Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
2.
Dev Dyn ; 251(10): 1728-1740, 2022 10.
Article in English | MEDLINE | ID: mdl-35429189

ABSTRACT

BACKGROUND: MLLT3 (AF9) is a nuclear transcription factor crucial for hematopoietic stem cell and progenitor cell maintenance, but its role during embryonic hematopoiesis remains uncertain. Here, we examine the role of mllt3 in developmental hematopoiesis during embryogenesis using zebrafish. RESULTS: Cloning, sequencing, phylogenetic, and synteny analyses showed high evolutionary conservation between important functional domains of the zebrafish orthologue of mllt3 and MLLT3 in humans. Quantitative reverse transcription-PCR and in situ hybridization analyses revealed that mllt3 is maternally supplied and zygotically expressed throughout embryonic development, and that expression is highest between 10 and 24 hours post-fertilization (hpf) coincident with enrichment in the intermediate cell mass (ICM) and posterior blood island, which are the sites of the primitive and transient definitive hematopoiesis in zebrafish, respectively. Further, we found co-expression of mllt3 with the early hematopoietic progenitor markers tal1, gata2, and gata1a in the posterior ICM. By investigating zebrafish hematopoietic mutants, we discovered that mllt3 is involved in erythroid precursor formation. By 48-72 hpf, mllt3 expression proved to be restricted to non-hematopoietic tissues including head structures, pronephric tubules, and liver primordium. CONCLUSIONS: These findings establish a link between mllt3 and primitive erythropoiesis and provide the basis for future functional investigations.


Subject(s)
Leukemia , Zebrafish , Animals , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Nuclear Proteins , Phylogeny , Transcription Factors/genetics , Zebrafish/genetics
3.
Pediatr Blood Cancer ; 69(1): e29344, 2022 01.
Article in English | MEDLINE | ID: mdl-34550633

ABSTRACT

Twelvepatients without therapy-related leukemia were studied after completing TOP2 poison chemotherapy in a high-risk neuroblastoma regimen. One patient harbored an inv(11) that was a KMT2A rearrangement. The KMT2A-MAML2 transcript was expressed at low level. The patient was prospectively followed. The inv(11) was undetectable in ensuing samples. Leukemia never developed after a 12.8-year follow-up period. Enriched etoposide-induced TOP2A cleavage in the relevant MAML2 genomic region supports a TOP2A DNA damage mechanism. After completing TOP2 poison chemotherapies, covert KMT2A-R clones may occur in a small minority of patients; however, not all KMT2A rearrangements herald a therapy-related leukemia diagnosis.


Subject(s)
Histone-Lysine N-Methyltransferase , Leukemia , Myeloid-Lymphoid Leukemia Protein , Neuroblastoma , Trans-Activators , Etoposide/administration & dosage , Follow-Up Studies , Gene Rearrangement , Humans , Leukemia/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Transcription Factors/genetics
4.
Blood ; 139(14): 2198-2211, 2022 04 07.
Article in English | MEDLINE | ID: mdl-34864916

ABSTRACT

KMT2A-rearranged (KMT2A-r) infant acute lymphoblastic leukemia (ALL) is a devastating malignancy with a dismal outcome, and younger age at diagnosis is associated with increased risk of relapse. To discover age-specific differences and critical drivers that mediate poor outcome in KMT2A-r ALL, we subjected KMT2A-r leukemias and normal hematopoietic cells from patients of different ages to single-cell multiomics analyses. We uncovered the following critical new insights: leukemia cells from patients <6 months have significantly increased lineage plasticity. Steroid response pathways are downregulated in the most immature blasts from younger patients. We identify a hematopoietic stem and progenitor-like (HSPC-like) population in the blood of younger patients that contains leukemic blasts and form an immunosuppressive signaling circuit with cytotoxic lymphocytes. These observations offer a compelling explanation for the ability of leukemias in young patients to evade chemotherapy and immune-mediated control. Our analysis also revealed preexisting lymphomyeloid primed progenitors and myeloid blasts at initial diagnosis of B-ALL. Tracking of leukemic clones in 2 patients whose leukemia underwent a lineage switch documented the evolution of such clones into frank acute myeloid leukemia (AML). These findings provide critical insights into KMT2A-r ALL and have clinical implications for molecularly targeted and immunotherapy approaches. Beyond infant ALL, our study demonstrates the power of single-cell multiomics to detect tumor intrinsic and extrinsic factors affecting rare but critical subpopulations within a malignant population that ultimately determines patient outcome.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Antineoplastic Agents/therapeutic use , Gene Rearrangement , Humans , Immunotherapy , Infant , Leukemia, Myeloid, Acute/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
5.
Oncogene ; 38(13): 2241-2262, 2019 03.
Article in English | MEDLINE | ID: mdl-30478448

ABSTRACT

The poor outcomes in infant acute lymphoblastic leukemia (ALL) necessitate new treatments. Here we discover that EIF4E protein is elevated in most cases of infant ALL and test EIF4E targeting by the repurposed antiviral agent ribavirin, which has anticancer properties through EIF4E inhibition, as a potential treatment. We find that ribavirin treatment of actively dividing infant ALL cells on bone marrow stromal cells (BMSCs) at clinically achievable concentrations causes robust proliferation inhibition in proportion with EIF4E expression. Further, we find that ribavirin treatment of KMT2A-rearranged (KMT2A-R) infant ALL cells and the KMT2A-AFF1 cell line RS4:11 inhibits EIF4E, leading to decreases in oncogenic EIF4E-regulated cell growth and survival proteins. In ribavirin-sensitive KMT2A-R infant ALL cells and RS4:11 cells, EIF4E-regulated proteins with reduced levels of expression following ribavirin treatment include MYC, MCL1, NBN, BCL2 and BIRC5. Ribavirin-treated RS4:11 cells exhibit impaired EIF4E-dependent nuclear to cytoplasmic export and/or translation of the corresponding mRNAs, as well as reduced phosphorylation of the p-AKT1, p-EIF4EBP1, p-RPS6 and p-EIF4E signaling proteins. This leads to an S-phase cell cycle arrest in RS4:11 cells corresponding to the decreased proliferation. Ribavirin causes nuclear EIF4E to re-localize to the cytoplasm in KMT2A-AFF1 infant ALL and RS4:11 cells, providing further evidence for EIF4E inhibition. Ribavirin slows increases in peripheral blasts in KMT2A-R infant ALL xenograft-bearing mice. Ribavirin cooperates with chemotherapy, particularly L-asparaginase, in reducing live KMT2A-AFF1 infant ALL cells in BMSC co-cultures. This work establishes that EIF4E is broadly elevated across infant ALL and that clinically relevant ribavirin exposures have preclinical activity and effectively inhibit EIF4E in KMT2A-R cases, suggesting promise in EIF4E targeting using ribavirin as a means of treatment.


Subject(s)
Eukaryotic Initiation Factor-4E/genetics , Molecular Targeted Therapy/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Ribavirin/therapeutic use , Cell Line, Tumor , Child, Preschool , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Eukaryotic Initiation Factor-4E/physiology , Gene Expression Profiling , Gene Expression Regulation, Leukemic/drug effects , Humans , Indoles , Infant , Microarray Analysis , Multigene Family/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Biosynthesis/drug effects , Pyrroles/therapeutic use , Signal Transduction/drug effects
6.
Genome Res ; 27(7): 1238-1249, 2017 07.
Article in English | MEDLINE | ID: mdl-28385713

ABSTRACT

Type II topoisomerases orchestrate proper DNA topology, and they are the targets of anti-cancer drugs that cause treatment-related leukemias with balanced translocations. Here, we develop a high-throughput sequencing technology to define TOP2 cleavage sites at single-base precision, and use the technology to characterize TOP2A cleavage genome-wide in the human K562 leukemia cell line. We find that TOP2A cleavage has functionally conserved local sequence preferences, occurs in cleavage cluster regions (CCRs), and is enriched in introns and lincRNA loci. TOP2A CCRs are biased toward the distal regions of gene bodies, and TOP2 poisons cause a proximal shift in their distribution. We find high TOP2A cleavage levels in genes involved in translocations in TOP2 poison-related leukemia. In addition, we find that a large proportion of genes involved in oncogenic translocations overall contain TOP2A CCRs. The TOP2A cleavage of coding and lincRNA genes is independently associated with both length and transcript abundance. Comparisons to ENCODE data reveal distinct TOP2A CCR clusters that overlap with marks of transcription, open chromatin, and enhancers. Our findings implicate TOP2A cleavage as a broad DNA damage mechanism in oncogenic translocations as well as a functional role of TOP2A cleavage in regulating transcription elongation and gene activation.


Subject(s)
DNA Damage , DNA Topoisomerases, Type II/metabolism , Genetic Loci , Leukemia/enzymology , Neoplasm Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Transcription Elongation, Genetic , DNA Topoisomerases, Type II/genetics , Humans , K562 Cells , Leukemia/genetics , Leukemia/pathology , Neoplasm Proteins/genetics , Poly-ADP-Ribose Binding Proteins/genetics , RNA, Long Noncoding/biosynthesis , RNA, Long Noncoding/genetics
7.
Pediatr Blood Cancer ; 63(7): 1175-80, 2016 07.
Article in English | MEDLINE | ID: mdl-26999444

ABSTRACT

BACKGROUND: Infant acute lymphoblastic leukemia (ALL) has never occurred in families except for the ∼100% concordant cases in monozygous twins attributed to twin-to-twin metastases. We report the first kindred with infant ALL in non-twin siblings. The siblings were diagnosed with MLL-rearranged (MLL-R) ALL 26 months apart. The second affected sibling had an unaffected dichorionic monozygous co-twin. Both had fatal outcomes. PROCEDURES: Translocations were characterized by karyotype, FISH, multiplex FISH, and MLL breakpoint cluster region (bcr) Southern blot analysis. Breakpoint junctions and fusion transcripts were cloned by PCR. TP53 mutation and NADPH quinone oxidorecuctase 1 (NQO1) C609T analyses were performed, and pedigree history and parental occupations were ascertained. The likelihood of chance occurrence of infant ALL in non-twin siblings was computed based on a binomial distribution. Zygosity was determined by single nucleotide polymorphism (SNP) array. RESULTS: The translocations were not related or vertically transmitted. The complex karyotype of the proband's ALL had chromosome 2, 3, 4, and 11 abnormalities causing a 5'-MLL-AFF1-3' fusion and a non-productive rearrangement of 3'MLL with a chromosome 3q intergenic region. The affected twin's ALL exhibited a simple t(4;11). The complex karyotype of the proband's ALL suggested a genotoxic insult, but no exposure was identified. There was no germline TP53 mutation. The NQO1 C609T risk allele was absent. The likelihood of infant ALL occurring in non-twin siblings by chance alone is one in 1.198 × 10(9) families. CONCLUSIONS: Whether because of a deleterious transplacental exposure, novel predisposition syndrome, or exceedingly rare chance occurrence, MLL-R infant ALL can occur in non-twin siblings. The discordant occurrence of infant ALL in the monozygous twins was likely because they were dichorionic.


Subject(s)
Chromosomes, Human/genetics , Gene Rearrangement , Histone-Lysine N-Methyltransferase/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Siblings , Translocation, Genetic , Twins, Dizygotic , Humans , Infant , Male , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology
8.
Hum Mol Genet ; 24(17): 4829-47, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26041819

ABSTRACT

Mitochondrial respiratory chain (RC) disease therapies directed at intra-mitochondrial pathology are largely ineffective. Recognizing that RC dysfunction invokes pronounced extra-mitochondrial transcriptional adaptations, particularly involving dysregulated translation, we hypothesized that translational dysregulation is itself contributing to the pathophysiology of RC disease. Here, we investigated the activities, and effects from direct inhibition, of a central translational regulator (mTORC1) and its downstream biological processes in diverse genetic and pharmacological models of RC disease. Our data identify novel mechanisms underlying the cellular pathogenesis of RC dysfunction, including the combined induction of proteotoxic stress, the ER stress response and autophagy. mTORC1 inhibition with rapamycin partially ameliorated renal disease in B6.Pdss2(kd/kd) mice with complexes I-III/II-III deficiencies, improved viability and mitochondrial physiology in gas-1(fc21) nematodes with complex I deficiency, and rescued viability across a variety of RC-inhibited human cells. Even more effective was probucol, a PPAR-activating anti-lipid drug that we show also inhibits mTORC1. However, directly inhibiting mTORC1-regulated downstream activities yielded the most pronounced and sustained benefit. Partial inhibition of translation by cycloheximide, or of autophagy by lithium chloride, rescued viability, preserved cellular respiratory capacity and induced mitochondrial translation and biogenesis. Cycloheximide also ameliorated proteotoxic stress via a uniquely selective reduction of cytosolic protein translation. RNAseq-based transcriptome profiling of treatment effects in gas-1(fc21) mutants provide further evidence that these therapies effectively restored altered translation and autophagy pathways toward that of wild-type animals. Overall, partially inhibiting cytosolic translation and autophagy offer novel treatment strategies to improve health across the diverse array of human diseases whose pathogenesis involves RC dysfunction.


Subject(s)
Autophagy , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Protein Biosynthesis , Animals , Autophagy/drug effects , Autophagy/genetics , Cell Survival/drug effects , Cycloheximide/pharmacology , Cytosol , Disease Models, Animal , Electron Transport , Endoplasmic Reticulum Stress/drug effects , Enzyme Activation , Gene Expression Profiling , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Multiprotein Complexes/metabolism , Phosphorylation , Probucol/pharmacology , Protein Biosynthesis/drug effects , Ribosomal Protein S6 Kinases/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Transcriptome
9.
Pediatr Blood Cancer ; 62(3): 419-26, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25399948

ABSTRACT

BACKGROUND: Infants with acute lymphoblastic leukemia (ALL) present with aggressive disease and a poor prognosis. Early relapse within 6-9 months of diagnosis is common. Approximately 75% of infants have MLL-rearranged (MLL-R) ALL with event free survival (EFS) ranging from 20% to 30%. Children's Oncology Group (COG) P9407 used shortened (46 weeks), intensified therapy to address early relapse and poor EFS. PROCEDURE: P9407 therapy was modified three times for induction toxicity resulting in three cohorts of therapy. One hundred forty-seven infants were enrolled in the third cohort. RESULTS: We report an overall 5-year EFS and OS of 42.3 ± 6% and 52.9 ± 6.5% respectively. Poor prognostic factors included age ≤90 days at diagnosis, MLL-R ALL and white cell count ≥50,000/µl. For infants ≤90 days of age, the 5-year EFS was 15.5 ± 10.1% and 48.5 ± 6.7% for those >90 days (P < 0.0001). Among infants >90 days of age, 5-year EFS rates were 43.8 ± 8% for MLL-R versus 69.1 ± 13.6% for MLL-germline ALL (P < 0.0001). CONCLUSIONS: Age ≤90 days at diagnosis was the most important prognostic factor. Despite shortened therapy with early intensification, EFS remained less than 50% overall in MLL-R ALL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Age Factors , Disease-Free Survival , Female , Follow-Up Studies , Gene Rearrangement , Histone-Lysine N-Methyltransferase , Humans , Infant , Infant, Newborn , Male , Myeloid-Lymphoid Leukemia Protein/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Recurrence , Risk Factors , Stem Cell Transplantation , Survival Rate
10.
Ann N Y Acad Sci ; 1310: 98-110, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24495080

ABSTRACT

Type II topoisomerases are essential enzymes that modulate DNA under- and overwinding, knotting, and tangling. Beyond their critical physiological functions, these enzymes are the targets for some of the most widely prescribed anticancer drugs (topoisomerase II poisons) in clinical use. Topoisomerase II poisons kill cells by increasing levels of covalent enzyme-cleaved DNA complexes that are normal reaction intermediates. Drugs such as etoposide, doxorubicin, and mitoxantrone are frontline therapies for a variety of solid tumors and hematological malignancies. Unfortunately, their use also is associated with the development of specific leukemias. Regimens that include etoposide or doxorubicin are linked to the occurrence of acute myeloid leukemias that feature rearrangements at chromosomal band 11q23. Similar rearrangements are seen in infant leukemias and are associated with gestational diets that are high in naturally occurring topoisomerase II-active compounds. Finally, regimens that include mitoxantrone and epirubicin are linked to acute promyelocytic leukemias that feature t(15;17) rearrangements. The first part of this article will focus on type II topoisomerases and describe the mechanism of enzyme and drug action. The second part will discuss how topoisomerase II poisons trigger chromosomal breaks that lead to leukemia and potential approaches for dissociating the actions of drugs from their leukemogenic potential.


Subject(s)
Cell Transformation, Neoplastic/genetics , Chromosome Breakage , DNA Topoisomerases, Type II/physiology , Leukemia/genetics , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Transformation, Neoplastic/chemically induced , Curcumin/pharmacology , DNA Topoisomerases, Type II/genetics , Genistein/pharmacology , Humans , Infant , Leukemia/enzymology , Neoplasms, Second Primary/chemically induced , Neoplasms, Second Primary/genetics , Translocation, Genetic/drug effects , Translocation, Genetic/genetics
11.
Blood ; 121(14): 2689-703, 2013 Apr 04.
Article in English | MEDLINE | ID: mdl-23393050

ABSTRACT

Survival in infants younger than 1 year who have acute lymphoblastic leukemia (ALL) is inferior whether MLL is rearranged (R) or germline (G). MLL translocations confer chemotherapy resistance, and infants experience excess complications. We characterized in vitro sensitivity to the pan-antiapoptotic BCL-2 family inhibitor obatoclax mesylate in diagnostic leukemia cells from 54 infants with ALL/bilineal acute leukemia because of the role of prosurvival BCL-2 proteins in resistance, their imbalanced expression in infant ALL, and evidence of obatoclax activity with a favorable toxicity profile in early adult leukemia trials. Overall, half maximal effective concentrations (EC50s) were lower than 176 nM (the maximal plasma concentration [Cmax] with recommended adult dose) in 76% of samples, whether in MLL-AF4, MLL-ENL, or other MLL-R or MLL-G subsets, and regardless of patients' poor prognostic features. However, MLL status and partner genes correlated with EC50. Combined approaches including flow cytometry, Western blot, obatoclax treatment with death pathway inhibition, microarray analyses, and/or electron microscopy indicated a unique killing mechanism involving apoptosis, necroptosis, and autophagy in MLL-AF4 ALL cell lines and primary MLL-R and MLL-G infant ALL cells. This in vitro obatoclax activity and its multiple killing mechanisms across molecular cytogenetic subsets provide a rationale to incorporate a similarly acting compound into combination strategies to combat infant ALL.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Pyrroles/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Leukemic/drug effects , Histone-Lysine N-Methyltransferase , Humans , Indoles , Infant , Infant, Newborn , Myeloid-Lymphoid Leukemia Protein/genetics , Necrosis/drug therapy , Oncogene Proteins, Fusion/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors
12.
Blood ; 119(8): 1872-81, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-22210879

ABSTRACT

Gene expression profiling was performed on 97 cases of infant ALL from Children's Oncology Group Trial P9407. Statistical modeling of an outcome predictor revealed 3 genes highly predictive of event-free survival (EFS), beyond age and MLL status: FLT3, IRX2, and TACC2. Low FLT3 expression was found in a group of infants with excellent outcome (n = 11; 5-year EFS of 100%), whereas differential expression of IRX2 and TACC2 partitioned the remaining infants into 2 groups with significantly different survivals (5-year EFS of 16% vs 64%; P < .001). When infants with MLL-AFF1 were analyzed separately, a 7-gene classifier was developed that split them into 2 distinct groups with significantly different outcomes (5-year EFS of 20% vs 65%; P < .001). In this classifier, elevated expression of NEGR1 was associated with better EFS, whereas IRX2, EPS8, and TPD52 expression were correlated with worse outcome. This classifier also predicted EFS in an independent infant ALL cohort from the Interfant-99 trial. When evaluating expression profiles as a continuous variable relative to patient age, we further identified striking differences in profiles in infants less than or equal to 90 days of age and those more than 90 days of age. These age-related patterns suggest different mechanisms of leukemogenesis and may underlie the differential outcomes historically seen in these age groups.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Leukemic , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Age Factors , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carrier Proteins/genetics , Cluster Analysis , Cohort Studies , DNA-Binding Proteins/genetics , Female , Gene Regulatory Networks , Homeodomain Proteins/genetics , Humans , Infant , Kaplan-Meier Estimate , Male , Models, Genetic , Myeloid-Lymphoid Leukemia Protein/genetics , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Fusion/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Prognosis , Transcription Factors/genetics , Transcriptional Elongation Factors , Treatment Outcome , Tumor Suppressor Proteins/genetics , fms-Like Tyrosine Kinase 3/genetics
13.
Br J Haematol ; 152(3): 307-21, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21155757

ABSTRACT

Infant leukaemia is an embryonal disease in which the underlying MLL translocations initiate in utero. Zebrafish offer unique potential to understand how MLL impacts haematopoiesis from the earliest embryonic timepoints and how translocations cause leukaemia as an embryonal process. In this study, a zebrafish mll cDNA syntenic to human MLL spanning the 5' to 3' UTRs, was cloned from embryos, and mll expression was characterized over the zebrafish lifespan. The protein encoded by the 35-exon ORF exhibited 46·4% overall identity to human MLL and 68-100% conservation in functional domains (AT-hooks, SNL, CXXC, PHD, bromodomain, FYRN, taspase1 sites, FYRC, SET). Maternally supplied transcripts were detected at 0-2 hpf. Strong ubiquitous early zygotic expression progressed to a cephalo-caudal gradient during later embryogenesis. mll was expressed in the intermediate cell mass (ICM) where primitive erythrocytes are produced and in the kidney where definitive haematopoiesis occurs in adults. mll exhibits high cross species conservation, is developmentally regulated in haematopoietic and other tissues and is expressed from the earliest embryonic timepoints throughout the zebrafish lifespan. Haematopoietic tissue expression validates using zebrafish for MLL haematopoiesis and leukaemia models.


Subject(s)
Hematopoietic System/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Zebrafish/metabolism , Aging/genetics , Aging/metabolism , Amino Acid Sequence , Animals , Base Sequence , Computational Biology , DNA, Complementary/genetics , Gene Expression Regulation, Developmental , Hematopoiesis/physiology , Humans , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein/genetics , Open Reading Frames , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Species Specificity , Zebrafish/genetics
15.
Blood ; 115(2): 326-30, 2010 Jan 14.
Article in English | MEDLINE | ID: mdl-19884644

ABSTRACT

Therapy-related acute promyelocytic leukemia (t-APL) with t(15;17)(q22;q21) involving the PML and RARA genes is associated with exposure to agents targeting topoisomerase II (topoII), particularly mitoxantrone and epirubicin. We previously have shown that mitoxantrone preferentially induces topoII-mediated DNA damage in a "hotspot region" within PML intron 6. To investigate mechanisms underlying epirubicin-associated t-APL, t(15;17) genomic breakpoints were characterized in 6 cases with prior breast cancer. Significant breakpoint clustering was observed in PML and RARA loci (P = .009 and P = .017, respectively), with PML breakpoints lying outside the mitoxantrone-associated hotspot region. Recurrent breakpoints identified in the PML and RARA loci in epirubicin-related t-APL were shown to be preferential sites of topoII-induced DNA damage, enhanced by epirubicin. Although site preferences for DNA damage differed between mitoxantrone and epirubicin, the observation that particular regions of the PML and RARA loci are susceptible to these agents may underlie their respective propensities to induce t-APL.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Breast Neoplasms/drug therapy , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 17/genetics , Epirubicin/adverse effects , Leukemia, Promyelocytic, Acute/genetics , Neoplasms, Second Primary/genetics , Translocation, Genetic/drug effects , Adult , Antibiotics, Antineoplastic/administration & dosage , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Chromosomes, Human, Pair 15/metabolism , Chromosomes, Human, Pair 17/metabolism , DNA Damage/drug effects , DNA Damage/genetics , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , Epirubicin/administration & dosage , Female , Humans , Introns/genetics , Leukemia, Promyelocytic, Acute/chemically induced , Leukemia, Promyelocytic, Acute/metabolism , Middle Aged , Mitoxantrone/pharmacology , Neoplasms, Second Primary/chemically induced , Neoplasms, Second Primary/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein , Quantitative Trait Loci , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Topoisomerase II Inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
17.
Methods Mol Biol ; 538: 85-114, 2009.
Article in English | MEDLINE | ID: mdl-19277575

ABSTRACT

Translocations and other rearrangements of the MLL gene at chromosome band 11q23 are biologically and clinically important molecular abnormalities in infant acute leukemias, leukemias associated with chemotherapeutic topoisomerase II poisons and, less often, acute leukemias in adults or myelodysplastic syndrome. Depending on the disease and the regimen, MLL-rearranged leukemias may be associated with inferior prognosis, and MLL rearrangements with some of the more than 60 known MLL-partner genes confer especially adverse effects as response to treatment (Blood 108:441-451, 2006). MLL rearrangements are usually evident as overt balanced chromosomal translocations by conventional cytogenetic analysis but up to one-third are cryptic rearrangements and occur in leukemias with del(11)(q23), a normal karyotype, or trisomy 11, the latter two of which sometimes are associated with partial tandem duplications of MLL itself (Proc Natl Acad Sci USA 97:2814-2819, 2000; Proc Natl Acad Sci USA 94:3899-3902, 1997). In addition, subsets of MLL rearrangements are complex at a cytogenetic level and/or molecular level, and fuse MLL with two different partner genes. Rapid and accurate methods to identify and characterize genomic breakpoint junctions and fusion transcripts resulting from the many types of MLL rearrangements are essential for risk group stratification, treatment protocol assignments, new partner gene discovery, understanding leukemia etiology and pathogenesis, and elucidating the impact of less common MLL-partner genes on biology and prognosis. Due to the vast heterogeneity in partner genes, typical gene-specific PCR based methods are not practical, especially when cytogenetics are normal or do not suggest involvement of a known partner gene of MLL. We have advanced seven different panhandle PCR based methods for cloning 5'-MLL-partner gene-3' and 5'-partner gene-MLL-3' genomic breakpoint junctions and identifying 5'-MLL-partner gene-3' fusion transcripts, all of which employ a stem-loop template shaped schematically like a pan with a handle and amplify the template without knowledge of the unknown partner sequence using primers all derived from MLL alone.


Subject(s)
Chromosome Breakage , Cloning, Molecular/methods , Leukemia/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Polymerase Chain Reaction/methods , Alternative Splicing/genetics , Chromosomes, Human, Pair 11/genetics , Exons/genetics , Histone-Lysine N-Methyltransferase , Humans
18.
Blood ; 112(8): 3383-90, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18650449

ABSTRACT

Therapy-related acute promyelocytic leukemia (t-APL) with t(15;17) translocation is a well-recognized complication of cancer treatment with agents targeting topoisomerase II. However, cases are emerging after mitoxantrone therapy for multiple sclerosis (MS). Analysis of 12 cases of mitoxantrone-related t-APL in MS patients revealed an altered distribution of chromosome 15 breakpoints versus de novo APL, biased toward disruption within PML intron 6 (11 of 12, 92% vs 622 of 1022, 61%: P = .035). Despite this intron spanning approximately 1 kb, breakpoints in 5 mitoxantrone-treated patients fell within an 8-bp region (1482-9) corresponding to the "hotspot" previously reported in t-APL, complicating mitoxantrone-containing breast cancer therapy. Another shared breakpoint was identified within the approximately 17-kb RARA intron 2 involving 2 t-APL cases arising after mitoxantrone treatment for MS and breast cancer, respectively. Analysis of PML and RARA genomic breakpoints in functional assays in 4 cases, including the shared RARA intron 2 breakpoint at 14 446-49, confirmed each to be preferential sites of topoisomerase IIalpha-mediated DNA cleavage in the presence of mitoxantrone. This study further supports the presence of preferential sites of DNA damage induced by mitoxantrone in PML and RARA genes that may underlie the propensity to develop this subtype of leukemia after exposure to this agent.


Subject(s)
Chromosomes, Human, Pair 15 , Chromosomes, Human, Pair 17 , Leukemia, Promyelocytic, Acute/chemically induced , Leukemia, Promyelocytic, Acute/genetics , Multiple Sclerosis/therapy , Translocation, Genetic , Adult , Antigens, Neoplasm/metabolism , DNA/chemistry , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Female , Humans , Introns , Male , Middle Aged , Mitoxantrone/adverse effects , Mitoxantrone/pharmacology , Models, Genetic
19.
Br J Haematol ; 141(6): 827-39, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18422996

ABSTRACT

Chemotherapy resistance from imbalanced apoptosis regulation may contribute to poor outcome in leukaemias with t(4;11). Anti-apoptotic BCL-2 expression and target modulation were characterized in cell lines with t(4;11) and BCL-2 expression was examined in MLL and non-MLL infant/paediatric leukaemia cases by Western blot analysis and/or real-time polymerase chain reaction. Cytotoxicity of Genasensetrade mark (Oblimersen Sodium, G3139) alone or combined with cytotoxic drugs was assessed by MTT [(3-4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide] assays of the cell lines, applying pharmacostatistical response surface modelling of drug interactions. Apoptosis and cell cycle were evaluated by flow cytometry in RS4:11 cells. Primary leukaemias and cell lines with t(4;11) expressed abundant BCL2 mRNA and protein. Variable, sometimes substantial BCL2 mRNA was detected in other leukaemia subtypes. G3139 reduced BCL2 mRNA and protein in RS4:11 cells. The most sensitive cell line to single-agent G3139 was RS4:11. Low G3139 concentrations sensitized RS4:11 and MV4-11 cells to select anti-leukaemia cytotoxic drugs. In RS4:11 cells, combining G3139 with doxorubicin (ADR) increased active caspase 3 and TUNEL staining compared to ADR alone, indicating greater apoptosis, and G3139 increased S-phase progression. The abundant BCL-2 affords a molecular target in leukaemias with t(4;11). G3139 exhibits preclinical activity and synergy with select cytotoxic agents in RS4:11 and MV4-11 cells, and these effects occur through apoptosis.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 4/genetics , Leukemia, Myeloid, Acute/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Translocation, Genetic , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Drug Screening Assays, Antitumor , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Thionucleotides/pharmacology , Tumor Cells, Cultured
20.
Blood ; 111(7): 3802-12, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18195096

ABSTRACT

We prospectively observed a child exposed to intensive multimodality therapy for metastatic neuroblastoma from emergence of a MLL translocation to disease diagnosis. The t(4;11)(p12;q23) was detected in the marrow 17 months after starting treatment following topoisomerase II poisons, alkylating agents, local radiation, hematopoietic stem cell transplantation, anti-GD2 monoclonal antibody with granulocyte macrophage-colony-stimulating factor, and a high cumulative dose of oral etoposide. Reciprocal genomic breakpoint junctions and fusion transcripts joined MLL with FRYL, the Drosophila melanogaster protein homologue of which regulates cell fate. Etoposide metabolites induced topoisomerase II cleavage complexes that could form both breakpoint junctions. Cells harboring the translocation replaced the marrow without clinical evidence of leukemia and differentiation appeared unaffected for 37 months. Subsequent bilineage dysplasia and increased blasts in addition to the translocation fulfilled criteria for MDS. The MEIS1 target gene of typical MLL fusion oncoproteins was underexpressed before and at MDS diagnosis. These results are consistent with repair of topoisomerase II cleavage from etoposide metabolites as the translocation mechanism, whereas other agents in the regimen may have contributed to progression of the clone with the translocation to MDS. MLL-FRYL did not increase MEIS1 expression, conferred a proliferative advantage without altering differentiation, and had protracted latency to disease.


Subject(s)
Gene Expression Regulation, Neoplastic , Homeodomain Proteins/biosynthesis , Myelodysplastic Syndromes/metabolism , Myeloid-Lymphoid Leukemia Protein/biosynthesis , Neoplasm Proteins/biosynthesis , Neuroblastoma/metabolism , Oncogene Proteins, Fusion/biosynthesis , Alkylating Agents/administration & dosage , Alkylating Agents/adverse effects , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bone Marrow/metabolism , Bone Marrow/pathology , Child, Preschool , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 12/metabolism , Chromosomes, Human, Pair 4/genetics , Chromosomes, Human, Pair 4/metabolism , Combined Modality Therapy/adverse effects , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Etoposide/administration & dosage , Etoposide/adverse effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Histone-Lysine N-Methyltransferase , Homeodomain Proteins/genetics , Humans , Male , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/etiology , Myelodysplastic Syndromes/pathology , Myeloid Ecotropic Viral Integration Site 1 Protein , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Proteins/genetics , Neuroblastoma/complications , Neuroblastoma/diagnosis , Neuroblastoma/pathology , Neuroblastoma/therapy , Oncogene Proteins, Fusion/genetics , Peripheral Blood Stem Cell Transplantation , Prospective Studies , Sequence Homology, Amino Acid , Topoisomerase II Inhibitors , Translocation, Genetic/genetics , Transplantation, Autologous
SELECTION OF CITATIONS
SEARCH DETAIL
...