Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Haemophilia ; 26(4): 735-740, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32420682

ABSTRACT

INTRODUCTION: The time-dependent nature of factor VIII (FVIII) inhibitors is well described, and the standard FVIII Bethesda assay used to measure inhibitors incorporates a 2-hour incubation. Despite case reports and reviews describing the immediate-acting nature of factor IX (FIX) inhibitors, many coagulation laboratories continue to use a traditional prolonged incubation for FIX Bethesda assays. To our knowledge, a comprehensive evaluation of the FIX Bethesda assay without incubation has not been reported. AIM: The goal of this study was to evaluate the performance of a rapid FIX Bethesda (ie no incubation) compared with the standard Bethesda assay (2-hour incubation). METHODS: The analysis used a Bethesda assay configured for either immediate testing or a 2-hour incubation. Samples from 14 haemophilia B patients with inhibitors and 9 non-human controls were tested. RESULTS: The two assays yielded similar performance overall. The average per cent difference in inhibitor titre between the rapid and standard FIX Bethesda assay was -3% (range -15% to +13%; P = .175) for patient samples and -2% (range -17% to +14%; P = .376) for controls. CONCLUSION: The rapid Bethesda assay showed good agreement with the standard Bethesda assay for determination of inhibitor levels in patients with severe haemophilia B. The rapid assay allows for faster assessment of inhibitors in patients with severe haemophilia B and has the potential to improve the ability of the coagulation laboratory to perform testing from a logistical viewpoint. Further studies involving larger numbers of patients would be important to confirm our findings.


Subject(s)
Blood Coagulation Factor Inhibitors/analysis , Blood Coagulation Tests/standards , Factor IX/antagonists & inhibitors , Hemophilia B/blood , Animals , Blood Coagulation/physiology , Blood Coagulation Tests/statistics & numerical data , Blood Coagulation Tests/trends , Factor IX/immunology , Factor IX/metabolism , Goats/blood , Hemophilia B/diagnosis , Humans , Indicators and Reagents/chemistry , Male , Mice/blood , Models, Animal , Reference Standards , Severity of Illness Index , Sheep/blood
2.
Br J Haematol ; 170(2): 200-7, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25891862

ABSTRACT

A jumping translocation (JT) is a rare cytogenetic aberration that can occur in haematological malignancy. It involves the translocation of the same fragment of donor chromosome onto two or more recipient chromosomes, typically in different cells. In this study, we describe the first series of chronic lymphocytic leukaemia (CLL) patients with JTs reported to date. Following a review of 878 CLL patient karyotypes, we identified 26 patients (3%) with 97 JTs. The most commonly occurring breakpoint in these translocations was 17p11.2. Loss of TP53 was identified prior to or at the same time as JT in 23 of 26 patients (88%). All patients eventually developed a complex karyotype. All but one patient has required treatment for CLL, with estimated median time to treatment of 11·5 months. This study establishes JTs as a recurrent abnormality found in CLL patients with aggressive disease. JTs contribute to complex karyotypes and, in many cases, are involved in chromosomal rearrangements that result in loss of the tumour suppressor gene TP53.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Translocation, Genetic , Adult , Aged , Chromosome Breakpoints , Chromosomes, Human, Pair 17 , Female , Genes, p53 , Humans , Karyotype , Karyotyping , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged , Neoplasm Staging
3.
Blood Cancer J ; 13(1): 59, 2023 04 24.
Article in English | MEDLINE | ID: mdl-37088803

ABSTRACT

Treated AML patients often have measurable residual disease (MRD) due to persisting low-level clones. This study assessed whether residual post-treatment somatic mutations, detected by NGS, were significantly prognostic for subsequent clinical outcomes. AML patients (n = 128) underwent both pre-and post-treatment testing with the same 42-gene MRD-validated NGS assay. After induction, 59 (46%) patients were mutation-negative (0.0024 VAF detection limit) and 69 (54%) had ≥1 persisting NGS-detectable mutation. Compared with NGS-negative patients, NGS-positive patients had shorter overall survival (17 months versus median not reached; P = 0.004; hazard ratio = 2.2 [95% CI: 1.3-3.7]) and a shorter time to relapse (14 months versus median not reached; P = 0.014; HR = 1.9 [95% CI: 1.1-3.1]). Among 95 patients with a complete morphologic remission (CR), 43 (45%) were MRD-positive by NGS and 52 (55%) were MRD-negative. These MRD-positive CR patients had a shorter overall survival (16.8 months versus median not reached; P = 0.013; HR = 2.1 [95% CI: 1.2-3.9]) than did the MRD-negative CR patients. Post-treatment persisting MRD positivity, defined by the same NGS-based test used at diagnosis, is thus a more sensitive biomarker for low-level leukemic clones compared to traditional non-molecular methods and is prognostic of subsequent relapse and death.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute , Humans , Prognosis , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Recurrence , Neoplasm, Residual/diagnosis
4.
PLoS One ; 16(4): e0243683, 2021.
Article in English | MEDLINE | ID: mdl-33909614

ABSTRACT

Identification of genomic mutations by molecular testing plays an important role in diagnosis, prognosis, and treatment of myeloid neoplasms. Next-generation sequencing (NGS) is an efficient method for simultaneous detection of clinically significant genomic mutations with high sensitivity. Various NGS based in-house developed and commercial myeloid neoplasm panels have been integrated into routine clinical practice. However, some genes frequently mutated in myeloid malignancies are particularly difficult to sequence with NGS panels (e.g., CEBPA, CARL, and FLT3). We report development and validation of a 48-gene NGS panel that includes genes that are technically challenging for molecular profiling of myeloid neoplasms including acute myeloid leukemia (AML), myelodysplastic syndrome (MDS), and myeloproliferative neoplasms (MPN). Target regions were captured by hybridization with complementary biotinylated DNA baits, and NGS was performed on an Illumina NextSeq500 instrument. A bioinformatics pipeline that was developed in-house was used to detect single nucleotide variations (SNVs), insertions/deletions (indels), and FLT3 internal tandem duplications (FLT3-ITD). An analytical validation study was performed on 184 unique specimens for variants with allele frequencies ≥5%. Variants identified by the 48-gene panel were compared to those identified by a 35-gene hematologic neoplasms panel using an additional 137 unique specimens. The developed assay was applied to a large cohort (n = 2,053) of patients with suspected myeloid neoplasms. Analytical validation yielded 99.6% sensitivity (95% CI: 98.9-99.9%) and 100% specificity (95% CI: 100%). Concordance of variants detected by the 2 tested panels was 100%. Among patients with suspected myeloid neoplasms (n = 2,053), 54.5% patients harbored at least one clinically significant mutation: 77% in AML patients, 48% in MDS, and 45% in MPN. Together, these findings demonstrate that the assay can identify mutations associated with diagnosis, prognosis, and treatment options of myeloid neoplasms even in technically challenging genes.


Subject(s)
Hematologic Neoplasms , High-Throughput Nucleotide Sequencing , Leukemia, Myeloid, Acute , Mutation , Myelodysplastic Syndromes , Neoplasm Proteins , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism
5.
Mol Cell Biol ; 26(6): 2373-86, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16508012

ABSTRACT

Overexpression of transferrin receptor 1 (TFRC1), a major mediator of iron uptake in mammalian cells, is a common feature of human malignancies. Therapeutic strategies designed to interfere with tumor iron metabolism have targeted TFRC1. The c-Myc oncogenic transcription factor stimulates proliferation and growth by activating thousands of target genes. Here we demonstrate that TFRC1 is a critical downstream target of c-Myc. Using in vitro and in vivo models of B-cell lymphoma, we show that TFRC1 expression is activated by c-Myc. Chromatin immunoprecipitation experiments reveal that c-Myc directly binds a conserved region of TFRC1. In light of these findings, we sought to determine whether TFRC1 is required for c-Myc-mediated cellular proliferation and cell size control. TFRC1 inhibition decreases cellular proliferation and results in G1 arrest without affecting cell size. Consistent with these findings, expression profiling reveals that TFRC1 depletion alters expression of genes that regulate the cell cycle. Furthermore, enforced TFRC1 expression confers a growth advantage to cells and significantly enhances the rate of c-Myc-mediated tumor formation in vivo. These findings provide a molecular basis for increased TFRC1 expression in human tumors, illuminate the role of TFRC1 in the c-Myc target gene network, and support strategies that target TFRC1 for cancer therapy.


Subject(s)
Antigens, CD/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Transferrin/metabolism , Animals , Antigens, CD/drug effects , Antigens, CD/genetics , Apoptosis/genetics , Carcinogenicity Tests , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Proliferation , Chromatin Immunoprecipitation , Humans , Iron Chelating Agents/pharmacology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Male , Mice , Mice, Nude , Phylogeny , Proto-Oncogene Proteins c-myc/genetics , Rats , Receptors, Transferrin/drug effects , Receptors, Transferrin/genetics , Tumor Cells, Cultured
6.
Br J Haematol ; 140(4): 420-8, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18162125

ABSTRACT

Langerhans cell histiocytosis (LCH) is a clonal, proliferative disorder of phenotypically immature CD1a(+) Langerhans cells (LC). The aetiology of LCH is unknown and data supporting an immune dysregulatory disorder as well as a clonal neoplasm have been reported. Telomere shortening has been associated with cancers and premalignant lesions as well as promoting chromosomal instability. To determine whether LCH LC have altered telomere lengths, we used dual detection of CD1a expression by immunofluorescence and telomere length by fluorescence in situ hybridization of LCH LC and lymphocytes in local, multisystem and systemic LCH and compared these with telomere lengths of LC and lymphocytes in reactive lymph nodes. LCH LC showed significantly shorter telomere lengths than LC from reactive lymph nodes or unaffected skin. Lymphocyte telomere lengths showed similar profiles among the different samples. These data show a significant telomere shortening in LCH LC in all stages of disease involvement compared with LC from reactive lymph nodes, suggesting that LCH may share mechanisms of telomere shortening and survival with clonal preneoplastic disorders and cancer, although an initiating infectious or immune event is still possible.


Subject(s)
Histiocytosis, Langerhans-Cell/genetics , Langerhans Cells/ultrastructure , Telomere/ultrastructure , Antigens, CD1/analysis , Histiocytosis, Langerhans-Cell/pathology , Humans , Image Processing, Computer-Assisted , In Situ Hybridization, Fluorescence , Lymph Nodes/ultrastructure , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Skin/ultrastructure
8.
Mol Cell Biol ; 24(17): 7779-94, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15314183

ABSTRACT

Although Jun upregulation and activation have been established as critical to oncogenesis, the relevant downstream pathways remain incompletely characterized. In this study, we found that c-Jun blocks erythroid differentiation in primary human hematopoietic progenitors and, correspondingly, that Jun factors block transcriptional activation by GATA-1, the central regulator of erythroid differentiation. Mutagenesis of c-Jun suggested that its repression of GATA-1 occurs through a transcriptional mechanism involving activation of downstream genes. We identified the hairy-enhancer-of-split-related factor HERP2 as a novel gene upregulated by c-Jun. HERP2 showed physical interaction with GATA-1 and repressed GATA-1 transcriptional activation. Furthermore, transduction of HERP2 into primary human hematopoietic progenitors inhibited erythroid differentiation. These results thus define a novel regulatory pathway linking the transcription factors c-Jun, HERP2, and GATA-1. Furthermore, these results establish a connection between the Notch signaling pathway, of which the HERP factors are a critical component, and the GATA family, which participates in programming of cellular differentiation.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Erythropoiesis/physiology , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Antigens, CD34 , Basic Helix-Loop-Helix Transcription Factors , Cell Cycle Proteins/genetics , Cell Differentiation , Erythroid-Specific DNA-Binding Factors , GATA1 Transcription Factor , Helix-Loop-Helix Motifs , Hematopoietic Stem Cells/cytology , Humans , K562 Cells , Proto-Oncogene Proteins c-jun/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Transcription, Genetic
9.
Lab Med ; 48(3): 253-257, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28520972

ABSTRACT

Mastocytosis is a clonal neoplasm with the potential to affect various organs within the body. It can range in clinical severity from benign to extremely aggressive. Mastocytosis can be separated into cutaneous, systemic, and leukemic forms, as well as mast-cell sarcoma and extracutaneous mastocytoma. It is most often an acquired condition but can be inherited; the most commonly identified genetic aberrations leading to mastocytosis are activating mutations involving codon 816 of the KIT gene. Herein, we present the case of a 30-year-old Caucasian man with systemic mastocytosis discovered to have a p.Arg634Trp mutation involving KIT. To our knowledge, this mutation has previously only been identified in children with familial urticarial pigmentosa. Ours is the the first case report in the literature of an adult with systemic mastocytosis likely due to a p.Arg634Trp KIT mutation.


Subject(s)
Mastocytosis, Systemic , Mutation/genetics , Proto-Oncogene Proteins c-kit/genetics , Adult , Biopsy , DNA Mutational Analysis , Humans , Male , Skin/pathology , Urticaria Pigmentosa
10.
PLoS One ; 10(7): e0131981, 2015.
Article in English | MEDLINE | ID: mdl-26147501

ABSTRACT

BACKGROUND: The self-renewing ability of HSCs is fundamental for the maintenance of a pool of bone marrow precursors throughout the life of an individual. The genetic mechanisms underlying such a complex process are still poorly understood. RESULTS AND SIGNIFICANCE: Here, we show that constitutive in vivo deletion of miR29ab1 leads to reduced number of HSCs and that miR29ab1 deficient bone marrow cannot repopulate the bone marrow of irradiated mice. An Affymetrix analysis of the miR29ab1 knockout mice identifies key proteins that could be responsible for this phenotype, as DNMT3a and b. Moreover, our findings reveal that whereas miR29b2c knockout mice do not exhibit any spontaneous abnormality, the double knock out--miR29ab1b2c--has marked generalized atrophy, raising the possibility that the two bi-cistrons might cooperate in order to maintain the stem cell number in general, not only limited to the bone marrow.


Subject(s)
Hematopoietic Stem Cells/metabolism , MicroRNAs/genetics , Animals , Atrophy/metabolism , Bone Marrow Transplantation , Hematopoietic Stem Cells/cytology , Liver/cytology , Liver/metabolism , Mice , Mice, Knockout , MicroRNAs/metabolism , Phenotype , Spleen/cytology , Spleen/metabolism
11.
Front Biosci ; 7: d978-85, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11897563

ABSTRACT

While regulating a wide variety of immunologic responses, the precise immunologic functions of CD1d-restricted (NK) T cells are not well defined. Notably, In vitro activation of human NK T cell clones results in the secretion of multiple cytokines important for the recruitment and differentiation of myeloid dendritic cells (DC). Once differentiated, these DC strongly activate NK T cells. In humans, CD1d is expressed by myeloid DC and on tumor cells of this lineage. Another specialized myeloid antigen presenting cell, the epithelioid histiocyte seen in granulomatous inflammation, also expresses CD1d. Because myeloid DC are important regulators of Th1/Th2 T cell responses, cross talk between human NK T cells and myeloid DC would be expected to have significant impact on many immune responses. Consistent with this hypothesis, NK T cells are required for myeloid DC-controlled antitumor responses in mice, and regulate diabetes in nonobese diabetic (NOD) mouse by locally controlling the frequency and function of DC subsets. Thus, regulation of myeloid DC by NK T cells controls both the transition from innate to adaptive immunity and the Th-phenotype of subsequent T cell responses.


Subject(s)
Antigens, CD1/metabolism , Dendritic Cells/immunology , Killer Cells, Natural/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD1d , Cell Differentiation , Diabetes Mellitus, Experimental/immunology , Humans , Mice , Models, Immunological , Myeloid Progenitor Cells/immunology , Neoplasms/immunology
12.
In Vivo ; 28(6): 1077-84, 2014.
Article in English | MEDLINE | ID: mdl-25398802

ABSTRACT

Cytochrome P450 2E1 (CYP2E1) has active roles in bioconversion and biotransformation in humans. Although predominantly present in hepatocytes, CYP2E1 has also been found in hematopoietic stem cells and subtypes of acute myeloid leukemia with unknown clinical significance except for the metabolism of anti-fungal drugs. In the present study, we demonstrated a novel role of CYP2E1 inducing megakaryocyte development in human hematopoietic stem cells and leukemia bipotent K562 cells. CYP2E1 was induced by phorbol-12-myristate-13-acetate in dose-dependent manner in K562 cells as well as in hematopoietic stem cells by thrombopoietin, and ingenol 3,20-dibenzoate (IDB), respectively. Overexpression of CYP2E1 was positively correlated with megakaryocytes and in megakaryocyte maturation. In addition, plasmid-driven expression of CYP2E1 in K562 cells led to morphological transformation of leukemic blasts to pro- and mature megakaryocytes. In contrast, knockout of CYP2E1 by specific interfering RNA diverted these cells to erythroid differentiation. Finally, treatment of K562 cells by a free radical scavenger, N-acetyl L-lysine significantly inhibited CYP2E1 and megakaryocyte differentiation. In summary, our data demonstrated that activation of CYP2E1 and reactive oxygen species signaling promotes megakaryocyte development.


Subject(s)
Cell Differentiation , Cytochrome P-450 CYP2E1/metabolism , Megakaryocytes/cytology , Megakaryocytes/metabolism , Thrombopoiesis , Butadienes/pharmacology , Cell Differentiation/genetics , Cytochrome P-450 CYP2E1/genetics , Erythropoietin/pharmacology , Gene Expression , Gene Expression Regulation/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , K562 Cells , Megakaryocytes/drug effects , Nitriles/pharmacology , Phenotype , Reactive Oxygen Species/metabolism , Thrombopoiesis/genetics
13.
Mol Cytogenet ; 7: 35, 2014.
Article in English | MEDLINE | ID: mdl-25177364

ABSTRACT

Advances in genome-wide molecular cytogenetics allow identification of novel submicroscopic DNA copy number alterations (aCNAs) and copy-neutral loss of heterozygosity (cnLOH) resulting in homozygosity for known gene mutations in myeloid neoplasms. We describe the use of an oligo-SNP array for genomic profiling of aCNA and cnLOH, together with sequence analysis of recurrently mutated genes, in a patient with myelodysplastic syndrome (MDS) presenting with normal karyotype and FISH results. Oligo-SNP array analysis revealed a hemizygous deletion of 896 kb at chromosome 5q31.2, representing the smallest 5q deletion reported to date. The deletion involved multiple genes, including two tumor suppressor candidate genes (CTNNA1 and HSPA9) that are associated with MDS/AML. The SNP-array study also detected 3 segments of somatic cnLOH: one involved the entire long arm of chromosome 4; the second involved the distal half of the long arm of chromosome 7, and the third encompassed the entire chromosome 22 (UPD 22). Sequence analysis revealed mutations in TET2 (4q), EZH2 (7q), ASXL1 (20q11.21), and RUNX1 (21q22.3). Coincidently, TET2 and EZH2 were located at segments of cnLOH resulting in their homozygosity. Loss of heterozygosity affecting these two chromosomes and mutations in TET2 and EZH2 are indicative of a myelodysplastic syndrome with a poor prognosis. Deletion of the tumor suppressor genes CTNNA1 and HSPA9 is also likely to contribute to a poor prognosis. Furthermore, the original cnLOHs in multiple chromosomes and additional cnLOH 14q in the follow-up study suggest genetic evolution of the disease and poor prognosis. This study attests to the fact that some patients with a myelodysplastic syndrome who exhibit a normal karyotype may have underlying genetic abnormalities detectable by chromosomal microarray and/or targeted mutation analyses.

14.
J Pathol Inform ; 4: 30, 2013.
Article in English | MEDLINE | ID: mdl-24392244

ABSTRACT

CONTEXT: Pathologists grade follicular lymphoma (FL) cases by selecting 10, random high power fields (HPFs), counting the number of centroblasts (CBs) in these HPFs under the microscope and then calculating the average CB count for the whole slide. Previous studies have demonstrated that there is high inter-reader variability among pathologists using this methodology in grading. AIMS: The objective of this study was to explore if newly available digital reading technologies can reduce inter-reader variability. SETTINGS AND DESIGN: IN THIS STUDY, WE CONSIDERED THREE DIFFERENT READING CONDITIONS (RCS) IN GRADING FL: (1) Conventional (glass-slide based) to establish the baseline, (2) digital whole slide viewing, (3) digital whole slide viewing with selected HPFs. Six board-certified pathologists from five different institutions read 17 FL slides in these three different RCs. RESULTS: Although there was relative poor consensus in conventional reading, with lack of consensus in 41.2% of cases, which was similar to previously reported studies; we found that digital reading with pre-selected fields improved the inter-reader agreement, with only 5.9% lacking consensus among pathologists. CONCLUSIONS: Digital whole slide RC resulted in the worst concordance among pathologists while digital whole slide reading selected HPFs improved the concordance. Further studies are underway to determine if this performance can be sustained with a larger dataset and our automated HPF and CB detection algorithms can be employed to further improve the concordance.

15.
Cancer Genet ; 205(12): 653-6, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23238285

ABSTRACT

Improvements in chemotherapy and medical support of patients treated with chemotherapy and radiation have led to an ever-increasing number of cancer survivors. Unfortunately, a small fraction of these patients develop secondary hematologic malignancies as a consequence of their exposure to genotoxic anti-cancer regimens. Most of these are myeloid malignancies, therapy-related acute myeloid leukemia (t-AML) or myelodysplasia (t-MDS); however, a small but growing body of literature exists, which describes therapy-related acute lymphoblastic leukemias (t-ALL). Nearly all these cases are reportedly associated with translocations involving chromosome 11q23, the site of the MLL gene. We herein report two cases of ALL occurring after chemotherapy for other malignancies that showed complex karyotypic abnormalities and distinct MLL amplification by fluorescence in situ hybridization analysis. Immunophenotypic analysis showed that both cases expressed a pro-B cell (CD10-) phenotype with aberrant myeloid antigen expression. Although MLL amplification has been reported in therapy-related myeloid disease, to our knowledge this is the first report of MLL amplification occurring in therapy-related B cell ALL.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Gene Amplification/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/chemically induced , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cells, B-Lymphoid/pathology , Aged, 80 and over , Bone Marrow/pathology , Humans , Karyotyping , Male , Middle Aged , Staining and Labeling
16.
PLoS One ; 7(12): e51059, 2012.
Article in English | MEDLINE | ID: mdl-23284657

ABSTRACT

Thrombopoiesis following severe bone marrow injury frequently is delayed, thereby resulting in life-threatening thrombocytopenia for which there are limited treatment options. The reasons for these delays in recovery are not well understood. Protein kinase C (PKC) agonists promote megakaryocyte differentiation in leukemia cell lines and primary cells. However, little is known about the megakaryopoietic effects of PKC agonists on primary CD34+ cells grown in culture or in vivo. Here we present evidence that the novel PKC isoform-selective agonist 3,20 ingenol dibenzoate (IDB) potently stimulates early megakaryopoiesis of human CD34+ cells. In contrast, broad spectrum PKC agonists failed to do so. In vivo, a single intraperitoneal injection of IDB selectively increased platelets in mice without affecting hemoglobin or white counts. Finally, IDB strongly mitigated radiation-induced thrombocytopenia, even when administered 24 hours after irradiation. Our data demonstrate that novel PKC isoform agonists such as IDB may represent a unique therapeutic strategy for accelerating the recovery of platelet counts following severe marrow injury.


Subject(s)
Calcium/metabolism , Diterpenes/pharmacology , Protein Kinase C/metabolism , Thrombopoiesis/drug effects , Animals , Antigens, CD34/metabolism , Biomarkers/metabolism , Blood Platelets/drug effects , Blood Platelets/metabolism , Cell Proliferation/drug effects , Female , Gene Expression Regulation/drug effects , Humans , Isoenzymes/metabolism , Megakaryocytes/cytology , Megakaryocytes/drug effects , Mice , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism
17.
Cancer Cell ; 22(5): 645-55, 2012 Nov 13.
Article in English | MEDLINE | ID: mdl-23153537

ABSTRACT

How inflammation causes cancer is unclear. Interleukin-15 (IL-15) is a pro-inflammatory cytokine elevated in human large granular lymphocyte (LGL) leukemia. Mice overexpressing IL-15 develop LGL leukemia. Here, we show that prolonged in vitro exposure of wild-type (WT) LGL to IL-15 results in Myc-mediated upregulation of aurora kinases, centrosome aberrancies, and aneuploidy. Simultaneously, IL-15 represses miR-29b via induction of Myc/NF-κBp65/Hdac-1, resulting in Dnmt3b overexpression and DNA hypermethylation. All this is validated in human LGL leukemia. Adoptive transfer of WT LGL cultured with IL-15 led to malignant transformation in vivo. Drug targeting that reverses miR-29b repression cures otherwise fatal LGL leukemia. We show how excessive IL-15 initiates cancer and demonstrate effective drug targeting for potential therapy of human LGL leukemia.


Subject(s)
Chromosomal Instability , DNA Methylation , Gene Expression Regulation, Neoplastic , Interleukin-15/genetics , Leukemia, Large Granular Lymphocytic/genetics , Aneuploidy , Animals , Cell Transformation, Neoplastic/genetics , Centrosome/physiology , Chromosome Segregation , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Humans , Interleukin-15/metabolism , Leukemia, Large Granular Lymphocytic/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , DNA Methyltransferase 3B
19.
Blood ; 111(7): 3849-58, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18245664

ABSTRACT

Constitutive activation of FMS-like tyrosine kinase 3 (FLT3) by internal tandem duplication (ITD) mutations is one of the most common molecular alterations known in acute myeloid leukemia (AML). To investigate the role FLT3/ITD mutations play in the development of leukemia, we generated a FLT3/ITD knock-in mouse model by inserting an ITD mutation into the juxtamembrane domain of murine Flt3. FLT3wt/ITD mice developed myeloproliferative disease, characterized by splenomegaly, leukocytosis, and myeloid hypercellularity, which progressed to mortality by 6 to 20 months. Bone marrow (BM) and spleen from FLT3wt/ITD mice had an increased fraction of granulocytes/monocytes and dendritic cells, and a decreased fraction of B-lymphocytes. No sign of acute leukemia was observed over the lifetime of these mice. BM from FLT3wt/ITD mice showed enhanced potential to generate myeloid colonies in vitro. BM from FLT3wt/ITD mice also produced more spleen colonies in the in vivo colony-forming unit (CFU)-spleen assay. In the long-term competitive repopulation assay, BM cells from FLT3wt/ITD mice outgrew the wild-type competitor cells and showed increased myeloid and reduced lymphoid expansion activity. In summary, our data indicate that expression of FLT3/ITD mutations alone is capable of conferring normal hematopoietic stem/progenitor cells (HSPCs) with enhanced myeloid expansion. It also appears to suppress B lymphoid maturation. Additional cooperative events appear to be required to progress to acute leukemia.


Subject(s)
Hematopoietic Stem Cells/metabolism , Mutation , Myelopoiesis , Myeloproliferative Disorders/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Membrane/pathology , Colony-Forming Units Assay , Disease Models, Animal , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Leukocytosis/genetics , Leukocytosis/metabolism , Leukocytosis/pathology , Mice , Mice, Transgenic , Myelopoiesis/genetics , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Protein Structure, Tertiary/genetics , Spleen/metabolism , Spleen/pathology , Splenomegaly/genetics , Splenomegaly/metabolism , Splenomegaly/pathology , Time Factors , fms-Like Tyrosine Kinase 3/genetics
20.
Blood ; 111(3): 1552-9, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17940205

ABSTRACT

The prognostic relevance of FLT3 D835/I836 mutations (FLT3-TKD) in cytogenetically normal acute myeloid leukemia (CN-AML) remains to be established. After excluding patients with FLT3 internal tandem duplications, we compared treatment outcome of 16 de novo CN-AML patients with FLT3-TKD with that of 123 patients with wild-type FLT3 (FLT3-WT), less than 60 years of age and similarly treated on Cancer and Leukemia Group B protocols. All FLT3-TKD(+) patients and 85% of FLT3-WT patients achieved a complete remission (P = .13). Disease-free survival (DFS) of FLT3-TKD(+) patients was worse than DFS of FLT3-WT patients (P = .01; estimated 3-year DFS rates, 31% vs 60%, respectively). In a multivariable analysis, FLT3-TKD was associated with worse DFS (P = .02) independent of NPM1 status and percentage of bone marrow blasts. To gain further biologic insights, a gene-expression signature differentiating FLT3-TKD(+) from FLT3-WT patients was identified. The signature (333 probe sets) included overexpression of VNN1, C3AR1, PTPN6, and multiple other genes involved in monocarboxylate transport activity, and underexpression of genes involved in signal transduction regulation. These associations with outcome, other prognostic markers, and the elucidated expression signature enhance our understanding of FLT3-TKD-associated biology and may lead to development of novel therapies that improve clinical outcome of CN-AML patients with FLT3-TKD.


Subject(s)
Gene Expression Regulation, Leukemic/genetics , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/pathology , fms-Like Tyrosine Kinase 3/metabolism , Adult , Aspartic Acid/genetics , Aspartic Acid/metabolism , Cytogenetics , Disease-Free Survival , Female , Humans , Isoleucine/genetics , Isoleucine/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Male , Middle Aged , Mutation/genetics , Nucleophosmin , Prognosis , Treatment Outcome , fms-Like Tyrosine Kinase 3/genetics
SELECTION OF CITATIONS
SEARCH DETAIL