Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Lab Hematol ; 43(4): 664-674, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34053184

ABSTRACT

INTRODUCTION: Reverse transcriptase quantitative PCR (RT-qPCR) is considered the method of choice for measurable residual disease (MRD) assessment in NPM1-mutated acute myeloid leukemia (AML). MRD can also be determined with DNA-based methods offering certain advantages. We here compared the DNA-based methods quantitative PCR (qPCR), droplet digital PCR (ddPCR), and targeted deep sequencing (deep seq) with RT-qPCR. METHODS: Of 110 follow-up samples from 30 patients with NPM1-mutated AML were analyzed by qPCR, ddPCR, deep seq, and RT-qPCR. To select DNA MRD cutoffs for bone marrow, we performed receiver operating characteristic analyses for each DNA method using prognostically relevant RT-qPCR cutoffs. RESULTS: The DNA-based methods showed strong intermethod correlation, but were less sensitive than RT-qPCR. A bone marrow cutoff at 0.1% leukemic DNA for qPCR or 0.05% variant allele frequency for ddPCR and deep seq offered optimal sensitivity and specificity with respect to 3 log10 reduction of NPM1 transcripts and/or 2% mutant NPM1/ABL. With these cutoffs, MRD results agreed in 95% (191/201) of the analyses. Although more sensitive, RT-qPCR failed to detect leukemic signals in 10% of samples with detectable leukemic DNA. CONCLUSION: DNA-based MRD techniques may complement RT-qPCR for assessment of residual leukemia. DNA-based methods offer high positive and negative predictive values with respect to residual leukemic NPM1 transcripts at levels of importance for response to treatment. However, moving to DNA-based MRD methods will miss a proportion of patients with residual leukemic RNA, but on the other hand some MRD samples with detectable leukemic DNA can be devoid of measurable leukemic RNA.


Subject(s)
DNA, Neoplasm/blood , Leukemia, Myeloid, Acute/blood , Mutation , Nuclear Proteins/metabolism , RNA, Neoplasm/blood , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Adult , Aged , DNA, Neoplasm/genetics , Female , Humans , Leukemia, Myeloid, Acute/genetics , Male , Middle Aged , Neoplasm, Residual/blood , Nuclear Proteins/genetics , Nucleophosmin , RNA, Neoplasm/genetics
2.
Biosci Rep ; 40(5)2020 05 29.
Article in English | MEDLINE | ID: mdl-32301496

ABSTRACT

Antibodies to the Vel blood group antigen can cause adverse hemolytic reactions unless Vel-negative blood units are transfused. Since the genetic background of Vel-negativity was discovered in 2013, DNA-based typing of the 17-bp deletion causing the phenotype has facilitated identification of Vel-negative blood donors. SMIM1, the gene underlying Vel, encodes a 78-amino acid erythroid transmembrane protein of unknown function. The transmembrane orientation of SMIM1 has been debated since experimental data supported both the N- and C-termini being extracellular. Likewise, computational predictions of its orientation were divided and potential alternatives such as monotopic or dual-topology have been discussed but not investigated. We used a cell-free system to explore the topology of SMIM1 when synthesized in the endoplasmic reticulum (ER). SMIM1 was tagged with an opsin-derived N-glycosylation reporter at either the N- or C-terminus and synthesized in vitro using rabbit reticulocyte lysate supplemented with canine pancreatic microsomes as a source of ER membrane. SMIM1 topology was then determined by assessing the N-glycosylation of its N- or C-terminal tags. Complementary experiments were carried out by expressing the same SMIM1 variants in HEK293T/17 cells and establishing their membrane orientation by immunoblotting and flow cytometry. Our data consistently indicate that SMIM1 has its short C-terminus located extracellularly and that it most likely belongs to the tail-anchored class of membrane proteins with the bulk of the polypeptide located in the cytoplasm. Having established its membrane orientation in an independent model system, future work can now focus on functional aspects of SMIM1 as a potential regulator of erythropoiesis.


Subject(s)
Endoplasmic Reticulum/metabolism , Intracellular Membranes/metabolism , Membrane Proteins/metabolism , Animals , Cell-Free System , Dogs , Glycosylation , HEK293 Cells , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Processing, Post-Translational , Protein Structure, Quaternary , Rabbits , Structure-Activity Relationship
3.
Blood ; 132(3): 334-338, 2018 07 19.
Article in English | MEDLINE | ID: mdl-29748255

ABSTRACT

The Xga blood group is differentially expressed on erythrocytes from men and women. The underlying gene, PBDX, was identified in 1994, but the molecular background for Xga expression remains undefined. This gene, now designated XG, partly resides in pseudoautosomal region 1 and encodes a protein of unknown function from the X chromosome. By comparing calculated Xga allele frequencies in different populations with 2612 genetic variants in the XG region, rs311103 showed the strongest correlation to the expected distribution. The same single-nucleotide polymorphism (SNP) had the most significant impact on XG transcript levels in whole blood (P = 2.0 × 10-22). The minor allele, rs311103C, disrupts a GATA-binding motif 3.7 kb upstream of the transcription start point. This silences erythroid XG messenger RNA expression and causes the Xg(a-) phenotype, a finding corroborated by SNP genotyping in 158 blood donors. Binding of GATA1 to biotinylated oligonucleotide probes with rs311103G but not rs311103C was observed by electrophoretic mobility shift assay and proven by mass spectrometry. Finally, a luciferase reporter assay indicated this GATA motif to be active for rs311103G but not rs311103C in HEL cells. By using an integrated bioinformatic and molecular biological approach, we elucidated the underlying genetic basis for the last unresolved blood group system and made Xga genotyping possible.


Subject(s)
Binding Sites , Blood Group Antigens/genetics , Cell Adhesion Molecules/genetics , GATA1 Transcription Factor/metabolism , Gene Expression Regulation , Nucleotide Motifs , Alleles , Blood Group Antigens/metabolism , Cell Adhesion Molecules/metabolism , Erythrocytes/metabolism , Female , Gene Frequency , Genes, Reporter , Genetic Association Studies , Genotype , Humans , Male , Phenotype , Polymorphism, Single Nucleotide , Transcription Initiation Site
4.
Blood ; 131(14): 1611-1616, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29438961

ABSTRACT

P1 and Pk are glycosphingolipid antigens synthesized by the A4GALT-encoded α1,4-galactosyltransferase, using paragloboside and lactosylceramide as acceptor substrates, respectively. In addition to the compatibility aspects of these histo-blood group molecules, both constitute receptors for multiple microbes and toxins. Presence or absence of P1 antigen on erythrocytes determines the common P1 (P1+Pk+) and P2 (P1-Pk+weak) phenotypes. A4GALT transcript levels are higher in P1 individuals and single-nucleotide polymorphisms (SNPs) in noncoding regions of A4GALT, particularly rs5751348, correlate with P1/P2 status. Despite these recent findings, the molecular mechanism underlying these phenotypes remains elusive. The In(Lu) phenotype is caused by Krüppel-like factor 1 (KLF1) haploinsufficiency and shows decreased P1 levels on erythrocytes. We therefore hypothesized KLF1 regulates A4GALT expression. Intriguingly, P1 -specific sequences including rs5751348 revealed potential binding sites for several hematopoietic transcription factors, including KLF1. However, KLF1 binding did not explain P1 -specific shifts in electrophoretic mobility-shift assays and small interfering RNA silencing of KLF1 did not affect A4GALT transcript levels. Instead, protein pull-down experiments using P1 but not P2 oligonucleotide probes identified runt-related transcription factor 1 (RUNX1) by mass spectrometry. Furthermore, RUNX1 binds P1 alleles selectively, and knockdown of RUNX1 significantly decreased A4GALT transcription. These data indicate that RUNX1 regulates A4GALT and thereby the expression of clinically important glycosphingolipids implicated in blood group incompatibility and host-pathogen interactions.


Subject(s)
Alleles , Core Binding Factor Alpha 2 Subunit/metabolism , Galactosyltransferases/biosynthesis , Globosides/biosynthesis , Haploinsufficiency , Transcription, Genetic , Cell Line , Core Binding Factor Alpha 2 Subunit/genetics , Galactosyltransferases/genetics , Gene Silencing , Globosides/genetics , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
5.
Oncotarget ; 8(50): 87136-87150, 2017 Oct 20.
Article in English | MEDLINE | ID: mdl-29152069

ABSTRACT

The Wilms' tumor gene 1 (WT1) is recurrently mutated in acute myeloid leukemia. Mutations and high expression of WT1 associate with a poor prognosis. In mice, WT1 cooperates with the RUNX1/RUNX1T1 (AML1/ETO) fusion gene in the induction of acute leukemia, further emphasizing a role for WT1 in leukemia development. Molecular mechanisms for WT1 are, however, incompletely understood. Here, we identify the transcriptional coregulator NAB2 as a target gene of WT1. Analysis of gene expression profiles of leukemic samples revealed a positive correlation between the expression of WT1 and NAB2, as well as a non-zero partial correlation. Overexpression of WT1 in hematopoietic cells resulted in increased NAB2 levels, while suppression of WT1 decreased NAB2 expression. WT1 bound and transactivated the proximal NAB2 promoter, as shown by ChIP and reporter experiments, respectively. ChIP experiments also revealed that WT1 can recruit NAB2 to the IRF8 promoter, thus modulating the transcriptional activity of WT1, as shown by reporter experiments. Our results implicate NAB2 as a previously unreported target gene of WT1 and that NAB2 acts as a transcriptional cofactor of WT1.

6.
Haematologica ; 102(2): 336-345, 2017 02.
Article in English | MEDLINE | ID: mdl-27612989

ABSTRACT

The zinc finger transcription factor Wilms tumor gene 1 (WT1) acts as an oncogene in acute myeloid leukemia. A naturally occurring alternative splice event between zinc fingers three and four, removing or retaining three amino acids (±KTS), is believed to change the DNA binding affinity of WT1, although there are conflicting data regarding the binding affinity and motifs of the different isoforms. Increased expression of the WT1 -KTS isoform at the expense of the WT1 +KTS isoform is associated with poor prognosis in acute myeloid leukemia. We determined the genome-wide binding pattern of WT1 -KTS and WT1 +KTS in leukemic K562 cells by chromatin immunoprecipitation and deep sequencing. We discovered that the WT1 -KTS isoform predominantly binds close to transcription start sites and to enhancers, in a similar fashion to other transcription factors, whereas WT1 +KTS binding is enriched within gene bodies. We observed a significant overlap between WT1 -KTS and WT1 +KTS target genes, despite the binding sites being distinct. Motif discovery revealed distinct binding motifs for the isoforms, some of which have been previously reported as WT1 binding sites. Additional analyses showed that both WT1 -KTS and WT1 +KTS target genes are more likely to be transcribed than non-targets, and are involved in cell proliferation, cell death, and development. Our study provides evidence that WT1 -KTS and WT1 +KTS share target genes yet still bind distinct locations, indicating isoform-specific regulation in transcription of genes related to cell proliferation and differentiation, consistent with the involvement of WT1 in acute myeloid leukemia.


Subject(s)
Alternative Splicing , Gene Expression Regulation, Leukemic , Leukemia/genetics , Leukemia/metabolism , WT1 Proteins/genetics , WT1 Proteins/metabolism , Binding Sites , Chromatin Immunoprecipitation , High-Throughput Nucleotide Sequencing , Humans , Nucleotide Motifs , Protein Binding , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Initiation Site
7.
Biochem Biophys Res Commun ; 482(4): 802-807, 2017 Jan 22.
Article in English | MEDLINE | ID: mdl-27889611

ABSTRACT

Wilms' tumor gene 1 (WT1) is a zinc finger transcription factor that has been implicated as an oncogene in leukemia and several other malignancies. When investigating possible gene expression network partners of WT1 in a large acute myeloid leukemia (AML) patient cohort, one of the genes with the highest correlation to WT1 was quinolinate phosphoribosyltransferase (QPRT), a key enzyme in the de novo nicotinamide adenine dinucleotide (NAD+) synthesis pathway. To investigate the possible relationship between WT1 and QPRT, we overexpressed WT1 in hematopoietic progenitor cells and cell lines, resulting in an increase of QPRT expression. WT1 knock-down gave a corresponding decrease in QPRT gene and protein expression. Chromatin-immunoprecipitation revealed WT1 binding to a conserved site in the first intron of the QPRT gene. Upon overexpression in leukemic K562 cells, QPRT conferred partial resistance to the anti-leukemic drug imatinib, indicating possible anti-apoptotic functions, consistent with previous reports on glioma cells. Interestingly, the rescue effect of QPRT overexpression was not correlated to increased NAD + levels, suggesting NAD + independent mechanisms. We conclude that QPRT, encoding a protein with anti-apoptotic properties, is a novel and direct target gene of WT1 in leukemic cells.


Subject(s)
Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , Pentosyltransferases/genetics , WT1 Proteins/genetics , Apoptosis , Base Sequence , Cell Line, Tumor , Genes, Wilms Tumor , Humans , Introns , K562 Cells , Leukemia, Myeloid, Acute/metabolism , NAD/metabolism , Pentosyltransferases/metabolism , Promoter Regions, Genetic , Protein Binding , Transcriptional Activation , WT1 Proteins/metabolism
8.
Leuk Res ; 40: 60-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26563595

ABSTRACT

The transcription factor interferon regulatory factor-8 (IRF8) is highly expressed in myeloid progenitors, while most myeloid leukemias show low or absent expression. Loss of IRF8 in mice leads to a myeloproliferative disorder, indicating a tumor-suppressive role of IRF8. The Wilms tumor gene 1 (WT1) protein represses the IRF8-promoter. The zinc finger protein ZNF224 can act as a transcriptional co-factor of WT1 and potentiate the cytotoxic response to the cytostatic drug cytarabine. We hypothesized that cytarabine upregulates IRF8 and that transcriptional control of IRF8 involves WT1 and ZNF224. Treatment of leukemic K562 cells with cytarabine upregulated IRF8 protein and mRNA, which was correlated to increased expression of ZNF224. Knock down of ZNF224 with shRNA suppressed both basal and cytarabine-induced IRF8 expression. While ZNF224 alone did not affect IRF8 promoter activity, ZNF224 partially reversed the suppressive effect of WT1 on the IRF8 promoter, as judged by luciferase reporter experiments. Coprecipitation revealed nuclear binding of WT1 and ZNF224, and by chromatin immunoprecipitation (ChIP) experiments it was demonstrated that WT1 recruits ZNF224 to the IRF8 promoter. We conclude that cytarabine-induced upregulation of the IRF8 in leukemic cells involves increased levels of ZNF224, which can counteract the repressive activity of WT1 on the IRF8-promoter.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Cytarabine/pharmacology , Interferon Regulatory Factors/physiology , Leukemia/pathology , Up-Regulation/drug effects , WT1 Proteins/metabolism , Zinc Fingers , Cell Line, Tumor , Humans
9.
Oncotarget ; 6(29): 28223-37, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26320177

ABSTRACT

The Kruppel-like protein ZNF224 is a co-factor of the Wilms' tumor 1 protein, WT1. We have previously shown that ZNF224 exerts a specific proapoptotic role in chronic myelogenous leukemia (CML) K562 cells and contributes to cytosine arabinoside-induced apoptosis, by modulating WT1-dependent transcription of apoptotic genes. Here we demonstrate that ZNF224 gene expression is down-regulated both in BCR-ABL positive cell lines and in primary CML samples and is restored after imatinib and second generation tyrosine kinase inhibitors treatment. We also show that WT1, whose expression is positively regulated by BCR-ABL, represses transcription of the ZNF224 gene. Finally, we report that ZNF224 is significantly down-regulated in patients with BCR-ABL positive chronic phase-CML showing poor response or resistance to imatinib treatment as compared to high-responder patients. Taken as a whole, our data disclose a novel pathway activated by BCR-ABL that leads to inhibition of apoptosis through the ZNF224 repression. ZNF224 could thus represent a novel promising therapeutic target in CML.


Subject(s)
Apoptosis/genetics , Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Neoplastic/genetics , Repressor Proteins/genetics , WT1 Proteins/genetics , Blotting, Western , Cell Line, Tumor , Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , Humans , Imatinib Mesylate/pharmacology , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Promoter Regions, Genetic/genetics , Protein Binding , Protein Kinase Inhibitors/pharmacology , RNA Interference , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , WT1 Proteins/metabolism
10.
Leuk Res ; 37(10): 1341-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23871158

ABSTRACT

The transcription factor Wilms' tumor gene 1 (WT1) is highly expressed in the majority of leukemias, suggesting a role in leukemogenesis. Acquired WT1 mutations are reported as an independent predictor of poor clinical outcome, and mutations resulting in deletion of the entire DNA-binding zinc-finger domain (WT1delZ), is the most common type. The aim of this study was to study cellular effects of WT1(delZ) that may contribute to an oncogenic phenotype. We found that expression of WT1(delZ) supported proliferation of human hematopoietic CD34(+) progenitor cells. Moreover, WT1(delZ) transduced cells expressed erythroid markers, including raised levels of STAT5, independently of addition of erythropoietin. At the global gene expression level, WT1(delZ) caused upregulation of genes related to cell division and genes associated with erythroid maturation, in the absence of added erythropoietin. Our results indicate that WT1(delZ) promotes cell proliferation and expansion of progenitor cells, consistent with a possible role in leukemogenesis.


Subject(s)
Hematopoietic Stem Cells/metabolism , Leukemia/genetics , Leukemia/metabolism , Mutant Proteins , WT1 Proteins/genetics , WT1 Proteins/metabolism , Antigens, CD34/metabolism , Cell Culture Techniques , Cell Proliferation , Colony-Forming Units Assay , Erythropoietin/metabolism , Gene Expression , Gene Expression Profiling , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Hemoglobin A/metabolism , Humans , STAT5 Transcription Factor/metabolism , Transduction, Genetic
11.
Hum Mol Genet ; 22(9): 1771-82, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23362234

ABSTRACT

The transcription factor Wilms' tumor gene 1, WT1, is implicated both in normal developmental processes and in the generation of a variety of solid tumors and hematological malignancies. Physical interactions of other cellular proteins with WT1 are known to modulate its function. We previously identified the Krüppel-like zinc-finger protein, ZNF224, as a novel human WT1-associating protein that enhances the transcriptional activation of the human vitamin D receptor promoter by WT1. Here, we have analyzed the effects of WT1-ZNF224 interaction on the expression of apoptosis-regulating genes in the chronic myelogenous leukemia (CML) K562 cell line. The results demonstrated that ZNF224 acts in fine tuning of WT1-dependent control of gene expression, acting as a co-activator of WT1 in the regulation of proapoptotic genes and suppressing WT1 mediated transactivation of antiapoptotitc genes. Moreover, the DNA damaging drug cytosine arabinoside (ara-C) induces expression of ZNF224 in K562 cells and this induction enhances cell apoptotic response to ara-C. These findings suggest that ZNF224 can be a mediator of DNA damage-induced apoptosis in leukemia cells.


Subject(s)
Apoptosis/genetics , DNA Damage , Repressor Proteins/metabolism , WT1 Proteins/metabolism , Chromatin Immunoprecipitation , Cytarabine/adverse effects , DNA-Binding Proteins , Epigenetic Repression , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , K562 Cells , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Repressor Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation , WT1 Proteins/genetics
12.
DNA Cell Biol ; 26(8): 589-97, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17688410

ABSTRACT

The Wilms' tumor gene 1 (WT1) protein is a transcriptional regulator that is highly expressed in immature hematopoietic progenitor cells and in the majority of patients with acute and chronic myeloid leukemia. However, it is still unclear how WT1 exerts its function(s) in hematopoietic cells. The aim of this work was to investigate the function of WT1 as a transcription factor in human hematopoietic progenitor cells. To this end, an oligonucleotide array approach was used to study the gene expression in CD34(+) cells from human cord blood retrovirally transduced with WT1 or a control vector. We found that the expression of the putative tumor suppressor gene N-myc downstream regulated gene 2 (NDRG2) mRNA was induced by WT1 in CD34(+) cells and also in leukemic U937 cells. Furthermore, a novel transcription start site in the NDRG2 gene was identified in WT1-transduced cells, in addition to two previously reported transcription start sites. These results show that the expression of the NDRG2 gene is directly or indirectly induced by WT1, and provide the first insights into transcriptional regulation of the NDRG2 gene, including demonstration of a novel splice variant.


Subject(s)
Genes, Wilms Tumor , Hematopoietic Stem Cells/metabolism , Tumor Suppressor Proteins/metabolism , Alternative Splicing/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Cells, Cultured , Gene Expression Regulation , Genes, Wilms Tumor/physiology , Hematopoietic Stem Cells/cytology , Humans , Promoter Regions, Genetic , Transcription, Genetic , Tumor Suppressor Proteins/genetics
13.
J Leukoc Biol ; 80(1): 196-203, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16684888

ABSTRACT

Bactericidal/permeability-increasing protein (BPI) neutralizes the proinflammatory effects of lipopolysaccharide and is of potential clinical use in the treatment of fulminant Gram-negative infections. BPI is a cationic protein with antibacterial activity stored in azurophil (primary) granules of neutrophil granulocytes. However, the absence of BPI in patients with specific granule deficiency indicates a transcriptional control of BPI, which is distinct from that of other azurophil granule proteins. Accordingly, we demonstrate in vivo that the BPI mRNA level peaks, together with mRNA for specific granule proteins, during the myelocytic and metamyelocytic stage of granulocytic maturation. The human promyelocytic cell line NB4 expresses several azurophil granule proteins, but expression of BPI is undetectable. We show that treatment of NB4 cells with all-trans retinoic acid (ATRA) induces BPI expression at mRNA and at protein level. The induction is dependent on de novo protein synthesis, as judged by sensitivity to cycloheximide. Previous investigations have indicated a potential role of CCAAT/enhancer-binding protein (C/EBP) transcription factors in the regulation of BPI expression. Here, we show that induction of NB4 cells with ATRA correlates to direct binding of C/EBPbeta and C/EBPepsilon to the proximal BPI promoter, as determined by electrophoretic mobility shift analysis and chromatin immunoprecipitation. The dependency on C/EBPbeta and C/EBPepsilon provides an explanation for delayed BPI mRNA expression, as compared with mRNA of other azurophil granule proteins.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Blood Proteins/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Membrane Proteins/metabolism , Myeloid Cells/drug effects , Promoter Regions, Genetic/physiology , Tretinoin/pharmacology , Antimicrobial Cationic Peptides/drug effects , Antimicrobial Cationic Peptides/genetics , Binding Sites , Blood Proteins/drug effects , Blood Proteins/genetics , CCAAT-Enhancer-Binding Protein-beta/drug effects , CCAAT-Enhancer-Binding Proteins/drug effects , Cell Differentiation , Cell Line, Tumor , Gene Expression Profiling , Humans , Membrane Proteins/drug effects , Membrane Proteins/genetics , Myeloid Cells/metabolism , Neutrophils/cytology , Promoter Regions, Genetic/drug effects , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Sensitivity and Specificity , Structure-Activity Relationship
14.
J Leukoc Biol ; 77(3): 369-77, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15590754

ABSTRACT

The bactericidal/permeability-increasing protein (BPI), stored in human neutrophil granulocytes, is cytotoxic against Gram-negative bacteria. Several genes related to BPI cluster on human chromosome 20 and on mouse chromosome 2, but expression and characterization of a BPI ortholog in the mouse have not been reported. We asked whether BPI is structurally and functionally conserved between humans and mice and whether murine BPI might be synthesized in neutrophils as well as in other tissues. We report the isolation of a murine full-length cDNA encoding a 54-kDa protein, showing 53% amino acid identity and 71% similarity, to human BPI. The murine BPI and human BPI genes show a similar exon-intron organization. Murine BPI mRNA was detected in testis, epididymis, and bone marrow, as well as in Sertoli and promyelocytic cell lines. Although levels of BPI mRNA in human and murine testis were comparable, expression in murine bone marrow cells was low as compared with that in human bone marrow. BPI protein showed a cytoplasmic, granular localization in mature neutrophils. BPI gene expression in Sertoli and promyelocytic cells was enhanced several-fold by all-trans retinoic acid. Overexpression of murine BPI in human embryonic kidney 293 cells resulted in antibacterial activity against Escherichia coli, comparable with that obtained with human BPI. In conclusion, it was demonstrated that mouse neutrophils store BPI with antibacterial activity and that murine BPI is also expressed in testis and epididymis.


Subject(s)
Blood Proteins/genetics , Blood Proteins/metabolism , Bone Marrow Cells/metabolism , Epididymis/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Proteins/genetics , Proteins/metabolism , Testis/metabolism , Amino Acid Sequence , Animals , Antimicrobial Cationic Peptides , Blood Proteins/pharmacology , Cloning, Molecular , DNA, Complementary/isolation & purification , DNA, Complementary/metabolism , Escherichia coli/drug effects , Humans , Immunohistochemistry , Male , Membrane Proteins/pharmacology , Mice , Molecular Sequence Data , Neutrophils/chemistry , Organ Specificity , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid
15.
Biochem Biophys Res Commun ; 311(4): 853-63, 2003 Nov 28.
Article in English | MEDLINE | ID: mdl-14623259

ABSTRACT

Bactericidal/permeability-increasing protein (BPI) is an antimicrobial protein in neutrophils, stored in azurophil granules. Expression of BPI is absent in neutrophils of newborns and patients with secondary granule deficiency (SGD), possibly contributing to dysfunction of neutrophils. We report two alternative transcription start sites at 52 and 22bp upstream of the translation start. A proximal 222bp promoter conferring expression in myeloid cells was identified, and critical cis-acting sites for myeloid expression were contained within the 159bp upstream of translation start. Within this region, direct binding and transactivation by AML-1, PU.1, and Sp3 were demonstrated, as judged by electrophoretic mobility shift analysis. Moreover, transient transfections of C/EBPalpha or C/EBPepsilon to HeLa cells resulted in increased promoter activity, indicating a direct or indirect role for C/EBP. In conclusion, we provide evidence for AML-1, PU.1, and Sp3 cooperatively and directly mediating BPI-expression during myeloid differentiation.


Subject(s)
Blood Proteins/genetics , Blood Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation/genetics , Membrane Proteins , Myeloid Cells/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Antimicrobial Cationic Peptides , Base Sequence , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line , Cells, Cultured , Core Binding Factor Alpha 2 Subunit , DNA-Binding Proteins/genetics , HL-60 Cells , Humans , K562 Cells , Molecular Sequence Data , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Sp3 Transcription Factor , Trans-Activators/genetics , Transcription Factor CHOP , Transcription Factors/genetics , Transcriptional Activation/genetics , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...