Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cytometry B Clin Cytom ; 94(2): 312-326, 2018 03.
Article in English | MEDLINE | ID: mdl-27784127

ABSTRACT

BACKGROUND: Bone marrow examination has been the confirmatory test for congenital dyserythropoietic anemia type II (CDAII). Occasional spherocytes on peripheral blood smear can confound the diagnosis. Since a screening test is still unavailable, we explored the feasibility of using flow cytometry as a preliminary screening method. METHODS: Thirteen monoclonal antibodies with specificities for eight erythrocyte membrane proteins were used in FACS analysis to probe the cellular features of red cells from CDAII, normal adults, hereditary spherocytosis (HS), and cord red cells. Confocal microscopy was performed on normal and CDAII to determine the overall distribution of CD44 and CD47. Their expression levels on cultured erythroblasts were also analyzed. RESULTS: The densely stained band 3 as seen in CDAII in gel electrophoresis was also obtained for Dantu phenotype. Likewise analysis of CDAII cases (n = 26) using the eosin-5'maleimide (EMA) binding test found 57% of patients giving results either positive or in the grey area for HS. Enhanced fluorescence of CD44 was detected in 96% of the CDAII patients, and anti-CD47 binding was also elevated to a lesser degree. Although RNA expressions of CD44 and CD47 in the cultured erythroblasts of normal controls and CDAII were similar, confocal microscopy revealed more CDAII red cells giving elevated fluorescence than normal red cells. CONCLUSIONS: A distinction between CDAII and HS can be made using the EMA Binding test and anti-CD44 binding. Confirmation of CDAII can subsequently be made based on clinical presentation together with either bone marrow examination or DNA sequencing of SEC23B. © 2016 International Clinical Cytometry Society.


Subject(s)
Anemia, Dyserythropoietic, Congenital/metabolism , Biomarkers/metabolism , Cell Differentiation/physiology , Hyaluronan Receptors/metabolism , Spherocytosis, Hereditary/metabolism , CD47 Antigen/metabolism , Erythrocytes/metabolism , Female , Flow Cytometry/methods , Humans , Male , Mass Screening , Middle Aged , Phenotype
2.
Blood ; 95(1): 12-8, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10607679

ABSTRACT

Antigens of the Rh blood group system are encoded by 2 homologous genes, RHD and RHCE, that produce 2 red cell membrane proteins. The D-negative phenotype is considered to result, almost invariably, from homozygosity for a complete deletion of RHD. The basis of all PCR tests for predicting fetal D phenotype from DNA obtained from amniocytes or maternal plasma is detection of the presence of RHD. These tests are used in order to ascertain the risk of hemolytic disease of the newborn. We have identified an RHD pseudogene (RHD psi) in Rh D-negative Africans. RHDpsi contains a 37 base pair (bp) insert in exon 4, which may introduce a stop codon at position 210. The insert is a sequence duplication across the boundary of intron 3 and exon 4. RHDpsi contains another stop codon in exon 6. The frequency of RHDpsi in black South Africans is approximately 0.0714. Of 82 D-negative black Africans, 66% had RHDpsi, 15% had the RHD-CE-D hybrid gene associated with the VS+ V- phenotype, and only 18% completely lacked RHD. RHDpsi is present in about 24% of D-negative African Americans and 17% of D-negative South Africans of mixed race. No RHD transcript could be detected in D-negative individuals with RHDpsi, probably as a result of nonsense-mediated mRNA decay. Existing PCR-based methods for predicting D phenotype from DNA are not suitable for testing Africans or any population containing a substantial proportion of people with African ethnicity. Consequently, we have developed a new test that detects the 37 bp insert in exon 4 of RHDpsi. (Blood. 2000; 95:12-18)


Subject(s)
Black People/genetics , Glycoproteins/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Oncogene Proteins, Fusion/genetics , Pseudogenes , Recombinant Fusion Proteins , Rh-Hr Blood-Group System , Amino Acid Sequence , Anemia, Hemolytic, Congenital/genetics , Base Sequence , Blood Donors , Ethnicity/genetics , Exons , Female , Ghana , Glycoproteins/chemistry , Humans , Huntingtin Protein , Infant, Newborn , Introns , Louisiana , Molecular Sequence Data , Mutation, Missense , Nerve Tissue Proteins/chemistry , Nuclear Proteins/chemistry , Phenotype , Polymerase Chain Reaction , Pregnancy , Pseudogenes/genetics , Repetitive Sequences, Nucleic Acid , Risk Factors , Sequence Alignment , Sequence Deletion , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , South Africa , Transcription, Genetic , Zimbabwe
3.
Cancer Res ; 59(14): 3454-60, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10416610

ABSTRACT

Severe immunodeficiency characterized by lymphopenia was found in two siblings, one of whom was examined in detail. The calcium flux, pattern of tyrosine phosphorylation of proteins, and interleukin 2 (IL-2) production and proliferation in response to mitogens suggested that the peripheral blood T cells activated normally. The peripheral blood T cells were shown to have an activated phenotype with increased expression of CD45RO+ and CD95/Fas. Increased spontaneous apoptosis occurred in unstimulated lymphocyte cultures. The elevated apoptosis was not due to alterations in expression or to mutations in Bcl-2, Bcl-X(L), or Flip, nor could the spontaneous apoptosis be prevented by blocking Fas, suggesting that it was independent of Fas signaling. This is the first inherited combined immunodeficiency associated with impaired lymphocyte survival. Fibroblasts derived from the patient showed appreciable radiosensitivity in clonal assays, but apoptosis was not elevated. Our results show that the fibroblasts represent a new radiosensitive phenotype not associated with cell cycle checkpoint defects, V(D)J recombination defects, or elevated chromosome breakage. We suggest that the affected gene plays a role in an undetermined damage response mechanism that results in elevated spontaneous apoptosis in lymphoid cells and radiosensitivity in fibroblasts.


Subject(s)
Apoptosis , Fibroblasts/radiation effects , Immunologic Deficiency Syndromes/pathology , Lymphocytes/radiation effects , Severe Combined Immunodeficiency/pathology , Apoptosis/radiation effects , Child , Child, Preschool , Chromosome Inversion , Chromosomes, Human, Pair 7/ultrastructure , DNA Damage , DNA Repair , DNA, Complementary/genetics , Female , Fibroblasts/pathology , Gamma Rays , Humans , Lymphocytes/pathology , Male , Radiation Tolerance , Severe Combined Immunodeficiency/genetics , Signal Transduction/physiology , Translocation, Genetic
4.
Mol Cell Biol ; 19(5): 3267-77, 1999 May.
Article in English | MEDLINE | ID: mdl-10207052

ABSTRACT

Ku is a heterodimeric protein with double-stranded DNA end-binding activity that operates in the process of nonhomologous end joining. Ku is thought to target the DNA-dependent protein kinase (DNA-PK) complex to the DNA and, when DNA bound, can interact and activate the DNA-PK catalytic subunit (DNA-PKcs). We have carried out a 3' deletion analysis of Ku80, the larger subunit of Ku, and shown that the C-terminal 178 amino acid residues are dispensable for DNA end-binding activity but are required for efficient interaction of Ku with DNA-PKcs. Cells expressing Ku80 proteins that lack the terminal 178 residues have low DNA-PK activity, are radiation sensitive, and can recombine the signal junctions but not the coding junctions during V(D)J recombination. These cells have therefore acquired the phenotype of mouse SCID cells despite expressing DNA-PKcs protein, suggesting that an interaction between DNA-PKcs and Ku, involving the C-terminal region of Ku80, is required for DNA double-strand break rejoining and coding but not signal joint formation. To gain further insight into important domains in Ku80, we report a point mutational change in Ku80 in the defective xrs-2 cell line. This residue is conserved among species and lies outside of the previously reported Ku70-Ku80 interaction domain. The mutational change nonetheless abrogates the Ku70-Ku80 interaction and DNA end-binding activity.


Subject(s)
Antigens, Nuclear , DNA Helicases , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/metabolism , Recombination, Genetic/genetics , Animals , CHO Cells , Cell Survival/genetics , Clone Cells/metabolism , Clone Cells/radiation effects , Cricetinae , DNA-Activated Protein Kinase , DNA-Binding Proteins/metabolism , Enzyme Activation/genetics , Gene Expression/genetics , Humans , Ku Autoantigen , Nuclear Proteins/metabolism , Point Mutation/genetics , Sequence Deletion/genetics , Transfection
5.
Nucleic Acids Res ; 26(8): 1965-73, 1998 Apr 15.
Article in English | MEDLINE | ID: mdl-9518490

ABSTRACT

The catalytic subunit of the DNA-dependent protein kinase (DNA-PKcs) is a member of a sub-family of phosphatidylinositol (PI) 3-kinases termed PIK-related kinases. A distinguishing feature of this sub-family is the presence of a conserved C-terminal region downstream of a PI 3-kinase domain. Mutants defective in DNA-PKcs are sensitive to ionising radiation and are unable to carry out V(D)J recombination. Irs-20 is a DNA-PKcs-defective cell line with milder gamma-ray sensitivity than two previously characterised mutants, V-3 and mouse scid cells. Here we show that the DNA-PKcs protein from irs-20 cells can bind to DNA but is unable to function as a protein kinase. To verify the defect in irs-20 cells and provide insight into the function and expression of DNA-PKcs in double-strand break repair and V(D)J recombination we introduced YACs encoding human and mouse DNA-PKcs into defective mutants and achieved complementation of the defective phenotypes. Furthermore, in irs-20 we identified a mutation in DNA-PKcs that causes substitution of a lysine for a glutamic acid in the fourth residue from the C-terminus. This represents a strong candidate for the inactivating mutation and provides supportive evidence that the extreme C-terminal motif is important for protein kinase activity.


Subject(s)
Cell Survival/radiation effects , DNA-Binding Proteins , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/metabolism , Animals , CHO Cells , Cell Line , Chromosomes, Artificial, Yeast , Cricetinae , DNA/metabolism , DNA Damage , DNA Nucleotidyltransferases/metabolism , DNA Repair , DNA-Activated Protein Kinase , Dose-Response Relationship, Radiation , Gamma Rays , Gene Library , Horses , Humans , Mice , Mice, SCID , Nuclear Proteins , Phosphatidylinositol 3-Kinases/metabolism , Polymerase Chain Reaction , Transfection , VDJ Recombinases
6.
J Mol Biol ; 279(2): 375-85, 1998 Jun 05.
Article in English | MEDLINE | ID: mdl-9642044

ABSTRACT

Mutations in genes controlling the correct functioning of the replicative, repair and recombination machineries may lead to genomic instability. A high level of spontaneous chromosomal aberrations amplified by treatment with DNA cross-linking agents is the hallmark of Fanconi anemia (FA), an inherited chromosomal instability syndrome associated with cancer proneness. Two of the eight FA genes have been cloned (FAA and FAC), but their function has not yet been defined. The lack of homology with known genes suggests the involvement of FA genes in a novel pathway specific to vertebrates. Using a DNA end-joining assay in cultured cells, we studied the processing of both blunt and cohesive-ended double strand breaks (DSB) in normal and FA cells. The results show that: (i) the overall ligation efficiency is normal in FA lymphoblasts; (ii) in FA-C, error-free processing of blunt-ended DSB is markedly decreased, resulting in a higher deletion frequency and larger deletion size; (iii) the fidelity of processing of blunt-DSB is completely restored in FACC cells (complemented with wild-type FAC gene) and the deletion size shifted to values similar to that observed in normal cells; (iv) the fidelity of cohesive end-joining is not affected in FA cells; (v) activities and/or expression of known factors involved in DSB processing, such as the components of the DNA-PK complex and XRCC4, are normal in FA cells. Our results provide strong evidence that the lack of a functional FAC gene results in loss of fidelity of end-joining, which likely accounts for the FA-C phenotype of chromosome instability. We conclude that FAC, and perhaps all FA gene products, are likely to play a role in the fidelity of end-joining of specific DSB.


Subject(s)
Cell Cycle Proteins , DNA Damage/genetics , DNA-Binding Proteins , DNA/genetics , DNA/metabolism , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Nuclear Proteins , Proteins/genetics , Proteins/metabolism , Amino Acid Sequence , Cell Line , Chromosome Aberrations , DNA Repair/genetics , DNA Replication , DNA-Activated Protein Kinase , Fanconi Anemia Complementation Group Proteins , Humans , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , Phenotype , Protein Serine-Threonine Kinases/metabolism , Recombination, Genetic , Sequence Deletion , Substrate Specificity
7.
Mol Cell Biol ; 17(3): 1264-73, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9032253

ABSTRACT

The gene product defective in radiosensitive CHO mutants belonging to ionizing radiation complementation group 5, which includes the extensively studied xrs mutants, has recently been identified as Ku80, a subunit of the Ku protein and a component of DNA-dependent protein kinase (DNA-PK). Several group 5 mutants, including xrs-5 and -6, lack double-stranded DNA end-binding and DNA-PK activities. In this study, we examined additional xrs mutants at the molecular and biochemical levels. All mutants examined have low or undetectable levels of Ku70 and Ku80 protein, end-binding, and DNA-PK activities. Only one mutant, xrs-6, has Ku80 transcript levels detectable by Northern hybridization, but Ku80 mRNA was detectable by reverse transcription-PCR in most other mutants. Two mutants, xrs-4 and -6, have altered Ku80 transcripts resulting from mutational changes in the genomic Ku80 sequence affecting RNA splicing, indicating that the defects in these mutants lie in the Ku80 gene rather than a gene controlling its expression. Neither of these two mutants has detectable wild-type Ku80 transcript. Since the mutation in both xrs-4 and xrs-6 cells results in severely truncated Ku80 protein, both are likely candidates to be null mutants. Azacytidine-induced revertants of xrs-4 and -6 carried both wild-type and mutant transcripts. The results with these revertants strongly support our model proposed earlier, that CHO-K1 cells carry a copy of the Ku80 gene (XRCC5) silenced by hypermethylation. Site-directed mutagenesis studies indicate that previously proposed ATP-binding and phosphorylation sites are not required for Ku80 activity, whereas N-terminal deletions of more than the first seven amino acids result in severe loss of activities.


Subject(s)
Antigens, Nuclear , CHO Cells , DNA Helicases , DNA-Binding Proteins/genetics , Mutation , Nuclear Proteins/genetics , Radiation Tolerance/genetics , Animals , Azacitidine/pharmacology , CHO Cells/radiation effects , Cricetinae , DNA/metabolism , DNA, Complementary/genetics , DNA-Activated Protein Kinase , DNA-Binding Proteins/metabolism , Gamma Rays , Gene Dosage , Genetic Complementation Test , Ku Autoantigen , Molecular Sequence Data , Nuclear Proteins/metabolism , Protein Binding , Protein Serine-Threonine Kinases/metabolism , RNA Splicing , RNA, Messenger/analysis , Sequence Analysis, DNA , Sequence Deletion
SELECTION OF CITATIONS
SEARCH DETAIL
...