Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Ann Oncol ; 34(9): 734-771, 2023 09.
Article in English | MEDLINE | ID: mdl-37343663

ABSTRACT

BACKGROUND: The European Society for Medical Oncology (ESMO)-Magnitude of Clinical Benefit Scale (MCBS) has been accepted as a robust tool to evaluate the magnitude of clinical benefit reported in trials for oncological therapies. However, the ESMO-MCBS hitherto has only been validated for solid tumours. With the rapid development of novel therapies for haematological malignancies, we aimed to develop an ESMO-MCBS version that is specifically designed and validated for haematological malignancies. METHODS: ESMO and the European Hematology Association (EHA) initiated a collaboration to develop a version for haematological malignancies (ESMO-MCBS:H). The process incorporated five landmarks: field testing of the ESMO-MCBS version 1.1 (v1.1) to identify shortcomings specific to haematological diseases, drafting of the ESMO-MCBS:H forms, peer review and revision of the draft based on re-scoring (resulting in a second draft), assessment of reasonableness of the scores generated, final review and approval by ESMO and EHA including executive boards. RESULTS: Based on the field testing results of 80 haematological trials and extensive review for feasibility and reasonableness, five amendments to ESMO-MCBS were incorporated in the ESMO-MCBS:H addressing the identified shortcomings. These concerned mainly clinical trial endpoints that differ in haematology versus solid oncology and the very indolent nature of nevertheless incurable diseases such as follicular lymphoma, which hampers presentation of mature data. In addition, general changes incorporated in the draft version of the ESMO-MCBS v2 were included, and specific forms for haematological malignancies generated. Here we present the final approved forms of the ESMO-MCBS:H, including instructions. CONCLUSION: The haematology-specific version ESMO-MCBS:H allows now full applicability of the scale for evaluating the magnitude of clinical benefit derived from clinical studies in haematological malignancies.


Subject(s)
Antineoplastic Agents , Hematologic Neoplasms , Lymphoma, Follicular , Neoplasms , Humans , Neoplasms/drug therapy , Medical Oncology , Hematologic Neoplasms/therapy , Societies, Medical , Lymphoma, Follicular/drug therapy , Antineoplastic Agents/therapeutic use
2.
Leukemia ; 32(2): 263-272, 2018 02.
Article in English | MEDLINE | ID: mdl-28674423

ABSTRACT

Prognostic gene expression signatures have been proposed as clinical tools to clarify therapeutic options in acute myeloid leukemia (AML). However, these signatures rely on measuring large numbers of genes and often perform poorly when applied to independent cohorts or those with older patients. Long intergenic non-coding RNAs (lincRNAs) are emerging as important regulators of cell identity and oncogenesis, but knowledge of their utility as prognostic markers in AML is limited. Here we analyze transcriptomic data from multiple cohorts of clinically annotated AML patients and report that (i) microarrays designed for coding gene expression can be repurposed to yield robust lincRNA expression data, (ii) some lincRNA genes are located in close proximity to hematopoietic coding genes and show strong expression correlations in AML, (iii) lincRNA gene expression patterns distinguish cytogenetic and molecular subtypes of AML, (iv) lincRNA signatures composed of three or four genes are independent predictors of clinical outcome and further dichotomize survival in European Leukemia Net (ELN) risk groups and (v) an analytical tool based on logistic regression analysis of quantitative PCR measurement of four lincRNA genes (LINC4) can be used to determine risk in AML.


Subject(s)
Leukemia, Myeloid, Acute/genetics , RNA, Long Noncoding/genetics , Transcriptome/genetics , Adolescent , Adult , Female , Gene Expression Profiling/methods , Humans , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Prognosis , Risk Assessment , Risk Factors , Young Adult
3.
Oncogene ; 35(3): 279-89, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-25893291

ABSTRACT

Growing evidence links abnormal epigenetic control to the development of hematological malignancies. Accordingly, inhibition of epigenetic regulators is emerging as a promising therapeutic strategy. The acetylation status of lysine residues in histone tails is one of a number of epigenetic post-translational modifications that alter DNA-templated processes, such as transcription, to facilitate malignant transformation. Although histone deacetylases are already being clinically targeted, the role of histone lysine acetyltransferases (KAT) in malignancy is less well characterized. We chose to study this question in the context of acute myeloid leukemia (AML), where, using in vitro and in vivo genetic ablation and knockdown experiments in murine models, we demonstrate a role for the epigenetic regulators CBP and p300 in the induction and maintenance of AML. Furthermore, using selective small molecule inhibitors of their lysine acetyltransferase activity, we validate CBP/p300 as therapeutic targets in vitro across a wide range of human AML subtypes. We proceed to show that growth retardation occurs through the induction of transcriptional changes that induce apoptosis and cell-cycle arrest in leukemia cells and finally demonstrate the efficacy of the KAT inhibitors in decreasing clonogenic growth of primary AML patient samples. Taken together, these data suggest that CBP/p300 are promising therapeutic targets across multiple subtypes in AML.


Subject(s)
E1A-Associated p300 Protein/genetics , Epigenesis, Genetic , Leukemia, Myeloid, Acute/genetics , Peptide Fragments/genetics , Sialoglycoproteins/genetics , Animals , Apoptosis/drug effects , Benzoates/administration & dosage , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , E1A-Associated p300 Protein/biosynthesis , Female , Gene Expression Regulation, Leukemic/drug effects , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Male , Mice , Nitrobenzenes , Peptide Fragments/biosynthesis , Pyrazoles/administration & dosage , Pyrazolones , Sialoglycoproteins/biosynthesis
4.
Leukemia ; 28(2): 311-20, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24220271

ABSTRACT

Recent evidence suggests that inhibition of bromodomain and extra-terminal (BET) epigenetic readers may have clinical utility against acute myeloid leukemia (AML). Here we validate this hypothesis, demonstrating the efficacy of the BET inhibitor I-BET151 across a variety of AML subtypes driven by disparate mutations. We demonstrate that a common 'core' transcriptional program, which is HOX gene independent, is downregulated in AML and underlies sensitivity to I-BET treatment. This program is enriched for genes that contain 'super-enhancers', recently described regulatory elements postulated to control key oncogenic driver genes. Moreover, our program can independently classify AML patients into distinct cytogenetic and molecular subgroups, suggesting that it contains biomarkers of sensitivity and response. We focus AML with mutations of the Nucleophosmin gene (NPM1) and show evidence to suggest that wild-type NPM1 has an inhibitory influence on BRD4 that is relieved upon NPM1c mutation and cytosplasmic dislocation. This leads to the upregulation of the core transcriptional program facilitating leukemia development. This program is abrogated by I-BET therapy and by nuclear restoration of NPM1. Finally, we demonstrate the efficacy of I-BET151 in a unique murine model and in primary patient samples of NPM1c AML. Taken together, our data support the use of BET inhibitors in clinical trials in AML.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Activation , Animals , Benzodiazepines/administration & dosage , Benzodiazepines/pharmacology , Cell Cycle Proteins , Cell Line, Tumor , Disease Models, Animal , Drug Evaluation, Preclinical , Gene Expression Profiling , Gene Expression Regulation, Leukemic/drug effects , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Mice , Nucleophosmin , Xenograft Model Antitumor Assays
5.
N Engl J Med ; 369(25): 2391-2405, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-24325359

ABSTRACT

BACKGROUND: Somatic mutations in the Janus kinase 2 gene (JAK2) occur in many myeloproliferative neoplasms, but the molecular pathogenesis of myeloproliferative neoplasms with nonmutated JAK2 is obscure, and the diagnosis of these neoplasms remains a challenge. METHODS: We performed exome sequencing of samples obtained from 151 patients with myeloproliferative neoplasms. The mutation status of the gene encoding calreticulin (CALR) was assessed in an additional 1345 hematologic cancers, 1517 other cancers, and 550 controls. We established phylogenetic trees using hematopoietic colonies. We assessed calreticulin subcellular localization using immunofluorescence and flow cytometry. RESULTS: Exome sequencing identified 1498 mutations in 151 patients, with medians of 6.5, 6.5, and 13.0 mutations per patient in samples of polycythemia vera, essential thrombocythemia, and myelofibrosis, respectively. Somatic CALR mutations were found in 70 to 84% of samples of myeloproliferative neoplasms with nonmutated JAK2, in 8% of myelodysplasia samples, in occasional samples of other myeloid cancers, and in none of the other cancers. A total of 148 CALR mutations were identified with 19 distinct variants. Mutations were located in exon 9 and generated a +1 base-pair frameshift, which would result in a mutant protein with a novel C-terminal. Mutant calreticulin was observed in the endoplasmic reticulum without increased cell-surface or Golgi accumulation. Patients with myeloproliferative neoplasms carrying CALR mutations presented with higher platelet counts and lower hemoglobin levels than patients with mutated JAK2. Mutation of CALR was detected in hematopoietic stem and progenitor cells. Clonal analyses showed CALR mutations in the earliest phylogenetic node, a finding consistent with its role as an initiating mutation in some patients. CONCLUSIONS: Somatic mutations in the endoplasmic reticulum chaperone CALR were found in a majority of patients with myeloproliferative neoplasms with nonmutated JAK2. (Funded by the Kay Kendall Leukaemia Fund and others.).


Subject(s)
Calreticulin/genetics , Mutation , Myelodysplastic Syndromes/genetics , Primary Myelofibrosis/genetics , Thrombocythemia, Essential/genetics , Amino Acid Sequence , Bone Marrow Diseases/genetics , Calreticulin/analysis , Exons , Humans , Janus Kinase 2/genetics , Leukemia, Myeloid/genetics , Molecular Sequence Data , Neoplasms/genetics , Polymerase Chain Reaction , Sequence Analysis, DNA
6.
Oncogene ; 32(48): 5471-80, 2013 Nov 28.
Article in English | MEDLINE | ID: mdl-23708655

ABSTRACT

The Lim Domain Only 2 (LMO2) leukaemia oncogene encodes an LIM domain transcriptional cofactor required for early haematopoiesis. During embryogenesis, LMO2 is also expressed in developing tail and limb buds, an expression pattern we now show to be recapitulated in transgenic mice by an enhancer in LMO2 intron 4. Limb bud expression depended on a cluster of HOX binding sites, while posterior tail expression required the HOX sites and two E-boxes. Given the importance of both LMO2 and HOX genes in acute leukaemias, we further demonstrated that the regulatory hierarchy of HOX control of LMO2 is activated in leukaemia mouse models as well as in patient samples. Moreover, Lmo2 knock-down impaired the growth of leukaemic cells, and high LMO2 expression at diagnosis correlated with poor survival in cytogenetically normal AML patients. Taken together, these results establish a regulatory hierarchy of HOX control of LMO2 in normal development, which can be resurrected during leukaemia development. Redeployment of embryonic regulatory hierarchies in an aberrant context is likely to be relevant in human pathologies beyond the specific example of ectopic activation of LMO2.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Gene Expression Regulation, Developmental/genetics , Genes, Homeobox , LIM Domain Proteins/genetics , Mesoderm/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/embryology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins/genetics , Adaptor Proteins, Signal Transducing/deficiency , Animals , Base Sequence , Cell Line, Tumor , Cell Proliferation , Chromatin/genetics , Conserved Sequence , E-Box Elements , Extremities/embryology , Gene Knockdown Techniques , Homeodomain Proteins/metabolism , Humans , Introns/genetics , LIM Domain Proteins/deficiency , Mice , Molecular Sequence Data , Phenotype , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins/deficiency , Transcriptional Activation/genetics
7.
Oncogene ; 29(43): 5796-808, 2010 Oct 28.
Article in English | MEDLINE | ID: mdl-20676125

ABSTRACT

The T-cell oncogene Lim-only 2 (LMO2) critically influences both normal and malignant haematopoiesis. LMO2 is not normally expressed in T cells, yet ectopic expression is seen in the majority of T-acute lymphoblastic leukaemia (T-ALL) patients with specific translocations involving LMO2 in only a subset of these patients. Ectopic lmo2 expression in thymocytes of transgenic mice causes T-ALL, and retroviral vector integration into the LMO2 locus was implicated in the development of clonal T-cell disease in patients undergoing gene therapy. Using array-based chromatin immunoprecipitation, we now demonstrate that in contrast to B-acute lymphoblastic leukaemia, human T-ALL samples largely use promoter elements with little influence from distal enhancers. Active LMO2 promoter elements in T-ALL included a previously unrecognized third promoter, which we demonstrate to be active in cell lines, primary T-ALL patients and transgenic mice. The ETS factors ERG and FLI1 previously implicated in lmo2-dependent mouse models of T-ALL bind to the novel LMO2 promoter in human T-ALL samples, while in return LMO2 binds to blood stem/progenitor enhancers in the FLI1 and ERG gene loci. Moreover, LMO2, ERG and FLI1 all regulate the +1 enhancer of HHEX/PRH, which was recently implicated as a key mediator of early progenitor expansion in LMO2-driven T-ALL. Our data therefore suggest that a self-sustaining triad of LMO2/ERG/FLI1 stabilizes the expression of important mediators of the leukaemic phenotype such as HHEX/PRH.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Metalloproteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Promoter Regions, Genetic/genetics , Adaptor Proteins, Signal Transducing , Animals , Chromatin Immunoprecipitation , Gene Expression , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , LIM Domain Proteins , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Proteins , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription, Genetic , Transcriptional Regulator ERG
9.
Best Pract Res Clin Haematol ; 14(3): 531-51, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11640868

ABSTRACT

The myeloproliferative disorders (MPDs) are a group of pre-leukaemic disorders characterized by proliferation of one or more lineages of the myelo-erythroid series. Unlike the Philadelphia chromosome in chronic myeloid leukaemia, there is no pathognomonic chromosomal abnormality associated with the MPDs. Chromosomal abnormalities are seen in 30-40% of patients with polycythaemia vera (PV) and idiopathic myelofibrosis (IMF) and seem to indicate a poor prognosis. On the other hand, chromosomal abnormalities are rare in essential thrombocythaemia. Consistent acquired changes seen at diagnosis include deletion of the long arm of chromosome 20, del(13q), trisomy 8 and 9 and duplication of parts of 1q. Furthermore del(20q), trisomy 8 and dupl(lq) all arise in multipotent progenitor cells. Molecular mapping of 20q deletions and, to some extent, 13q deletions has identified a number of candidate target genes, although no mutations have yet been found. Finally, translocations associated with the rare 8p11 myeloproliferative syndrome and other atypical myeloproliferative disorders have permitted the identification of a number of novel fusion proteins involving fibroblast growth factor receptor-1.


Subject(s)
Myeloproliferative Disorders/genetics , Chromosome Aberrations/classification , Chromosome Mapping , Cytogenetic Analysis , Humans , Models, Genetic , Myeloproliferative Disorders/classification , Myeloproliferative Disorders/etiology , Oncogene Proteins, Fusion/genetics
10.
Best Pract Res Clin Haematol ; 14(3): 553-71, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11640869

ABSTRACT

The standard Philadelphia (Ph) translocation t(9;22), its variants and a proportion of Ph-negative cases are positive for the BCR-ABL fusion gene, as determined by molecular analysis. Extensive deletions of chromosome 9 and 22 derived sequences around the translocation breakpoints on the derivative 9 are seen in 10-30% of patients at diagnosis and may confer a worse prognosis. Additional cytogenetic changes can occur in the few months before or during disease progression and are often specific for blast morphology; however, the molecular basis of the most common additional cytogenetic abnormalities is largely unknown. Cytogenetics is important for monitoring patient response to treatment but is increasingly being replaced by the more sensitive and less invasive techniques of RT-PCR and FISH.


Subject(s)
Cytogenetic Analysis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Antineoplastic Agents , Bone Marrow Transplantation , Chromosome Aberrations , Disease Progression , Fusion Proteins, bcr-abl/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Philadelphia Chromosome , Prognosis
11.
Blood ; 98(6): 1732-8, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11535505

ABSTRACT

Chronic myeloid leukemia (CML) is characterized by formation of the BCR-ABL fusion gene, usually as a consequence of the Philadelphia (Ph) translocation between chromosomes 9 and 22. Large deletions on the derivative chromosome 9 have recently been reported, but it was unclear whether deletions arose during disease progression or at the time of the Ph translocation. Fluorescence in situ hybridization (FISH) analysis was used to assess the deletion status of 253 patients with CML. The strength of deletion status as a prognostic indicator was then compared to the Sokal and Hasford scoring systems. The frequency of deletions was similar at diagnosis and after disease progression but was significantly increased in patients with variant Ph translocations. In patients with a deletion, all Ph(+) metaphases carried the deletion. The median survival of patients with and without deletions was 38 months and 88 months, respectively (P =.0001). By contrast the survival difference between Sokal or Hasford high-risk and non-high-risk patients was of only borderline significance (P =.057 and P =.034). The results indicate that deletions occur at the time of the Ph translocation. An apparently simple reciprocal translocation may therefore result in considerable genetic heterogeneity ab initio, a concept that is likely to apply to other malignancies associated with translocations. Deletion status is also a powerful and independent prognostic factor for patients with CML. The prognostic significance of deletion status should now be studied prospectively and, if confirmed, should be incorporated into management decisions and the analysis of clinical trials.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 9 , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Philadelphia Chromosome , Adult , Chromosomes, Human, Pair 9/ultrastructure , Female , Humans , In Situ Hybridization, Fluorescence , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Male , Middle Aged , Prognosis , Survival Rate
12.
Br J Haematol ; 113(2): 439-42, 2001 May.
Article in English | MEDLINE | ID: mdl-11380413

ABSTRACT

A range of fluorescent in situ hybridization techniques have been used to reveal hidden variant Philadelphia translocations in two cases of Ph-positive chronic-phase chronic myeloid leukaemia. In one patient, a highly complex variant Ph translocation affecting four chromosomes had resulted in the formation of structures with the appearance of i(17q) and +8. Misinterpretation of these karyotypes has direct clinical relevance. Our findings illustrate that even established cytogenetic abnormalities may contain cryptic abnormalities beyond the resolution of conventional cytogenetic methods.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Philadelphia Chromosome , Translocation, Genetic , Aged , Chromosome Banding , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 22 , Chromosomes, Human, Pair 9 , Diagnosis, Differential , Female , Humans , In Situ Hybridization, Fluorescence , Metaphase
13.
Br J Haematol ; 115(4): 872-80, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11843821

ABSTRACT

Polycythaemia vera (PV) is a myeloproliferative disorder (MPD) thought to result from transformation of a haemopoietic stem cell. Transforming growth factor beta1 (TGF-beta1) is a negative regulator of haemopoietic stem cells, an effect mediated by direct binding to TGF-beta receptor II (TGF-beta RII). Reduced levels of TGF-beta RII mRNA or protein have been reported in several MPDs including PV, suggesting a role for TGF-beta RII in PV. No mutational analysis of the TGF-beta RII gene has yet been performed in PV. To investigate whether genetic or epigenetic alteration of the TGF-beta RII gene contributes to the pathogenesis of PV, we performed mutation and methylation analysis in 15 PV patients. The promoter, all seven exons and all intron/exon junctions were studied using single-strand conformation polymorphism (SSCP) and heteroduplex analysis (HA). In total, three single nucleotide polymorphisms (SNPs) were identified. These were located in the promoter, intron 2 and exon 5. No acquired mutations were detected in any patient sample. We also present a novel method, termed methylation-specific strand extension (MSSE), for the detection of methylated CpG dinucleotides. The combination of bisulphite modification and MSSE permits rapid analysis of the methylation status of CpG dinucleotides in multiple samples. We analysed the methylation status of the promoter and of a CpG island within exon 1 in 15 PV patients. No aberrant methylation was detected in either of these regions. These data demonstrate that neither mutation nor abnormal methylation of the TGF-beta RII gene is associated with the pathogenesis of PV. Furthermore, MSSE is a rapid and robust approach for assessing the methylation status of a given genomic region.


Subject(s)
CpG Islands , Polycythemia Vera/genetics , Promoter Regions, Genetic , Receptors, Transforming Growth Factor beta/genetics , Adult , Aged , Aged, 80 and over , DNA Methylation , Heteroduplex Analysis , Humans , Middle Aged , Polymorphism, Single-Stranded Conformational , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II
14.
Br J Haematol ; 110(4): 839-46, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11054066

ABSTRACT

We have used interphase fluorescence in situ hybridization (IFISH) to detect trisomy 8, trisomy 9 and 20q deletion in circulating granulocytes from patients with polycythaemia vera (PV). Out of 64 PV patients, 15 (23%) exhibited an abnormality. Two patients had trisomy 9, three had trisomy 8 and 10 patients had hemizygous deletion of D20S108 (a locus in the 20q common deleted region). Aberrant nuclei ranged from 10% to 80% in these 15 cases. There was no correlation between the presence of a marker and sex, age, interval between presentation and IFISH analysis, neutrophil or platelet count or therapy. Conventional marrow cytogenetic karyotype results were available in 23 cases and there was concurrence between these and blood IFISH in 16 cases (13 normal and three with 20q/D20S108 deletion by both methods). Three patients with D20S108 deletion by IFISH were normal by previous marrow cytogenetic testing and four cases with 20q deletion by previous marrow cytogenetics had normal blood granulocytes according to IFISH. Thus, we confirm that trisomies 8 and 9 and deletion of 20q are diagnostically useful markers of PV. IFISH analysis of blood granulocytes is a practical method for detecting these markers, but as an adjunct to, not as a substitute for, conventional marrow cytogenetics.


Subject(s)
Chromosomes, Human, Pair 8 , Chromosomes, Human, Pair 9 , Gene Deletion , Polycythemia Vera/genetics , Trisomy , Adult , Aged , Aged, 80 and over , Cytogenetic Analysis , Female , Genetic Markers , Granulocytes , Humans , In Situ Hybridization, Fluorescence , Interphase , Male , Middle Aged
15.
Oncogene ; 19(34): 3902-13, 2000 Aug 10.
Article in English | MEDLINE | ID: mdl-10952764

ABSTRACT

Deletion of the long arm of chromosome 20 represents the most common chromosomal abnormality associated with the myeloproliferative disorders (MPDs) and is also found in other myeloid malignancies including myelodysplastic syndromes (MDS) and acute myeloid leukaemia (AML). Previous studies have identified a common deleted region (CDR) spanning approximately 8 Mb. We have now used G-banding, FISH or microsatellite PCR to analyse 113 patients with a 20q deletion associated with a myeloid malignancy. Our results define a new MPD CDR of 2.7 Mb, an MDS/AML CDR of 2.6 Mb and a combined 'myeloid' CDR of 1.7 Mb. We have also constructed the most detailed physical map of this region to date--a bacterial clone map spanning 5 Mb of the chromosome which contains 456 bacterial clones and 202 DNA markers. Fifty-one expressed sequences were localized within this contig of which 37 lie within the MPD CDR and 20 within the MDS/AML CDR. Of the 16 expressed sequences (six genes and 10 unique ESTs) within the 'myeloid' CDR, five were expressed in both normal bone marrow and purified CD34 positive cells. These data identify a set of genes which are both positional and expression candidates for the target gene(s) on 20q.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 20 , Contig Mapping , Myelodysplastic Syndromes/genetics , Myeloproliferative Disorders/genetics , Antigens, CD34/metabolism , Bone Marrow Cells/physiology , Chromosome Banding , Chromosomes, Bacterial , Expressed Sequence Tags , Gene Expression Profiling , Humans , Leukemia, Myeloid/genetics , Reverse Transcriptase Polymerase Chain Reaction
17.
Blood Rev ; 10(4): 220-30, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9012919
18.
Brain ; 119 ( Pt 4): 1277-87, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8813290

ABSTRACT

A general impairment of cognitive performance occurs during acute insulin-induced hypoglycaemia, but little objective evidence is available for disruption of more specific cognitive processes. The effect of controlled hypoglycaemia on the early stages of visual information processing and contrast sensitivity was examined in a homogeneous group of 20 nondiabetic human subjects. Hypoglycaemia caused a significant disruption in general cognitive performance as assessed by a digit symbol task (P < 0.001) and the trail making B task (P < 0.05). Hypoglycaemia also produced a highly significant deterioration in performance on all of the visual information processing tasks, namely inspection time (IT) (P = 0.01), visual change detection (VCD) (P < 0.005) and visual movement detection (VMD) (P < 0.005). A significant deterioration in contrast sensitivity was observed during hypoglycaemia (P < 0.005). In contrast, no significant effect of hypoglycaemia was demonstrated on standard clinical measures of visual acuity or stereoscopic vision. Thus, although hypoglycaemia caused no detectable deterioration in visual acuity as measured by Snellen-type tests, a marked deterioration occurred in the speed of visual information processing and in contrast sensitivity. As many decisions are made under conditions of limited perceptual time and low visual contrast (e.g. when driving), the disruptive effect of moderate insulin-induced hypoglycaemia on visual perception will have important practical implications in diabetic humans exposed to this metabolic stress. The present results are congruent with other evidence which shows that the early stages of visual information processing are susceptible to deterioration by general cerebral insults.


Subject(s)
Hypoglycemia/physiopathology , Visual Acuity/physiology , Visual Pathways/physiology , Adult , Blood Glucose/analysis , Female , Humans , Male , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...