Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
2.
Mol Cell ; 81(16): 3323-3338.e14, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34352207

ABSTRACT

The emerging "epitranscriptomics" field is providing insights into the biological and pathological roles of different RNA modifications. The RNA methyltransferase METTL1 catalyzes N7-methylguanosine (m7G) modification of tRNAs. Here we find METTL1 is frequently amplified and overexpressed in cancers and is associated with poor patient survival. METTL1 depletion causes decreased abundance of m7G-modified tRNAs and altered cell cycle and inhibits oncogenicity. Conversely, METTL1 overexpression induces oncogenic cell transformation and cancer. Mechanistically, we find increased abundance of m7G-modified tRNAs, in particular Arg-TCT-4-1, and increased translation of mRNAs, including cell cycle regulators that are enriched in the corresponding AGA codon. Accordingly, Arg-TCT expression is elevated in many tumor types and is associated with patient survival, and strikingly, overexpression of this individual tRNA induces oncogenic transformation. Thus, METTL1-mediated tRNA modification drives oncogenic transformation through a remodeling of the mRNA "translatome" to increase expression of growth-promoting proteins and represents a promising anti-cancer target.


Subject(s)
Carcinogenesis/genetics , Methyltransferases/genetics , Neoplasms/genetics , tRNA Methyltransferases/genetics , Guanosine/analogs & derivatives , Guanosine/genetics , Humans , Methylation , Neoplasms/pathology , Oncogenes/genetics , RNA Processing, Post-Transcriptional/genetics , RNA, Messenger/genetics , RNA, Transfer/genetics
3.
Blood Adv ; 5(9): 2412-2425, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33956058

ABSTRACT

Advances in cancer genomics have revealed genomic classes of acute myeloid leukemia (AML) characterized by class-defining mutations, such as chimeric fusion genes or in genes such as NPM1, MLL, and CEBPA. These class-defining mutations frequently synergize with internal tandem duplications in FLT3 (FLT3-ITDs) to drive leukemogenesis. However, ∼20% of FLT3-ITD-positive AMLs bare no class-defining mutations, and mechanisms of leukemic transformation in these cases are unknown. To identify pathways that drive FLT3-ITD mutant AML in the absence of class-defining mutations, we performed an insertional mutagenesis (IM) screening in Flt3-ITD mice, using Sleeping Beauty transposons. All mice developed acute leukemia (predominantly AML) after a median of 73 days. Analysis of transposon insertions in 38 samples from Flt3-ITD/IM leukemic mice identified recurrent integrations at 22 loci, including Setbp1 (20/38), Ets1 (11/38), Ash1l (8/38), Notch1 (8/38), Erg (7/38), and Runx1 (5/38). Insertions at Setbp1 led exclusively to AML and activated a transcriptional program similar, but not identical, to those of NPM1-mutant and MLL-rearranged AMLs. Guide RNA targeting of Setbp1 was highly detrimental to Flt3ITD/+/Setbp1IM+, but not to Flt3ITD/+/Npm1cA/+, AMLs. Also, analysis of RNA-sequencing data from hundreds of human AMLs revealed that SETBP1 expression is significantly higher in FLT3-ITD AMLs lacking class-defining mutations. These findings propose that SETBP1 overexpression collaborates with FLT3-ITD to drive a subtype of human AML. To identify genetic vulnerabilities of these AMLs, we performed genome-wide CRISPR-Cas9 screening in Flt3ITD/+/Setbp1IM+ AMLs and identified potential therapeutic targets, including Kdm1a, Brd3, Ezh2, and Hmgcr. Our study gives new insights into epigenetic pathways that can drive AMLs lacking class-defining mutations and proposes therapeutic approaches against such cases.


Subject(s)
Leukemia, Myeloid, Acute , Acute Disease , Animals , DNA-Binding Proteins , Histone-Lysine N-Methyltransferase , Leukemia, Myeloid, Acute/genetics , Mice , Mutation , Nuclear Proteins/genetics , Nucleophosmin
4.
Nature ; 593(7860): 597-601, 2021 05.
Article in English | MEDLINE | ID: mdl-33902106

ABSTRACT

N6-methyladenosine (m6A) is an abundant internal RNA modification1,2 that is catalysed predominantly by the METTL3-METTL14 methyltransferase complex3,4. The m6A methyltransferase METTL3 has been linked to the initiation and maintenance of acute myeloid leukaemia (AML), but the potential of therapeutic applications targeting this enzyme remains unknown5-7. Here we present the identification and characterization of STM2457, a highly potent and selective first-in-class catalytic inhibitor of METTL3, and a crystal structure of STM2457 in complex with METTL3-METTL14. Treatment of tumours with STM2457 leads to reduced AML growth and an increase in differentiation and apoptosis. These cellular effects are accompanied by selective reduction of m6A levels on known leukaemogenic mRNAs and a decrease in their expression consistent with a translational defect. We demonstrate that pharmacological inhibition of METTL3 in vivo leads to impaired engraftment and prolonged survival in various mouse models of AML, specifically targeting key stem cell subpopulations of AML. Collectively, these results reveal the inhibition of METTL3 as a potential therapeutic strategy against AML, and provide proof of concept that the targeting of RNA-modifying enzymes represents a promising avenue for anticancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Methyltransferases/antagonists & inhibitors , Adenosine/analogs & derivatives , Animals , Apoptosis , Cell Differentiation , Cell Line, Tumor , Female , Gene Expression Regulation, Leukemic/drug effects , Humans , Mice , Mice, Inbred C57BL , Molecular Structure , Xenograft Model Antitumor Assays
5.
Leukemia ; 35(4): 1012-1022, 2021 04.
Article in English | MEDLINE | ID: mdl-32764680

ABSTRACT

Histone acetyltransferases (HATs) catalyze the transfer of an acetyl group from acetyl-CoA to lysine residues of histones and play a central role in transcriptional regulation in diverse biological processes. Dysregulation of HAT activity can lead to human diseases including developmental disorders and cancer. Through genome-wide CRISPR-Cas9 screens, we identified several HATs of the MYST family as fitness genes for acute myeloid leukemia (AML). Here we investigate the essentiality of lysine acetyltransferase KAT7 in AMLs driven by the MLL-X gene fusions. We found that KAT7 loss leads to a rapid and complete loss of both H3K14ac and H4K12ac marks, in association with reduced proliferation, increased apoptosis, and differentiation of AML cells. Acetyltransferase activity of KAT7 is essential for the proliferation of these cells. Mechanistically, our data propose that acetylated histones provide a platform for the recruitment of MLL-fusion-associated adaptor proteins such as BRD4 and AF4 to gene promoters. Upon KAT7 loss, these factors together with RNA polymerase II rapidly dissociate from several MLL-fusion target genes that are essential for AML cell proliferation, including MEIS1, PBX3, and SENP6. Our findings reveal that KAT7 is a plausible therapeutic target for this poor prognosis AML subtype.


Subject(s)
Gene Rearrangement , Genetic Predisposition to Disease , Histone Acetyltransferases/genetics , Histone-Lysine N-Methyltransferase/genetics , Leukemia, Myeloid, Acute/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Apoptosis/genetics , Biomarkers, Tumor , Cell Differentiation , Cell Line, Tumor , Disease Management , Epigenesis, Genetic , Gene Knockout Techniques , Genetic Association Studies , Histone Acetyltransferases/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/therapy , Myeloid Cells/metabolism , Myeloid Cells/pathology , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Promoter Regions, Genetic , Protein Binding
6.
Nat Commun ; 11(1): 4132, 2020 08 17.
Article in English | MEDLINE | ID: mdl-32807781

ABSTRACT

Precise genome editing using CRISPR-Cas9 is a promising therapeutic avenue for genetic diseases, although off-target editing remains a significant safety concern. Guide RNAs shorter than 16 nucleotides in length effectively recruit Cas9 to complementary sites in the genome but do not permit Cas9 nuclease activity. Here we describe CRISPR Guide RNA Assisted Reduction of Damage (CRISPR GUARD) as a method for protecting off-targets sites by co-delivery of short guide RNAs directed against off-target loci by competition with the on-target guide RNA. CRISPR GUARD reduces off-target mutagenesis while retaining on-target editing efficiencies with Cas9 and base editor. However, we discover that short guide RNAs can also support base editing if they contain cytosines within the deaminase activity window. We explore design rules and the universality of this method through in vitro studies and high-throughput screening, revealing CRISPR GUARD as a rapidly implementable strategy to improve the specificity of genome editing for most genomic loci. Finally, we create an online tool for CRISPR GUARD design.


Subject(s)
Gene Editing/methods , RNA, Guide, Kinetoplastida/metabolism , CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/physiology , Humans , Mutagenesis/genetics , Mutagenesis/physiology , RNA, Guide, Kinetoplastida/genetics
7.
Nat Commun ; 9(1): 5378, 2018 12 19.
Article in English | MEDLINE | ID: mdl-30568163

ABSTRACT

We recently identified the splicing kinase gene SRPK1 as a genetic vulnerability of acute myeloid leukemia (AML). Here, we show that genetic or pharmacological inhibition of SRPK1 leads to cell cycle arrest, leukemic cell differentiation and prolonged survival of mice transplanted with MLL-rearranged AML. RNA-seq analysis demonstrates that SRPK1 inhibition leads to altered isoform levels of many genes including several with established roles in leukemogenesis such as MYB, BRD4 and MED24. We focus on BRD4 as its main isoforms have distinct molecular properties and find that SRPK1 inhibition produces a significant switch from the short to the long isoform at the mRNA and protein levels. This was associated with BRD4 eviction from genomic loci involved in leukemogenesis including BCL2 and MYC. We go on to show that this switch mediates at least part of the anti-leukemic effects of SRPK1 inhibition. Our findings reveal that SRPK1 represents a plausible new therapeutic target against AML.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Cell Cycle Checkpoints , Cell Cycle Proteins , Cell Differentiation , Chromatin/metabolism , Epigenesis, Genetic , HL-60 Cells , Hematopoiesis , Humans , K562 Cells , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA Splicing
8.
Cell Stem Cell ; 23(5): 700-713.e6, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30388424

ABSTRACT

Clonal hematopoiesis (CH), in which stem cell clones dominate blood production, becomes increasingly common with age and can presage malignancy development. The conditions that promote ascendancy of particular clones are unclear. We found that mutations in PPM1D (protein phosphatase Mn2+/Mg2+-dependent 1D), a DNA damage response regulator that is frequently mutated in CH, were present in one-fifth of patients with therapy-related acute myeloid leukemia or myelodysplastic syndrome and strongly correlated with cisplatin exposure. Cell lines with hyperactive PPM1D mutations expand to outcompete normal cells after exposure to cytotoxic DNA damaging agents including cisplatin, and this effect was predominantly mediated by increased resistance to apoptosis. Moreover, heterozygous mutant Ppm1d hematopoietic cells outcompeted their wild-type counterparts in vivo after exposure to cisplatin and doxorubicin, but not during recovery from bone marrow transplantation. These findings establish the clinical relevance of PPM1D mutations in CH and the importance of studying mutation-treatment interactions. VIDEO ABSTRACT.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Clone Cells/drug effects , Doxorubicin/pharmacology , Hematopoiesis/drug effects , Leukemia, Myeloid, Acute/drug therapy , Mutation , Protein Phosphatase 2C/genetics , Aged , Animals , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cisplatin/chemistry , Doxorubicin/chemistry , Drug Screening Assays, Antitumor , Female , HEK293 Cells , Hematopoiesis/genetics , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Middle Aged , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Protein Phosphatase 2C/metabolism
9.
EMBO Mol Med ; 10(10)2018 10.
Article in English | MEDLINE | ID: mdl-30190333

ABSTRACT

Metastatic progression remains a major burden for cancer patients and is associated with eventual resistance to prevailing therapies such as chemotherapy. Here, we reveal how chemotherapy induces an extracellular matrix (ECM), wound healing, and stem cell network in cancer cells via the c-Jun N-terminal kinase (JNK) pathway, leading to reduced therapeutic efficacy. We find that elevated JNK activity in cancer cells is linked to poor clinical outcome in breast cancer patients and is critical for tumor initiation and metastasis in xenograft mouse models of breast cancer. We show that JNK signaling enhances expression of the ECM and stem cell niche components osteopontin, also called secreted phosphoprotein 1 (SPP1), and tenascin C (TNC), that promote lung metastasis. We demonstrate that both SPP1 and TNC are direct targets of the c-Jun transcription factor. Exposure to multiple chemotherapies further exploits this JNK-mediated axis to confer treatment resistance. Importantly, JNK inhibition or disruption of SPP1 or TNC expression sensitizes experimental mammary tumors and metastases to chemotherapy, thus providing insights to consider for future treatment strategies against metastatic breast cancer.


Subject(s)
Breast Neoplasms/physiopathology , Drug Resistance, Neoplasm , Neoplasm Metastasis/physiopathology , Signal Transduction , Animals , Cell Movement , Cell Proliferation , Disease Models, Animal , Extracellular Matrix/metabolism , Female , Heterografts , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Neoplasm Transplantation , Neoplastic Stem Cells/physiology
10.
Nat Genet ; 50(6): 883-894, 2018 06.
Article in English | MEDLINE | ID: mdl-29736013

ABSTRACT

The histone H3 Lys27-specific demethylase UTX (or KDM6A) is targeted by loss-of-function mutations in multiple cancers. Here, we demonstrate that UTX suppresses myeloid leukemogenesis through noncatalytic functions, a property shared with its catalytically inactive Y-chromosome paralog, UTY (or KDM6C). In keeping with this, we demonstrate concomitant loss/mutation of KDM6A (UTX) and UTY in multiple human cancers. Mechanistically, global genomic profiling showed only minor changes in H3K27me3 but significant and bidirectional alterations in H3K27ac and chromatin accessibility; a predominant loss of H3K4me1 modifications; alterations in ETS and GATA-factor binding; and altered gene expression after Utx loss. By integrating proteomic and genomic analyses, we link these changes to UTX regulation of ATP-dependent chromatin remodeling, coordination of the COMPASS complex and enhanced pioneering activity of ETS factors during evolution to AML. Collectively, our findings identify a dual role for UTX in suppressing acute myeloid leukemia via repression of oncogenic ETS and upregulation of tumor-suppressive GATA programs.


Subject(s)
Chromatin/genetics , Enhancer Elements, Genetic , GATA Transcription Factors/genetics , Histone Demethylases/genetics , Leukemia, Myeloid/genetics , Proto-Oncogene Proteins c-ets/genetics , Animals , Cell Line , Chromatin Assembly and Disassembly/genetics , Gene Expression Regulation, Leukemic , HEK293 Cells , Histones/genetics , Humans , Mice , Mice, Inbred C57BL , Proteomics/methods , Regulatory Sequences, Nucleic Acid/genetics , Transcriptional Activation
11.
Nature ; 552(7683): 126-131, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29186125

ABSTRACT

N6-methyladenosine (m6A) is an abundant internal RNA modification in both coding and non-coding RNAs that is catalysed by the METTL3-METTL14 methyltransferase complex. However, the specific role of these enzymes in cancer is still largely unknown. Here we define a pathway that is specific for METTL3 and is implicated in the maintenance of a leukaemic state. We identify METTL3 as an essential gene for growth of acute myeloid leukaemia cells in two distinct genetic screens. Downregulation of METTL3 results in cell cycle arrest, differentiation of leukaemic cells and failure to establish leukaemia in immunodeficient mice. We show that METTL3, independently of METTL14, associates with chromatin and localizes to the transcriptional start sites of active genes. The vast majority of these genes have the CAATT-box binding protein CEBPZ present at the transcriptional start site, and this is required for recruitment of METTL3 to chromatin. Promoter-bound METTL3 induces m6A modification within the coding region of the associated mRNA transcript, and enhances its translation by relieving ribosome stalling. We show that genes regulated by METTL3 in this way are necessary for acute myeloid leukaemia. Together, these data define METTL3 as a regulator of a chromatin-based pathway that is necessary for maintenance of the leukaemic state and identify this enzyme as a potential therapeutic target for acute myeloid leukaemia.


Subject(s)
Adenosine/analogs & derivatives , Gene Expression Regulation, Neoplastic/genetics , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Methyltransferases/metabolism , Promoter Regions, Genetic/genetics , Protein Biosynthesis , Adenosine/genetics , Adenosine/metabolism , Animals , CRISPR-Cas Systems , Cell Line, Tumor , Cell Proliferation/genetics , Chromatin/genetics , Chromatin/metabolism , Female , Genes, Neoplasm/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Methyltransferases/chemistry , Methyltransferases/deficiency , Methyltransferases/genetics , Mice , Protein Biosynthesis/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/metabolism , Transcription Initiation Site
12.
Leuk Lymphoma ; 58(2): 257-265, 2017 02.
Article in English | MEDLINE | ID: mdl-27401303

ABSTRACT

The p15 gene (also known as CDKN2B, INK4B, p15INK4B), located in band 9p21, encodes a protein that induces a G1-phase cell cycle arrest through inhibition of CDK4/6 (cyclin-dependent kinase 4/6). It also plays an important role in the regulation of cellular commitment of hematopoietic progenitor cells and myeloid cell differentiation. p15 can be silenced by several mechanisms, including deletion and hypermethylation of its promoter. Homozygous p15 deletion is rare in acute myeloblastic leukemia (AML) and myelodysplastic syndromes (MDS) but frequent in acute lymphoblastic leukemia (ALL). On the contrary, methylation of the p15 promoter is identified in some 50% of the patients with AML and MDS, but is less frequent in ALL. The analysis of the 28 studies available in the literature revealed conflicting results (unfavorable, favorable or no impact) that can be due, at least in part, to methodological and/or biological pitfalls. Among those, are the heterogeneity of the methylation patterns of the p15 gene and the lack of a comprehensive analysis including transcriptional and translational inactivation that have major impact on its expression. Therefore, detection of the p15 mRNA expression (quantitative or not) may represent a more appropriate method to determine the prognostic impact of the p15 gene.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p15/genetics , Genetic Variation , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Cyclin-Dependent Kinase Inhibitor p15/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/drug effects , Gene Frequency , Genetic Loci , Genetic Predisposition to Disease , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Molecular Targeted Therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Prognosis
13.
Methods Mol Biol ; 1541: 311-331, 2017.
Article in English | MEDLINE | ID: mdl-27910033

ABSTRACT

The main databases devoted stricto sensu to cancer cytogenetics are the "Mitelman Database of Chromosome Aberrations and Gene Fusions in Cancer" ( http://cgap.nci.nih.gov/Chromosomes/Mitelman ), the "Atlas of Genetics and Cytogenetics in Oncology and Haematology" ( http://atlasgeneticsoncology.org ), and COSMIC ( http://cancer.sanger.ac.uk/cosmic ).However, being a complex multistep process, cancer cytogenetics are broadened to "cytogenomics," with complementary resources on: general databases (nucleic acid and protein sequences databases; cartography browsers: GenBank, RefSeq, UCSC, Ensembl, UniProtKB, and Entrez Gene), cancer genomic portals associated with recent international integrated programs, such as TCGA or ICGC, other fusion genes databases, array CGH databases, copy number variation databases, and mutation databases. Other resources such as the International System for Human Cytogenomic Nomenclature (ISCN), the International Classification of Diseases for Oncology (ICD-O), and the Human Gene Nomenclature Database (HGNC) allow a common language.Data within the scientific/medical community should be freely available. However, most of the institutional stakeholders are now gradually disengaging, and well-known databases are forced to beg or to disappear (which may happen!).


Subject(s)
Computational Biology/methods , Cytogenetics/methods , Databases, Genetic , Web Browser , Chromosome Aberrations , Comparative Genomic Hybridization , Cytogenetic Analysis/methods , Gene Rearrangement , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Mutation , Neoplasms/diagnosis , Neoplasms/genetics , Software , User-Computer Interface
14.
Cell Rep ; 17(4): 1193-1205, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27760321

ABSTRACT

Acute myeloid leukemia (AML) is an aggressive cancer with a poor prognosis, for which mainstream treatments have not changed for decades. To identify additional therapeutic targets in AML, we optimize a genome-wide clustered regularly interspaced short palindromic repeats (CRISPR) screening platform and use it to identify genetic vulnerabilities in AML cells. We identify 492 AML-specific cell-essential genes, including several established therapeutic targets such as DOT1L, BCL2, and MEN1, and many other genes including clinically actionable candidates. We validate selected genes using genetic and pharmacological inhibition, and chose KAT2A as a candidate for downstream study. KAT2A inhibition demonstrated anti-AML activity by inducing myeloid differentiation and apoptosis, and suppressed the growth of primary human AMLs of diverse genotypes while sparing normal hemopoietic stem-progenitor cells. Our results propose that KAT2A inhibition should be investigated as a therapeutic strategy in AML and provide a large number of genetic vulnerabilities of this leukemia that can be pursued in downstream studies.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Genetic Testing , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Molecular Targeted Therapy , Adult , Apoptosis , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Humans , Reproducibility of Results
15.
Expert Rev Anticancer Ther ; 15(9): 1093-102, 2015.
Article in English | MEDLINE | ID: mdl-26211807

ABSTRACT

In 1963, Jean Bernard, a French hematologist, opened a new chapter in hematology called geographic hematology ('Hématologie Géographique'). He distinguished two research avenues. One dealt with the differences between the various populations (ethnic hematology), the other with various environmental factors (environmental hematology). In recent years, focus has been put on analyzing the genetic susceptibility in cancer and hematological malignancies, particularly in childhood acute lymphoblastic leukemia, using specific gene or (genome-wide association study) approach. However, almost 30 years ago, it was suggested by a few workers that chromosomal abnormalities observed in leukemia could have a geographic and/or ethnic distribution. In this review, we analyze the literature on chromosomal and molecular abnormalities in several types of leukemia.


Subject(s)
Genetic Predisposition to Disease , Leukemia/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Child , Chromosome Aberrations , Genome-Wide Association Study , Humans , Leukemia/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
16.
Future Oncol ; 11(11): 1675-86, 2015.
Article in English | MEDLINE | ID: mdl-26043219

ABSTRACT

The EVI1 gene, located in chromosomal band 3q26, is a transcription factor that has stem cell-specific expression pattern and is essential for the regulation of self-renewal of hematopoietic stem cells. It is now recognized as one of the dominant oncogenes associated with myeloid leukemia. EVI1 overexpression is associated with minimal to no response to chemotherapy and poor clinical outcome. Several chromosomal rearrangements involving band 3q26 are known to induce EVI1 overexpression. They are mainly found in acute myeloid leukemia and blastic phase of Philadelphia chromosome-positive chronic myeloid leukemia, more rarely in myelodysplastic syndromes. They include inv(3)(q21q26), t(3;3)(q21;q26), t(3;21)(q26;q22), t(3;12)(q26;p13) and t(2;3)(p15-23;q26). However, many other chromosomal rearrangements involving 3q26/EVI1 have been identified. The precise molecular event has not been elucidated in the majority of these chromosomal abnormalities and most gene partners remain unknown.


Subject(s)
DNA-Binding Proteins/genetics , Gene Rearrangement , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myeloid, Acute/genetics , Proto-Oncogenes/genetics , Transcription Factors/genetics , Chromosome Breakpoints , Chromosomes, Human, Pair 3 , Gene Expression , Humans , MDS1 and EVI1 Complex Locus Protein
18.
Expert Rev Hematol ; 7(3): 347-57, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24720386

ABSTRACT

The t(15;17)(q24;q21), generating a PML-RARA fusion gene, is the hallmark of acute promyelocytic leukemia (APL). At present, eight other genes fusing with RARA have been identified. The resulting fusion proteins retain domains of the RARA protein allowing binding to retinoic acid response elements (RARE) and dimerization with the retinoid X receptor protein (RXRA). They participate in protein-protein interactions, associating with RXRA to form hetero-oligomeric complexes that can bind to RARE. They have a dominant-negative effect on wild-type RARA/RXRA transcriptional activity. Moreover, RARA fusion proteins can homodimerize, conferring the ability to regulate an expanded repertoire of genes normally not affected by RARA. RARA fusion proteins behave as potent transcriptional repressors of retinoic acid signalling, inducing a differentiation blockage at the promyelocyte stage which can be overcome with therapeutic doses of ATRA or arsenic trioxide. However, resistance to these two drugs is a major problem, which necessitates development of new therapies.


Subject(s)
Gene Fusion , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/genetics , Receptors, Retinoic Acid/genetics , Animals , Cell Differentiation/genetics , Chromosome Aberrations , Humans , Leukemia, Promyelocytic, Acute/pathology , Response Elements , Retinoic Acid Receptor alpha
19.
Future Oncol ; 10(3): 475-95, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24559452

ABSTRACT

In humans, class I homeobox genes (HOX genes) are distributed in four clusters. Upstream regulators include transcriptional activators and members of the CDX family of transcription factors. HOX genes encode proteins and need cofactor interactions, to increase their specificity and selectivity. HOX genes contribute to the organization and regulation of hematopoiesis by controlling the balance between proliferation and differentiation. Changes in HOX gene expression can be associated with chromosomal rearrangements generating fusion genes, such as those involving MLL and NUP98, or molecular defects, such as mutations in NPM1 and CEBPA for example. Several miRNAs are involved in the control of HOX gene expression and their expression correlates with HOX gene dysregulation. HOX genes dysregulation is a dominant mechanism of leukemic transformation. A better knowledge of their target genes and the mechanisms by which their dysregulated expression contributes to leukemogenesis could lead to the development of new drugs.


Subject(s)
Gene Expression Regulation, Leukemic , Genes, Homeobox , Leukemia, Myeloid, Acute/genetics , Animals , Genes, Neoplasm , Humans , Leukemia, Myeloid, Acute/metabolism , Multigene Family , Nucleophosmin , Oncogene Proteins, Fusion/genetics
20.
Expert Rev Mol Diagn ; 14(2): 127-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24437978

ABSTRACT

Since the first specific chromosomal abnormality was identified in leukemia more than 50 years ago, technology has much evolved, now allowing the deciphering of cancer genomes in ever-greater detail. However, much has still to be learned as we have not yet completely dissected all the genomic aberrations driving the genesis and the evolution of malignant hemopathies. The first techniques that have been developed allowed 'gross' chromosomal abnormalities to be identified. They include conventional and molecular cytogenetics and microarray-based techniques. However, these techniques can only reveal part of the problem, as genes can be altered in a number of ways (mutations, methylation and so on). This led to the development of what is now known as next-generation sequencing (NGS). Each method has advantages and limits. At present, no single method can decipher all the mechanisms involved in leukemogenesis. Therefore, in our view, it is unlikely that a particular technique will become the 'gold standard'.


Subject(s)
Cytogenetic Analysis/methods , High-Throughput Nucleotide Sequencing/methods , Leukemia/diagnosis , Leukemia/genetics , Cytogenetic Analysis/trends , DNA Methylation , Genome , High-Throughput Nucleotide Sequencing/trends , Humans , In Situ Hybridization, Fluorescence , Mutation , Oligonucleotide Array Sequence Analysis , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...