Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
Add more filters

Publication year range
1.
Hum Immunol ; 82(10): 758-766, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34353675

ABSTRACT

In Italy, an HLA-matched unrelated donor is currently the primary donor when a HLA matched sibling is not found for allogeneic haematopoietic stem cell transplantation (HSCT). Better outcomes for transplantation require optimal matching between donor and recipient at least at the HLA-A, -B, -C, and -DRB1 loci; therefore, the availability of HLA-matched unrelated donors is important. The enormous HLA polymorphism has always necessitated registries with a large number of individuals in order to be able to provide well-matched donors to a substantial percentage of patients. In order to increase the efficiency of the Italian Bone Marrow Donor Registry (IBMDR) in providing Italian patients with a suitable donor, the probability of finding an HLA-A, -B, -C, and -DRB1 allele-matched (8/8) or a single mismatch unrelated donor (7/8) was estimated in this study according to IBMDR size. Using a biostatistical approach based on HLA haplotype frequencies of more than 100,000 Italian donors enrolled in the IBMDR and HLA-typed at high-resolution level, the probability of finding an 8/8 HLA-matched donor was 23.8%; 33.4%; and 41.4% in simulated registry sizes of 200,000; 500,000; and 1,000,000 donors; respectively. More than 2 million recruited donors are needed to increase the likelihood of identifying an HLA 8/8 matched donor for 50% of Italian patients. If one single mismatch at HLA I class loci was accepted, the probability of finding a 7/8 HLA-matched donor was 62.8%; 73.7%; and 80.3% in 200,000 donors; 500,000; and 1,000,000 donors; respectively. Using the regional haplotype frequencies of IBMDR donors, the probability of recruiting a donor with a new HLA phenotype, in the different Italian regions, was also calculated. Our findings are highly relevant in estimating the optimal size of the national registry, in planning a cost-effective strategy for donor recruitment in Italy, and determining the regional priority setting of recruitment activity in order to increase the phenotypic variability of IBMDR as well as its efficiency.


Subject(s)
Alleles , Genetics, Population , HLA Antigens/genetics , Haplotypes , Registries , Tissue Donors , Algorithms , Gene Frequency , Hematopoietic Stem Cell Transplantation , Histocompatibility Testing/methods , Humans , Italy , Likelihood Functions , Models, Theoretical , Probability , Unrelated Donors
2.
J Cell Biol ; 150(5): 939-48, 2000 Sep 04.
Article in English | MEDLINE | ID: mdl-10973986

ABSTRACT

Dentato-rubral and pallido-luysian atrophy (DRPLA) is one of the family of neurodegenerative diseases caused by expansion of a polyglutamine tract. The drpla gene product, atrophin-1, is widely expressed, has no known function or activity, and is found in both the nuclear and cytoplasmic compartments of neurons. Truncated fragments of atrophin-1 accumulate in neuronal nuclei in a transgenic mouse model of DRPLA, and may underlie the disease phenotype. Using the yeast two-hybrid system, we identified ETO/MTG8, a component of nuclear receptor corepressor complexes, as an atrophin-1-interacting protein. When cotransfected into Neuro-2a cells, atrophin-1 and ETO/MTG8 colocalize in discrete nuclear structures that contain endogenous mSin3A and histone deacetylases. These structures are sodium dodecyl sulfate-soluble and associated with the nuclear matrix. Cotransfection of ETO/MTG8 with atrophin-1 recruits atrophin-1 to the nuclear matrix, while atrophin-1 and ETO/MTG8 cofractionate in nuclear matrix preparations from brains of DRPLA transgenic mice. Furthermore, in a cell transfection-based assay, atrophin-1 represses transcription. Together, these results suggest that atrophin-1 associates with nuclear receptor corepressor complexes and is involved in transcriptional regulation. Emerging links between disease-associated polyglutamine proteins, nuclear receptors, translocation-leukemia proteins, and the nuclear matrix may have important repercussions for the pathobiology of this family of neurodegenerative disorders.


Subject(s)
DNA-Binding Proteins/metabolism , Globus Pallidus/pathology , Nerve Tissue Proteins/metabolism , Nuclear Matrix/metabolism , Proto-Oncogene Proteins , Transcription Factors/metabolism , Transcription, Genetic , Animals , Atrophy , Cloning, Molecular , Histone Deacetylases/analysis , Histone Deacetylases/metabolism , Humans , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nuclear Matrix/ultrastructure , Peptide Fragments/metabolism , RUNX1 Translocation Partner 1 Protein , Recombinant Proteins/metabolism , Repressor Proteins/analysis , Repressor Proteins/metabolism , Sin3 Histone Deacetylase and Corepressor Complex , Transfection , Tumor Cells, Cultured
3.
Science ; 231(4736): 379-82, 1986 Jan 24.
Article in English | MEDLINE | ID: mdl-3941901

ABSTRACT

Human probes identifying the cellular homologs of the v-ets gene, Hu-ets-1 and Hu-ets-2, and two panels of rodent-human cell hybrids were used to study specific translocations occurring in acute leukemias. The human ets-1 gene was found to translocate from chromosome 11 to 4 in the t(4;11)(q21;23), a translocation characteristic of a subtype of leukemia that represents the expansion of a myeloid/lymphoid precursor cell. Similarly, the human ets-2 gene was found to translocate from chromosome 21 to chromosome 8 in the t(8;21)(q22;q22), a nonrandom translocation commonly found in patients with acute myeloid leukemia with morphology M2 (AML-M2). Both translocations are associated with expression different from the expression in normal lymphoid cells of ets genes, raising the possibility that these genes play a role in the pathogenesis of these leukemias.


Subject(s)
Leukemia/genetics , Oncogenes , Translocation, Genetic , Animals , Cell Line , Chromosomes, Human, 21-22 and Y , Chromosomes, Human, 6-12 and X , Cricetinae , Cricetulus , Humans , Hybrid Cells , Nucleic Acid Hybridization
4.
Bone Marrow Transplant ; 41 Suppl 2: S75-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18545249

ABSTRACT

During the last 10 years, the number of alternative Haematopoietic stem cell transplantations (HSCTs) performed on children in Europe has increased significantly and has reached 61% of the allografts. In this paper, we provide practical guidelines to help define an algorithm for the treatment of children relapsing during or after first-line chemotherapy for ALL and lacking a matched sibling donor. A simultaneous search for an unrelated donor and for a cord blood unit should be started. This study focuses mainly on the effects of some factors on survival in an effort to highlight the influence that these factors have on our choices. Matching the patient for HLA-A, -B, -C and -DRB1 alleles remains the top priority: a single HLA class I or II allele mismatch has no influence on survival, while multiple mismatching for more than one class I allele and simultaneous disparities in class I and II alleles increase mortality. The impact of additional mismatches for HLA-DQ and -DP loci on survival is still controversial. Young donor age is the most important factor that has a significant effect on better survival from among several other factors, including CMV sero-status, gender and ABO. An 18- to 30-year-old, 8/8 allele-matched donor (excluding allele matching at DQB1) or for many teams 10/10 allele-matched donor; or a 4 out of 6 (considering Ag HLA-A, -B and allelic typing of DRB1) CB unit containing more than 3.0 x 10(7) nuclear cells is considered by most institutions. The choice should be made on the basis of urgency. If a donor or a CB unit is not found within an appropriate time frame, generally less than 3 months after obtention of remission, haploidentical HSCT should be offered. Some institutions consider haploidentical HSCT the second therapeutic option when a matched donor is not available.


Subject(s)
Hematopoietic Stem Cell Transplantation , Neoplasm Recurrence, Local/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Tissue Donors , Algorithms , Child , Cord Blood Stem Cell Transplantation , Haplotypes , Hematopoietic Stem Cell Transplantation/methods , Histocompatibility Testing , Humans , Remission Induction , Transplantation, Homologous
5.
Bone Marrow Transplant ; 42(1): 9-14, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18362904

ABSTRACT

The ability to identify unrelated haematopoietic stem cell donors in one country for recipients in another country requires cooperation and standardization in many areas. The donor assessment and testing are very important issues affecting quality and safety of donation. This special report details the World Marrow Donor Association's recommended procedures regarding the medical evaluation of donors, with the intent to protect the volunteer from the risk to damage his health and to offer the recipient the appropriate quality of stem cells. This document describes criteria for permanent or temporary deferral, guidelines for risk evaluation of infectious disease, examples of conditions requiring assessment and questionnaires designed to elicit relevant information about a donor's medical history and general health.


Subject(s)
Donor Selection/standards , Hematopoietic Stem Cell Transplantation , Living Donors , Registries , Tissue and Organ Procurement/standards , Donor Selection/methods , Health Surveys , Physical Examination , Tissue Banks/standards , Transplantation, Homologous
6.
HLA ; 2018 May 17.
Article in English | MEDLINE | ID: mdl-29770606

ABSTRACT

HLA-C*02:10:03 identified in a Venezuelan patient and characterized using next generation sequencing.

7.
Oncogene ; 25(9): 1400-7, 2006 Mar 02.
Article in English | MEDLINE | ID: mdl-16261163

ABSTRACT

Retinoic acid (RA) is a signaling molecule that plays a pivotal role in major cellular processes and vertebrate development. RA action is mediated by specialized transcription factors, the nuclear RA receptors (RARs), which regulate the transcription of genes containing a RA-responsive element (RARE). Here we demonstrate that the genes for the RA-receptor RARbeta2 and the cytochrome P450 RA-specific hydrolase Cyp26a1 involved in RA catabolism are coordinately regulated by RA. We found that both RARbeta2 and Cyp26a1 genes are epigenetically silenced in the absence of DNA methylation in RAC65, a P19 embryocarcinoma cell line derivative carrying a dominant-negative RARalpha mutant and resistant to the growth-inhibitory and differentiation effects of RA. In response to RA, RARbeta2 transcription is epigenetically regulated by RARalpha. Similarly, we found that Cyp26a1 transcription is epigenetically regulated by RARbeta2. Knocking down RARbeta2 transcription by RNA interference in wild-type P19 cells, with an intact RARalpha, induced Cyp26a1 transcriptional repression in the absence of DNA methylation. Concomitantly, cells developed RA resistance and did not undergo RA-induced neuron differentiation. Apparently, RARalpha, RARbeta2 and Cyp26a1 are components of a RA-regulated gene network. Factors affecting an upstream gene of the network can trigger repressive chromatin changes -- which are propagated in a domino fashion - at downstream genes of the network. This study also shows that chromatin inactivity, and consequent transcriptional silencing, can be achieved in the absence of DNA methylation.


Subject(s)
Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/pathology , Cytochrome P-450 Enzyme System/biosynthesis , Receptors, Retinoic Acid/biosynthesis , Tretinoin/physiology , Animals , Cell Differentiation , Chromatin , Cytochrome P-450 Enzyme System/physiology , DNA Methylation , Epigenesis, Genetic , Gene Silencing , Mice , Neurons , Receptors, Retinoic Acid/physiology , Retinoic Acid 4-Hydroxylase , Tretinoin/metabolism
8.
Bone Marrow Transplant ; 39(6): 341-6, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17277788

ABSTRACT

We assessed the long-term outcome of patients with relapsed acute myeloid (n=86) or acute lymphoid leukemia (n=66), undergoing an allogeneic hemopoietic stem cell transplantation in our unit. The median blast count in the marrow was 30%. Conditioning regimen included total body irradiation (TBI) (10-12 Gy) in 115 patients. The donor was a matched donor (n=132) or a family mismatched donor (n=20). Twenty-two patients (15%) survive disease free, with a median follow-up of 14 years: 18 are off medications. The cumulative incidence of transplant related mortality is 40% and the cumulative incidence of relapse related death (RRD) is 45%. In multivariate analysis of survival, favorable predictors were chronic graft-versus-host disease (GvHD) (P=0.0003), donor other than family mismatched (P=0.02), donor age less than 34 years (P=0.02) and blast count less than 30% (P=0.07). Patients with all four favorable predictors had a 54% survival. In multivariate analysis of relapse, protective variables were the use of TBI (P=0.005) and cGvHD (P=0.01). This study confirms that a fraction of relapsed leukemias is cured with an allogeneic transplant: selection of patients with a blast count <30%, identification of young, human leukocyte antigen-matched donors and the use of total body radiation may significantly improve the outcome.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid/therapy , Neoplasm Recurrence, Local/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Acute Disease , Adolescent , Adult , Bone Marrow Examination , Child , Female , Follow-Up Studies , Graft Survival , Graft vs Host Disease , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid/complications , Male , Middle Aged , Patient Selection , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Prognosis , Survivors , Transplantation, Homologous
9.
Bone Marrow Transplant ; 38(2): 111-7, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16751783

ABSTRACT

A bone marrow harvest is filtered either in the operating room, in the laboratory or during infusion to the patient. Filters are usually discarded. Little is known of haemopoietic progenitor cells (HPCs) trapped in the filters. The aim of the study was to evaluate HPC content in the filters and to assess the outcome of transplants with filter-discarded or filter-recovered cells. Haemopoietic progenitors were grown from filters of 19 marrow transplants. We then compared the outcome of 39 filter-recovered transplants from HLA-identical siblings (years 2001-2004) with a matched cohort of 43 filter-discarded marrow grafts (years 1997-2000). Filters contained on average 21% long-term culture-initiating cells (LTC-IC) and 15% fibroblasts colony-forming units (CFU-F) of the total progenitor cell content. Filter-discarded transplants had significantly more grade II-IV graft-versus-host disease (GvHD) (42 vs 15%, P=0.008) as compared to filter-recovered transplants, and more transplant-related mortality (TRM) (20 vs 3%, P=0.04). The actuarial survival at 5 years is 69 vs 87%, respectively (P=0.15). This study suggests that a significant proportion of LTC-IC is lost in the filters together with CFU-F. Recovery and add back of progenitors trapped in the filters may reduce GvHD and TRM.


Subject(s)
Bone Marrow Transplantation/methods , Filtration/methods , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Adolescent , Adult , Bone Marrow Transplantation/adverse effects , Cells, Cultured , Female , Flow Cytometry/methods , Humans , Male , Middle Aged , Retrospective Studies , Survival Rate , Treatment Outcome
10.
J Natl Cancer Inst ; 92(16): 1303-7, 2000 Aug 16.
Article in English | MEDLINE | ID: mdl-10944551

ABSTRACT

BACKGROUND: Retinoic acid plays an important role in lung development and differentiation, acting primarily via nuclear receptors encoded by the retinoic acid receptor-beta (RARbeta) gene. Because receptor isoforms RARbeta2 and RARbeta4 are repressed in human lung cancers, we investigated whether methylation of their promoter, P2, might lead to silencing of the RARbeta gene in human lung tumors and cell lines. METHODS: Methylation of the P2 promoter from small-cell lung cancer (SCLC) and non-small-cell lung cancer (NSCLC) cell lines and tumor samples was analyzed by the methylation-specific polymerase chain reaction (PCR). Expression of RARbeta2 and RARbeta4 was analyzed by reverse transcription-PCR. Loss of heterozygosity (LOH) was analyzed by PCR amplification followed by electrophoretic separation of PCR products. Statistical differences were analyzed by Fisher's exact test with continuity correction. RESULTS: The P2 promoter was methylated in 72% (63 of 87) of SCLC and in 41% (52 of 127) of NSCLC tumors and cell lines, and the difference was statistically significant (two-sided P:<.001). By contrast, in 57 of 58 control samples, we observed only the unmethylated form of the gene. Four tumor cell lines with unmethylated promoter regions expressed both RARbeta2 and RARbeta4. Four tumor lines with methylated promoter regions lacked expression of these isoforms, but demethylation by exposure to 5-aza-2'-deoxycytidine restored their expression. LOH at chromosome 3p24 was observed in 100% (13 of 13) of SCLC lines and 67% (12 of 18) of NSCLC cell lines, and the difference was statistically significant (two-sided P: =.028). CONCLUSIONS: Methylation of the RARbeta P2 promoter is one mechanism that silences RARbeta2 and RARbeta4 expression in many lung cancers, particularly SCLC. Chemical demethylation is a potential approach to lung cancer therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Small Cell/metabolism , Loss of Heterozygosity , Lung Neoplasms/metabolism , Promoter Regions, Genetic , Receptors, Retinoic Acid/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Small Cell/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Methylation , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
11.
Oncogene ; 6(11): 2149-54, 1991 Nov.
Article in English | MEDLINE | ID: mdl-1945417

ABSTRACT

The genes of the ets family are thought to code for a novel class of transcriptional factors. These proteins have a specific DNA-binding domain different from the basic domain of both the helix-loop-helix and leucine zipper families of DNA-binding proteins. The ets-1 gene product has been shown to bind to the enhancer region of the human T-cell receptor alpha gene during thymocyte ontogeny. This finding explains the high expression of ets-1 observed in T cells and the correlation between ets-1 expression and the expression of the T-cell receptor gene during fetal development. The ets-1 gene is also possibly biologically active in neural cells. By using RNA in situ hybridization analysis, we demonstrate the presence of ets-1 transcripts in cells of peripheral embryonal neuroectodermal tumors, specifically neuroepithelioma and neuroblastoma. In addition, the gene is found transcribed in Ewing's sarcoma, postulated to be ontogenetically related to tumors derived from the neural crest.


Subject(s)
Nerve Tissue/metabolism , Peripheral Nervous System Neoplasms/metabolism , Retroviridae Proteins, Oncogenic/biosynthesis , Gene Expression , Humans , Lymphocytes/metabolism , Microscopy, Fluorescence , Neuroblastoma/metabolism , Neuroectodermal Tumors, Primitive, Peripheral/metabolism , Nucleic Acid Hybridization , RNA, Messenger/analysis , Sarcoma, Ewing/metabolism
12.
Oncogene ; 12(2): 437-44, 1996 Jan 18.
Article in English | MEDLINE | ID: mdl-8570222

ABSTRACT

AML1, a gene encoding a protein of the PEBP2/CBF family of transcription factors is disrupted by translocations associated with human leukemia. In the t(8;21) acute myelogenous leukemia (AML), AML1 was found fused to a gene on chromosome 8 that we designated CDR (also known as ETO and MTG8). Immunoprecipitation experiments followed by immunoblotting using a combination of antibodies against different epitopes of one of the predicted chimeric proteins encoded by a fully characterized fusion transcript enabled us to visualize a chimeric protein in the t(8;21) Kasumi-1 cell line. The estimated size of this protein is 64 kDa. Immunoblotting of leukemic blasts containing the t(8;21) detected a protein of the same size. Immunofluorescence experiments indicate that the chimeric protein is localized in the nucleus. A normal AML1 protein of 27 kDa was also detected in t(8;21) Kasumi-1 cells. It remains to be established by which mechanism the mutant AML1 isoform may contribute to the leukemogenesis process of t(8;21)-positive acute myeloid leukemia.


Subject(s)
Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , DNA-Binding Proteins , Leukemia, Myeloid, Acute/genetics , Neoplasm Proteins/analysis , Proto-Oncogene Proteins , Recombinant Fusion Proteins/analysis , Transcription Factors/analysis , Translocation, Genetic , Amino Acid Sequence , Core Binding Factor Alpha 2 Subunit , Humans , Leukemia, Myeloid, Acute/metabolism , Molecular Sequence Data
13.
Oncogene ; 16(20): 2609-15, 1998 May.
Article in English | MEDLINE | ID: mdl-9632137

ABSTRACT

The t(8;21) translocation associated with acute myeloid leukemia (AML) disrupts two genes, the AML1 gene also known as the core binding factor A2 (CBFA2) on chromosome 21, and a gene on chromosome 8, hereafter referred to as MTG8, but also known as CDR and ETO. Extensive information is available on AML1, a member of the CBF family of transcription factors, containing a highly conserved domain, the runt box, of the Drosophila segmentation gene runt. This gene is essential for the hematopoietic development and is found disrupted in several leukemias. In contrast, the function of the MTG8 gene is poorly understood. The predicted protein sequence shows two unusual, putative zinc-fingers, three proline-rich regions, a PEST domain and several phosphorylation sites. In addition, we found a region encompassing aa 443-514 predicted to have a significant propensity to form coiled coil structures. MTG8 displays a high degree of similarity with nervy, a homeotic target gene of Drosophila, expressed in the nervous system. Human and mouse wild-type MTG8 are also highly expressed in brain relative to other tissues. For these reasons, we set out to investigate the expression and subcellular localization of the MTG8 protein in neural cells. Immunohistochemical experiments in a 12.5-day-old mouse embryo clearly showed that the protein was expressed in the neural cells of the developing brain and the spinal cord. In primary cultures of hippocampal neurons of 2-3 day-old mice, MTG8 was found in the nucleus, in the cytoplasm and as fine granules in the neurites. Cytoplasmic localization of the protein was observed in Purkinje cells of both human and mouse cerebellum. The molecular mass of MTG8 in total human and mouse brain was analysed by immunoblotting and determined to be between 70 and 90 kDa. Isoforms with the same molecular mass were demonstrated in synaptosomes isolated from mouse forebrain. The evidence of MTG8 in the nucleus and cytoplasm of neural cells suggests a specific mechanism regulating the subcellular localization of the protein.


Subject(s)
DNA-Binding Proteins/metabolism , Neurons/metabolism , Proto-Oncogene Proteins , Transcription Factors/metabolism , Animals , Brain/embryology , Brain/metabolism , COS Cells , Cell Nucleus/metabolism , Cells, Cultured , Cytoplasm/metabolism , Humans , Immunohistochemistry , Mice , Purkinje Cells/metabolism , RUNX1 Translocation Partner 1 Protein , Synaptosomes/metabolism
14.
Oncogene ; 19(12): 1556-63, 2000 Mar 16.
Article in English | MEDLINE | ID: mdl-10734315

ABSTRACT

Retinoic acid (RA)-resistance in breast cancer cells has been associated with irreversible loss of retinoic acid receptor beta, RARbeta, gene expression. Search of the causes affecting RARbeta gene activity has been oriented at identifying possible differences either at the level of one of the RARbeta promoters, RARbeta2, or at regulatory factors. We hypothesized that loss of RARbeta2 activity occurs as a result of multiple factors, including epigenetic modifications, which can pattern RARbeta2 chromatin state. Using methylation-specific PCR, we found hypermethylation at RARbeta2 in a significant proportion of both breast cancer cell lines and primary breast tumors. Treatment of cells with a methylated RARbeta2 promoter, by means of the DNA methyltransferase inhibitor 5-Aza-2'-deoxycytidine (5-Aza-CdR), led to demethylation within RARbeta2 and expression of RARbeta indicating that DNA methylation is at least one factor, contributing to RARbeta inactivity. However, identically methylated promoters can differentially respond to RA, suggesting that RARbeta2 activity may be associated to different repressive chromatin states. This supposition is supported by the finding that the more stable repressive RARbeta2 state in the RA-resistant MDA-MB-231 cell line can be alleviated by the HDAC inhibitor, trichostatin A (TSA), with restoration of RA-induced RARbeta transcription. Thus, chromatin-remodeling drugs might provide a strategy to restore RARbeta activity, and help to overcome the hurdle of RA-resistance in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Chromatin/genetics , Receptors, Retinoic Acid/genetics , Antimetabolites, Antineoplastic , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Breast/cytology , Breast Neoplasms/drug therapy , Cell Line , CpG Islands , DNA Methylation , Decitabine , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Female , Gene Expression Regulation/drug effects , Histone Deacetylase Inhibitors , Humans , Hydroxamic Acids/pharmacology , Promoter Regions, Genetic , Receptors, Estrogen/metabolism , Receptors, Retinoic Acid/drug effects , Receptors, Retinoic Acid/metabolism , Tretinoin/pharmacology , Tumor Cells, Cultured
15.
Leukemia ; 10(6): 984-90, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8667656

ABSTRACT

Truncated AML1 proteins are predicted to be expressed from out-of-frame AML1 transcripts present in myeloid leukemia cells harboring t(8;21) and t(3;21). To test whether these proteins, consisting of almost exclusively an N-terminal AML1 DNA-binding domain, interfere with myeloid differentiation we expressed a similar truncated AML1 protein in 32D cl3 myeloid cells. In all clones examined, the ectopically expressed truncated AML1 protein prevented binding of endogenous PEBP2/CBFs to DNA, possibly by interacting with all available CBF beta subunits. However, compared to control clones, the 32D cl3 clones expressing truncated AML1 remained IL-3 dependent for survival, proliferated similarly in low and high concentrations of IL-3, and differentiated similarly upon transfer to G-CSF. Thus, truncated AML1 proteins may contribute to myeloid leukemogeneis by inhibiting PEBP2/CBF activities, although contributions from other oncoproteins are likely required as well.


Subject(s)
Cell Transformation, Neoplastic/genetics , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 3 , Chromosomes, Human, Pair 8 , DNA, Neoplasm/metabolism , DNA-Binding Proteins/metabolism , Leukemia, Myeloid/genetics , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Translocation, Genetic , Animals , Base Sequence , Binding Sites , Blotting, Western , Cell Differentiation , Core Binding Factor Alpha 2 Subunit , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Interleukin-3/pharmacology , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Mice , Molecular Sequence Data , Transcription Factor AP-2 , Tumor Cells, Cultured/pathology
16.
Leukemia ; 12(1): 96-101, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9436927

ABSTRACT

The translocation (8;21) is a chromosome abnormality associated with acute myeloid leukemia (AML). As a consequence of the translocation the AML1 (CBFA2) gene in the 21q22 region is fused to the ETO(CDR,MTG8) gene in the 8q22 region, resulting in one transcriptionally active gene on the 8q- derivative chromosome. In this report we demonstrate the use of a highly specific dual-colour FISH method for the detection of t(8;21) on interphase cells. Genomic probes able to detect the chimeric AML1/ETO gene on the 8q- derivative chromosome were assayed on both normal and leukemic bone marrow and peripheral blood samples. Cut-off values were established by independent analysis of 15 bone marrow specimens negative for the translocation. The cut-off value of positive nuclei was determined to be 2% and the cut-off value for both positive nuclei and nuclei of uncertain classification, 4%. Persistence of cells above these cut-off values was interpreted as persistence of the mutated clone. A total of 36 samples at different disease stages were tested. Interphase cytogenetics detected the translocation at the onset and relapse in the BM or the PB of 14 AML patients with t(8;21). The technique appears to be an alternative tool to both conventional cytogenetics and reverse transcription polymerase chain reaction (RT-PCR) for the monitoring of disease during patients' follow-up. By enabling the analysis of individual cells, interphase FISH is ideal for clonality studies both for clinical and experimental applications.


Subject(s)
Chromosome Aberrations , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , DNA-Binding Proteins , Leukemia, Myeloid/genetics , Proto-Oncogene Proteins , Translocation, Genetic , Acute Disease , Adolescent , Adult , Aged , Bone Marrow/pathology , Child , Chromosome Mapping , Core Binding Factor Alpha 2 Subunit , Disease-Free Survival , Exons , Female , Humans , In Situ Hybridization, Fluorescence/methods , Interphase , Leukemia, Myeloid/blood , Leukemia, Myeloid/mortality , Leukemia, Myeloid/pathology , Male , Middle Aged , Prognosis , Proto-Oncogenes , Survival Rate , Time Factors , Transcription Factors/genetics
17.
Leukemia ; 11(2): 202-5, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9009081

ABSTRACT

Bone marrow from six patients with acute myeloid leukemia (AML) and t(8;21) (q22;q22) or a variant t(8;13;21) was studied by simultaneous analysis of cell morphology and karyotype. Combination of May-GrĆ¼nwald-Giemsa (MGG) and fluorescence in situ hybridization (FISH) using probes specific for the breakpoint regions of chromosome 8 and 21 allowed us to establish the extent of cell-lineage involvement of the translocation. The translocation was found in all myeloid blasts and in high percentages of the more mature neutrophilic cells. In one patient we could demonstrate the translocation in the eosinophils as well. Erythroblasts and lymphocytes did not show the t(8;21) abnormality. These results indicate that the t(8;21) in AML is restricted to the myeloid (granulocytic) lineage.


Subject(s)
Chromosomes, Human, Pair 21/ultrastructure , Chromosomes, Human, Pair 8/ultrastructure , Clone Cells/pathology , Hematopoietic Stem Cells/pathology , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/pathology , Translocation, Genetic , Adolescent , Adult , Bone Marrow/pathology , Cell Differentiation/drug effects , Female , Humans , In Situ Hybridization, Fluorescence , Leukemia, Myeloid, Acute/genetics , Male , Middle Aged
18.
Leukemia ; 2(1): 12-8, 1988 Jan.
Article in English | MEDLINE | ID: mdl-2448555

ABSTRACT

The cellular ets-1 gene homologous to the 5' region of the v-ets sequence of the E26 retrovirus codes for a 6.8-kb mRNA that is translated into a 51-kDa protein in human cells. A survey of mRNA from human tissues showed the thymus as the tissue with the highest level of ets-1 transcription, within other hematopoietic organs and tissues, including spleen, fetal liver, lymph nodes, bone marrow, and peripheral lymphocytes exhibiting low or undetectable levels of hybridization. A high level of ets-1 expression was found in murine thymocyte mRNA as well. Investigation of the ets-1 expression levels in human leukemic samples showed that primary malignant T cells (T-ALLs), corresponding to intrathymic stages of maturation, have a much higher level of ets-1 mRNA than malignant T lymphoid cells with a mature phenotype, such as adult T cell leukemias (ATLs). T-ALLs were also higher in ets-1 expression than the other lymphoid (pre-T-ALL, c-ALL, pre-B-ALL) malignant cells analyzed. Insignificant amounts of the specific ets-1 mRNA were detected in several acute myeloid leukemias representing various degrees of maturation. The elevated ets-1 mRNA in thymocytes suggests a biological role for the ets-1 product in these cells that could be explored to investigate ets-1 function. Finally, the exhibited expression of ets-1 in lymphoid cells and absence from malignant myeloid cells makes it a candidate marker for phenotyping human hematopoietic tumors.


Subject(s)
Gene Expression Regulation , Leukemia/genetics , Proto-Oncogenes , T-Lymphocytes/physiology , Thymus Gland/physiology , Cell Line , Deltaretrovirus Infections/genetics , Deltaretrovirus Infections/metabolism , Fetus , Humans , Leukemia/metabolism , Leukemia/pathology , Phenotype , RNA/analysis , RNA, Messenger/metabolism , T-Lymphocytes/metabolism , Thymus Gland/cytology , Transcription, Genetic
19.
Leukemia ; 9(2): 282-7, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7869765

ABSTRACT

Fluorescence in situ hybridization (FISH) and/or RNA-based polymerase chain reaction (RT-PCR) were used to analyze the breakpoints within the AML1 gene and the AML1 fusion transcripts in t(8;21) acute myeloid leukemia (AML). Twenty-two patients presented with the simple t(8;21)(q22;q22) and one with a complex variant t(8;2;16;21). In eight cases we used FISH with AML1 cosmid probes on metaphase chromosomes as well as RT-PCR to detect the junctions of MAL1/CDR (ETO,MTG8). Five cases were analyzed by FISH alone and ten cases by RT-PCR alone. By FISH we could identify three groups according to the distribution of the fluorescent signal. Signals were found in group 1 on chromosomes 21 and 21q+, in group 2 on chromosomes 21, 21q+ and 8q- and in group 3 on chromosomes 21 and 8q-. In all groups we could detect an identical AML1/CDR fusion transcript. This transcript showed splicing of AML1 exon 5 onto CDR. Thus regardless of the heterogeneity suggested by FISH, all the breakpoints in the AML1 gene were clustered in the same intro between exons 5 and 6. Our results bring to over one hundred the number of t(8;21) cases in which an identical translocation could be detected at molecular level by RT-PCR. The high sensitivity of the technique makes it suitable for the diagnosis of this translocation in different stages of the disease. The impact of the molecular detection of t(8;21) cells in clinical remission as far as the treatment and the management of the disease are concerned deserves further discussion.


Subject(s)
Chromosomes, Human, Pair 21/ultrastructure , Chromosomes, Human, Pair 8/ultrastructure , DNA-Binding Proteins , Leukemia, Myeloid/genetics , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins , Transcription Factors , Translocation, Genetic , Acute Disease , Adolescent , Adult , Aged , Aged, 80 and over , Base Sequence , Blast Crisis/genetics , Blast Crisis/pathology , Child , Child, Preschool , Chromosomes, Human, Pair 16/ultrastructure , Chromosomes, Human, Pair 2/ultrastructure , Cohort Studies , Core Binding Factor Alpha 2 Subunit , Female , Humans , In Situ Hybridization, Fluorescence , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myeloid/pathology , Male , Middle Aged , Molecular Sequence Data , Polymerase Chain Reaction
20.
Leukemia ; 16(11): 2228-37, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12399966

ABSTRACT

Aims of this study were to verify whether reduction in transplant-related mortality (TRM) of children with acute lymphoblastic leukemia (ALL) in second complete remission (CR) given allogeneic hematopoietic stem cell transplantation (HSCT) from unrelated volunteers has occurred over time and to investigate the role of other variables on the probabilities of relapse, TRM and event-free survival (EFS). We compared results obtained in 26 children given HSCT before January 1998 with those of 37 patients transplanted beyond that date. In all donor-recipient pairs, histocompatibility was determined by serology for HLA-A and -B antigens and by high-resolution DNA typing for DRB1 antigen. High-resolution molecular typing of HLA class I antigens was employed in 20 of the 37 children transplanted more recently. Probability of both acute and chronic GVHD was comparable in the two groups of patients. In multivariate analysis, children transplanted before January 1998, those with T-lineage ALL and those experiencing grade II-IV acute GVHD had a higher relative risk of TRM at 6 months after transplantation. Relapse rate was unfavorably affected by a time interval between diagnosis and relapse <30 months. The 2-year probability of EFS for children transplanted before and after 1 January 1998 was 27% (10-44) and 58% (42-75), respectively (P = 0.02), this difference remaining significant in multivariate analysis. EFS of unrelated donor HSCT in children with ALL in second CR has improved in the last few years, mainly due to a decreased TRM. This information is of value for counseling of patients with relapsed ALL.


Subject(s)
Hematopoietic Stem Cell Transplantation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adolescent , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Asparaginase/administration & dosage , Child , Child, Preschool , Cyclophosphamide/administration & dosage , Cytarabine/administration & dosage , DNA, Neoplasm/analysis , Daunorubicin/administration & dosage , Disease-Free Survival , Female , Graft vs Host Disease , HLA-A Antigens/immunology , HLA-B Antigens/immunology , HLA-DR Antigens/genetics , HLA-DRB1 Chains , Humans , Infant , Living Donors , Male , Mercaptopurine/administration & dosage , Methotrexate/administration & dosage , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Prednisone/administration & dosage , Registries , Remission Induction , Survival Rate , Time Factors , Transplantation, Homologous , Treatment Outcome , Vincristine/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL