conformational modifications in the chromatin landscape, nucleosomes, histones and interacting components via the cofactor–TF interplay is critical to optimal TF activity. Mutations or translocations of cofactor genes are expected to alter cofactor–TF interactions, which may be causative for the pathogenesis of various hematologic disorders. Blocking TF oncogenic activity in hematologic disorders through targeting cofactors in aberrant complexes has been an exciting therapeutic strategy. In this review, we summarize the current knowledge regarding the models and functions of cofactor–TF interplay in physiological hematopoiesis and highlight their implications in the etiology of hematological malignancies. This review presents a deep insight into the physiological and pathological implications of transcription machinery in the blood system. Signal Transduction and Targeted Therapy (2021) ;6:24 https://doi.org/10.1038/s41392-020-00422-1 1234567890();,: INTRODUCTION by their ATPase subunits into four major families, including the Hematopoiesisisacomplexhierarchicaldifferentiationprocessthat SWI/SNF, ISWI, Mi-2/NuRD, and INO80/SWR1 families.
    [Show full text]
  • Characterization of a Pediatric T-Cell Acute Lymphoblastic Leukemia Patient with Simultaneous LYL1 and LMO2 Rearrangements
    Articles and Brief Reports Acute Lymphoblastic Leukemia Characterization of a pediatric T-cell acute lymphoblastic leukemia patient with simultaneous LYL1 and LMO2 rearrangements Irene Homminga, 1 Maartje J. Vuerhard, 1 Anton W. Langerak, 2 Jessica Buijs-Gladdines, 1 Rob Pieters, 1 and Jules P.P. Meijerink 1 1Department of Pediatric Oncology/Hematology, Erasmus MC/Sophia Children’s Hospital, Rotterdam; and 2Department of Immunology, Erasmus MC, Rotterdam, The Netherlands ABSTRACT Translocation of the LYL1 oncogene are rare in T-cell acute consistently clustered along with cases having TAL1 or lymphoblastic leukemia, whereas the homologous TAL1 LMO2 rearrangements. Therefore, LYL1 -rearranged cases are gene is rearranged in approximately 20% of patients. not necessarily associated with immature T-cell develop - Previous gene-expression studies have identified an imma - ment, despite high LYL1 levels, but elicit a TALLMO expres - ture T-cell acute lymphoblastic leukemia subgroup with high sion signature. LYL1 expression in the absence of chromosomal aberrations. Molecular characterization of a t(7;19)(q34;p13) in a pediatric Key words: T-ALL, pediatric, LYL1, LMO2, rearrangements. T-cell acute lymphoblastic leukemia patient led to the identi - fication of a translocation between the TRB@ and LYL1 loci. Citation: Homminga I, Vuerhard MJ, Langerak AW, Buijs- Similar to incidental T-cell acute lymphoblastic leukemia Gladdines J, Pieters R, and Meijerink JPP. Characterization of a cases with synergistic, double translocations affecting pediatric T-cell acute lymphoblastic leukemia patient with simul - TAL1/2 and LMO1/2 oncogenes, this LYL1 -translocated taneous LYL1 and LMO2 rearrangements. Haematologica patient also had an LMO2 rearrangement pointing to onco - 2012;97(2):258-261.
    [Show full text]