Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Mol Cell Biol ; 30(19): 4575-94, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20679491

ABSTRACT

The interferon consensus sequence binding protein (ICSBP) is an interferon regulatory transcription factor, also referred to as IRF8. ICSBP acts as a suppressor of myeloid leukemia, although few target genes explaining this effect have been identified. In the current studies, we identified the gene encoding growth arrest specific 2 (GAS2) as an ICSBP target gene relevant to leukemia suppression. We find that ICSBP, Tel, and histone deacetylase 3 (HDAC3) bind to a cis element in the GAS2 promoter and repress transcription in myeloid progenitor cells. Gas2 inhibits calpain protease activity, and beta-catenin is a calpain substrate in these cells. Consistent with this, ICSBP decreases beta-catenin protein and activity in a Gas2- and calpain-dependent manner. Conversely, decreased ICSBP expression increases beta-catenin protein and activity by the same mechanism. This is of interest, because decreased ICSBP expression and increased beta-catenin activity are associated with poor prognosis and blast crisis in chronic myeloid leukemia (CML). We find that the expression of Bcr/abl (the CML oncoprotein) increases Gas2 expression in an ICSBP-dependent manner. This results in decreased calpain activity and a consequent increase in beta-catenin activity in Bcr/abl-positive (Bcr/abl(+)) cells. Therefore, these studies have identified a Gas2/calpain-dependent mechanism by which ICSBP influences beta-catenin activity in myeloid leukemia.


Subject(s)
Interferon Regulatory Factors/metabolism , Microfilament Proteins/metabolism , Myeloid Cells/metabolism , beta Catenin/metabolism , Animals , Blotting, Western , Cells, Cultured , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Gene Expression , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Interferon Regulatory Factors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Microscopy, Confocal , Promoter Regions, Genetic/genetics , Protein Binding , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , RNA Interference , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , U937 Cells , beta Catenin/genetics , ETS Translocation Variant 6 Protein
2.
J Biol Chem ; 284(48): 33242-54, 2009 Nov 27.
Article in English | MEDLINE | ID: mdl-19801548

ABSTRACT

The interferon consensus sequence binding protein (ICSBP) is an interferon regulatory transcription factor with leukemia-suppressor activity. ICSBP regulates genes that are involved in phagocyte function, proliferation, and apoptosis. In murine models ICSBP deficiency results in a myeloproliferative disorder (MPD) with increased mature neutrophils. Over time this MPD progresses to acute myeloid leukemia (AML), suggesting that ICSBP deficiency is adequate for MPD, but additional genetic lesions are required for AML. The hypothesis of these studies is that dysregulation of key target genes predisposes to disease progression under conditions of decreased ICSBP expression. To investigate this hypothesis, we used chromatin co-immunoprecipitation to identify genes involved the ICSBP-leukemia suppressor effect. In the current studies, we identify the gene encoding Fanconi F (FANCF) as an ICSBP target gene. FancF participates in a repair of cross-linked DNA. We identify a FANCF promoter cis element, which is activated by ICSBP in differentiating myeloid cells. We also determine that DNA cross-link repair is impaired in ICSBP-deficient myeloid cells in a FancF-dependent manner. This effect is observed in differentiating cells, suggesting that ICSBP protects against the genotoxic stress of myelopoiesis. Decreased ICSBP expression is found in human AML and chronic myeloid leukemia during blast crisis (CML-BC). Our studies suggest that ICSBP deficiency may be functionally important for accumulation of chromosomal abnormalities during disease progression in these myeloid malignancies.


Subject(s)
Cell Differentiation , Fanconi Anemia Complementation Group F Protein/metabolism , Interferon Regulatory Factors/metabolism , Transcriptional Activation , Animals , Blotting, Western , Cells, Cultured , Chromatin Immunoprecipitation , DNA Repair , Fanconi Anemia Complementation Group F Protein/genetics , Humans , Interferon Regulatory Factors/deficiency , Interferon Regulatory Factors/genetics , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transfection , U937 Cells
3.
J Biol Chem ; 284(4): 2549-67, 2009 Jan 23.
Article in English | MEDLINE | ID: mdl-19022774

ABSTRACT

The homeodomain transcription factor HoxA10 is maximally expressed in myeloid progenitor cells. Sustained HoxA10 expression during differentiation has been described in poor prognosis human acute myeloid leukemia (AML). Consistent with this, engineered overexpression of HoxA10 in murine bone marrow induces a myeloproliferative disorder that progresses to AML over time. This murine model suggests that HoxA10 overexpression is sufficient for myeloproliferation but that differentiation block, and therefore AML, requires acquisition of additional mutations. In myeloid progenitor cells, HoxA10 represses transcription of genes that encode phagocyte effector proteins such as gp91PHOX and p67PHOX. Tyrosine phosphorylation of HoxA10 during myelopoiesis decreases binding to these target genes. In immature myeloid cells, HoxA10 also activates transcription of the DUSP4 gene that encodes Mkp2, an anti-apoptotic protein. HoxA10 binding to the DUSP4 promoter decreases during myelopoiesis. Therefore, both myeloid-specific gene repression and DUSP4 activation by HoxA10 decrease during myelopoiesis. This results in phenotypic differentiation and facilitates apoptosis as differentiation proceeds. HoxA10 is de-phosphorylated by SHP2 protein-tyrosine phosphatase in myeloid progenitors. This mechanism maintains HoxA10 in a nonphosphorylated state in immature, but not differentiating, myeloid cells. Constitutively active SHP2 mutants have been described in human AML, which dephosphorylate HoxA10 throughout myelopoiesis. In this study, we hypothesize that constitutive SHP2 activation synergizes with HoxA10 overexpression to accelerate progression to AML. Because both HoxA10 overexpression and constitutive SHP2 activation are found in poor prognosis human AML, these studies contribute to understanding biochemical aspects of disease progression in myeloid malignancy.


Subject(s)
Homeodomain Proteins/metabolism , Leukemia, Myeloid, Acute/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cytokines/pharmacology , Enzyme Activation , Gene Expression Regulation, Neoplastic , Homeobox A10 Proteins , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Mutation/genetics , Myeloid Cells/cytology , Myeloid Cells/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Tyrosine/genetics , Tyrosine/metabolism
4.
J Clin Invest ; 118(3): 853-67, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18246201

ABSTRACT

Myeloproliferative disorders (MPDs) are characterized by cytokine hypersensitivity and apoptosis resistance. Development of a block in myeloid differentiation is associated with progression of MPD to acute myeloid leukemia (AML) and portends poor prognosis. Identifying molecular markers of this transition may suggest targets for therapeutic intervention. Interferon consensus sequence binding protein (ICSBP, also known as IRF8) is an interferon-regulatory transcription factor that functions as a leukemia tumor suppressor. In mice, ICSBP deficiency induces an MPD that progresses to AML over time, suggesting that ICSBP deficiency is sufficient for myeloproliferation, but additional genetic lesions are necessary for AML. Since activity of ICSBP is influenced by tyrosine phosphorylation state, we hypothesized that mutations in molecular pathways that regulate this process might synergize with ICSBP deficiency for progression to AML. Consistent with this, we found that constitutive activation of SHP2 protein tyrosine phosphatase synergized with ICSBP haploinsufficiency to facilitate cytokine-induced myeloproliferation, apoptosis resistance, and rapid progression to AML in a murine bone marrow transplantation model. Constitutive SHP2 activation cooperated with ICSBP deficiency to increase the number of progenitors in the bone marrow and myeloid blasts in circulation, indicating a block in differentiation. Since SHP2 activation and ICSBP deficiency may coexist in human myeloid malignancies, our studies have identified a molecular mechanism potentially involved in disease progression in such diseases.


Subject(s)
Interferon Regulatory Factors/physiology , Leukemia, Myeloid, Acute/etiology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Animals , Apoptosis , Cell Differentiation , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/physiology , Humans , Interferon Regulatory Factors/chemistry , Interferon Regulatory Factors/deficiency , Mice , Mice, Inbred C57BL , Myeloproliferative Disorders/complications , Phosphorylation , Transcription, Genetic , Tyrosine/metabolism , U937 Cells
5.
Mol Cell Biol ; 26(17): 6311-32, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16914719

ABSTRACT

Deficiency in either the interferon consensus sequence binding protein (ICSBP) or neurofibromin 1 (Nf1) increases the proliferative response of myeloid progenitor cell to hematopoietic cytokines. Consistent with this, we previously demonstrated that ICSBP activates transcription of the gene encoding Nf1 (the NF1 gene). In the studies presented here, we determine that ICSBP tyrosine phosphorylation is necessary for the activation of NF1 transcription. Since ICSBP is tyrosine phosphorylated in response to hematopoietic cytokines, these studies identify a novel pathway by which cytokine-induced posttranslational modification of ICSBP results in NF1 transcription. Nf1 subsequently inactivates cytokine-activated Ras, thereby creating a negative feedback mechanism for cytokine-induced proliferation. In these studies, we also determine that ICSBP is a substrate for SHP2 protein tyrosine phosphatase (SHP2-PTP). We find that wild-type SHP2-PTP dephosphorylates ICSBP only in undifferentiated myeloid cells. In contrast, a leukemia-associated, constitutively activated mutant form of SHP2-PTP dephosphorylates ICSBP in both myeloid progenitors and differentiating myeloid cells. Activated SHP2-PTP mutants thereby inhibit ICSBP-dependent NF1 transcription, impairing this negative feedback mechanism on cytokine-activated Ras. Therefore, these studies suggest that leukemia-associated ICSBP deficiency cooperates with leukemia-associated activating mutants of SHP2-PTP to contribute to the proliferative phenotype in myeloid malignancies.


Subject(s)
Interferon Regulatory Factors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Leukemia, Myeloid/metabolism , Mutant Proteins/metabolism , Neurofibromin 1/genetics , Protein Tyrosine Phosphatases/metabolism , Transcriptional Activation/genetics , Animals , Bone Marrow/drug effects , Cell Differentiation/drug effects , Glutamine/genetics , Humans , Interferon-gamma/pharmacology , Lysine/genetics , Mice , Mice, Inbred C57BL , Myeloid Cells/cytology , Myeloid Cells/drug effects , Phosphotyrosine/metabolism , Protein Binding/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Regulatory Sequences, Nucleic Acid/drug effects , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , U937 Cells
6.
Int J Hematol ; 80(2): 146-54, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15481443

ABSTRACT

The bone marrow of patients with myelodysplastic syndromes (MDS) shows excessive intramedullary apoptosis, particularly in S-phase cells. In the light of previous reports that showed a link between experimental overexpression of the E2F1 transcription factor and apoptosis in the S phase, we compared the status of E2F1 protein in bone marrow mononuclear cells of MDS patients with that of healthy donors. Nearly 67% of MDS marrow samples showed higher expression of E2F1 transcription factor than in healthy donors. The retinoblastoma gene product, Rb, is a major negative regulator of E2F1 activity; however, Rb protein levels were found to be normal in MDS marrow samples. Amplification of genomic DNA by the polymerase chain reaction (PCR) showed no E2F1 gene amplification or mutation in the Rb-binding region of E2F1 in MDS patients, nor was transcriptional up-regulation noted when E2F1 messenger RNA (mRNA) levels were estimated with real-time reverse transcriptase-PCR. Furthermore, the overexpression of E2F1 was paralleled by its increased transcriptional activity, as reflected by the increased mRNA levels for one of its target genes, dihydrofolate reductase. Importantly, in a subset of the studied MDS patients for whom a simultaneous measurement of apoptosis in S-phase cells was possible, the E2F1 protein levels showed a significant positive correlation with this phenomenon. Previously, increased E2F1 activity in human disease had been found primarily as a consequence of Rb derailment. Hence, the observation in MDS of increased E2F1 activity in the presence of normal Rb levels is novel and unique, and E2F1 activity in association with apoptosis in S-phase cells may thus have significant therapeutic implications.


Subject(s)
Bone Marrow Cells/pathology , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Myelodysplastic Syndromes/pathology , Transcription Factors/genetics , Aged , Bone Marrow Cells/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , Female , Humans , Karyotyping , Male , Middle Aged , Myelodysplastic Syndromes/genetics , Reference Values , Retinoblastoma Protein/genetics , S Phase , Tetrahydrofolate Dehydrogenase/genetics , Transcription, Genetic/genetics
7.
J Biol Chem ; 279(49): 50874-85, 2004 Dec 03.
Article in English | MEDLINE | ID: mdl-15371411

ABSTRACT

Deficiency of the interferon consensus sequence-binding protein (ICSBP) is associated with increased myeloid cell proliferation in response to hematopoietic cytokines. However, previously identified ICSBP target genes do not indicate a mechanism for this "cytokine hypersensitivity." In these studies, we identify the gene encoding neurofibromin 1 (Nf1) as an ICSBP target gene, by chromatin immunoprecipitation. Additionally, we find decreased Nf1 expression in bone marrow-derived myeloid cells from ICSBP-/- mice. Since Nf1 deficiency is also associated with cytokine hypersensitivity, our results suggested that NF1 is a functionally significant ICSBP target gene. Consistent with this, we find that the hypersensitivity of ICSBP-/- myeloid cells to granulocyte monocyte colony-stimulating factor (GM-CSF) is reversed by expression of the Nf1 GAP-related domain. We also find that treatment of ICSBP-deficient myeloid cells with monocyte colony-stimulating factor (M-CSF) results in sustained Ras activation, ERK phosphorylation, and proliferation associated with impaired Nf1 expression. These M-CSF effects are reversed by ICSBP expression in ICSBP-/- cells. Consistent with this, we find that ICSBP activates the NF1 promoter in myeloid cell line transfectants and identify an ICSBP-binding NF1 cis element. Therefore, the absence of ICSBP leads to Nf1 deficiency, impairing down-regulation of Ras activation by GM-CSF or M-CSF. These results suggest that one mechanism of increased myeloid proliferation, in ICSBP-deficient cells, is decreased NF1 gene transcription. This novel ICSBP function provides insight into regulation of myelopoiesis under normal conditions and in myeloproliferative disorders.


Subject(s)
Neurofibromin 1/chemistry , Neurofibromin 1/genetics , Repressor Proteins/chemistry , Repressor Proteins/physiology , Transcription, Genetic , Animals , Blotting, Western , Bone Marrow Cells/cytology , Cell Nucleus/metabolism , Cell Proliferation , Cells, Cultured , Chromatin Immunoprecipitation , Cloning, Molecular , Cytokines/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Genes, Reporter , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Immunoprecipitation , Interferon Regulatory Factors , Macrophage Colony-Stimulating Factor/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 3/metabolism , Oligonucleotides/chemistry , Phosphorylation , Plasmids/metabolism , Promoter Regions, Genetic , Protein Structure, Tertiary , RNA/metabolism , Retroviridae/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , U937 Cells , ras Proteins/metabolism
8.
Cell Biol Int ; 27(9): 761-8, 2003.
Article in English | MEDLINE | ID: mdl-12972282

ABSTRACT

Tamoxifen at a dose of 400 microg/kg/day has been reported to reduce the fertility of adult male rats and alter the pattern of cauda sperm motility from forward progressive to circular yawing type. Since sperm motility is powered by mitochondria, the effect of tamoxifen on mitochondrial function was studied. Tamoxifen treatment significantly increased rhodamine 123 fluorescent dye uptake by sperm mitochondria, reflecting an altered mitochondrial membrane potential. ATP and DAG levels, activities of glycolytic enzymes, creatine kinase and PKC all remained unaffected by tamoxifen. This is also the first report describing the presence of PKC alpha and beta in rat sperm. Morphological and biochemical integrity of sperm membranes was determined by electron microscopy and malondialdehyde levels, which were unaltered after tamoxifen treatment. This study indicates that the altered sperm motility induced by tamoxifen is accompanied by changes in mitochondrial membrane potential, but in the absence of any detectable change in membrane integrity, lipid peroxidation, ATP levels and activities of glycolytic enzymes, creatine kinase and PKC.


Subject(s)
Mitochondria/drug effects , Protein Kinase C/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Spermatozoa/drug effects , Tamoxifen/pharmacology , Adenosine Triphosphate/metabolism , Animals , Biological Transport , Creatine Kinase/metabolism , Fluorescent Dyes/metabolism , Glycolysis , Male , Malondialdehyde/metabolism , Mitochondria/metabolism , Mitochondria/ultrastructure , Rats , Rhodamine 123/metabolism , Sperm Motility/drug effects , Spermatozoa/enzymology , Spermatozoa/ultrastructure
9.
J Hematother Stem Cell Res ; 12(4): 443-50, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12965081

ABSTRACT

An unusually high incidence of apoptosis in S-phase cells is characteristically found in the bone marrow (BM) of patients with myelodysplastic syndromes (MDS). Previously, E2F1, c-myc, and Cyclin D1 have been shown to bring about both S-phase changes and/or apoptotic changes. We have already found a stoichiometric imbalance between pRb and E2F1 causing deregulated E2F1 activity in these disorders. In the present study, we investigated the status of Cyclin D1 in relation to E2F1 and apoptosis in 19 patients with a confirmed diagnosis of MDS in comparison with 6 healthy donors. Cyclin D1 was localized immunohistochemically using a specific monoclonal antibody (1:150 dilution) in plastic-embedded BM sections. The nuclear localization of Cyclin D1 graded on a subjective rating scale of 0 (negligible staining) to 8+ (highest), demonstrated negligible levels in normal marrows (median 1+), and in 11/19 evaluable MDS marrows. In contrast, 8/19 MDS biopsies showed an almost four-fold increase in Cyclin D1 localization (p< or =0.001). A western blot analysis of E2F1 in corresponding bone marrow (BM) aspirate mononuclear cells (MNC) demonstrated that the MDS patients with elevated Cyclin D1 expression also had a significant increase in E2F1 protein (p< or =0.03). Additionally, these patients revealed higher levels of mRNA of one of the E2F1 transcriptional target genes, dihydrofolate reductase (DHFR, p=0.01). Subsequently, the relationship of Cyclin D1 with apoptosis was elucidated in a colocalization experiment in BM biopsy sections using immunohistochemistry for Cyclin D1 and in situ end labeling of DNA (ISEL) for apoptosis. The percentage of ISEL-positive apoptotic cells was several fold higher in MDS as compared to normal BMs (p=0.009). Interestingly, 7-41% (median 20%) of the apoptotic cells in different MDS BMs revealed co-localization of Cyclin D1 in their nucleus, whereas in normal BMs co-localization was virtually absent (p=0.008). Thus, it is possible that in a subset of MDS patients, apoptotic death of bone marrow cells may involve Cyclin D1/E2F1 pathway.


Subject(s)
Apoptosis , Cell Cycle Proteins , Cyclin D1/biosynthesis , DNA-Binding Proteins , Myelodysplastic Syndromes/pathology , Transcription Factors/biosynthesis , Adult , Aged , Antibodies, Monoclonal/metabolism , Blotting, Western , Bone Marrow Cells/cytology , Cell Nucleus/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , Female , Humans , Immunohistochemistry , Male , Middle Aged , Myelodysplastic Syndromes/metabolism , RNA, Messenger/metabolism , Retinoblastoma Protein/metabolism , Reverse Transcriptase Polymerase Chain Reaction , S Phase , Transcription, Genetic
10.
FASEB J ; 17(6): 569-74, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12665469

ABSTRACT

E2F transcription factors may play a pivotal role in the transcriptional regulation of several cellular processes far beyond the originally described cell cycle and proliferation. Among the six E2F family members, only E2F1 is noted for its role in apoptosis. The pocket protein family members Rb, p107, and p130 act as the main regulators of E2F activity. Nonetheless, in recent years other protein-protein interactions have been described for E2Fs. The post-translational modifications resulting from such protein interactions may have significant implications in the stability, half-life, and functional activity of E2Fs. In human diseases the significance of E2Fs is still under appreciated and is primarily recognized only as a consequence of the impairment in retinoblastoma gene product (Rb). However, with increasing knowledge of other protein interactions, the derailment of E2F activity could be anticipated to stem from an abnormality of any node in the complex network governing their availability and activity. The present review is intended to provide a perspective on the diversity of biochemical mechanisms underlying abnormal E2F expression and activity, understanding of which may have significant clinical implications.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle/physiology , DNA-Binding Proteins , Transcription Factors/physiology , Animals , Cell Cycle/genetics , Cell Cycle Proteins/genetics , E2F Transcription Factors , E2F1 Transcription Factor , Gene Expression Regulation , Humans , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Transcription Factors/genetics
11.
Int J Hematol ; 75(3): 289-97, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11999358

ABSTRACT

Labeling index (LI), apoptosis, levels of 2 pro-apoptotic cytokines tumor necrosis factor-alpha (TNF-alpha) and transforming growth factor-beta(TGF-beta), and the number of monocyte/macrophage cells that are the likely source of the cytokines were simultaneously measured in plastic-embedded bone marrow (BM) biopsy sections of 145 patients with myelodysplastic syndromes (MDS). TNF-alpha was correlated with TGF-beta (P = .001) and with monocyte/macrophage cells (P = .003). Patients with excess blasts in their marrows had a higher TGF-beta level (P = .01) and monocyte/macrophage number (P = .05). In a linear regression model,TGF-beta emerged as the most significant biological difference between patients who have excess of blasts and those who do not (P = .01). We conclude that in addition to TNF-alpha, TGF-beta also plays a significant role in the initiation and pathogenesis of MDS, and that a more precise definition of its role will likely identify better preventive and therapeutic strategies.


Subject(s)
Apoptosis , Bone Marrow Cells/pathology , Cytokines/analysis , Macrophages/pathology , Monocytes/pathology , Myelodysplastic Syndromes/pathology , Anemia, Refractory/pathology , Anemia, Refractory, with Excess of Blasts/pathology , Animals , Cell Division , Female , Humans , Leukemia, Myelomonocytic, Chronic/pathology , Male , Regression Analysis , S Phase , Transforming Growth Factor beta/analysis , Tumor Necrosis Factor-alpha/analysis
12.
Br J Haematol ; 116(3): 564-75, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11849212

ABSTRACT

Mitochondria (mt) play an important role in both apoptosis and haem synthesis. The present study was conducted to determine DNA mutations in mitochondrial encoded cytochrome c-oxidase I and II genes. Bone marrow (BM) biopsy and aspirate, peripheral blood (PB) and buccal smear samples were collected from 20 myelodysplastic syndrome (MDS) patients and 10 age-matched controls. Cytochrome c-oxidase I (CO I) and II (CO II) genes were amplified using polymerase chain reaction and sequenced. CO I mutations were found in 13/20 MDS patients and the CO II gene in 2/10 normal and 12/20 MDS samples, irrespective of MDS subtype. Mutations were substitutional, deletional and insertional. CO I mutations were most common at nucleotide positions 7264 (25%) and 7289 (15%), and CO II mutations were most common at nucleotide positions 7595 (40%) and 7594 (30%), suggesting the presence of potential 'hot-spots'. Mutations were not found in buccal smears of MDS patients and were significantly higher in MDS samples compared with age-matched controls in all cell fractions (P < 0.05), with bone marrow high-density fraction (BMHDF) showing a higher mutation rate than other fractions (P < 0.05). MDS marrows showed higher levels of apoptosis than normal controls (P < 0.05), and apoptosis in BMHDF was directly related to cytochrome c-oxidase I gene mutations (P < 0.05). Electron microscopy revealed apoptosis affecting all haematopoietic lineages with highly abnormal, iron-laden mitochondria. These results suggest a role for mt-DNA mutations in the excessive apoptosis and resulting cytopenias of MDS patients.


Subject(s)
DNA, Mitochondrial/genetics , Electron Transport Complex IV/genetics , Mitochondria/enzymology , Mutation , Myelodysplastic Syndromes/genetics , Adult , Aged , Aged, 80 and over , Apoptosis , Bone Marrow Cells/enzymology , Bone Marrow Cells/ultrastructure , Female , Humans , In Situ Nick-End Labeling , Male , Middle Aged , Mitochondria/ultrastructure , Myelodysplastic Syndromes/enzymology , Myelodysplastic Syndromes/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...